101
|
Zhang T, Gaffrey MJ, Li X, Qian WJ. Characterization of cellular oxidative stress response by stoichiometric redox proteomics. Am J Physiol Cell Physiol 2020; 320:C182-C194. [PMID: 33264075 DOI: 10.1152/ajpcell.00040.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The thiol redox proteome refers to all proteins whose cysteine thiols are subjected to various redox-dependent posttranslational modifications (PTMs) including S-glutathionylation (SSG), S-nitrosylation (SNO), S-sulfenylation (SOH), and S-sulfhydration (SSH). These modifications can impact various aspects of protein function such as activity, binding, conformation, localization, and interactions with other molecules. To identify novel redox proteins in signaling and regulation, it is highly desirable to have robust redox proteomics methods that can provide global, site-specific, and stoichiometric quantification of redox PTMs. Mass spectrometry (MS)-based redox proteomics has emerged as the primary platform for broad characterization of thiol PTMs in cells and tissues. Herein, we review recent advances in MS-based redox proteomics approaches for quantitative profiling of redox PTMs at physiological or oxidative stress conditions and highlight some recent applications. Considering the relative maturity of available methods, emphasis will be on two types of modifications: 1) total oxidation (i.e., all reversible thiol modifications), the level of which represents the overall redox state, and 2) S-glutathionylation, a major form of reversible thiol oxidation. We also discuss the significance of stoichiometric measurements of thiol PTMs as well as future perspectives toward a better understanding of cellular redox regulatory networks in cells and tissues.
Collapse
Affiliation(s)
- Tong Zhang
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Matthew J Gaffrey
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Xiaolu Li
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington.,Bioproducts Sciences and Engineering Laboratory, Department of Biological Systems Engineering, Washington State University, Richland, Washington
| | - Wei-Jun Qian
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| |
Collapse
|
102
|
Papanikolaou K, Veskoukis AS, Draganidis D, Baloyiannis I, Deli CK, Poulios A, Jamurtas AZ, Fatouros IG. Redox-dependent regulation of satellite cells following aseptic muscle trauma: Implications for sports performance and nutrition. Free Radic Biol Med 2020; 161:125-138. [PMID: 33039652 DOI: 10.1016/j.freeradbiomed.2020.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/26/2020] [Accepted: 10/01/2020] [Indexed: 12/21/2022]
Abstract
Skeletal muscle satellite cells (SCs) are indispensable for tissue regeneration, remodeling and growth. Following myotrauma, SCs are activated, and assist in tissue repair. Exercise-induced muscle damage (EIMD) is characterized by a pronounced inflammatory response and the production of reactive oxygen species (ROS). Experimental evidence suggests that SCs kinetics (the propagation from a quiescent to an activated/proliferative state) following EIMD is redox-dependent and interconnected with changes in the SCs microenvironment (niche). Animal studies have shown that following aseptic myotrauma, antioxidant and/or anti-inflammatory supplementation leads to an improved recovery and skeletal muscle regeneration through enhanced SCs kinetics, suggesting a redox-dependent molecular mechanism. Although evidence suggests that antioxidant/anti-inflammatory compounds may prevent performance deterioration and enhance recovery, there is lack of information regarding the redox-dependent regulation of SCs responses following EIMD in humans. In this review, SCs kinetics following aseptic myotrauma, as well as the intrinsic redox-sensitive molecular mechanisms responsible for SCs responses are discussed. The role of redox status on SCs function should be further investigated in the future with human clinical trials in an attempt to elucidate the molecular pathways responsible for muscle recovery and provide information for potential nutritional strategies aiming at performance recovery.
Collapse
Affiliation(s)
- Konstantinos Papanikolaou
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Aristidis S Veskoukis
- Department of Nutrition and Dietetics, University of Thessaly, Argonafton 1, 42132, Trikala, Greece; Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500, Larissa, Greece
| | - Dimitrios Draganidis
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Ioannis Baloyiannis
- Department of Surgery, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Chariklia K Deli
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Athanasios Poulios
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Athanasios Z Jamurtas
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Ioannis G Fatouros
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece.
| |
Collapse
|
103
|
Jackson MJ. On the mechanisms underlying attenuated redox responses to exercise in older individuals: A hypothesis. Free Radic Biol Med 2020; 161:326-338. [PMID: 33099002 PMCID: PMC7754707 DOI: 10.1016/j.freeradbiomed.2020.10.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/08/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
Responding appropriately to exercise is essential to maintenance of skeletal muscle mass and function at all ages and particularly during aging. Here, a hypothesis is presented that a key component of the inability of skeletal muscle to respond effectively to exercise in aging is a denervation-induced failure of muscle redox signalling. This novel hypothesis proposes that an initial increase in oxidation in muscle mitochondria leads to a paradoxical increase in the reductive state of specific cysteines of signalling proteins in the muscle cytosol that suppresses their ability to respond to normal oxidising redox signals during exercise. The following are presented for consideration:Transient loss of integrity of peripheral motor neurons occurs repeatedly throughout life and is normally rapidly repaired by reinnervation, but this repair process becomes less efficient with aging. Each transient loss of neuromuscular integrity leads to a rapid, large increase in mitochondrial peroxide production in the denervated muscle fibers and in neighbouring muscle fibers. This peroxide may initially act to stimulate axonal sprouting and regeneration, but also stimulates retrograde mitonuclear communication to increase expression of a range of cytoprotective proteins in an attempt to protect the fiber and neighbouring tissues against oxidative damage. The increased peroxide within mitochondria does not lead to an increased cytosolic peroxide, but the increases in adaptive cytoprotective proteins include some located to the muscle cytosol which modify the local cytosol redox environment to induce a more reductive state in key cysteines of specific signalling proteins. Key adaptations of skeletal muscle to exercise involve transient peroxiredoxin oxidation as effectors of redox signalling in the cytosol. This requires sensitive oxidation of key cysteine residues. In aging, the chronic change to a more reductive cytosolic environment prevents the transient oxidation of peroxiredoxin 2 and hence prevents essential adaptations to exercise, thus contributing to loss of muscle mass and function. Experimental approaches suitable for testing the hypothesis are also outlined.
Collapse
Affiliation(s)
- Malcolm J Jackson
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK.
| |
Collapse
|
104
|
Foret MK, Lincoln R, Do Carmo S, Cuello AC, Cosa G. Connecting the "Dots": From Free Radical Lipid Autoxidation to Cell Pathology and Disease. Chem Rev 2020; 120:12757-12787. [PMID: 33211489 DOI: 10.1021/acs.chemrev.0c00761] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Our understanding of lipid peroxidation in biology and medicine is rapidly evolving, as it is increasingly implicated in various diseases but also recognized as a key part of normal cell function, signaling, and death (ferroptosis). Not surprisingly, the root and consequences of lipid peroxidation have garnered increasing attention from multiple disciplines in recent years. Here we "connect the dots" between the fundamental chemistry underpinning the cascade reactions of lipid peroxidation (enzymatic or free radical), the reactive nature of the products formed (lipid-derived electrophiles), and the biological targets and mechanisms associated with these products that culminate in cellular responses. We additionally bring light to the use of highly sensitive, fluorescence-based methodologies. Stemming from the foundational concepts in chemistry and biology, these methodologies enable visualizing and quantifying each reaction in the cascade in a cellular and ultimately tissue context, toward deciphering the connections between the chemistry and physiology of lipid peroxidation. The review offers a platform in which the chemistry and biomedical research communities can access a comprehensive summary of fundamental concepts regarding lipid peroxidation, experimental tools for the study of such processes, as well as the recent discoveries by leading investigators with an emphasis on significant open questions.
Collapse
Affiliation(s)
- Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 1Y6
| | - Richard Lincoln
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A 0B8
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 1Y6
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 1Y6.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada H3A 0C7.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Gonzalo Cosa
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A 0B8
| |
Collapse
|
105
|
Ma C, Hoffmann PR. Selenoproteins as regulators of T cell proliferation, differentiation, and metabolism. Semin Cell Dev Biol 2020; 115:54-61. [PMID: 33214077 DOI: 10.1016/j.semcdb.2020.11.006] [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] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Selenium (Se) is an essential micronutrient that plays a key role in regulating the immune system. T cells are of particular interest due to their important role in promoting adaptive immunity against pathogens and cancer as well as regulating tolerance, all of which are influenced by dietary Se levels. The biological effects of Se are mainly exerted through the actions of the proteins into which it is inserted, i.e. selenoproteins. Thus, the roles that selenoproteins play in regulating T cell biology and molecular mechanisms involved have emerged as important areas of research for understanding how selenium affects immunity. Members of this diverse family of proteins exhibit a wide variety of functions within T cells that include regulating calcium flux induced by T cell receptor (TCR) engagement, shaping the redox tone of T cells before, during, and after activation, and linking TCR-induced activation to metabolic reprogramming required for T cell proliferation and differentiation. This review summarizes recent insights into the roles that selenoproteins play in these processes and their implications in understanding how Se may influence immunity.
Collapse
Affiliation(s)
- Chi Ma
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, Hawaii 96813 USA
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, Hawaii 96813 USA.
| |
Collapse
|
106
|
Cobley JN. Mechanisms of Mitochondrial ROS Production in Assisted Reproduction: The Known, the Unknown, and the Intriguing. Antioxidants (Basel) 2020; 9:E933. [PMID: 33003362 PMCID: PMC7599503 DOI: 10.3390/antiox9100933] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
The consensus that assisted reproduction technologies (ART), like in vitro fertilization, to induce oxidative stress (i.e., the known) belies how oocyte/zygote mitochondria-a major presumptive oxidative stressor-produce reactive oxygen species (ROS) with ART being unknown. Unravelling how oocyte/zygote mitochondria produce ROS is important for disambiguating the molecular basis of ART-induced oxidative stress and, therefore, to rationally target it (e.g., using site-specific mitochondria-targeted antioxidants). I review the known mechanisms of ROS production in somatic mitochondria to critique how oocyte/zygote mitochondria may produce ROS (i.e., the unknown). Several plausible site- and mode-defined mitochondrial ROS production mechanisms in ART are proposed. For example, complex I catalyzed reverse electron transfer-mediated ROS production is conceivable when oocytes are initially extracted due to at least a 10% increase in molecular dioxygen exposure (i.e., the intriguing). To address the term oxidative stress being used without recourse to the underlying chemistry, I use the species-specific spectrum of biologically feasible reactions to define plausible oxidative stress mechanisms in ART. Intriguingly, mitochondrial ROS-derived redox signals could regulate embryonic development (i.e., their production could be beneficial). Their potential beneficial role raises the clinical challenge of attenuating oxidative damage while simultaneously preserving redox signaling. This discourse sets the stage to unravel how mitochondria produce ROS in ART, and their biological roles from oxidative damage to redox signaling.
Collapse
Affiliation(s)
- James N Cobley
- Redox Biology Group, Institute for Health Sciences, University of the Highlands and Islands, Old Perth Road, Inverness IV2 3JH, UK
| |
Collapse
|
107
|
Chang YD, Li CH, Tsai CH, Cheng YW, Kang JJ, Lee CC. Aryl hydrocarbon receptor deficiency enhanced airway inflammation and remodeling in a murine chronic asthma model. FASEB J 2020; 34:15300-15313. [PMID: 32959404 DOI: 10.1096/fj.202001529r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 01/05/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-dependent-activated transcriptional factor that regulates the metabolism of xenobiotic and endogenous compounds. Recent studies have shown that AhR is a novel master regulator of the mucosal immune system, including lungs and intestine. To elucidate the role of AhR in chronic severe asthma, AhR wild-type and knockout mice (AhR-/- ) were sensitized and challenged with ovalbumin for 4 weeks. To uncover the underlying mechanisms, inflammatory cells profile and cytokines production were analyzed in bronchial lavage fluid (BALF) and lung tissue. Compared to wild-type mice, AhR-/- mice had exacerbated asthma symptoms, including airway inflammation, mucus production, airway hyperresponsiveness, and airway remodeling. BALF monocytes, neutrophils, eosinophils, and lymphocytes were all enhanced in OVA-immunized AhR-/- mice. In OVA-immunized AhR-/- mice, T helper (Th) 17 cell-specific cytokine IL-17A, as well as airway remodeling factors, including epithelial-mesenchymal transition (EMT) markers and vascular endothelial growth factor (VEGF), were all enhanced in lung tissue. Moreover, human cohort studies showed that AhR gene expression in bronchial epithelial cells decreases in severe asthma patients. Loss of AhR leads to worsening of allergic asthma symptoms, indicating its importance in maintaining normal lung function and mediating disease severity.
Collapse
Affiliation(s)
- Yu-Di Chang
- Department of Microbiology and Immunology, School of Medicine, China Medicine University, Taichung, Taiwan
| | - Ching-Hao Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Hao Tsai
- School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Wen Cheng
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jaw-Jou Kang
- School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Chen Lee
- Department of Microbiology and Immunology, School of Medicine, China Medicine University, Taichung, Taiwan
| |
Collapse
|
108
|
Li H, Xu X, Zhang Y, Tang X, Li W. Tetrandrine enhances antitumor effects of the histone deacetylase inhibitor MS-275 in human cancer in a Bax- and p53-dependent manner. Eur J Pharmacol 2020; 888:173575. [PMID: 32950498 DOI: 10.1016/j.ejphar.2020.173575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022]
Abstract
MS-275 (Entinostat), is an oral histone deacetylase (HDAC) inhibitor with a high specificity for class 1 HDACs. As single agent, MS-275 exerts only modest antitumor activity against most solid malignancies. The use of MS-275 in combination with other anticancer agents is currently being evaluated to determine whether this approach can achieve superior therapeutic efficacy. Tetrandrine, a bisbenzylisoquinoline alkaloid isolated from the root of a Chinese medicinal herb, is safe and exhibits low toxicity, showing great potential to enhance chemotherapeutic efficacy. In the present study, we investigated the synergistic antitumor effects of MS-275 in combination with tetrandrine. Based on the results of in vitro experiments, the application of MS-275 in combination with tetrandrine induced selective apoptotic death in various cancer cells but spared normal cells. Mechanistically, the combination treatment induced a dramatic accumulation of reactive oxygen species (ROS), and a pretreatment with the ROS scavenger N-acetyl-L-cysteine (NAC) significantly prevented the cellular apoptosis induced by MS-275/tetrandrine. Moreover, molecular assays indicated that Bax and p53 were the key regulators of MS-275/tetrandrine induced apoptosis. The results of the in vivo studies were consistent with the results of the in vitro studies. Based on our findings, tetrandrine enhanced the antitumor effects of MS-275 in a Bax- and p53-dependent manner. The combination of MS-275 and tetrandrine may represent a novel and promising therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Han Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Xiaoqing Xu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Yudi Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Xianying Tang
- College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, PR China
| | - Wenhua Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, PR China.
| |
Collapse
|
109
|
Radhakrishnan S, Norley J, Wendt S, LeRoy N, Hall H, Norcross S, Doan S, Snaider J, MacVicar BA, Weake VM, Huang L, Tantama M. Neuron Activity Dependent Redox Compartmentation Revealed with a Second Generation Red-Shifted Ratiometric Sensor. ACS Chem Neurosci 2020; 11:2666-2678. [PMID: 32786310 PMCID: PMC7526680 DOI: 10.1021/acschemneuro.0c00342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Oxidative stress is a hallmark of several aging and trauma related neurological disorders, but the precise details of how altered neuronal activity elicits subcellular redox changes have remained difficult to resolve. Current redox sensitive dyes and fluorescent proteins can quantify spatially distinct changes in reactive oxygen species levels, but multicolor probes are needed to accurately analyze compartment-specific redox dynamics in single cells that can be masked by population averaging. We previously engineered genetically encoded red-shifted redox-sensitive fluorescent protein sensors using a Förster resonance energy transfer relay strategy. Here, we developed a second-generation excitation ratiometric sensor called rogRFP2 with improved red emission for quantitative live-cell imaging. Using this sensor to measure activity-dependent redox changes in individual cultured neurons, we observed an anticorrelation in which mitochondrial oxidation was accompanied by a concurrent reduction in the cytosol. This behavior was dependent on the activity of Complex I of the mitochondrial electron transport chain and could be modulated by the presence of cocultured astrocytes. We also demonstrated that the red fluorescent rogRFP2 facilitates ratiometric one- and two-photon redox imaging in rat brain slices and Drosophila retinas. Overall, the proof-of-concept studies reported here demonstrate that this new rogRFP2 redox sensor can be a powerful tool for understanding redox biology both in vitro and in vivo across model organisms.
Collapse
Affiliation(s)
- Saranya Radhakrishnan
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
- Purdue Institute for Integrative Neuroscience, 560 Oval Drive, West Lafayette, IN 47907, United States
- Purdue Interdisciplinary Life Sciences Graduate Program, 560 Oval Drive, West Lafayette, IN 47907, United States
| | - Jacob Norley
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
| | - Stefan Wendt
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Nathan LeRoy
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
| | - Hana Hall
- Department of Biochemistry, 175 South University Street, West Lafayette, IN 47907, United States
| | - Stevie Norcross
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
- Purdue Interdisciplinary Life Sciences Graduate Program, 560 Oval Drive, West Lafayette, IN 47907, United States
| | - Sara Doan
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
| | - Jordan Snaider
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
| | - Brian A. MacVicar
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Vikki M. Weake
- Department of Biochemistry, 175 South University Street, West Lafayette, IN 47907, United States
- Purdue Center for Cancer Research, 175 South University Street, West Lafayette, IN 47907, United States
| | - Libai Huang
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
| | - Mathew Tantama
- Department of Chemistry, 560 Oval Drive, West Lafayette, IN 47907, United States
- Purdue Institute for Integrative Neuroscience, 560 Oval Drive, West Lafayette, IN 47907, United States
- Purdue Interdisciplinary Life Sciences Graduate Program, 560 Oval Drive, West Lafayette, IN 47907, United States
- Department of Chemistry, Wellesley College, 106 Central Street, Wellesley, MA 02481, United States
- Biochemistry Program, Wellesley College, 106 Central Street, Wellesley, MA 02481, United States
| |
Collapse
|
110
|
Roles of Reactive Oxygen Species in Cardiac Differentiation, Reprogramming, and Regenerative Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2102841. [PMID: 32908625 PMCID: PMC7475763 DOI: 10.1155/2020/2102841] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in mechanisms of heart development and regenerative therapies such as the use of pluripotent stem cells. The roles of ROS mediating cell fate are dependent on the intensity of stimuli, cellular context, and metabolic status. ROS mainly act through several targets (such as kinases and transcription factors) and have diverse roles in different stages of cardiac differentiation, proliferation, and maturation. Therefore, further detailed investigation and characterization of redox signaling will help the understanding of the molecular mechanisms of ROS during different cellular processes and enable the design of targeted strategies to foster cardiac regeneration and functional recovery. In this review, we focus on the roles of ROS in cardiac differentiation as well as transdifferentiation (direct reprogramming). The potential mechanisms are discussed in regard to ROS generation pathways and regulation of downstream targets. Further methodological optimization is required for translational research in order to robustly enhance the generation efficiency of cardiac myocytes through metabolic modulations. Additionally, we highlight the deleterious effect of the host's ROS on graft (donor) cells in a paracrine manner during stem cell-based implantation. This knowledge is important for the development of antioxidant strategies to enhance cell survival and engraftment of tissue engineering-based technologies. Thus, proper timing and level of ROS generation after a myocardial injury need to be tailored to ensure the maximal efficacy of regenerative therapies and avoid undesired damage.
Collapse
|
111
|
Gorni D, Finco A. Oxidative stress in elderly population: A prevention screening study. Aging Med (Milton) 2020; 3:205-213. [PMID: 33103041 PMCID: PMC7574639 DOI: 10.1002/agm2.12121] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022] Open
Abstract
Background Aging is a multifactorial phenomenon, characterized by a progressive decline in the efficiency of biochemical and physiological processes and an increased susceptibility to disease. There is increasing evidence that aging and age‐related disease are correlated with an oxidative stress (OS) condition. The latter is characterized by an imbalance between reactive species (RS), in particular reactive oxygen species (ROS) and antioxidant reserve. Objectives The aim of this study is to evaluate the two main markers of oxidative stress, plasmatic peroxide concentration (through d‐ROMs FAST test, derivates‐Reactive Oxygen Metabolites) and plasmatic antioxidant power measured by iron‐reducing power (PAT test, Plasma Antioxidant Test) in 290 apparently healthy volunteers over 60, and their possible correlation with age and gender. Materials and methods Human capillary blood samples from healthy volunteers were used in this observational study for the evaluation of the markers of OS. Results The data obtained broadly demonstrate that the majority of elderly people display an OS condition characterized by increased levels of peroxides and a slight reduction in antioxidant reserve. Conclusions Seniors have a greater propensity to develop a condition of oxidative stress, and therefore it is important to associate the monitoring of oxidative stress markers and, if necessary, antioxidant supplementation, with a healthy lifestyle.
Collapse
Affiliation(s)
- Davide Gorni
- Department of Oxidation Research Cor. Con. International Srl Parma Italy
| | - Annarosa Finco
- Department of Oxidation Research Cor. Con. International Srl Parma Italy
| |
Collapse
|
112
|
Mason SA, Trewin AJ, Parker L, Wadley GD. Antioxidant supplements and endurance exercise: Current evidence and mechanistic insights. Redox Biol 2020; 35:101471. [PMID: 32127289 PMCID: PMC7284926 DOI: 10.1016/j.redox.2020.101471] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 01/07/2023] Open
Abstract
Antioxidant supplements are commonly consumed by endurance athletes to minimize exercise-induced oxidative stress, with the intention of enhancing recovery and improving performance. There are numerous commercially available nutritional supplements that are targeted to athletes and health enthusiasts that allegedly possess antioxidant properties. However, most of these compounds are poorly investigated with respect to their in vivo redox activity and efficacy in humans. Therefore, this review will firstly provide a background to endurance exercise-related redox signalling and the subsequent adaptations in skeletal muscle and vascular function. The review will then discuss commonly available compounds with purported antioxidant effects for use by athletes. N-acetyl cysteine may be of benefit over the days prior to an endurance event; while chronic intake of combined 1000 mg vitamin C + vitamin E is not recommended during periods of heavy training associated with adaptations in skeletal muscle. Melatonin, vitamin E and α-lipoic acid appear effective at decreasing markers of exercise-induced oxidative stress. However, evidence on their effects on endurance performance are either lacking or not supportive. Catechins, anthocyanins, coenzyme Q10 and vitamin C may improve vascular function, however, evidence is either limited to specific sub-populations and/or does not translate to improved performance. Finally, additional research should clarify the potential benefits of curcumin in improving muscle recovery post intensive exercise; and the potential hampering effects of astaxanthin, selenium and vitamin A on skeletal muscle adaptations to endurance training. Overall, we highlight the lack of supportive evidence for most antioxidant compounds to recommend to athletes.
Collapse
Affiliation(s)
- Shaun A Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Adam J Trewin
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Lewan Parker
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia.
| |
Collapse
|
113
|
Jackson MJ, Stretton C, McArdle A. Hydrogen peroxide as a signal for skeletal muscle adaptations to exercise: What do concentrations tell us about potential mechanisms? Redox Biol 2020; 35:101484. [PMID: 32184060 PMCID: PMC7284923 DOI: 10.1016/j.redox.2020.101484] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 12/26/2022] Open
Abstract
Hydrogen peroxide appears to be the key reactive oxygen species involved in redox signalling, but comparisons of the low concentrations of hydrogen peroxide that are calculated to exist within cells with those previously shown to activate common signalling events in vitro indicate that direct oxidation of key thiol groups on "redox-sensitive" signalling proteins is unlikely to occur. A number of potential mechanisms have been proposed to explain how cells overcome this block to hydrogen peroxide-stimulated redox signalling and these will be discussed in the context of the redox-stimulation of specific adaptations of skeletal muscle to contractile activity and exercise. It is argued that current data implicate a role for currently unidentified effector molecules (likely to be highly reactive peroxidases) in propagation of the redox signal from sites of hydrogen peroxide generation to common adaptive signalling pathways.
Collapse
Affiliation(s)
- Malcolm J Jackson
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L87TX, UK.
| | - Clare Stretton
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L87TX, UK
| | - Anne McArdle
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L87TX, UK
| |
Collapse
|
114
|
Margaritelis NV, Paschalis V, Theodorou AA, Kyparos A, Nikolaidis MG. Redox basis of exercise physiology. Redox Biol 2020; 35:101499. [PMID: 32192916 PMCID: PMC7284946 DOI: 10.1016/j.redox.2020.101499] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/20/2020] [Accepted: 03/05/2020] [Indexed: 12/15/2022] Open
Abstract
Redox reactions control fundamental processes of human biology. Therefore, it is safe to assume that the responses and adaptations to exercise are, at least in part, mediated by redox reactions. In this review, we are trying to show that redox reactions are the basis of exercise physiology by outlining the redox signaling pathways that regulate four characteristic acute exercise-induced responses (muscle contractile function, glucose uptake, blood flow and bioenergetics) and four chronic exercise-induced adaptations (mitochondrial biogenesis, muscle hypertrophy, angiogenesis and redox homeostasis). Based on our analysis, we argue that redox regulation should be acknowledged as central to exercise physiology.
Collapse
Affiliation(s)
- N V Margaritelis
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece; Dialysis Unit, 424 General Military Hospital of Thessaloniki, Thessaloniki, Greece.
| | - V Paschalis
- School of Physical Education and Sport Science, National and Kapodistrian University of Athens, Athens, Greece
| | - A A Theodorou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - A Kyparos
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - M G Nikolaidis
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
115
|
Duan J, Zhang T, Gaffrey MJ, Weitz KK, Moore RJ, Li X, Xian M, Thrall BD, Qian WJ. Stochiometric quantification of the thiol redox proteome of macrophages reveals subcellular compartmentalization and susceptibility to oxidative perturbations. Redox Biol 2020; 36:101649. [PMID: 32750668 PMCID: PMC7397701 DOI: 10.1016/j.redox.2020.101649] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/24/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
Posttranslational modifications of protein cysteine thiols play a significant role in redox regulation and the pathogenesis of human diseases. Herein, we report the characterization of the cellular redox landscape in terms of quantitative, site-specific occupancies of both S-glutathionylation (SSG) and total reversible thiol oxidation (total oxidation) in RAW 264.7 macrophage cells under basal conditions. The occupancies of thiol modifications for ~4000 cysteine sites were quantified, revealing a mean site occupancy of 4.0% for SSG and 11.9% for total oxidation, respectively. Correlations between site occupancies and structural features such as pKa, relative residue surface accessibility, and hydrophobicity were observed. Proteome-wide site occupancy analysis revealed that the average occupancies of SSG and total oxidation in specific cellular compartments correlate well with the expected redox potentials of respective organelles in macrophages, consistent with the notion of redox compartmentalization. The lowest average occupancies were observed in more reducing organelles such as the mitochondria (non-membrane) and nucleus, while the highest average occupancies were found in more oxidizing organelles such as endoplasmic reticulum (ER) and lysosome. Furthermore, a pattern of subcellular susceptibility to redox changes was observed under oxidative stress induced by exposure to engineered metal oxide nanoparticles. Peroxisome, ER, and mitochondria (membrane) are the organelles which exhibit the most significant redox changes; while mitochondria (non-membrane) and Golgi were observed as the organelles being most resistant to oxidative stress. Finally, it was observed that Cys residues at enzymatic active sites generally had a higher level of occupancy compared to non-active Cys residues within the same proteins, suggesting site occupancy as a potential indicator of protein functional sites. The raw data are available via ProteomeXchange with identifier PXD019913.
Collapse
Affiliation(s)
- Jicheng Duan
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Tong Zhang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Matthew J Gaffrey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Xiaolu Li
- Department of Biological Systems Engineering, Washington State University, Richland, WA, USA
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - Brian D Thrall
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
116
|
The vicious cycle between ferritinophagy and ROS production triggered EMT inhibition of gastric cancer cells was through p53/AKT/mTor pathway. Chem Biol Interact 2020; 328:109196. [PMID: 32687844 DOI: 10.1016/j.cbi.2020.109196] [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: 05/04/2020] [Revised: 06/23/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
Cancer metastasis and resistance for chemotherapeutic agent correlate with epithelial-mesenchymal transition (EMT), while ROS production also involves in the EMT process, However, how autophagy mediated ROS production affects EMT remains unclear. Previous study showed that DpdtC (2,2'-di-pyridylketone hydrazone dithiocarbamate) could induce ferritinophagy in HepG2 cell. To insight into more details that how ferritinophagy affects cellular feature, the SGC-7901and BGC-823 gastric cancer cell lines were used. Interestingly DpdtC treatment resulted in EMT inhibition and was ROS dependent. Similar situation occurred in TGF-β1 induced EMT model, supporting that DpdtC was able to inhibit EMT. Next the ability of DpdtC in ferritinophagy induction was further evaluated. As expected, DpdtC induced ferritinophagy in the absence and presence of TGF-β1. The correlation analysis revealed that an enhanced ferritinophagic flux contributed to the EMT inhibition. In addition, ferritinophagy triggers Fenton reaction, resulting in ROS production which give rise of p53 response, thus the role of p53 was further investigated. DpdtC treatment resulted in upregulation of p53, but, the addition of p53 inhibitor, PFT-α could significantly neutralize the action of DpdtC on ferritinophagy induction and EMT inhibition. Furthermore, autophagy inhibitors or NAC could counteract the action of DpdtC, indicating that ferrtinophagy-mediated ROS played an important role in the cellular events. In addition to upregulation of p53, its down-stream targets, AKT/mTor were also downregulated, supporting that DpdtC induced EMT inhibition was achieved through ferritinophagy-ROS vicious cycle mediated p53/AKT/mTor pathway. And the activation of ferritinophagic flux was the dominant driving force in action of DpdtC in gastric cancer cells.
Collapse
|
117
|
Khan SA, Choudhury R, Majumdar M, Nandi NB, Roy S, Misra TK. Gluconate‐Stabilized Silver Nanoparticles as pH Dependent Dual‐Nanosensor for Quantitative Evaluation of Methionine and Cysteine. ChemistrySelect 2020. [DOI: 10.1002/slct.202001654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Shamim Ahmed Khan
- Department of ChemistryNational Institute of Technology Agartala Agartala Tripura 799046 India
| | - Rupasree Choudhury
- Department of ChemistryNational Institute of Technology Agartala Agartala Tripura 799046 India
| | - Moumita Majumdar
- Department of ChemistryNational Institute of Technology Agartala Agartala Tripura 799046 India
| | | | - Shaktibrata Roy
- Department of ChemistryNational Institute of Technology Agartala Agartala Tripura 799046 India
| | - Tarun Kumar Misra
- Department of ChemistryNational Institute of Technology Agartala Agartala Tripura 799046 India
| |
Collapse
|
118
|
Khan SA, Choudhury R, Majumdar M, Misra TK. Development of dual-tool nanosensor for cysteine based on N-(1-naphthyl)ethylenediamine cation functionalized silver nanoparticles. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 234:118240. [PMID: 32172188 DOI: 10.1016/j.saa.2020.118240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 06/10/2023]
Abstract
In an accomplishment of development of silver nanoparticles (AgNPs) based nanosensor for cysteine in its anionic and neutral forms, we have preferred N-(1-naphthyl)ethylenediamine cation (NEDA+) stabilized AgNPs (NEDA-AgNPs), because NEDA+ is a fluorescent active ion and it imparts excellent stability to AgNPs. Surface Plasmon resonance (SPR) of AgNPs and fluorescence property of NEDA+ are thus useful for presenting NEDA-AgNPs as a dual-tool nanosensor for cysteine molecules. The surface adsorbed NEDA+ cations interact selectively with cysteine as a consequence, the particles get aggregated, which was monitored using spectrophotometric method. The fluorescence property of NEDA+ is heavily quenched in NEDA-AgNPs, which could be reversed in presence of cysteine. The spectrofluorimetric method was thus used for quantification of cysteine as well. The detection limits (LOD to LOL) of anionic cysteine are 0.1784-1.598 μM and 0.0842-2.0 μM, respectively in spectrophotometric and spectrofluorimetric methods. From a real sample matrix, the recovery results are excellent, >95%. For neutral cysteine, the sensitivity is a bit low; 0.308-2.8 μM for spectrophotometric and 0.131-2.8 μM for spectrofluorimetric methods. It is found that the anionic cysteine (Kasso = 2.24 × 105 M-1/4.02 × 105 M-1) binds surface adsorbed NEDA+ cations strongly than that of neutral cysteine (Kasso = 3.69 × 104 M-1/1.24 × 105 M-1). Thus, NEDA-AgNPs show its potentials for being a dual-tool nanosensor as well as dual-form nanosensor for quantification of cysteine in a sample which may be the attractive system to an analyst.
Collapse
Affiliation(s)
- Shamim Ahmed Khan
- Department of Chemistry, National Institute of Technology Agartala, Agartala, Tripura 799046, India
| | - Rupasree Choudhury
- Department of Chemistry, National Institute of Technology Agartala, Agartala, Tripura 799046, India
| | - Moumita Majumdar
- Department of Chemistry, National Institute of Technology Agartala, Agartala, Tripura 799046, India
| | - Tarun Kumar Misra
- Department of Chemistry, National Institute of Technology Agartala, Agartala, Tripura 799046, India.
| |
Collapse
|
119
|
Science and Healthy Meals in the World: Nutritional Epigenomics and Nutrigenetics of the Mediterranean Diet. Nutrients 2020; 12:nu12061748. [PMID: 32545252 PMCID: PMC7353392 DOI: 10.3390/nu12061748] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
The Mediterranean Diet (MD), UNESCO Intangible Cultural Heritage of Humanity, has become a scientific topic of high interest due to its health benefits. The aim of this review is to pick up selected studies that report nutrigenomic or nutrigenetic data and recapitulate some of the biochemical/genomic/genetic aspects involved in the positive health effects of the MD. These include (i) the antioxidative potential of its constituents with protective effects against several diseases; (ii) the epigenetic and epigenomic effects exerted by food components, such as Indacaxanthin, Sulforaphane, and 3-Hydroxytyrosol among others, and their involvement in the modulation of miRNA expression; (iii) the existence of predisposing or protective human genotypes due to allelic diversities and the impact of the MD on disease risk. A part of the review is dedicated to the nutrigenomic effects of the main cooking methods used in the MD and also to a comparative analysis of the nutrigenomic properties of the MD and other diet regimens and non-MD-related aliments. Taking all the data into account, the traditional MD emerges as a diet with a high antioxidant and nutrigenomic modulation power, which is an example of the “Environment-Livings-Environment” relationship and an excellent patchwork of interconnected biological actions working toward human health.
Collapse
|
120
|
Guo P, Chen S, Li D, Zhang J, Luo J, Zhang A, Yu D, Bloom MS, Chen L, Chen W. SFPQ is involved in regulating arsenic-induced oxidative stress by interacting with the miRNA-induced silencing complexes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 261:114160. [PMID: 32066060 DOI: 10.1016/j.envpol.2020.114160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 06/10/2023]
Abstract
Arsenic exposure contributed to the development of human diseases. Arsenic exerted multiple organ toxicities mainly by triggering oxidative stress. However, the signaling pathway underlying oxidative stress is unclear. We previously found that the expression of SFPQ, a splicing factor, was positively associated with urinary arsenic concentration in an arsenic-exposed population, suggesting an oxidative stress regulatory role for SFPQ. To test this hypothesis, we established cell models of oxidative stress in human hepatocyte cells (L02) treated with NaAsO2. Reactive oxygen species (ROS) synthesis displayed a time- and dose-dependent increase with NaAsO2 treatment. SFPQ suppression resulted in a 36%-53% decrease in ROS generation, leading to enhanced cellular damage determined by 8-OHdG, comet tail moment, and micronucleus analysis. Particularly, SFPQ deficiency attenuated expression of the oxidase genes DUOX1, DUOX2, NCF2, and NOX2. A fluorescent-based RNA electrophoretic mobility shift assay (FREMSA) and dual-luciferase reporter system revealed that miR-92b-5p targeted DUOX2 mRNA degradation. An RNA immunoprecipitation assay showed an interaction between SFPQ and miR-92b-5p of the miRNA-induced silencing complex (miRISC). Notably, NaAsO2 treatment diminished the interaction between SFPQ and miR92b-5p, accompanied by decreased binding between miR-92b-5p and 3'-UTR of DUOX2. However, SFPQ deficiency suppressed the dissociation of miR-92b-5p from 3'-UTR of DUOX2, indicating that miR-92b-5p regulated the SFPQ-dependent DUOX2 expression. Taken together, we reveal that SFPQ responds to arsenic-induced oxidative stress by interacting with the miRISC. These findings offer new insight into the potential role of SFPQ in regulating cellular stress response.
Collapse
Affiliation(s)
- Ping Guo
- The Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shen Chen
- The Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Daochuan Li
- The Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jinmiao Zhang
- The Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Aihua Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Michael S Bloom
- Departments of Environmental Health Sciences & Epidemiology and Biostatistics, University at Albany, State University of New York, Rensselaer, NY 12144, USA
| | - Liping Chen
- The Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Wen Chen
- The Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
121
|
Tadi S, Sharp JS. Top-Down ETD-MS Provides Unreliable Quantitation of Methionine Oxidation. J Biomol Tech 2020; 30:50-57. [PMID: 31662705 DOI: 10.7171/jbt.19-3004-002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Methionine oxidation plays a critical role in many processes of biologic and biomedical importance, including cellular redox responses and stability of protein pharmaceuticals. Bottom-up methods for analysis of methionine oxidation can suffer from incomplete sequence coverage, as well as an inability to readily detect correlated oxidation between 2 or more methionines. However, the methodology for quantifying protein oxidation in top-down analyses is lacking. Previous work has shown that electron transfer dissociation (ETD)-based tandem mass spectrometry (MS/MS) fragmentation offers accurate and precise quantification of amino acid oxidation in peptides, even in complex samples. However, the ability of ETD-based MS/MS fragmentation to accurately quantify amino acid oxidation of proteins in a top-down manner has not been reported. Using apomyoglobin and calmodulin as model proteins, we partially converted methionines into methionine sulfoxide by incubation in H2O2. Using top-down ETD-based fragmentation, we quantified the amount of oxidation of various ETD product ions and compared the quantified values with those from traditional bottom-up analysis. We find that overall quantification of methionine oxidation by top-down MS/MS ranges from good agreement with traditional bottom-up methods to vast differences between the 2 techniques, including missing oxidized product ions and large differences in measured oxidation quantities. Care must be taken in transitioning ETD-based quantitation of oxidation from the peptide level to the intact protein level.
Collapse
Affiliation(s)
- Surendar Tadi
- Department of Biomolecular Sciences, University of Mississippi, Oxford, Mississippi, 38677, USA
| | - Joshua S Sharp
- Department of Biomolecular Sciences, University of Mississippi, Oxford, Mississippi, 38677, USA
| |
Collapse
|
122
|
Ebrahimi SO, Reiisi S, Shareef S. miRNAs, oxidative stress, and cancer: A comprehensive and updated review. J Cell Physiol 2020; 235:8812-8825. [PMID: 32394436 DOI: 10.1002/jcp.29724] [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: 09/12/2019] [Accepted: 04/07/2020] [Indexed: 01/17/2023]
Abstract
Oxidative stress refers to elevated levels of intracellular reactive oxygen species (ROS). ROS homeostasis functions as a signaling pathway for normal cell survival and appropriate cell signaling. Chronic inflammation induced by imbalanced levels of ROS contributes to many diseases and different types of cancer. ROS can alter the expression of oncogenes and tumor suppressor genes through epigenetic modifications, transcription factors, and non-coding RNAs. MicroRNAs (miRNAs) are small non-coding RNAs that play a key role in most biological pathways. Each miRNA regulates hundreds of target genes by inhibiting protein translation and/or promoting messenger RNA degradation. In normal conditions, miRNAs play a physiological role in cell proliferation, differentiation, and apoptosis. However, different factors that can dysregulate cell signaling and cellular homeostasis can also affect miRNA expression. The alteration of miRNA expression can work against disturbing factors or mediate their effects. Oxidative stress is one of these factors. Considering the complex interplay between ROS level and miRNA regulation and both of these with cancer development, we review the role of miRNAs in cancer, focusing on their function in oxidative stress.
Collapse
Affiliation(s)
- Seyed Omar Ebrahimi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Somayeh Reiisi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Salar Shareef
- Department of Medical Laboratory Science, College of Sciences, University of Raparin, Ranya, Kurdistan Region, Iraq
| |
Collapse
|
123
|
Andreadou I, Schulz R, Papapetropoulos A, Turan B, Ytrehus K, Ferdinandy P, Daiber A, Di Lisa F. The role of mitochondrial reactive oxygen species, NO and H 2 S in ischaemia/reperfusion injury and cardioprotection. J Cell Mol Med 2020; 24:6510-6522. [PMID: 32383522 PMCID: PMC7299678 DOI: 10.1111/jcmm.15279] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/04/2020] [Accepted: 03/08/2020] [Indexed: 12/12/2022] Open
Abstract
Redox signalling in mitochondria plays an important role in myocardial ischaemia/reperfusion (I/R) injury and in cardioprotection. Reactive oxygen and nitrogen species (ROS/RNS) modify cellular structures and functions by means of covalent changes in proteins including among others S‐nitros(yl)ation by nitric oxide (NO) and its derivatives, and S‐sulphydration by hydrogen sulphide (H2S). Many enzymes are involved in the mitochondrial formation and handling of ROS, NO and H2S under physiological and pathological conditions. In particular, the balance between formation and removal of reactive species is impaired during I/R favouring their accumulation. Therefore, various interventions aimed at decreasing mitochondrial ROS accumulation have been developed and have shown cardioprotective effects in experimental settings. However, ROS, NO and H2S play also a role in endogenous cardioprotection, as in the case of ischaemic pre‐conditioning, so that preventing their increase might hamper self‐defence mechanisms. The aim of the present review was to provide a critical analysis of formation and role of reactive species, NO and H2S in mitochondria, with a special emphasis on mechanisms of injury and protection that determine the fate of hearts subjected to I/R. The elucidation of the signalling pathways of ROS, NO and H2S is likely to reveal novel molecular targets for cardioprotection that could be modulated by pharmacological agents to prevent I/R injury.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Andreas Daiber
- Molecular Cardiology, Center for Cardiology 1, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Fabio Di Lisa
- Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| |
Collapse
|
124
|
Davies MJ, Hawkins CL. The Role of Myeloperoxidase in Biomolecule Modification, Chronic Inflammation, and Disease. Antioxid Redox Signal 2020; 32:957-981. [PMID: 31989833 DOI: 10.1089/ars.2020.8030] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Significance: The release of myeloperoxidase (MPO) by activated leukocytes is critical in innate immune responses. MPO produces hypochlorous acid (HOCl) and other strong oxidants, which kill bacteria and other invading pathogens. However, MPO also drives the development of numerous chronic inflammatory pathologies, including atherosclerosis, neurodegenerative disease, lung disease, arthritis, cancer, and kidney disease, which are globally responsible for significant patient mortality and morbidity. Recent Advances: The development of imaging approaches to precisely identify the localization of MPO and the molecular targets of HOCl in vivo is an important advance, as typically the involvement of MPO in inflammatory disease has been inferred by its presence, together with the detection of biomarkers of HOCl, in biological fluids or diseased tissues. This will provide valuable information in regard to the cell types responsible for releasing MPO in vivo, together with new insight into potential therapeutic opportunities. Critical Issues: Although there is little doubt as to the value of MPO inhibition as a protective strategy to mitigate tissue damage during chronic inflammation in experimental models, the impact of long-term inhibition of MPO as a therapeutic strategy for human disease remains uncertain, in light of the potential effects on innate immunity. Future Directions: The development of more targeted MPO inhibitors or a treatment regimen designed to reduce MPO-associated host tissue damage without compromising pathogen killing by the innate immune system is therefore an important future direction. Similarly, a partial MPO inhibition strategy may be sufficient to maintain adequate bacterial activity while decreasing the propagation of inflammatory pathologies.
Collapse
Affiliation(s)
- Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
125
|
Cobley JN, Husi H. Immunological Techniques to Assess Protein Thiol Redox State: Opportunities, Challenges and Solutions. Antioxidants (Basel) 2020; 9:E315. [PMID: 32326525 PMCID: PMC7222201 DOI: 10.3390/antiox9040315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
To understand oxidative stress, antioxidant defense, and redox signaling in health and disease it is essential to assess protein thiol redox state. Protein thiol redox state is seldom assessed immunologically because of the inability to distinguish reduced and reversibly oxidized thiols by Western blotting. An underappreciated opportunity exists to use Click PEGylation to realize the transformative power of simple, time and cost-efficient immunological techniques. Click PEGylation harnesses selective, bio-orthogonal Click chemistry to separate reduced and reversibly oxidized thiols by selectively ligating a low molecular weight polyethylene glycol moiety to the redox state of interest. The resultant ability to disambiguate reduced and reversibly oxidized species by Western blotting enables Click PEGylation to assess protein thiol redox state. In the present review, to enable investigators to effectively harness immunological techniques to assess protein thiol redox state we critique the chemistry, promise and challenges of Click PEGylation.
Collapse
Affiliation(s)
- James Nathan Cobley
- Centre for Health Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK;
| | | |
Collapse
|
126
|
Benhar M. Oxidants, Antioxidants and Thiol Redox Switches in the Control of Regulated Cell Death Pathways. Antioxidants (Basel) 2020; 9:antiox9040309. [PMID: 32290499 PMCID: PMC7222211 DOI: 10.3390/antiox9040309] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022] Open
Abstract
It is well appreciated that biological reactive oxygen and nitrogen species such as hydrogen peroxide, superoxide and nitric oxide, as well as endogenous antioxidant systems, are important modulators of cell survival and death in diverse organisms and cell types. In addition, oxidative stress, nitrosative stress and dysregulated cell death are implicated in a wide variety of pathological conditions, including cancer, cardiovascular and neurological diseases. Therefore, much effort is devoted to elucidate the molecular mechanisms linking oxidant/antioxidant systems and cell death pathways. This review is focused on thiol redox modifications as a major mechanism by which oxidants and antioxidants influence specific regulated cell death pathways in mammalian cells. Growing evidence indicates that redox modifications of cysteine residues in proteins are involved in the regulation of multiple cell death modalities, including apoptosis, necroptosis and pyroptosis. In addition, recent research suggests that thiol redox switches play a role in the crosstalk between apoptotic and necrotic forms of regulated cell death. Thus, thiol-based redox circuits provide an additional layer of control that determines when and how cells die.
Collapse
Affiliation(s)
- Moran Benhar
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
127
|
Dustin CM, Heppner DE, Lin MCJ, van der Vliet A. Redox regulation of tyrosine kinase signalling: more than meets the eye. J Biochem 2020; 167:151-163. [PMID: 31599960 DOI: 10.1093/jb/mvz085] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Protein kinases are essential mediators of cellular signal transduction and are often dysregulated in disease. Among these, protein tyrosine kinases (PTKs) have received specific interest due to their common roles in various diseases including cancer, and emerging observations indicating that PTK signalling pathways are susceptible to regulation by reactive oxygen species (ROS), which are also frequently implicated in disease pathology. While it is well recognized that ROS can impact on tyrosine kinase signalling by inhibiting tyrosine phosphatases, more recent studies highlight additional modes of redox-based regulation of tyrosine kinase signalling by direct redox modification of non-catalytic cysteines within tyrosine kinases or other protein components of this signalling pathway. In this review, we will present recent advancements with respect to redox-based mechanisms in regulating PTK signalling, with a specific focus on recent studies demonstrating direct redox regulation of Src-family kinases and epidermal growth factor receptor kinases. Importantly, redox-based modulation of tyrosine kinases may be relevant for many other kinases and has implications for current approaches to develop pharmacological inhibitors for these proteins.
Collapse
Affiliation(s)
- Christopher M Dustin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David E Heppner
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Miao-Chong J Lin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| |
Collapse
|
128
|
Janssen-Heininger Y, Reynaert NL, van der Vliet A, Anathy V. Endoplasmic reticulum stress and glutathione therapeutics in chronic lung diseases. Redox Biol 2020; 33:101516. [PMID: 32249209 PMCID: PMC7251249 DOI: 10.1016/j.redox.2020.101516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
129
|
Jahandideh F, Wu J. Perspectives on the Potential Benefits of Antihypertensive Peptides towards Metabolic Syndrome. Int J Mol Sci 2020; 21:E2192. [PMID: 32235782 PMCID: PMC7139547 DOI: 10.3390/ijms21062192] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
In addition to the regulation of blood pressure, the renin-angiotensin system (RAS) also plays a key role in the onset and development of insulin resistance, which is central to metabolic syndrome (MetS). Due to the interplay between RAS and insulin resistance, antihypertensive compounds may exert beneficial effects in the management of MetS. Food-derived bioactive peptides with RAS blocking properties can potentially improve adipose tissue dysfunction, glucose intolerance, and insulin resistance involved in the pathogenesis of MetS. This review discusses the pathophysiology of hypertension and the association between RAS and pathogenesis of the MetS. The effects of bioactive peptides with RAS modulating effects on other components of the MetS are discussed. While the in vivo reports on the effectiveness of antihypertensive peptides against MetS are encouraging, the exact mechanism by which these peptides infer their effects on glucose and lipid handling is mostly unknown. Therefore, careful design of experiments along with standardized physiological models to study the effect of antihypertensive peptides on insulin resistance and obesity could help to clarify this relationship.
Collapse
Affiliation(s)
- Forough Jahandideh
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
130
|
The Role of Reactive Oxygen Species in the Life Cycle of the Mitochondrion. Int J Mol Sci 2020; 21:ijms21062173. [PMID: 32245255 PMCID: PMC7139706 DOI: 10.3390/ijms21062173] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/12/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023] Open
Abstract
Currently, it is known that, in living systems, free radicals and other reactive oxygen and nitrogen species play a double role, because they can cause oxidative damage and tissue dysfunction and serve as molecular signals activating stress responses that are beneficial to the organism. It is also known that mitochondria, because of their capacity to produce free radicals, play a major role in tissue oxidative damage and dysfunction and provide protection against excessive tissue dysfunction through several mechanisms, including the stimulation of permeability transition pore opening. This process leads to mitoptosis and mitophagy, two sequential processes that are a universal route of elimination of dysfunctional mitochondria and is essential to protect cells from the harm due to mitochondrial disordered metabolism. To date, there is significant evidence not only that the above processes are induced by enhanced reactive oxygen species (ROS) production, but also that such production is involved in the other phases of the mitochondrial life cycle. Accumulating evidence also suggests that these effects are mediated through the regulation of the expression and the activity of proteins that are engaged in processes such as genesis, fission, fusion, and removal of mitochondria. This review provides an account of the developments of the knowledge on the dynamics of the mitochondrial population, examining the mechanisms governing their genesis, life, and death, and elucidating the role played by free radicals in such processes.
Collapse
|
131
|
VanHecke GC, Abeywardana MY, Huang B, Ahn YH. Isotopically Labeled Clickable Glutathione to Quantify Protein S-Glutathionylation. Chembiochem 2020; 21:853-859. [PMID: 31560820 PMCID: PMC7078011 DOI: 10.1002/cbic.201900528] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Indexed: 12/28/2022]
Abstract
Protein S-glutathionylation is one of the important cysteine oxidation events that regulate various redox-mediated biological processes. Despite several existing methods, there are few proteomic approaches to identify and quantify specific cysteine residues susceptible to S-glutathionylation. We previously developed a clickable glutathione approach that labels intracellular glutathione with azido-Ala by using a mutant form of glutathione synthetase. In this study, we developed a quantification strategy with clickable glutathione by using isotopically labeled heavy and light derivatives of azido-Ala, which provides the relative quantification of glutathionylated peptides in mass spectrometry-based proteomic analysis. We applied isotopically labeled clickable glutathione to HL-1 cardiomyocytes, quantifying relative levels of 1398 glutathionylated peptides upon addition of hydrogen peroxide. Importantly, we highlight elevated levels of glutathionylation on sarcomere-associated muscle proteins while validating glutathionylation of two structural proteins, α-actinin and desmin. Our report provides a chemical proteomic strategy to quantify specific glutathionylated cysteines.
Collapse
Affiliation(s)
- Garrett C. VanHecke
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, MI 48202, USA
| | | | - Bo Huang
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, MI 48202, USA
| | - Young-Hoon Ahn
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, MI 48202, USA
| |
Collapse
|
132
|
Sexual hormones regulate the redox status and mitochondrial function in the brain. Pathological implications. Redox Biol 2020; 31:101505. [PMID: 32201220 PMCID: PMC7212485 DOI: 10.1016/j.redox.2020.101505] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 02/11/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
Compared to other organs, the brain is especially exposed to oxidative stress. In general, brains from young females tend to present lower oxidative damage in comparison to their male counterparts. This has been attributed to higher antioxidant defenses and a better mitochondrial function in females, which has been linked to neuroprotection in this group. However, these differences usually disappear with aging, and the incidence of brain pathologies increases in aged females. Sexual hormones, which suffer a decrease with normal aging, have been proposed as the key factors involved in these gender differences. Here, we provide an overview of redox status and mitochondrial function regulation by sexual hormones and their influence in normal brain aging. Furthermore, we discuss how sexual hormones, as well as phytoestrogens, may play an important role in the development and progression of several brain pathologies, including neurodegenerative diseases such as Alzheimer's and Parkinson's diseases, stroke or brain cancer. Sex hormones are reduced with aging, especially in females, affecting redox balance. Normal aging is associated to a worse redox homeostasis in the brain. Young females show better mitochondrial function and higher antioxidant defenses. Development of brain pathologies is influenced by sex hormones and phytoestrogens.
Collapse
|
133
|
Otoupalova E, Smith S, Cheng G, Thannickal VJ. Oxidative Stress in Pulmonary Fibrosis. Compr Physiol 2020; 10:509-547. [PMID: 32163196 DOI: 10.1002/cphy.c190017] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress has been linked to various disease states as well as physiological aging. The lungs are uniquely exposed to a highly oxidizing environment and have evolved several mechanisms to attenuate oxidative stress. Idiopathic pulmonary fibrosis (IPF) is a progressive age-related disorder that leads to architectural remodeling, impaired gas exchange, respiratory failure, and death. In this article, we discuss cellular sources of oxidant production, and antioxidant defenses, both enzymatic and nonenzymatic. We outline the current understanding of the pathogenesis of IPF and how oxidative stress contributes to fibrosis. Further, we link oxidative stress to the biology of aging that involves DNA damage responses, loss of proteostasis, and mitochondrial dysfunction. We discuss the recent findings on the role of reactive oxygen species (ROS) in specific fibrotic processes such as macrophage polarization and immunosenescence, alveolar epithelial cell apoptosis and senescence, myofibroblast differentiation and senescence, and alterations in the acellular extracellular matrix. Finally, we provide an overview of the current preclinical studies and clinical trials targeting oxidative stress in fibrosis and potential new strategies for future therapeutic interventions. © 2020 American Physiological Society. Compr Physiol 10:509-547, 2020.
Collapse
Affiliation(s)
- Eva Otoupalova
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sam Smith
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Guangjie Cheng
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
134
|
DpdtC-Induced EMT Inhibition in MGC-803 Cells Was Partly through Ferritinophagy-Mediated ROS/p53 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9762390. [PMID: 32256964 PMCID: PMC7091554 DOI: 10.1155/2020/9762390] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 02/14/2020] [Indexed: 12/22/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular process in which epithelial cells are partially transformed into stromal cells, which endows the polarized epithelium cells more invasive feature and contributes cancer metastasis and drug resistance. Ferritinophagy is an event of ferritin degradation in lysosomes, which contributes Fenton-mediated ROS production. In addition, some studies have shown that ROS participates in EMT process, but the effect of ROS stemmed from ferritin degradation on EMT has not been fully established. A novel iron chelator, DpdtC (2,2'-di-pyridylketone dithiocarbamate), which could induce ferritinophagy in HepG2 cell in our previous study, was used to investigate its effect on EMT in gastric cancer cells. The proliferation assay showed that DpdtC treatment resulted in growth inhibition and morphologic alteration in MGC-803 cell (IC50 = 3.1 ± 0.3 μM), and its action involved ROS production that was due to the occurrence of ferritinophagy. More interestingly, DpdtC could also inhibit EMT, leading to the upregulation of E-cadherin and the downregulation of vimentin; however, the addition of NAC and 3-MA could attenuate (or neutralize) the action of DpdtC on ferritinophagy induction and EMT inhibition, supporting that the enhanced ferritinophagic flux contributed to the EMT inhibition. Since the degradation of ferritin may trigger the production of ROS and induce the response of p53, we next studied the role of p53 in the above two-cell events. As expected, an upregulation of p53 was observed after DpdtC insulting; however, the addition of a p53 inhibitor, PFT-α, could significantly attenuate the action of DpdtC on ferritinophagy induction and EMT inhibition. In addition, autophagy inhibitors or NAC could counteract the effect of DpdtC and restore the level of p53 to the control group, indicating that the upregulation of p53 was caused by ferritinophagy-mediated ROS production. In conclusion, our data demonstrated that the inhibition of EMT induced by DpdtC was realized through ferritinophagy-mediated ROS/p53 pathway, which supported that the activation of ferritinophagic flux was the main driving force in EMT inhibition in gastric cancer cells, and further strengthening the concept that NCOA4 participates in EMT process.
Collapse
|
135
|
Jackson MJ. Mechanistic models to guide redox investigations and interventions in musculoskeletal ageing. Free Radic Biol Med 2020; 149:2-7. [PMID: 31981622 DOI: 10.1016/j.freeradbiomed.2020.01.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 10/25/2022]
Abstract
Age is the greatest risk factor for the major chronic musculoskeletal disorders, osteoarthritis, osteoporosis and age-related loss of skeletal muscle mass and function (sarcopenia). Dramatic advances in understanding of the fundamental mechanisms underlying the ageing process are being exploited to understand the causes of these age-related disorders and identify approaches to prevent or treat these disorders. This review will focus on one of these fundamental mechanisms, redox regulation, and the role of redox changes in age-related loss of skeletal muscle mass and function (sarcopenia). Key to understanding the role of such pathways has been the development and study of experimental models of musculoskeletal ageing that are designed to examine the effect of modification of ROS regulatory enzymes. These have primarily involved genetic deletion of regulatory enzymes for ROS in mice. Many of the models studied show increased oxidative damage in tissues, but no clear relationship with skeletal muscle aging has been seen The exception to this has been mice with disruption of the superoxide dismutases and, in particular, deletion of Cu,ZnSOD (SOD1) localised in the cytosol and mitochondrial intermembrane space. Studies of tissue specific models lacking SOD1 have highlighted the potential role that disrupted redox pathways can play in muscle loss and weakness and have demonstrated the need to study both motor neurons and muscle to understand age-related loss of skeletal muscle. The complex interplay that has been identified between changes in redox homeostasis in the motor neuron and skeletal muscle and their role in premature loss of muscle mass and function illustrates the utility of modifiable models to establish key pathways that may contribute to age-related changes and identify potential logical approaches to intervention.
Collapse
Affiliation(s)
- Malcolm J Jackson
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing & Chronic Disease, University of Liverpool, Liverpool, L78TX, UK.
| |
Collapse
|
136
|
Woodroofe CC, Ivanic J, Monti S, Levine RL, Swenson RE. Repurposing the Pummerer Rearrangement: Determination of Methionine Sulfoxides in Peptides. Chembiochem 2020; 21:508-516. [PMID: 31365170 PMCID: PMC7065062 DOI: 10.1002/cbic.201900463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Indexed: 12/29/2022]
Abstract
The reversible oxidation of methionine residues in proteins has emerged as a biologically important post-translational modification. However, detection and quantitation of methionine sulfoxide in proteins is difficult. Our aim is to develop a method for specifically derivatizing methionine sulfoxide residues. We report a Pummerer rearrangement of methionine sulfoxide treated sequentially with trimethylsilyl chloride and then 2-mercaptoimidazole or pyridine-2-thiol to produce a dithioacetal product. This derivative is stable to standard mass spectrometry conditions, and its formation identified oxidized methionine residues. The scope and requirements of dithioacetal formation are reported for methionine sulfoxide and model substrates. The reaction intermediates have been investigated by computational techniques and by 13 C NMR spectroscopy. These provide evidence for an α-chlorinated intermediate. The derivatization allows for detection and quantitation of methionine sulfoxide in proteins by mass spectrometry and potentially by immunochemical methods.
Collapse
Affiliation(s)
- Carolyn C. Woodroofe
- Imaging Probe Development CenterNational Heart, Lung and Blood InstituteNational Institutes of Health9800 Medical Center DriveRockvilleMD20850USA
| | - Joseph Ivanic
- Advanced Biomedical Computational Sciences GroupFrederick National Laboratory for Cancer Research operated byLeidos Biomedical Research, Inc.1011 Beasley DriveFrederickMD21701USA
| | - Sarah Monti
- Laboratory of Biochemistry, National Heart, Lung, and Blood InstituteNational Institutes of Health50 South DriveBethesdaMD20892USA
| | - Rodney L. Levine
- Laboratory of Biochemistry, National Heart, Lung, and Blood InstituteNational Institutes of Health50 South DriveBethesdaMD20892USA
| | - Rolf E. Swenson
- Imaging Probe Development CenterNational Heart, Lung and Blood InstituteNational Institutes of Health9800 Medical Center DriveRockvilleMD20850USA
| |
Collapse
|
137
|
|
138
|
Secchi C, Orecchioni M, Carta M, Galimi F, Turrini F, Pantaleo A. Signaling Response to Transient Redox Stress in Human Isolated T Cells: Molecular Sensor Role of Syk Kinase and Functional Involvement of IL2 Receptor and L-Selectine. SENSORS 2020; 20:s20020466. [PMID: 31947584 PMCID: PMC7013990 DOI: 10.3390/s20020466] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/04/2020] [Accepted: 01/08/2020] [Indexed: 01/04/2023]
Abstract
Reactive oxygen species (ROS) are central effectors of inflammation and play a key role in cell signaling. Previous reports have described an association between oxidative events and the modulation of innate immunity. However, the role of redox signaling in adaptive immunity is still not well understood. This work is based on a novel investigation of diamide, a specific oxidant of sulfhydryl groups, and it is the first performed in purified T cell tyrosine phosphorylation signaling. Our data show that ex vivo T cells respond to –SH group oxidation with a distinctive tyrosine phosphorylation response and that these events elicit specific cellular responses. The expression of two essential T-cell receptors, CD25 and CD62L, and T-cell cytokine release is also affected in a specific way. Experiments with Syk inhibitors indicate a major contribution of this kinase in these phenomena. This pilot work confirms the presence of crosstalk between oxidation of cysteine residues and tyrosine phosphorylation changes, resulting in a series of functional events in freshly isolated T cells. Our experiments show a novel role of Syk inhibitors in applying their anti-inflammatory action through the inhibition of a ROS-generated reaction.
Collapse
Affiliation(s)
- Christian Secchi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
- Department of Biomedical Sciences, University of Sassari, I-07100 Sassari, Italy; (M.C.); (F.G.)
- Istituto Nazionale Biostrutture e Biosistemi, University of Sassari, I-07100 Sassari, Italy
- Correspondence: (C.S.); (A.P.); Tel./Fax: +39-079-228-651 (A.P.)
| | - Marco Orecchioni
- La Jolla Institute of Immunology, La Jolla, CA 92093, USA;
- Department of Chemistry and Pharmacy, University of Sassari, I-07100 Sassari, Italy
| | - Marissa Carta
- Department of Biomedical Sciences, University of Sassari, I-07100 Sassari, Italy; (M.C.); (F.G.)
| | - Francesco Galimi
- Department of Biomedical Sciences, University of Sassari, I-07100 Sassari, Italy; (M.C.); (F.G.)
- Istituto Nazionale Biostrutture e Biosistemi, University of Sassari, I-07100 Sassari, Italy
| | | | - Antonella Pantaleo
- Department of Biomedical Sciences, University of Sassari, I-07100 Sassari, Italy; (M.C.); (F.G.)
- Correspondence: (C.S.); (A.P.); Tel./Fax: +39-079-228-651 (A.P.)
| |
Collapse
|
139
|
Ramírez-Moreno IG, Ibarra-Sánchez A, Castillo-Arellano JI, Blank U, González-Espinosa C. Mast Cells Localize in Hypoxic Zones of Tumors and Secrete CCL-2 under Hypoxia through Activation of L-Type Calcium Channels. THE JOURNAL OF IMMUNOLOGY 2020; 204:1056-1068. [PMID: 31900336 DOI: 10.4049/jimmunol.1801430] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/05/2019] [Indexed: 01/19/2023]
Abstract
Hypoxia is a condition that together with low pH, high amounts of reactive oxygen species (ROS), and increased adenosine levels characterize tumor microenvironment. Mast cells (MCs) are part of tumor microenvironment, but the effect of hypoxia on the production of MC-derived cytokines has not been fully described. Using the hypoxia marker pimonidazole in vivo, we found that MCs were largely located in the low-oxygen areas within B16-F1 mice melanoma tumors. In vitro, hypoxia promoted ROS production, a ROS-dependent increase of intracellular calcium, and the production of MCP 1 (CCL-2) in murine bone marrow-derived MCs. Hypoxia-induced CCL-2 production was sensitive to the antioxidant trolox and to nifedipine, a blocker of L-type voltage-dependent Ca2+ channels (LVDCCs). Simultaneously with CCL-2 production, hypoxia caused the ROS-dependent glutathionylation and membrane translocation of the α1c subunit of Cav1.2 LVDCCs. Relationship between ROS production, calcium rise, and CCL-2 synthesis was also observed when cells were treated with H2O2 In vivo, high CCL-2 production was detected on hypoxic zones of melanoma tumors (where tryptase-positive MCs were also found). Pimonidazole and CCL-2 positive staining diminished when B16-F1 cell-inoculated animals were treated with trolox, nifedipine, or the adenosine receptor 2A antagonist KW6002. Our results show that MCs are located preferentially in hypoxic zones of melanoma tumors, hypoxia-induced CCL-2 production in MCs requires calcium rise mediated by glutathionylation and membrane translocation of LVDCCs, and this mechanism of CCL-2 synthesis seems to operate in other cells inside melanoma tumors, with the participation of the adenosine receptor 2A.
Collapse
Affiliation(s)
- Itzel G Ramírez-Moreno
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Tlalpan, 14330 Mexico City, Mexico
| | - Alfredo Ibarra-Sánchez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Tlalpan, 14330 Mexico City, Mexico
| | - Jorge Ivan Castillo-Arellano
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510 Mexico City, Mexico; and
| | - Ulrich Blank
- Inserm U1149, CNRS ERL 8252, Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site X. bichat, Laboratorie d'excellence INFLAMEX, 75018 Paris, France
| | - Claudia González-Espinosa
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Tlalpan, 14330 Mexico City, Mexico;
| |
Collapse
|
140
|
Pervaiz S, Bellot GL, Lemoine A, Brenner C. Redox signaling in the pathogenesis of human disease and the regulatory role of autophagy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 352:189-214. [DOI: 10.1016/bs.ircmb.2020.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
141
|
Bonzon-Kulichenko E, Camafeita E, López JA, Gómez-Serrano M, Jorge I, Calvo E, Núñez E, Trevisan-Herraz M, Bagwan N, Bárcena JA, Peral B, Vázquez J. Improved integrative analysis of the thiol redox proteome using filter-aided sample preparation. J Proteomics 2019; 214:103624. [PMID: 31874222 DOI: 10.1016/j.jprot.2019.103624] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 02/08/2023]
Abstract
Changes in the oxidation state of protein Cys residues are involved in cell signalling and play a key role in a variety of pathophysiological states. We had previously developed GELSILOX, an in-gel method that enables the large-scale, parallel analysis of dynamic alterations to the redox state of Cys sites and protein abundance changes. Here we present FASILOX, a further development of the GELSILOX approach featuring: i) significantly increased peptide recovery, ii) enhanced sensitivity for the detection of Cys oxidative alterations, and iii) streamlined workflow that results in shortened assay duration. In mitochondria isolated from the adipose tissue of obese, diabetic patients, FASILOX revealed a sexually dimorphic trait of Cys oxidation involving mainly mitochondrial oxidative phosphorylation complexes. These results provide the first evidence for a decreased efficiency in the antioxidant response of men as compared to women.
Collapse
Affiliation(s)
- Elena Bonzon-Kulichenko
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Emilio Camafeita
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - Juan Antonio López
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - María Gómez-Serrano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM), Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Inmaculada Jorge
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Enrique Calvo
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Estefanía Núñez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Marco Trevisan-Herraz
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Navratan Bagwan
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José Antonio Bárcena
- Dept. Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Belén Peral
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM), Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
142
|
Hawkins CL, Davies MJ. Detection, identification, and quantification of oxidative protein modifications. J Biol Chem 2019; 294:19683-19708. [PMID: 31672919 PMCID: PMC6926449 DOI: 10.1074/jbc.rev119.006217] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Exposure of biological molecules to oxidants is inevitable and therefore commonplace. Oxidative stress in cells arises from both external agents and endogenous processes that generate reactive species, either purposely (e.g. during pathogen killing or enzymatic reactions) or accidentally (e.g. exposure to radiation, pollutants, drugs, or chemicals). As proteins are highly abundant and react rapidly with many oxidants, they are highly susceptible to, and major targets of, oxidative damage. This can result in changes to protein structure, function, and turnover and to loss or (occasional) gain of activity. Accumulation of oxidatively-modified proteins, due to either increased generation or decreased removal, has been associated with both aging and multiple diseases. Different oxidants generate a broad, and sometimes characteristic, spectrum of post-translational modifications. The kinetics (rates) of damage formation also vary dramatically. There is a pressing need for reliable and robust methods that can detect, identify, and quantify the products formed on amino acids, peptides, and proteins, especially in complex systems. This review summarizes several advances in our understanding of this complex chemistry and highlights methods that are available to detect oxidative modifications-at the amino acid, peptide, or protein level-and their nature, quantity, and position within a peptide sequence. Although considerable progress has been made in the development and application of new techniques, it is clear that further development is required to fully assess the relative importance of protein oxidation and to determine whether an oxidation is a cause, or merely a consequence, of injurious processes.
Collapse
Affiliation(s)
- Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
143
|
Lim JKM, Leprivier G. The impact of oncogenic RAS on redox balance and implications for cancer development. Cell Death Dis 2019; 10:955. [PMID: 31852884 PMCID: PMC6920345 DOI: 10.1038/s41419-019-2192-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
Abstract
The RAS family of proto-oncogenes comprises HRAS, KRAS, and NRAS, which are among the most mutated genes in human cancers. The RAS family genes encode small GTPases that coordinate key signaling pathways in response to growth factors. Mutations in RAS result in a constitutively active form of the protein that supports cellular transformation and tumorigenesis. The mechanisms of oncogenic RAS-mediated transformation encompass uncontrolled proliferation and inhibition of cell death through overactivation of the RAF-MEK-ERK and the PI3K-AKT pathways, respectively. In addition, the control of redox balance by RAS has also been proposed to play a role in its oncogenic properties. However, the exact role of redox balance in mediating mutant RAS transformation is still under debate. Here, we present, on one hand, the involvement of pro-oxidant components in oncogenic RAS transformation, such as NADPH oxidases and mitochondrial reactive oxygen species, and how these promote transformation. On the other hand, we describe the contribution of antioxidant components to mutant RAS transformation, including Nrf2, glutathione biosynthesis and xCT, as well as the mechanisms by which antioxidant programs drive transformation. Finally, we aim to reconcile the seemingly opposite effects of oncogenic RAS on redox balance and discuss a model for the complementary role of both pro-oxidant and antioxidant pathways in mutant RAS-driven tumor progression.
Collapse
Affiliation(s)
- Jonathan K M Lim
- Institute for Neuropathology, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Gabriel Leprivier
- Institute for Neuropathology, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
144
|
Gellert M, Hossain MF, Berens FJF, Bruhn LW, Urbainsky C, Liebscher V, Lillig CH. Substrate specificity of thioredoxins and glutaredoxins - towards a functional classification. Heliyon 2019; 5:e02943. [PMID: 31890941 PMCID: PMC6928294 DOI: 10.1016/j.heliyon.2019.e02943] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
The spatio-temporal reduction and oxidation of protein thiols is an essential mechanism in signal transduction in all kingdoms of life. Thioredoxin (Trx) family proteins efficiently catalyze thiol-disulfide exchange reactions and the proteins are widely recognized for their importance in the operation of thiol switches. Trx family proteins have a broad and at the same time very distinct substrate specificity – a prerequisite for redox switching. Despite of multiple efforts, the true nature for this specificity is still under debate. Here, we comprehensively compare the classification/clustering of various redoxins from all domains of life based on their similarity in amino acid sequence, tertiary structure, and their electrostatic properties. We correlate these similarities to the existence of common interaction partners, identified in various previous studies and suggested by proteomic screenings. These analyses confirm that primary and tertiary structure similarity, and thereby all common classification systems, do not correlate to the target specificity of the proteins as thiol-disulfide oxidoreductases. Instead, a number of examples clearly demonstrate the importance of electrostatic similarity for their target specificity, independent of their belonging to the Trx or glutaredoxin subfamilies.
Collapse
Affiliation(s)
- Manuela Gellert
- Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Germany
| | - Md Faruq Hossain
- Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Germany
| | - Felix Jacob Ferdinand Berens
- Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Germany.,Institute for Mathematics and Informatics, University of Greifswald, Germany
| | - Lukas Willy Bruhn
- Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Germany.,Institute for Mathematics and Informatics, University of Greifswald, Germany
| | - Claudia Urbainsky
- Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Germany
| | - Volkmar Liebscher
- Institute for Mathematics and Informatics, University of Greifswald, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Germany
| |
Collapse
|
145
|
Whitehall JC, Greaves LC. Aberrant mitochondrial function in ageing and cancer. Biogerontology 2019; 21:445-459. [PMID: 31802313 PMCID: PMC7347693 DOI: 10.1007/s10522-019-09853-y] [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: 09/17/2019] [Accepted: 11/23/2019] [Indexed: 12/12/2022]
Abstract
Alterations in mitochondrial metabolism have been described as one of the major hallmarks of both ageing cells and cancer. Age is the biggest risk factor for the development of a significant number of cancer types and this therefore raises the question of whether there is a link between age-related mitochondrial dysfunction and the advantageous changes in mitochondrial metabolism prevalent in cancer cells. A common underlying feature of both ageing and cancer cells is the presence of somatic mutations of the mitochondrial genome (mtDNA) which we postulate may drive compensatory alterations in mitochondrial metabolism that are advantageous for tumour growth. In this review, we discuss basic mitochondrial functions, mechanisms of mtDNA mutagenesis and their metabolic consequences, and review the evidence for and against a role for mtDNA mutations in cancer development.
Collapse
Affiliation(s)
- Julia C Whitehall
- The Medical School, Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Laura C Greaves
- The Medical School, Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
146
|
Yura Y, Chong BSH, Johnson RD, Watanabe Y, Tsukahara Y, Ferran B, Murdoch CE, Behring JB, McComb ME, Costello CE, Janssen-Heininger YMW, Cohen RA, Bachschmid MM, Matsui R. Endothelial cell-specific redox gene modulation inhibits angiogenesis but promotes B16F0 tumor growth in mice. FASEB J 2019; 33:14147-14158. [PMID: 31647879 PMCID: PMC6894059 DOI: 10.1096/fj.201900786r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/17/2019] [Indexed: 01/26/2023]
Abstract
Glutaredoxin-1 (Glrx) is a small cytosolic enzyme that removes S-glutathionylation, glutathione adducts of protein cysteine residues, thus modulating redox signaling and gene transcription. Although Glrx up-regulation prevented endothelial cell (EC) migration and global Glrx transgenic mice had impaired ischemic vascularization, the effects of cell-specific Glrx overexpression remained unknown. Here, we examined the role of EC-specific Glrx up-regulation in distinct models of angiogenesis; namely, hind limb ischemia and tumor angiogenesis. EC-specific Glrx transgenic (EC-Glrx TG) overexpression in mice significantly impaired EC migration in Matrigel implants and hind limb revascularization after femoral artery ligation. Additionally, ECs migrated less into subcutaneously implanted B16F0 melanoma tumors as assessed by decreased staining of EC markers. Despite reduced angiogenesis, EC-Glrx TG mice unexpectedly developed larger tumors compared with control mice. EC-Glrx TG mice showed higher levels of VEGF-A in the tumors, indicating hypoxia, which may stimulate tumor cells to form vascular channels without EC, referred to as vasculogenic mimicry. These data suggest that impaired ischemic vascularization does not necessarily associate with suppression of tumor growth, and that antiangiogenic therapies may be ineffective for melanoma tumors because of their ability to implement vasculogenic mimicry during hypoxia.-Yura, Y., Chong, B. S. H., Johnson, R. D., Watanabe, Y., Tsukahara, Y., Ferran, B., Murdoch, C. E., Behring, J. B., McComb, M. E., Costello, C. E., Janssen-Heininger, Y. M. W., Cohen, R. A., Bachschmid, M. M., Matsui, R. Endothelial cell-specific redox gene modulation inhibits angiogenesis but promotes B16F0 tumor growth in mice.
Collapse
Affiliation(s)
- Yoshimitsu Yura
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Brian S. H. Chong
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ryan D. Johnson
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yosuke Watanabe
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yuko Tsukahara
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Beatriz Ferran
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Colin E. Murdoch
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jessica B. Behring
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Mark E. McComb
- Cardiovascular Proteomics Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Catherine E. Costello
- Cardiovascular Proteomics Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Richard A. Cohen
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Markus M. Bachschmid
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Reiko Matsui
- Department of Medicine, Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
147
|
Wilhelm EA, Soares PS, Reis AS, Barth A, Freitas BG, Motta KP, Lemos BB, Vogt AG, da Fonseca CAR, Araujo DR, Barcellos AM, Perin G, Luchese C. Se - [(2,2-Dimethyl-1,3-dioxolan-4-yl) methyl] 4-chlorobenzoselenolate reduces the nociceptive and edematogenic response by chemical noxious stimuli in mice: Implications of multi-target actions. Pharmacol Rep 2019; 71:1201-1209. [PMID: 31669884 DOI: 10.1016/j.pharep.2019.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/03/2019] [Accepted: 07/18/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND The present study evaluated the antioxidant, antinociceptive and anti-edematogenic effects of Se-[(2,2-dimethyl-1,3-dioxolan-4-yl) methyl] 4-chlorobenzoselenolate (Se-DMC). METHODS In vitro experiments were carried out to evaluate Se-DMC antioxidant action. Thiobarbituric acid reactive species levels, 2,2'-diphenyl-1-picrylhydrazyl and 2,2'-azino-bis(3-thylbenzthiazoline-6-sulfonic acid) radicals scavenging and glutathione S-transferase-like activity were determined. Male Swiss mice were orally pretreated with Se-DMC (1, 10 and 50 mg/kg), meloxicam (50 mg/kg) or vehicle 30 min prior to acetic acid or glutamate test. To extend our knowledge of the pharmacological properties of this compound, it was tested in an inflammatory model through ear edema induced by croton oil. The contribution of glutamatergic and serotonergic systems was also investigated. RESULTS In vitro experiments revealed that Se-DMC exerts antioxidant activity. Nociception induced by glutamate or acetic acid was reduced by Se-DMC or meloxicam. Se-DMC diminished the paw edema formation induced by glutamate, while meloxicam did not show any effect. Se-DMC and meloxicam decreased the ear edema formation and protected against the increase in myeloperoxidase activity in mice ear induced by croton oil. The pretreatment of animals with MK-801 did not alter antinociception caused by Se-DMC in the glutamate test. The antinociceptive effect exerted by Se-DMC in the acetic acid test was reverted by the pretreatment of mice with different serotonergic antagonists (WAY100635, ketanserin and pindolol). CONCLUSIONS Data presented here showed that the modulation of serotonergic and glutamatergic systems and the anti-inflammatory and antioxidant actions could contribute to the antinociceptive and anti-edematogenic effects of Se-DMC and it supported the therapeutic potential of this compound.
Collapse
Affiliation(s)
- Ethel A Wilhelm
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil.
| | - Paola S Soares
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Angélica S Reis
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Anelise Barth
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Barbara G Freitas
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Ketlyn P Motta
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Briana B Lemos
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Ane G Vogt
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Caren A R da Fonseca
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil
| | - Daniela R Araujo
- Laboratório de Síntese Orgânica Limpa, CCQFA - Universidade Federal de Pelotas - UFPel, Pelotas, RS, Brazil
| | - Angelita M Barcellos
- Laboratório de Síntese Orgânica Limpa, CCQFA - Universidade Federal de Pelotas - UFPel, Pelotas, RS, Brazil
| | - Gelson Perin
- Laboratório de Síntese Orgânica Limpa, CCQFA - Universidade Federal de Pelotas - UFPel, Pelotas, RS, Brazil
| | - Cristiane Luchese
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil.
| |
Collapse
|
148
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
149
|
Fujitani N, Yoneda A, Takahashi M, Takasawa A, Aoyama T, Miyazaki T. Silencing of Glutathione S-Transferase Pi Inhibits Cancer Cell Growth via Oxidative Stress Induced by Mitochondria Dysfunction. Sci Rep 2019; 9:14764. [PMID: 31611630 PMCID: PMC6791853 DOI: 10.1038/s41598-019-51462-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
Antitumor drug development based on the concept of intervening in the antioxidant system of cancer cells has been gaining increased interest. In this study, we propose a promising strategy for cancer treatment using modulation of oxidative stress by suppression of glutathione S-transferases (GSTs), a typical antioxidant enzyme. siRNA which can be applied to the development of nucleic acid drugs, enabling them to eliminate unwanted side effects, increase specificity, and avoid the problem of drug resistance, was employed for GSTP-silencing at the transcriptional level. The silencing of the pi class of GST (GSTP) that displayed the most characteristic expression profile in 13 kinds of cancer cell lines has shown significant impairment in the growth of cancer cells due to oxidative stress caused by excess ROS accumulation. Comparative proteomics between normal cells and GSTP-silenced pancreatic cancer cell PANC-1 suggested that GSTP-silencing facilitated the mitochondrial dysfunction. These findings show promise for the development of strategies toward cancer therapy based on the mechanism that allows genetic silencing of GSTP to promote oxidative stress through mitochondria dysfunction.
Collapse
Affiliation(s)
- Naoki Fujitani
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Sapporo, 060-8556, Japan
| | - Akihiro Yoneda
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for the Promotion of Business-Regional Collaboration, Hokkaido University, N21W11, Sapporo, 001-0021, Japan.
| | - Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Sapporo, 060-8556, Japan
| | - Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, S1W17, Sapporo, 060-8556, Japan
| | - Tomoyuki Aoyama
- Department of Pathology, Sapporo Medical University School of Medicine, S1W17, Sapporo, 060-8556, Japan.,Department of Surgical Pathology, Sapporo Medical University School of Medicine, S1W16, Sapporo, 060-8556, Japan
| | - Tadaaki Miyazaki
- Department of Probiotics Immunology, Institute for Genetic Medicine, Hokkaido University, N15W7, Sapporo, 001-0015, Japan
| |
Collapse
|
150
|
Sandalio LM, Gotor C, Romero LC, Romero-Puertas MC. Multilevel Regulation of Peroxisomal Proteome by Post-Translational Modifications. Int J Mol Sci 2019; 20:E4881. [PMID: 31581473 PMCID: PMC6801620 DOI: 10.3390/ijms20194881] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 01/10/2023] Open
Abstract
Peroxisomes, which are ubiquitous organelles in all eukaryotes, are highly dynamic organelles that are essential for development and stress responses. Plant peroxisomes are involved in major metabolic pathways, such as fatty acid β-oxidation, photorespiration, ureide and polyamine metabolism, in the biosynthesis of jasmonic, indolacetic, and salicylic acid hormones, as well as in signaling molecules such as reactive oxygen and nitrogen species (ROS/RNS). Peroxisomes are involved in the perception of environmental changes, which is a complex process involving the regulation of gene expression and protein functionality by protein post-translational modifications (PTMs). Although there has been a growing interest in individual PTMs in peroxisomes over the last ten years, their role and cross-talk in the whole peroxisomal proteome remain unclear. This review provides up-to-date information on the function and crosstalk of the main peroxisomal PTMs. Analysis of whole peroxisomal proteomes shows that a very large number of peroxisomal proteins are targeted by multiple PTMs, which affect redox balance, photorespiration, the glyoxylate cycle, and lipid metabolism. This multilevel PTM regulation could boost the plasticity of peroxisomes and their capacity to regulate metabolism in response to environmental changes.
Collapse
Affiliation(s)
- Luisa M Sandalio
- Department of Biochemistry and Cellular and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| | - Cecilia Gotor
- Institute of Plant Biochemistry and Photosynthesis, CSIC and the University of Seville, 41092 Seville, Spain.
| | - Luis C Romero
- Institute of Plant Biochemistry and Photosynthesis, CSIC and the University of Seville, 41092 Seville, Spain.
| | - Maria C Romero-Puertas
- Department of Biochemistry and Cellular and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| |
Collapse
|