151
|
Targeting Reactive Oxygen Species in Cancer via Chinese Herbal Medicine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9240426. [PMID: 31583051 PMCID: PMC6754955 DOI: 10.1155/2019/9240426] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023]
Abstract
Recently, reactive oxygen species (ROS), a class of highly bioactive molecules, have been extensively studied in cancers. Cancer cells typically exhibit higher levels of basal ROS than normal cells, primarily due to their increased metabolism, oncogene activation, and mitochondrial dysfunction. This moderate increase in ROS levels facilitates cancer initiation, development, and progression; however, excessive ROS concentrations can lead to various types of cell death. Therefore, therapeutic strategies that either increase intracellular ROS to toxic levels or, conversely, decrease the levels of ROS may be effective in treating cancers via ROS regulation. Chinese herbal medicine (CHM) is a major type of natural medicine and has greatly contributed to human health. CHMs have been increasingly used for adjuvant clinical treatment of tumors. Although their mechanism of action is unclear, CHMs can execute a variety of anticancer effects by regulating intracellular ROS. In this review, we summarize the dual roles of ROS in cancers, present a comprehensive analysis of and update the role of CHM—especially its active compounds and ingredients—in the prevention and treatment of cancers via ROS regulation and emphasize precautions and strategies for the use of CHM in future research and clinical trials.
Collapse
|
152
|
Michán C, Chicano-Gálvez E, Fuentes-Almagro CA, Alhama J. Redox and global interconnected proteome changes in mice exposed to complex environmental hazards surrounding Doñana National Park. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 252:427-439. [PMID: 31158671 DOI: 10.1016/j.envpol.2019.05.085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/30/2019] [Accepted: 05/16/2019] [Indexed: 06/09/2023]
Abstract
Natural environments are receiving an increasing number of contaminants. Therefore, the evaluation and identification of early responses to pollution in these complex habitats is an urgent and challenging task. Doñana National Park (DNP, SW Spain) has been widely used as a model area for environmental studies because, despite its strictly protected core, it is surrounded by numerous threat sources from agricultural, mining and industrial activities. Since many pollutants often induce oxidative stress, redox proteomics was used to detect redox-based variations within the proteome of Mus spretus mice captured in DNP and the surrounding areas. Functional analysis showed that most differentially oxidized proteins are involved in the maintenance of homeostasis, by eliciting mechanisms to respond to toxic substances and oxidative stress, such as antioxidant and biotransformation processes, immune and inflammatory responses, and blood coagulation. Furthermore, changes in the overall protein abundance were also analysed by label-free quantitative proteomics. The upregulation of phase I and II biotransformation enzymes in mice from Lucio del Palacio may be an alert for organic pollution in the area located at the heart of DNP. Metabolic processes involved in protein turnover (proteolysis, amino acid catabolism, new protein biosynthesis and folding) were activated in response to oxidative damage to these biomolecules. Consequently, aerobic respiratory metabolism increased to address the greater ATP demands. Alterations of cholesterol metabolism that could cause hepatic steatosis were also detected. The proteomic detection of globally altered metabolic and physiological processes offers a complete view of the main biological changes caused by environmental pollution in complex habitats.
Collapse
Affiliation(s)
- Carmen Michán
- Department of Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain
| | | | | | - José Alhama
- Department of Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain.
| |
Collapse
|
153
|
What sustains the multidrug resistance phenotype beyond ABC efflux transporters? Looking beyond the tip of the iceberg. Drug Resist Updat 2019; 46:100643. [PMID: 31493711 DOI: 10.1016/j.drup.2019.100643] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022]
Abstract
Identification of multidrug (MDR) efflux transporters that belong to the ATP-Binding Cassette (ABC) superfamily, represented an important breakthrough for understanding cancer multidrug resistance (MDR) and its possible overcoming. However, recent data indicate that drug resistant cells have a complex intracellular physiology that involves constant changes in energetic and oxidative-reductive metabolic pathways, as well as in the molecular circuitries connecting mitochondria, endoplasmic reticulum (ER) and lysosomes. The aim of this review is to discuss the key molecular mechanisms of cellular reprogramming that induce and maintain MDR, beyond the presence of MDR efflux transporters. We specifically highlight how cancer cells characterized by high metabolic plasticity - i.e. cells able to shift the energy metabolism between glycolysis and oxidative phosphorylation, to survive both the normoxic and hypoxic conditions, to modify the cytosolic and mitochondrial oxidative-reductive metabolism, are more prone to adapt to exogenous stressors such as anti-cancer drugs and acquire a MDR phenotype. Similarly, we discuss how changes in mitochondria dynamics and mitophagy rates, changes in proteome stability ensuring non-oncogenic proteostatic mechanisms, changes in ubiquitin/proteasome- and autophagy/lysosome-related pathways, promote the cellular survival under stress conditions, along with the acquisition or maintenance of MDR. After dissecting the complex intracellular crosstalk that takes place during the development of MDR, we suggest that mapping the specific adaptation pathways underlying cell survival in response to stress and targeting these pathways with potent pharmacologic agents may be a new approach to enhance therapeutic efficacy against MDR tumors.
Collapse
|
154
|
Ferritinophagic Flux Activation in CT26 Cells Contributed to EMT Inhibition Induced by a Novel Iron Chelator, DpdtpA. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8753413. [PMID: 31320987 PMCID: PMC6610730 DOI: 10.1155/2019/8753413] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/07/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022]
Abstract
Epithelial-mesenchymal transition (EMT) contributes to metastasis and drug resistance; inhibition of EMT may attenuate metastasis and drug resistance. It has been demonstrated that ferritinophagy involves the process of many diseases; however, the relationship between EMT and ferritinophagy was not fully established. Some iron chelators show the ability to inhibit EMT, but whether ferritinophagy plays a role in EMT is largely unknown. To this end, we investigated the effect of a novel iron chelator, DpdtpA (2,2 ′-di-pyridylketone dithiocarbamate propionic acid), on EMT in the CT26 cell line. The DpdtpA displayed excellent antitumor (IC50 = 1.5 ± 0.2 μM), leading to ROS production and apoptosis occurrence. Moreover, the ROS production correlated with ferritin degradation. The upregulation of LC3-II and NCOA4 from immunofluorescence and Western blotting analysis revealed that the occurrence of ferritinophagy contributed to ROS production. Furthermore, DpdtpA could induce an alteration both in morphology and in epithelial-mesenchymal markers, displaying significant EMT inhibition. The correlation analysis revealed that DpdtpA-induced ferritinophagy contributed to the EMT inhibition, implying that NCOA4 involved EMT process, which was firstly reported. To reinforce this concept, the ferritinophagic flux (NCOA4/ferritin) in either treated by TGF-β1 or combined with DpdtpA was determined. The results indicated that activating ferritinophagic flux would enhance ROS production which accordingly suppressed EMT or implementing the EMT suppression seemed to be through “fighting fire with fire” strategy. Taken together, our data demonstrated that ferritinophagic flux was a dominating driving force in EMT proceeding, and the new finding definitely will enrich our knowledge of ferritinophagy in EMT process.
Collapse
|
155
|
Rajendran S, Shen X, Glawe J, Kolluru GK, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Growth and Remodeling. Compr Physiol 2019; 9:1213-1247. [PMID: 31187898 DOI: 10.1002/cphy.c180026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic vascular remodeling occurs in response to stenosis or arterial occlusion leading to a change in blood flow and tissue perfusion. Altered blood flow elicits a cascade of molecular and cellular physiological responses leading to vascular remodeling of the macro- and micro-circulation. Although cellular mechanisms of vascular remodeling such as arteriogenesis and angiogenesis have been studied, therapeutic approaches in these areas have had limited success due to the complexity and heterogeneous constellation of molecular signaling events regulating these processes. Understanding central molecular players of vascular remodeling should lead to a deeper understanding of this response and aid in the development of novel therapeutic strategies. Hydrogen sulfide (H2 S) and nitric oxide (NO) are gaseous signaling molecules that are critically involved in regulating fundamental biochemical and molecular responses necessary for vascular growth and remodeling. This review examines how NO and H2 S regulate pathophysiological mechanisms of angiogenesis and arteriogenesis, along with important chemical and experimental considerations revealed thus far. The importance of NO and H2 S bioavailability, their synthesis enzymes and cofactors, and genetic variations associated with cardiovascular risk factors suggest that they serve as pivotal regulators of vascular remodeling responses. © 2019 American Physiological Society. Compr Physiol 9:1213-1247, 2019.
Collapse
Affiliation(s)
| | - Xinggui Shen
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - John Glawe
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Gopi K Kolluru
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Christopher G Kevil
- Departments of Pathology, LSU Health Sciences Center, Shreveport.,Departments of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport.,Departments of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport
| |
Collapse
|
156
|
Bacchetti T, Ferretti G, Sahebkar A. The role of paraoxonase in cancer. Semin Cancer Biol 2019; 56:72-86. [PMID: 29170064 DOI: 10.1016/j.semcancer.2017.11.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/20/2017] [Accepted: 11/18/2017] [Indexed: 12/15/2022]
Abstract
The paraoxonase (PON) gene family includes three proteins, PON1, PON2 and PON3. PON1 and PON3 are both associated with high-density lipoprotein (HDL) particles and exert anti-oxidant and anti-inflammatory properties. PON2 and PON3 are intracellular enzymes which modulate mitochondrial superoxide anion production and endoplasmic reticulum (ER) stress-induced apoptosis. The pleiotropic roles exerted by PONs have been mainly investigated in cardiovascular and neurodegenerative diseases. In recent years, overexpression of PON2 and PON3 has been observed in cancer cells and it has been proposed that both enzymes could be involved in tumor survival and stress resistance. Moreover, a lower activity of serum PON1 has been reported in cancer patients. This review summarizes literature data on the role of PONs in human cancers and their potential role as a target for antitumor drugs.
Collapse
Affiliation(s)
- Tiziana Bacchetti
- Department of Life and Environmental Sciences (DiSVA), Polytechnic University of Marche, Ancona, Italy.
| | - Gianna Ferretti
- Department of Clinical Science and Odontostomatology, Polytechnic University of Marche, Ancona, Italy.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
157
|
Biomarkers of Oxidative Stress in Metabolic Syndrome and Associated Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8267234. [PMID: 31191805 PMCID: PMC6525823 DOI: 10.1155/2019/8267234] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 02/08/2019] [Accepted: 03/19/2019] [Indexed: 12/11/2022]
Abstract
Metabolic syndrome (MS) represents worldwide public health issue characterized by a set of cardiovascular risk factors including obesity, diabetes, dyslipidemia, hypertension, and impaired glucose tolerance. The link between the MS and the associated diseases is represented by oxidative stress (OS) and by the intracellular redox imbalance, both caused by the persistence of chronic inflammatory conditions that characterize MS. The increase in oxidizing species formation in MS has been accepted as a major underlying mechanism for mitochondrial dysfunction, accumulation of protein and lipid oxidation products, and impairment of the antioxidant systems. These oxidative modifications are recognized as relevant OS biomarkers potentially able to (i) clarify the role of reactive oxygen and nitrogen species in the etiology of the MS, (ii) contribute to the diagnosis/evaluation of the disease's severity, and (iii) evaluate the utility of possible therapeutic strategies based on natural antioxidants. The antioxidant therapies indeed could be able to (i) counteract systemic as well as mitochondrial-derived OS, (ii) enhance the endogenous antioxidant defenses, (iii) alleviate MS symptoms, and (iv) prevent the complications linked to MS-derived cardiovascular diseases. The focus of this review is to summarize the current knowledge about the role of OS in the development of metabolic alterations characterizing MS, with particular regard to the occurrence of OS-correlated biomarkers, as well as to the use of therapeutic strategies based on natural antioxidants.
Collapse
|
158
|
Fang Y, Vadlamudi M, Huang Y, Guo X. Lipid-Coated, pH-Sensitive Magnesium Phosphate Particles for Intracellular Protein Delivery. Pharm Res 2019; 36:81. [DOI: 10.1007/s11095-019-2607-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/10/2019] [Indexed: 12/13/2022]
|
159
|
Chakraborty P, Chatterjee S, Kesarwani P, Thyagarajan K, Iamsawat S, Dalheim A, Nguyen H, Selvam SP, Nasarre P, Scurti G, Hardiman G, Maulik N, Ball L, Gangaraju V, Rubinstein MP, Klauber-DeMore N, Hill EG, Ogretmen B, Yu XZ, Nishimura MI, Mehrotra S. Thioredoxin-1 improves the immunometabolic phenotype of antitumor T cells. J Biol Chem 2019; 294:9198-9212. [PMID: 30971427 DOI: 10.1074/jbc.ra118.006753] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/25/2019] [Indexed: 12/16/2022] Open
Abstract
Adoptive transfer of tumor epitope-reactive T cells has emerged as a promising strategy to control tumor growth. However, chronically-stimulated T cells expanded for adoptive cell transfer are susceptible to cell death in an oxidative tumor microenvironment. Because oxidation of cell-surface thiols also alters protein functionality, we hypothesized that increasing the levels of thioredoxin (Trx), an antioxidant molecule facilitating reduction of proteins through cysteine thiol-disulfide exchange, in T cells will promote their sustained antitumor function. Using pre-melanosome protein (Pmel)-Trx1 transgenic mouse-derived splenic T cells, flow cytometry, and gene expression analysis, we observed here that higher Trx expression inversely correlated with reactive oxygen species and susceptibility to T-cell receptor restimulation or oxidation-mediated cell death. These Trx1-overexpressing T cells exhibited a cluster of differentiation 62Lhi (CD62Lhi) central memory-like phenotype with reduced glucose uptake (2-NBDGlo) and decreased effector function (interferon γlo). Furthermore, culturing tumor-reactive T cells in the presence of recombinant Trx increased the dependence of T cells on mitochondrial metabolism and improved tumor control. We conclude that strategies for increasing the antioxidant capacity of antitumor T cells modulate their immunometabolic phenotype leading to improved immunotherapeutic control of established tumors.
Collapse
Affiliation(s)
| | | | | | | | | | - Annika Dalheim
- the Department of Surgery, Loyola University, Maywood, Illinois 60153, and
| | | | | | | | - Gina Scurti
- the Department of Surgery, Loyola University, Maywood, Illinois 60153, and
| | | | - Nilanjana Maulik
- the Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut 06030
| | | | | | | | | | - Elizabeth G Hill
- Public Health, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | | | | | | | | |
Collapse
|
160
|
Antonucci S, Mulvey JF, Burger N, Di Sante M, Hall AR, Hinchy EC, Caldwell ST, Gruszczyk AV, Deshwal S, Hartley RC, Kaludercic N, Murphy MP, Di Lisa F, Krieg T. Selective mitochondrial superoxide generation in vivo is cardioprotective through hormesis. Free Radic Biol Med 2019; 134:678-687. [PMID: 30731114 PMCID: PMC6607027 DOI: 10.1016/j.freeradbiomed.2019.01.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 01/14/2023]
Abstract
Reactive oxygen species (ROS) have an equivocal role in myocardial ischaemia reperfusion injury. Within the cardiomyocyte, mitochondria are both a major source and target of ROS. We evaluate the effects of a selective, dose-dependent increase in mitochondrial ROS levels on cardiac physiology using the mitochondria-targeted redox cycler MitoParaquat (MitoPQ). Low levels of ROS decrease the susceptibility of neonatal rat ventricular myocytes (NRVMs) to anoxia/reoxygenation injury and also cause profound protection in an in vivo mouse model of ischaemia/reperfusion. However higher doses of MitoPQ resulted in a progressive alteration of intracellular [Ca2+] homeostasis and mitochondrial function in vitro, leading to dysfunction and death at high doses. Our data show that a primary increase in mitochondrial ROS can alter cellular function, and support a hormetic model in which low levels of ROS are cardioprotective while higher levels of ROS are cardiotoxic.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis
- Disease Models, Animal
- Herbicides/pharmacology
- Hormesis
- Male
- Mice
- Mice, Inbred C57BL
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Paraquat/pharmacology
- Rats
- Rats, Wistar
- Superoxides/metabolism
Collapse
Affiliation(s)
- Salvatore Antonucci
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - John F Mulvey
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Nils Burger
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Moises Di Sante
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Andrew R Hall
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Elizabeth C Hinchy
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | | | - Anja V Gruszczyk
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Soni Deshwal
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | | | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), 35131, Padova, Italy
| | - Michael P Murphy
- Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy; Neuroscience Institute, National Research Council of Italy (CNR), 35131, Padova, Italy.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
161
|
Little AC, Hristova M, van Lith L, Schiffers C, Dustin CM, Habibovic A, Danyal K, Heppner DE, Lin MCJ, van der Velden J, Janssen-Heininger YM, van der Vliet A. Dysregulated Redox Regulation Contributes to Nuclear EGFR Localization and Pathogenicity in Lung Cancer. Sci Rep 2019; 9:4844. [PMID: 30890751 PMCID: PMC6425021 DOI: 10.1038/s41598-019-41395-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/01/2019] [Indexed: 12/14/2022] Open
Abstract
Lung cancers are frequently characterized by inappropriate activation of epidermal growth factor receptor (EGFR)-dependent signaling and epigenetic silencing of the NADPH oxidase (NOX) enzyme DUOX1, both potentially contributing to worse prognosis. Based on previous findings linking DUOX1 with redox-dependent EGFR activation, the present studies were designed to evaluate whether DUOX1 silencing in lung cancers may be responsible for altered EGFR regulation. In contrast to normal epithelial cells, EGF stimulation of lung cancer cell lines that lack DUOX1 promotes EGF-induced EGFR internalization and nuclear localization, associated with induction of EGFR-regulated genes and related tumorigenic outcomes. Each of these outcomes could be reversed by overexpression of DUOX1 or enhanced by shRNA-dependent DUOX1 silencing. EGF-induced nuclear EGFR localization in DUOX1-deficient lung cancer cells was associated with altered dynamics of cysteine oxidation of EGFR, and an overall reduction of EGFR cysteines. These various outcomes could also be attenuated by silencing of glutathione S-transferase P1 (GSTP1), a mediator of metabolic alterations and drug resistance in various cancers, and a regulator of cysteine oxidation. Collectively, our findings indicate DUOX1 deficiency in lung cancers promotes dysregulated EGFR signaling and enhanced GSTP1-mediated turnover of EGFR cysteine oxidation, which result in enhanced nuclear EGFR localization and tumorigenic properties.
Collapse
Affiliation(s)
- Andrew C Little
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA.,Rogel Cancer Center, Department of Internal Medicine Hematology-Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Loes van Lith
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Caspar Schiffers
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - David E Heppner
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Miao-Chong J Lin
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Yvonne M Janssen-Heininger
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
162
|
Viola HM, Hool LC. Impaired calcium handling and mitochondrial metabolic dysfunction as early markers of hypertrophic cardiomyopathy. Arch Biochem Biophys 2019; 665:166-174. [PMID: 30885674 DOI: 10.1016/j.abb.2019.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder, characterised by myocyte remodeling, disorganisation of sarcomeric proteins, impaired energy metabolism and altered cardiac contractility. Gene mutations encoding cardiac contractile proteins account for 60% of HCM aetiology. Current drug therapy including L-type calcium channel antagonists, are used to manage symptoms in patients with overt HCM, but no treatment exists that can reverse or prevent the cardiomyopathy. Design of effective drug therapy will require a clear understanding of the early pathophysiological mechanisms of the disease. Numerous studies have investigated specific aspects of HCM pathophysiology. This review brings these findings together, in order to develop a holistic understanding of the early pathophysiological mechanisms of the disease. We focus on gene mutations in cardiac myosin binding protein-C, β-cardiac myosin heavy chain, cardiac troponin I, and cardiac troponin T, that comprise the majority of all HCM sarcomeric gene mutations. We find that although some similarities exist, each mutation leads to mutation-specific alterations in calcium handling, myofilament calcium sensitivity and mitochondrial metabolic function. This may contribute to the observed clinical phenotypic variability in sarcomeric-related HCM. An understanding of early mutation-specific mechanisms of the disease may provide useful markers of disease progression, and inform therapeutic design.
Collapse
Affiliation(s)
- Helena M Viola
- School of Human Sciences (Physiology), The University of Western Australia, Crawley, WA, Australia
| | - Livia C Hool
- School of Human Sciences (Physiology), The University of Western Australia, Crawley, WA, Australia; Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
163
|
Abstract
Cancer has long been viewed as a disease of altered metabolism. Although it has long been recognized that the majority of cancer cells display increased dependence on glycolysis, the metabolism of "cancer stem-like cells" (CSCs) that drive tumor growth and metastasis is less well characterized. In this chapter, we review the current state of knowledge of CSC metabolism with an emphasis on the development of therapeutic strategies to exploit the metabolic vulnerabilities of these cells. We outline emerging evidence indicating distinct metabolic pathways active in the proliferative, epithelial- (E) and quiescent, mesenchymal-like (M) CSC states in triple negative breast cancer. These CSC states are characterized by their different redox potentials and divergent sensitivities to inhibitors of glycolysis and redox metabolism. We highlight the roles of two redox-regulated signaling pathways, hypoxia-inducible factor 1α and nuclear factor erythroid 2-related factor 2, in regulating CSC epithelial-mesenchymal plasticity during metabolic and/or oxidative stress, and discuss clinical strategies using combinations of pro-oxidant-based therapeutics simultaneously targeting E- and M-like CSCs. By specifically targeting CSCs of both states, these strategies have the potential to increase the therapeutic efficacy of traditional chemotherapy and radiation therapy.
Collapse
|
164
|
McArdle A, Pollock N, Staunton CA, Jackson MJ. Aberrant redox signalling and stress response in age-related muscle decline: Role in inter- and intra-cellular signalling. Free Radic Biol Med 2019; 132:50-57. [PMID: 30508577 PMCID: PMC6709668 DOI: 10.1016/j.freeradbiomed.2018.11.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/29/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022]
Abstract
Age-associated frailty is predominantly due to loss of muscle mass and function. The loss of muscle mass is also associated with a greater loss of muscle strength, suggesting that the remaining muscle fibres are weaker than those of adults. The mechanisms by which muscle is lost with age are unclear, but in this review we aim to pull together various strands of evidence to explain how muscle contractions support proteostasis in non-muscle tissues, particularly focussed on the production and potential transfer of Heat Shock Proteins (HSPs) and how this may fail during ageing, Furthermore we will identify logical approaches, based on this hypothesis, by which muscle loss in ageing may be reduced. Skeletal muscle generates superoxide and nitric oxide at rest and this generation is increased by contractile activity. In adults, this increased generation of reactive oxygen and nitrogen species (RONS) activate redox-sensitive transcription factors such as nuclear factor κB (NFκB), activator protein-1 (AP1) and heat shock factor 1 (HSF1), resulting in increases in cytoprotective proteins such as the superoxide dismutases, catalase and heat shock proteins that prevent oxidative damage to tissues and facilitate remodelling and proteostasis in both an intra- and inter-cellular manner. During ageing, the ability of skeletal muscle from aged organisms to respond to an increase in ROS generation by increased expression of cytoprotective proteins through activation of redox-sensitive transcription factors is severely attenuated. This age-related lack of physiological adaptations to the ROS induced by contractile activity appears to contribute to a loss of ROS homeostasis, increased oxidative damage and age-related dysfunction in skeletal muscle and potentially other tissues.
Collapse
Affiliation(s)
- Anne McArdle
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, University of Liverpool, L7 8TX, United Kingdom.
| | - Natalie Pollock
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, University of Liverpool, L7 8TX, United Kingdom
| | - Caroline A Staunton
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, University of Liverpool, L7 8TX, United Kingdom
| | - Malcolm J Jackson
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, University of Liverpool, L7 8TX, United Kingdom
| |
Collapse
|
165
|
Cobley JN, Sakellariou GK, Husi H, McDonagh B. Proteomic strategies to unravel age-related redox signalling defects in skeletal muscle. Free Radic Biol Med 2019; 132:24-32. [PMID: 30219702 DOI: 10.1016/j.freeradbiomed.2018.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 01/06/2023]
Abstract
Increased oxidative damage and disrupted redox signalling are consistently associated with age-related loss of skeletal muscle mass and function. Redox signalling can directly regulate biogenesis and degradation pathways and indirectly via activation of key transcription factors. Contracting skeletal muscle fibres endogenously generate free radicals (e.g. superoxide) and non-radical derivatives (e.g. hydrogen peroxide). Exercise induced redox signalling can promote beneficial adaptive responses that are disrupted by age-related redox changes. Identifying and quantifying the redox signalling pathways responsible for successful adaptation to exercise makes skeletal muscle an attractive physiological model for redox proteomic approaches. Site specific identification of the redox modification and quantification of site occupancy in the context of protein abundance remains a crucial concept for redox proteomics approaches. Notwithstanding, the technical limitations associated with skeletal muscle for proteomic analysis, we discuss current approaches for the identification and quantification of transient and stable redox modifications that have been employed to date in ageing research. We also discuss recent developments in proteomic approaches in skeletal muscle and potential implications and opportunities for investigating disrupted redox signalling in skeletal muscle ageing.
Collapse
Affiliation(s)
- James N Cobley
- Free Radical Laboratory, Departments of Diabetes and Cardiovascular Sciences, Centre for Health Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK
| | | | - Holger Husi
- Free Radical Laboratory, Departments of Diabetes and Cardiovascular Sciences, Centre for Health Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, NUI Galway, Ireland.
| |
Collapse
|
166
|
Nethi SK, Barui AK, Mukherjee S, Patra CR. Engineered Nanoparticles for Effective Redox Signaling During Angiogenic and Antiangiogenic Therapy. Antioxid Redox Signal 2019; 30:786-809. [PMID: 29943661 DOI: 10.1089/ars.2017.7383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Redox signaling plays a vital role in regulating various cellular signaling pathways and disease biology. Recently, nanomedicine (application of nanotechnology in biology and medicine) has been demonstrated to regulate angiogenesis through redox signaling. A complete understanding of redox signaling pathways influenced angiogenesis/antiangiogenesis triggered by therapeutic nanoparticles is extensively reviewed in this article. Recent Advances: In recent times, nanomedicines are regarded as the Trojan horses that could be employed for successful drug delivery, gene delivery, peptide delivery, disease diagnosis, and others, conquering barriers associated with conventional theranostic approaches. CRITICAL ISSUES Physiological angiogenesis is a tightly regulated process maintaining a balance between proangiogenic and antiangiogenic factors. The redox signaling is one of the main factors that contribute to this physiological balance. An aberrant redox signaling cascade can be caused by several exogenous and endogenous factors and leads to reduced or augmented angiogenesis that ultimately results in several disease conditions. FUTURE DIRECTIONS Redox signaling-based nanomedicine approach has emerged as a new platform for angiogenesis-related disease therapy, where nanoparticles promote angiogenesis via controlled reactive oxygen species (ROS) production and antiangiogenesis by triggering excessive ROS formation. Recently, investigators have identified different efficient nano-candidates, which modulate angiogenesis by controlling intracellular redox molecules. Considering the importance of angiogenesis in health care a thorough understanding of nanomedicine-regulated redox signaling would inspire researchers to design and develop more novel nanomaterials that could be used as an alternative strategy for the treatment of various diseases, where angiogenesis plays a vital role.
Collapse
Affiliation(s)
- Susheel Kumar Nethi
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Ayan Kumar Barui
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Sudip Mukherjee
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Chitta Ranjan Patra
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| |
Collapse
|
167
|
Anjo SI, Melo MN, Loureiro LR, Sabala L, Castanheira P, Grãos M, Manadas B. oxSWATH: An integrative method for a comprehensive redox-centered analysis combined with a generic differential proteomics screening. Redox Biol 2019; 22:101130. [PMID: 30737169 PMCID: PMC6435957 DOI: 10.1016/j.redox.2019.101130] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022] Open
Abstract
Most of the redox proteomics strategies are focused on the identification and relative quantification of cysteine oxidation without considering the variation in the total levels of the proteins. However, protein synthesis and protein degradation also belong to the regulatory mechanisms of the cells, being therefore important to consider the changes in total protein levels in PTMs-focused analyses, such as cysteine redox characterization. Therefore, a novel integrative approach combining the SWATH-MS method with differential alkylation using a combination of commonly available alkylating reagents (oxSWATH) is presented, by which it is possible to integrate the information regarding relative cysteine oxidation with the analysis of the total protein levels in a cost-effective high-throughput approach. The proposed method was tested using a redox-regulated protein and further applied to a comparative analysis of secretomes obtained from cells cultured under control or oxidative stress conditions to strengthen the importance of considering the overall proteome changes. Using the OxSWATH method it was possible to determine both the relative proportion of reduced and reversible oxidized oxoforms, as well as the total levels of each oxoform by taking into consideration the total levels of the protein. Therefore, using OxSWATH the comparative analyses can be performed at two different levels by considering the relative proportion or the total levels at both peptide and protein level. Moreover, since samples are acquired in SWATH-MS mode, besides the redox centered analysis, a generic differential protein expression analysis can also be performed, allowing a truly comprehensive evaluation of proteomics changes upon the oxidative stimulus. Data are available via ProteomeXchange and SWATHAtlas with the identifiers PXD006802, PXD006802, and PASS01210. Determination of redox changes considering protein total levels. Integrative redoxomics and common differential proteomics in a single analysis. Differential alkylation strategy using commonly available alkylating agents. First untargeted label-free quantitative method to study cysteine oxidation.
Collapse
Affiliation(s)
- Sandra I Anjo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - Matilde N Melo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Liliana R Loureiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Lúcia Sabala
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | | | - Mário Grãos
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Biocant, Technology Transfer Association, Cantanhede, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
168
|
Treatment of Infected Wounds in the Age of Antimicrobial Resistance: Contemporary Alternative Therapeutic Options. Plast Reconstr Surg 2019; 142:1082-1092. [PMID: 30252823 DOI: 10.1097/prs.0000000000004799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
As antibiotic resistance increases and antimicrobial options diminish, there is a pressing need to identify and develop new and/or alternative (non-antimicrobial-based) wound therapies. The authors describe the implications of antibiotic resistance on their current wound treatment paradigms and review the most promising non-antibiotic-based antimicrobial agents currently in research and development, with a focus on preclinical and human studies of therapeutic bacteriophages, antimicrobial peptides, cold plasma treatment, photodynamic therapy, honey, silver, and bioelectric dressings.
Collapse
|
169
|
Staurengo-Ferrari L, Badaro-Garcia S, Hohmann MSN, Manchope MF, Zaninelli TH, Casagrande R, Verri WA. Contribution of Nrf2 Modulation to the Mechanism of Action of Analgesic and Anti-inflammatory Drugs in Pre-clinical and Clinical Stages. Front Pharmacol 2019; 9:1536. [PMID: 30687097 PMCID: PMC6337248 DOI: 10.3389/fphar.2018.01536] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
Despite the progress that has occurred in recent years in the development of therapies to treat painful and inflammatory diseases, there is still a need for effective and potent analgesics and anti-inflammatory drugs. It has long been known that several types of antioxidants also possess analgesic and anti-inflammatory properties, indicating a strong relationship between inflammation and oxidative stress. Understanding the underlying mechanisms of action of anti-inflammatory and analgesic drugs, as well as essential targets in disease physiopathology, is essential to the development of novel therapeutic strategies. The Nuclear factor-2 erythroid related factor-2 (Nrf2) is a transcription factor that regulates cellular redox status through endogenous antioxidant systems with simultaneous anti-inflammatory activity. This review summarizes the molecular mechanisms and pharmacological actions screened that link analgesic, anti-inflammatory, natural products, and other therapies to Nrf2 as a regulatory system based on emerging evidences from experimental disease models and new clinical trial data.
Collapse
Affiliation(s)
- Larissa Staurengo-Ferrari
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Stephanie Badaro-Garcia
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Miriam S. N. Hohmann
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Marília F. Manchope
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Tiago H. Zaninelli
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Rubia Casagrande
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Estadual de Londrina, Londrina, Brazil
| | - Waldiceu A. Verri
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
170
|
The Emerging Role of Estrogens in Thyroid Redox Homeostasis and Carcinogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2514312. [PMID: 30728883 PMCID: PMC6343143 DOI: 10.1155/2019/2514312] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022]
Abstract
Reactive oxygen species (ROS) are the most critical class of free radicals or reactive metabolites produced by all living organisms. ROS regulate several cellular functions through redox-dependent mechanisms, including proliferation, differentiation, hormone synthesis, and stress defense response. However, ROS overproduction or lack of appropriate detoxification is harmful to cells and can be linked to the development of several diseases, such as cancer. Oxidative damage in cellular components, especially in DNA, can promote the malignant transformation that has already been described in thyroid tissue. In thyrocyte physiology, NADPH oxidase enzymes produce large amounts of ROS that are necessary for hormone biosynthesis and might contribute to the high spontaneous mutation rate found in this tissue. Thyroid cancer is the most common endocrine malignancy, and its incidence is significantly higher in women than in men. Several lines of evidence suggest the sex hormone estrogen as a risk factor for thyroid cancer development. Estrogen in turn, besides being a potent growth factor for both normal and tumor thyroid cells, regulates different mechanisms of ROS generation. Our group demonstrated that the thyroid gland of adult female rats exhibits higher hydrogen peroxide (H2O2) production and lower enzymatic antioxidant defense in comparison with male glands. In this review, we discuss the possible involvement of thyroid redox homeostasis and estrogen in the development of thyroid carcinogenesis.
Collapse
|
171
|
Pasquali MA, Harlow BL, Soares CN, Otto MW, Cohen LS, Minuzzi L, Gelain DP, Moreira JCF, Frey BN. A longitudinal study of neurotrophic, oxidative, and inflammatory markers in first-onset depression in midlife women. Eur Arch Psychiatry Clin Neurosci 2018; 268:771-781. [PMID: 28550365 DOI: 10.1007/s00406-017-0812-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 05/19/2017] [Indexed: 12/15/2022]
Abstract
Prospective studies have shown during the years preceding and following menopause, also known as "menopause transition", that midlife women are at higher risk for developing first-onset major depressive disorder (MDD). The biological factors associated with risk and resilience in this population are, however, largely unknown. Considering the growing body of evidence suggesting that inflammation, oxidative stress, and brain-derived neurotrophic factor (BDNF) are associated with the pathophysiology of MDD, we investigated serum levels of protein carbonyl, lipid peroxidation (thiobarbituric acid reactive substances-TBARS), thiol group content, BDNF, 3-nitrotyrosine, and heat shock protein 70 (HSP70) in a longitudinal cohort of first-onset MDD. One hundred and forty-eight women from the Harvard Study of Moods and Cycles, a prospective study of midlife women monitored throughout the transition to menopause, were studied. Within- and between-groups analyses of these peripheral markers were conducted in 37 women who developed and 111 women that did not develop MDD during the 3-year follow-up period. In women who developed MDD, HSP70 and 3-nitrotyrosine were elevated at baseline, whereas TBARS were elevated 6 months prior to development of MDD, as compared to those who did not develop MDD. Within-group analyses showed that HSP70, 3-nitrotyrosine, and BDNF decreased over time, whereas protein carbonyl was elevated only at 12 months prior to development of MDD. In women who did not develop MDD, HSP70 and thiol decreased over time. The development of MDD in midlife women may be associated with a systemic cascade of pro-oxidative and pro-inflammatory events including increased HSP70, 3-nitrotyrosine, protein carbonyl, and lipid peroxidation and decreased BDNF.
Collapse
Affiliation(s)
- Matheus A Pasquali
- Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada.,Mood Disorders Program, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada.,Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bernard L Harlow
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Claudio N Soares
- Department of Psychiatry, Queen's University School of Medicine, Kingston, Canada
| | - Michael W Otto
- Department of Psychological and Brain Sciences, Boston University, Boston, USA
| | - Lee S Cohen
- Center for Women's Mental Health, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Luciano Minuzzi
- Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada.,Mood Disorders Program, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, 100 West 5th Street, Suite C124, Hamilton, ON, L8N 3K7, Canada
| | - Daniel P Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jose Claudio F Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Benicio N Frey
- Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada. .,Mood Disorders Program, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada. .,Department of Psychiatry and Behavioural Neurosciences, McMaster University, 100 West 5th Street, Suite C124, Hamilton, ON, L8N 3K7, Canada.
| |
Collapse
|
172
|
Chung TH, Wu YP, Chew CY, Lam CH, Tan KT. Imaging and Quantification of Secreted Peroxynitrite at the Cell Surface by a Streptavidin-Biotin-Controlled Binding Probe. Chembiochem 2018; 19:2584-2590. [PMID: 30352141 DOI: 10.1002/cbic.201800542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Indexed: 12/13/2022]
Abstract
The ability to detect and image secreted peroxynitrite (ONOO- ) along the extracellular surface of a single cell is biologically significant, as ONOO- generally exerts its function for host defense and signal transductions at the plasma membrane. However, as a result of the short lifetime and fast diffusion rate of small ONOO- , precise determination of the ONOO- level at the cell surface remains a challenging task. In this paper, the use of a membrane-anchored streptavidin-biotin-controlled binding probe (CBP), ONOO-CBP, to determine quantitatively the ONOO- level at the cell surface and to investigate the effect of different stimulants on the production of ONOO- along the plasma membrane of macrophages is reported. Our results revealed that the combination of NO synthase (iNOS) and NADPH oxidase (NOX) activators was highly effective in inducing ONOO- secretion, achieving more than a 25-fold increase in ONOO- relative to untreated cells. After 1 h of phorbol-12-myristate-13-acetate (PMA) stimulation, the amount of ONOO- secreted by RAW264.7 macrophages was similar to the condition treated with 25 μm 3-morpholinosydnonimine hydrochloride (SIN-1), which was estimated to release about 20 μm of ONOO- into Dulbecco's modified Eagle's medium (DMEM) in 1 h. This novel approach should open up new opportunities to image various reactive oxygen and nitrogen species secreted at the plasma membrane that cannot be simply achieved by conventional analytical methods.
Collapse
Affiliation(s)
- Tzu-Hsuan Chung
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Yung-Peng Wu
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Chee Ying Chew
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Chak Hin Lam
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| | - Kui-Thong Tan
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan.,Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan
| |
Collapse
|
173
|
Mitoproteomics: Tackling Mitochondrial Dysfunction in Human Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1435934. [PMID: 30533169 PMCID: PMC6250043 DOI: 10.1155/2018/1435934] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Mitochondria are highly dynamic and regulated organelles that historically have been defined based on their crucial role in cell metabolism. However, they are implicated in a variety of other important functions, making mitochondrial dysfunction an important axis in several pathological contexts. Despite that conventional biochemical and molecular biology approaches have provided significant insight into mitochondrial functionality, innovative techniques that provide a global view of the mitochondrion are still necessary. Proteomics fulfils this need by enabling accurate, systems-wide quantitative analysis of protein abundance. More importantly, redox proteomics approaches offer unique opportunities to tackle oxidative stress, a phenomenon that is intimately linked to aging, cardiovascular disease, and cancer. In addition, cutting-edge proteomics approaches reveal how proteins exert their functions in complex interaction networks where even subtle alterations stemming from early pathological states can be monitored. Here, we describe the proteomics approaches that will help to deepen the role of mitochondria in health and disease by assessing not only changes to mitochondrial protein composition but also alterations to their redox state and how protein interaction networks regulate mitochondrial function and dynamics. This review is aimed at showing the reader how the application of proteomics approaches during the last 20 years has revealed crucial mitochondrial roles in the context of aging, neurodegenerative disorders, metabolic disease, and cancer.
Collapse
|
174
|
Peng J, Huang N, Huang S, Li L, Ling Z, Jin S, Huang A, Lin K, Zou X. [Effect of miR-21 down-regulated by H 2O 2 on osteogenic differentiation of MC3T3-E1 cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:276-284. [PMID: 29806275 DOI: 10.7507/1002-1892.201707030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To explore the effect and mechanism of miR-21 down-regulated which was induced by H 2O 2 on osteogenic differentiation of MC3T3-E1 cells. Methods MC3T3-E1 cells were cultured and passaged, and the 7th generation cells were harvested to use in experiment. The MC3T3-E1 cells were treated with different concentrations (0, 40, 80, 160, and 320 μmol/L) of H 2O 2. The expression of miR-21 was detected by real-time quantitative PCR (RT-PCR) and the cell viability was determined by MTS. Then the appropriate concentration of H 2O 2 was obtained. To analyze the effect of H 2O 2 on osteogenic differentiation of MC3T3-E1 cells, the MC3T3-E1 cells were divided into blank control group (group A), H 2O 2 group (group B), osteogenic induction group (group C), and H 2O 2+osteogenic induction group (group D). The expression of miR-21 and the osteogenesis related genes expressions of Runx2, osteopontin (OPN), and collagen type Ⅰ alpha 1 (Col1a1) were detected by RT-PCR. The expression of phosphatase and tensin homolog (PTEN) was detected by Western blot. The extracellular calcium deposition was detected by alizarin red staining. To analyze the effect on osteogenic differentiation of MC3T3-E1 cells after the transfection of miR-21 inhibitor and siRNA-PTEN, the MC3T3-E1 cells were divided into H 2O 2 group (group A1), H 2O 2+osteogenic induction group (group B1), H 2O 2+osteogenic induction+miR-21 inhibitor group (group C1), and H 2O 2+osteogenic induction+miR-21 inhibitor negative control group (group D1); and H 2O 2 group (group A2), H 2O 2+osteogenic induction group (group B2), H 2O 2+osteogenic induction+siRNA-PTEN negative control group (group C2), and H 2O 2+osteogenic induction+siRNA-PTEN group (group D2). The osteogenesis related genes were detected by RT-PCR and the extracellular calcium deposition was detected by alizarin red staining. Results The results of MTS and RT-PCR showed that the appropriate concentration of H 2O 2 was 160 μmol/L. The expression of miR-21 was significantly lower in group B than in group A at 1 and 2 weeks ( P<0.05). The expression of miR-21 was significantly lower in group D than in group C at 1 and 2 weeks ( P<0.05). The expression of PTEN protein was significantly lower in group C than in groups A and D ( P<0.05). The mRNA expressions of Runx2, OPN, and Col1a1 were significantly lower in group D than in group C at 1 and 2 weeks ( P<0.05). The extracellular calcium deposition in group D was obviously less than that in group C. The expression of PTEN protein was significantly higher in group C1 than in group D1 ( P<0.05). The mRNA expressions of Runx2 and OPN were significantly lower in group C1 than in groups B1 and D1 at 1 and 2 weeks ( P<0.05). The mRNA expression of Col1a1 was significantly lower in group C1 than in groups B1 and D1 at 2 weeks ( P<0.05). The extracellular calcium deposition in group C1 was obviously less than those in groups B1 and D1. The mRNA expressions of OPN and Col1a1 were significantly higher in group D2 than in groups B2 and C2 at 1 week ( P<0.05). The extracellular calcium deposition in group D2 was obviously more than those in groups B2 and C2. Conclusion H 2O 2 inhibits the osteogenic differentiation of MC3T3-E1 cells, which may be induced by down-regulating the expression of miR-21.
Collapse
Affiliation(s)
- Jianqiang Peng
- Department of Spine Surgery, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen Guangdong, 518033, P.R.China
| | | | - Sheng Huang
- Department of Orthopedics, the First Affiliated Hospital of Nanchang University, Nanchang Jiangxi, 330006, P.R.China
| | - Liangping Li
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, 510080, P.R.China
| | - Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, 510080, P.R.China
| | - Song Jin
- Department of Spine Surgery, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen Guangdong, 518033, P.R.China
| | - Aijun Huang
- Department of Spine Surgery, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen Guangdong, 518033, P.R.China
| | - Kun Lin
- Department of Spine Surgery, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen Guangdong, 518033, P.R.China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, 510080,
| |
Collapse
|
175
|
Wiśniewski K, Jóźwik-Pruska J, Bieńkowski M, Bobeff EJ, Bryl M, Kałużna-Czaplińska J, Jaskólski DJ. Isoprostanes as potential cerebral vasospasm biomarkers. Neurol Neurochir Pol 2018; 52:643-651. [PMID: 30314904 DOI: 10.1016/j.pjnns.2018.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/01/2018] [Accepted: 09/25/2018] [Indexed: 11/29/2022]
Abstract
Despite enormous progress in medicine, symptomatic cerebral vasospasm (CVS), remains an unexplained clinical problem, which leaves both physicians and patients helpless and relying on chance, due to the lack of specific marker indicative of imminent danger as well as the lack of specific treatment. In our opinion CVS occurrence depends on dynamic disbalance between free radicals' formation (oxidative stress) and antioxidant activity. Isoprostanes are products of free-radical peroxidation of polyunsaturated fatty acids, and seem to mark a promising path for the research aiming to unravel its possible mechanism. Not only are they the biomarkers of oxidative stress in vivo and in vitro, but also have manifold biological effects (including vasoactive, inflammatory and mitogenic) via activation of the thromboxane A2 receptor (TBXA2R), both in physiological and pathophysiological processes. This review addresses the importance of isoprostanes in CVS in quest of appropriate biomarkers.
Collapse
Affiliation(s)
- Karol Wiśniewski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcińskiego 22, 90-153 Lodz, Poland.
| | - Jagoda Jóźwik-Pruska
- Institute of General and Ecological Chemistry, Department of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
| | - Michał Bieńkowski
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| | - Ernest J Bobeff
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcińskiego 22, 90-153 Lodz, Poland
| | - Maciej Bryl
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcińskiego 22, 90-153 Lodz, Poland
| | - Joanna Kałużna-Czaplińska
- Institute of General and Ecological Chemistry, Department of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
| | - Dariusz J Jaskólski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcińskiego 22, 90-153 Lodz, Poland
| |
Collapse
|
176
|
Karabulutoglu M, Finnon R, Imaoka T, Friedl AA, Badie C. Influence of diet and metabolism on hematopoietic stem cells and leukemia development following ionizing radiation exposure. Int J Radiat Biol 2018; 95:452-479. [PMID: 29932783 DOI: 10.1080/09553002.2018.1490042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE The review aims to discuss the prominence of dietary and metabolic regulators in maintaining hematopoietic stem cell (HSC) function, long-term self-renewal, and differentiation. RESULTS Most adult stem cells are preserved in a quiescent, nonmotile state in vivo which acts as a "protective state" for stem cells to reduce endogenous stress provoked by DNA replication and cellular respiration as well as exogenous environmental stress. The dynamic balance between quiescence, self-renewal and differentiation is critical for supporting a functional blood system throughout life of an organism. Stress-conditions, for example ionizing radiation exposure can trigger the blood forming HSCs to proliferate and migrate through extramedullary tissues to expand the number of HSCs and increase hematopoiesis. In addition, a wealth of investigation validated that deregulation of this balance plays a critical pathogenic role in various different hematopoietic diseases including the leukemia development. CONCLUSION The review summarizes the current knowledge on how alterations in dietary and metabolic factors could alter the risk of leukemia development following ionizing radiation exposure by inhibiting or even reversing the leukemic progression. Understanding the influence of diet, metabolism, and epigenetics on radiation-induced leukemogenesis may lead to the development of practical interventions to reduce the risk in exposed populations.
Collapse
Affiliation(s)
- Melis Karabulutoglu
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK.,b CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
| | - Rosemary Finnon
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| | - Tatsuhiko Imaoka
- c Department of Radiation Effects Research, National Institute of Radiological Sciences , National Institutes for Quantum and Radiological Science and Technology , Chiba , Japan
| | - Anna A Friedl
- d Department of Radiation Oncology , University Hospital, LMU Munich , Munich , Germany
| | - Christophe Badie
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| |
Collapse
|
177
|
Redox state affects fecundity and insecticide susceptibility in Anopheles gambiae. Sci Rep 2018; 8:13054. [PMID: 30158658 PMCID: PMC6115382 DOI: 10.1038/s41598-018-31360-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/19/2018] [Indexed: 01/02/2023] Open
Abstract
Redox reactions play a central role in the metabolism of an organism. It is vital to maintain redox homeostasis in response to the fluctuation of redox shift in various biological contexts. NADPH-dependent reducing capacity is one of the key factors contributing to the redox homeostasis. To understand the redox capacity and its impact on mosquito fecundity and susceptibility to insecticides in Anopheles gambiae, we examined the dynamics of elevated oxidative state via induction by paraquat (PQ) and the inhibition of NADPH regeneration by 6-aminonicotinamide (6AN). In naïve conditions, inherent oxidative capacity varies between individuals, as measured by GSSG/GSH ratio. The high GSSG/GSH ratio was negatively correlated with fecundity. Both PQ and 6AN feeding increased GSSG/GSH ratio and elevated protein carbonylation, a marker of oxidative damage. Both pro-oxidants lowered egg production. Co-feeding the pro-oxidants with antioxidant lycopene attenuated the adverse effects on fecundity, implying that oxidative stress was the cause of this phenotype. Pre-feeding with 6AN increased insecticide susceptibility in DDT resistant mosquitoes. These results indicate that oxidative state is delicate in mosquitoes, manipulation of NADPH pool may adversely affect fecundity and insecticide detoxification capacity. This knowledge can be exploited to develop novel vector control strategies targeting fecundity and insecticide resistance.
Collapse
|
178
|
Oxidative stress in chronic lung disease: From mitochondrial dysfunction to dysregulated redox signaling. Mol Aspects Med 2018; 63:59-69. [PMID: 30098327 DOI: 10.1016/j.mam.2018.08.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 12/31/2022]
Abstract
The lung is a delicate organ with a large surface area that is continuously exposed to the external environment, and is therefore highly vulnerable to exogenous sources of oxidative stress. In addition, each of its approximately 40 cell types can also generate reactive oxygen species (ROS), as byproducts of cellular metabolism and in a more regulated manner by NOX enzymes with functions in host defense, immune regulation, and cell proliferation or differentiation. To effectively regulate the biological actions of exogenous and endogenous ROS, various enzymatic and non-enzymatic antioxidant defense systems are present in all lung cell types to provide adequate protection against their injurious effects and to allow for appropriate ROS-mediated biological signaling. Acute and chronic lung diseases are commonly thought to be associated with increased oxidative stress, evidenced by altered cellular or extracellular redox status, increased irreversible oxidative modifications in proteins or DNA, mitochondrial dysfunction, and altered expression or activity of NOX enzymes and antioxidant enzyme systems. However, supplementation strategies with generic antioxidants have been minimally successful in prevention or treatment of lung disease, most likely due to their inability to distinguish between harmful and beneficial actions of ROS. Recent studies have attempted to identify specific redox-based mechanisms that may mediate chronic lung disease, such as allergic asthma or pulmonary fibrosis, which provide opportunities for selective redox-based therapeutic strategies that may be useful in treatment of these diseases.
Collapse
|
179
|
Mandato A, Chai YC. Regulation of antigen 85C activity by reversible S-glutathionylation. IUBMB Life 2018; 70:1111-1114. [DOI: 10.1002/iub.1923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/19/2018] [Accepted: 07/04/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Alysia Mandato
- Department of Chemistry; John Carroll University; University Heights OH
| | - Yuh-Cherng Chai
- Department of Chemistry; John Carroll University; University Heights OH
| |
Collapse
|
180
|
Wang M, Li K, Zou Z, Li L, Zhu L, Wang Q, Gao W, Wang Y, Huang W, Liu R, Yao K, Liu Q. Piperidine nitroxide Tempol enhances cisplatin-induced apoptosis in ovarian cancer cells. Oncol Lett 2018; 16:4847-4854. [PMID: 30250550 PMCID: PMC6144655 DOI: 10.3892/ol.2018.9289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 05/11/2018] [Indexed: 12/14/2022] Open
Abstract
A nitroxide radical, Tempol (Tempol, TPL), is usually used as an antioxidative agent clinically, whereas the mechanism underlying its pro-oxidative effect has not been thoroughly investigated. The present study investigated the pro-oxidative effect of TPL on the inhibition of cellular proliferation and its role in enhancing the effect of anticancer drug cisplatin (DDP) on the induction of apoptosis in ovarian cancer cells. Cell viability and proliferation were evaluated by MTT assay. Cell apoptosis was analyzed by flow cytometry (FCM) following staining with Annexin V/propidium iodide. Western blot analysis was performed to determine the expression levels of anti-apoptotic protein B-cell lymphoma-2 (Bcl-2) and pro-apoptotic protein Bcl-2-associated X protein (Bax), and the Bcl-2:Bax expression ratio. Cellular reactive oxygen species (ROS) were labeled with dichlorofluorescin-diacetate and analyzed by FCM. The results revealed that cell viabilities of OVCAR3 and SKOV3 cells were decreased by TPL in dose-dependent manner at concentrations of 2 to 10 mM after 48 h incubation. The cell proliferation rates of OVCAR3 and SKOV3 cells were suppressed by TPL at lower toxic concentrations of 1.5 and 1 mM, respectively, compared with the control group. The MTT assay indicated that the combination therapy significantly inhibited the cell proliferation of OVCAR3 cells compared with treatment with DDP alone. FCM demonstrated that the combination treatment increased the proportion of early apoptotic cells in OVCAR3 cells compared with single DDP treatment. Western blot analysis revealed that the combination treatment markedly decreased the Bcl-2:Bax expression ratio compared with treatment with DDP alone. Detection of cellular ROS expression levels demonstrated that the combination therapy significantly increased cellular ROS generation compared with the DDP-only therapy. These data indicated that TPL increased the effect of DDP on inducing apoptosis in OVCAR3 cells.
Collapse
Affiliation(s)
- Meng Wang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Keyi Li
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhiwei Zou
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Linlin Li
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Lingqun Zhu
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qianli Wang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wenwen Gao
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ying Wang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wenhua Huang
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ruiyuan Liu
- Science Department of Chemistry, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Kaitai Yao
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiuzhen Liu
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Guangzhou Key Laboratory of Tumor Immunology Research, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
181
|
Anathy V, Lahue KG, Chapman DG, Chia SB, Casey DT, Aboushousha R, van der Velden JLJ, Elko E, Hoffman SM, McMillan DH, Jones JT, Nolin JD, Abdalla S, Schneider R, Seward DJ, Roberson EC, Liptak MD, Cousins ME, Butnor KJ, Taatjes DJ, Budd RC, Irvin CG, Ho YS, Hakem R, Brown KK, Matsui R, Bachschmid MM, Gomez JL, Kaminski N, van der Vliet A, Janssen-Heininger YMW. Reducing protein oxidation reverses lung fibrosis. Nat Med 2018; 24:1128-1135. [PMID: 29988126 PMCID: PMC6204256 DOI: 10.1038/s41591-018-0090-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis is characterized by excessive deposition of collagen in the lung, leading to chronically impaired gas exchange and death1-3. Oxidative stress is believed to be critical in this disease pathogenesis4-6, although the exact mechanisms remain enigmatic. Protein S-glutathionylation (PSSG) is a post-translational modification of proteins that can be reversed by glutaredoxin-1 (GLRX)7. It remains unknown whether GLRX and PSSG play a role in lung fibrosis. Here, we explored the impact of GLRX and PSSG status on the pathogenesis of pulmonary fibrosis, using lung tissues from subjects with idiopathic pulmonary fibrosis, transgenic mouse models and direct administration of recombinant Glrx to airways of mice with existing fibrosis. We demonstrate that GLRX enzymatic activity was strongly decreased in fibrotic lungs, in accordance with increases in PSSG. Mice lacking Glrx were far more susceptible to bleomycin- or adenovirus encoding active transforming growth factor beta-1 (AdTGFB1)-induced pulmonary fibrosis, whereas transgenic overexpression of Glrx in the lung epithelium attenuated fibrosis. We furthermore show that endogenous GLRX was inactivated through an oxidative mechanism and that direct administration of the Glrx protein into airways augmented Glrx activity and reversed increases in collagen in mice with TGFB1- or bleomycin-induced fibrosis, even when administered to fibrotic, aged animals. Collectively, these findings suggest the therapeutic potential of exogenous GLRX in treating lung fibrosis.
Collapse
Affiliation(s)
- Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David G Chapman
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Dylan T Casey
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jos L J van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Sidra M Hoffman
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David H McMillan
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jane T Jones
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - James D Nolin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Sarah Abdalla
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Robert Schneider
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David J Seward
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | | - Matthew D Liptak
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Morgan E Cousins
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Kelly J Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Ralph C Budd
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Charles G Irvin
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Razq Hakem
- Department of Medical Biophysics and Immunology, University of Toronto, and the Ontario Cancer Institute/University Health Network, Toronto, Ontario, Canada
| | - Kevin K Brown
- Department of Medicine, Pulmonary, Critical Care and Sleep Section, National Jewish Health and the University of Colorado, Denver, CO, USA
| | - Reiko Matsui
- Department of Medicine, Boston University, Boston, MA, USA
| | | | - Jose L Gomez
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
182
|
Wilson C, Muñoz-Palma E, González-Billault C. From birth to death: A role for reactive oxygen species in neuronal development. Semin Cell Dev Biol 2018; 80:43-49. [DOI: 10.1016/j.semcdb.2017.09.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
|
183
|
Sulforaphane protects granulosa cells against oxidative stress via activation of NRF2-ARE pathway. Cell Tissue Res 2018; 374:629-641. [PMID: 30032437 DOI: 10.1007/s00441-018-2877-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 06/20/2018] [Indexed: 01/06/2023]
Abstract
Sulforaphane (SFN) has been considered as an indirect antioxidant and potential inducer of the Nrf2-ARE pathway. This study was conducted to investigate the protective role of SFN against oxidative stress in bovine granulosa cells (GCs). GCs were collected from antral follicles (4-8 mm) and cultured according to the experimental design where group 1 = control, group 2 = treated with SFN, group 3 = treated with hydrogen peroxide (H2O2), group 4 = pretreated with SFN and then with H2O2 (protective) and group 5 = treated with H2O2 followed by SFN treatment (rescuing). Results showed that SFN pretreatment significantly increases cell viability and reduces cytotoxicity in GCs under oxidative stress. Following H2O2 exposure, expression of NRF2 was found to be significantly increased (p < 0.05) in SFN-pretreated cells, while no significant differences were observed between group 3 and group 5, although the expression was significantly increased compared to the control group. Moreover, the relative abundance of the NRF2 downstream target antioxidant genes (CAT, PRDX1, SOD1 and TXN1) were higher (fold change ranged from 7 to 14, p < 0.05) in sulforaphane pretreated GCs. Low level of ROS and lipid accumulation and higher mitochondrial activity were observed in GCs pretreated with SFN, whereas no such changes were observed in GCs treated with SFN after exposure to oxidative stress (group 5). Thus, we suggest that SFN pretreatment effectively protects GCs against oxidative damage through the activation of the NRF2-ARE pathway, whereas addition of SFN during oxidative insult failed to rescue GCs.
Collapse
|
184
|
Zhang Y, Xiong Y, Zhou J, Xin N, Zhu Z, Wu Y. FoxO1 expression in osteoblasts modulates bone formation through resistance to oxidative stress in mice. Biochem Biophys Res Commun 2018; 503:1401-1408. [PMID: 30025894 DOI: 10.1016/j.bbrc.2018.07.055] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 02/05/2023]
Abstract
Accumulation of reactive oxygen species (ROS) induced by oxidative stress (OS) affects cell survival, cell function and even results in cell death. As a major transcription factor of forkhead O (FoxOs) family, FoxO1 orchestrates multiple osteoblastic biological processes, thus regulating osteoblast physiology and bone metabolism. However, the outcome of osteoblast behaviors varies under different physiological and pathological conditions. Also, the underlying impact of FoxO1 on oxidative stress and further on bone metabolism still remains unclear. In this study, using osteoblast-specific FoxO1 knockout (FoxO1OB-/-) mice, we investigated the potential roles of FoxO1 on bone formation and osteoblast bioactivity under physiological condition. We show herein that FoxO1-knockout decreased bone volume and bone formation rate in FoxO1OB-/- mice, which might be related to the decreased osteoblasts number. We also found that FoxO1-knockout increased apoptosis-related caspase-3 activity of osteoblasts, and inhibited the expression of osteogenic phenotypic markers (i.e. Runx2, Osx, ALP and OPN), leading to reduced osteoblasts differentiation. The alterations of bone formation and osteoblasts bioactivity were further testified to be linked to the elevated intracellular oxidative stress levels in FoxO1-deficient osteoblasts. Besides, administration of the antioxidant N-acetyl-l-cysteine (NAC) normalized the increased ROS levels in FoxO1-deficient osteoblasts, restoring the decreased osteoblasts differentiation, suppressing apoptosis-related caspase-3 activity, and promoting the expression of osteogenic markers in FoxO1-deficient osteoblasts. These results together illustrated that as a major regulator in redox homeostasis and osteoblast physiology, FoxO1 provides a favorable intracellular environment for osteoblast functions by defensing against the adverse effects of oxidative stress.
Collapse
Affiliation(s)
- Yixin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, China; Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiaqi Zhou
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhimin Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, China; Department of Oral Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yingying Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
185
|
Baranowska M, Suliborska K, Chrzanowski W, Kusznierewicz B, Namieśnik J, Bartoszek A. The relationship between standard reduction potentials of catechins and biological activities involved in redox control. Redox Biol 2018; 17:355-366. [PMID: 29803149 PMCID: PMC6007051 DOI: 10.1016/j.redox.2018.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/01/2018] [Accepted: 05/12/2018] [Indexed: 01/03/2023] Open
Abstract
Redox homeostasis involves factors that ensure proper function of cells. The excess reactive oxygen species (ROS) leads to oxidative stress and increased risk of oxidative damage to cellular components. In contrast, upon reductive stress, insufficient ROS abundance may result in faulty cell signalling. It may be expected that dietary antioxidants, depending on their standard reduction potentials (E°), will affect both scenarios. In our study, for the first time, we systematically tested the relationship among E°, chemical properties, and biological effects in HT29 cells for a series of structurally different catechins and a major endogenous antioxidant - glutathione (GSH), at both physiological and dietary concentrations. Among chemical antioxidant activity tests, the strongest correlation with E° was seen using a DPPH assay. The values of E° were also highly correlated with cellular antioxidant activity (CAA) values determined in HT29 cells. Our results indicated that physiological concentrations (1-10 µM) of tested catechins stabilized the redox status of cells, which was not exhibited at higher concentrations. This stabilization of redox homeostasis was mirrored by constant, dose and E° independent CAA values, uninhibited growth of HT29 cells, modulation of hydrogen peroxide-induced DNA damage, as well as effects at the genomic level, where either up-regulation of three redox-related genes (ALB, CCL5, and HSPA1A) out of 84 in the array (1 µM) or no effect (10 µM) was observed for catechins. Higher catechin concentrations (over 10 µM) increased CAA values in a dose- and E°-dependent manner, caused cell growth inhibition, but surprisingly did not protect HT29 cells against reactive oxygen species (ROS)-induced DNA fragmentation. In conclusion, dose-dependent effects of dietary antioxidants and biological functions potentially modulated by them may become deregulated upon exposure to excessive doses.
Collapse
Affiliation(s)
- Monika Baranowska
- Department of Food Chemistry, Technology and Biotechnology, Gdansk University of Technology, Gdansk, Poland.
| | - Klaudia Suliborska
- Department of Physical Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Wojciech Chrzanowski
- Department of Physical Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Barbara Kusznierewicz
- Department of Food Chemistry, Technology and Biotechnology, Gdansk University of Technology, Gdansk, Poland
| | - Jacek Namieśnik
- Department of Analytical Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Agnieszka Bartoszek
- Department of Food Chemistry, Technology and Biotechnology, Gdansk University of Technology, Gdansk, Poland
| |
Collapse
|
186
|
Dong Y, Xu S, Liu J, Ponnusamy M, Zhao Y, Zhang Y, Wang Q, Li P, Wang K. Non-coding RNA-linked epigenetic regulation in cardiac hypertrophy. Int J Biol Sci 2018; 14:1133-1141. [PMID: 29989099 PMCID: PMC6036733 DOI: 10.7150/ijbs.26215] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
Cardiac hypertrophy is an adaptive enlargement of myocardium in response to pressure overload caused various pathological insults, which is accompanied by alteration of a complex cascade of signaling pathways. During the hypertrophy process, many changes occur at cellular level including gene reprogramming by turning off chromatin regulators. Studies from the past decade have demonstrated that the abnormal epigenetic modifications, such as DNA methylation, histone modification, and oxidative modification of nucleic acid, could lead to changes in chromosome structure and cardiac dysfunction. Increasing evidence indicates that non-coding RNAs (ncRNAs) have functional significance in modulating the gene expression during those pathological events in the heart. Emerging evidences have highlighted that ncRNAs might serve as a signal for changing the state of chromatin, however, the knowledge about the ncRNA-linked epigenetic regulatory mechanisms in cardiac pathologies is still largely unexplored. In this review, we summarize the current information on association between ncRNAs and epigenetic modifications in cardiac hypertrophy, and we have discussed their crosstalk. In addition, this review provides insights into their therapeutic and diagnostic potential for treating hypertrophic heart disease.
Collapse
Affiliation(s)
- Yanhan Dong
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Sheng Xu
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Jing Liu
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yanfang Zhao
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yanhui Zhang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Qi Wang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Kun Wang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| |
Collapse
|
187
|
Robb EL, Hall AR, Prime TA, Eaton S, Szibor M, Viscomi C, James AM, Murphy MP. Control of mitochondrial superoxide production by reverse electron transport at complex I. J Biol Chem 2018; 293:9869-9879. [PMID: 29743240 PMCID: PMC6016480 DOI: 10.1074/jbc.ra118.003647] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/08/2018] [Indexed: 12/17/2022] Open
Abstract
The generation of mitochondrial superoxide (O2̇̄) by reverse electron transport (RET) at complex I causes oxidative damage in pathologies such as ischemia reperfusion injury, but also provides the precursor to H2O2 production in physiological mitochondrial redox signaling. Here, we quantified the factors that determine mitochondrial O2̇̄ production by RET in isolated heart mitochondria. Measuring mitochondrial H2O2 production at a range of proton-motive force (Δp) values and for several coenzyme Q (CoQ) and NADH pool redox states obtained with the uncoupler p-trifluoromethoxyphenylhydrazone, we show that O2̇̄ production by RET responds to changes in O2 concentration, the magnitude of Δp, and the redox states of the CoQ and NADH pools. Moreover, we determined how expressing the alternative oxidase from the tunicate Ciona intestinalis to oxidize the CoQ pool affected RET-mediated O2̇̄ production at complex I, underscoring the importance of the CoQ pool for mitochondrial O2̇̄ production by RET. An analysis of O2̇̄ production at complex I as a function of the thermodynamic forces driving RET at complex I revealed that many molecules that affect mitochondrial reactive oxygen species production do so by altering the overall thermodynamic driving forces of RET, rather than by directly acting on complex I. These findings clarify the factors controlling RET-mediated mitochondrial O2̇̄ production in both pathological and physiological conditions. We conclude that O2̇̄ production by RET is highly responsive to small changes in Δp and the CoQ redox state, indicating that complex I RET represents a major mode of mitochondrial redox signaling.
Collapse
Affiliation(s)
- Ellen L Robb
- From the Medical Research Council Mitochondrial Biology Unit, Hills Road, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Andrew R Hall
- From the Medical Research Council Mitochondrial Biology Unit, Hills Road, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Tracy A Prime
- From the Medical Research Council Mitochondrial Biology Unit, Hills Road, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Simon Eaton
- the UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Marten Szibor
- the Faculty of Medicine and Life Sciences, University of Tampere, Tampere FI-33014, Finland, and
- the Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Carlo Viscomi
- From the Medical Research Council Mitochondrial Biology Unit, Hills Road, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Andrew M James
- From the Medical Research Council Mitochondrial Biology Unit, Hills Road, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Michael P Murphy
- From the Medical Research Council Mitochondrial Biology Unit, Hills Road, University of Cambridge, Cambridge CB2 0XY, United Kingdom,
| |
Collapse
|
188
|
Synapse Pruning: Mitochondrial ROS with Their Hands on the Shears. Bioessays 2018; 40:e1800031. [DOI: 10.1002/bies.201800031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/27/2018] [Indexed: 12/27/2022]
|
189
|
Pomatto LCD, Cline M, Woodward N, Pakbin P, Sioutas C, Morgan TE, Finch CE, Forman HJ, Davies KJA. Aging attenuates redox adaptive homeostasis and proteostasis in female mice exposed to traffic-derived nanoparticles ('vehicular smog'). Free Radic Biol Med 2018; 121:86-97. [PMID: 29709705 PMCID: PMC5987225 DOI: 10.1016/j.freeradbiomed.2018.04.574] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 01/02/2023]
Abstract
Environmental toxicants are catalysts for protein damage, aggregation, and the aging process. Fortunately, evolution selected adaptive homeostasis as a system to mitigate such damage by expanding the normal capacity to cope with toxic stresses. Little is known about the subcellular degradative responses to proteins oxidatively damaged by air pollution. To better understand the impact of environmental toxicants upon the adaptive homeostatic response, female C57BL/6 mice were exposed for 10 weeks to filtered air or reaerosolized vehicular-derived nano-scale particulate matter (nPM), at which point tissues from young (6 month) and middle-aged (21 month) mice were studied. We found significant increases of proteolytic capacity in lung, liver, and heart. Up to two-fold increases were seen in the 20S Proteasome, the Immunoproteasome, the mitochondrial Lon protease, and NF-E2-related factor 2 (Nrf2), a major transcriptional factor for these and other stress-responsive genes. The responses were equivalent in all organs, despite the indirect input of inhaled particles to heart and liver which are downstream of lung. To our knowledge, this is the first exploration of proteostatic responses to oxidative damage by air pollution. Although, middle-aged mice had higher basal levels, their Nrf2-responsive-genes exhibited no response to nanoparticulate exposure. We also found a parallel age-associated rise in the Nrf2 transcriptional inhibitors, Bach1 and c-Myc which appear to attenuate adaptive responses in older mammals, possibly explaining the 'age-ceiling effect.' This report extends prior findings in male mice by demonstrating the involvement of proteolytic responses to traffic-related air pollution in lung, liver, and heart of female mice, with an age-dependent loss of adaptive homeostasis.
Collapse
Affiliation(s)
- Laura C D Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Mayme Cline
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Nicholas Woodward
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Payam Pakbin
- Department of Civil and Environmental Engineering of the Viterbi School of Engineering, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering of the Viterbi School of Engineering, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Todd E Morgan
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, the University of Southern California, Los Angeles, CA 90089-0191, USA; Molecular and Computational Biology Program, Department of Biological Sciences of the Dornsife College of Letters, Arts & Sciences, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Henry Jay Forman
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, the University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, the University of Southern California, Los Angeles, CA 90089-0191, USA; Molecular and Computational Biology Program, Department of Biological Sciences of the Dornsife College of Letters, Arts & Sciences, the University of Southern California, Los Angeles, CA 90089-0191, USA; Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, the University of Southern California, Los Angeles, CA 90089-0191, USA.
| |
Collapse
|
190
|
Frontiñán-Rubio J, Santiago-Mora RM, Nieva-Velasco CM, Ferrín G, Martínez-González A, Gómez MV, Moreno M, Ariza J, Lozano E, Arjona-Gutiérrez J, Gil-Agudo A, De la Mata M, Pesic M, Peinado JR, Villalba JM, Pérez-Romasanta L, Pérez-García VM, Alcaín FJ, Durán-Prado M. Regulation of the oxidative balance with coenzyme Q10 sensitizes human glioblastoma cells to radiation and temozolomide. Radiother Oncol 2018; 128:236-244. [PMID: 29784452 DOI: 10.1016/j.radonc.2018.04.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 04/05/2018] [Accepted: 04/29/2018] [Indexed: 10/16/2022]
Abstract
OBJECTIVES To investigate how the modulation of the oxidative balance affects cytotoxic therapies in glioblastoma, in vitro. MATERIAL AND METHODS Human glioblastoma U251 and T98 cells and normal astrocytes C8D1A were loaded with coenzyme Q10 (CoQ). Mitochondrial superoxide ion (O2-) and H2O2 were measured by fluorescence microscopy. OXPHOS performance was assessed in U251 cells with an oxytherm Clark-type electrode. Radio- and chemotherapy cytotoxicity was assessed by immunostaining of γH2AX (24 h), annexin V and nuclei morphology, at short (72 h) and long (15 d) time. Hif-1α, SOD1, SOD2 and NQO1 were determined by immunolabeling. Catalase activity was measured by classic enzymatic assay. Glutathione levels and total antioxidant capacity were quantified using commercial kits. RESULTS CoQ did not affect oxygen consumption but reduced the level of O2- and H2O2 while shifted to a pro-oxidant cell status mainly due to a decrease in catalase activity and SOD2 level. Hif-1α was dampened, echoed by a decrease lactate and several key metabolites involved in glutathione synthesis. CoQ-treated cells were twofold more sensitive than control to radiation-induced DNA damage and apoptosis in short and long-term clonogenic assays, potentiating TMZ-induced cytotoxicity, without affecting non-transformed astrocytes. CONCLUSIONS CoQ acts as sensitizer for cytotoxic therapies, disarming GBM cells, but not normal astrocytes, against further pro-oxidant injuries, being potentially useful in clinical practice for this fatal pathology.
Collapse
Affiliation(s)
- Javier Frontiñán-Rubio
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Raquel María Santiago-Mora
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Consuelo María Nieva-Velasco
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Gustavo Ferrín
- Instituto Maimónides de Investigación Biomédica en Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Córdoba, Spain
| | | | - María Victoria Gómez
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies and Instituto Regional de Investigación Científica Aplicada, University of Castilla-La Mancha, Ciudad Real, Spain
| | - María Moreno
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies and Instituto Regional de Investigación Científica Aplicada, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Julia Ariza
- Department of Cell Biology, Physiology and Immunology, Faculty of Sciences, University of Córdoba, Agrifood Campus of International Excellence ceiA3, Córdoba, Spain
| | - Eva Lozano
- Radiotherapy Unit, University Hospital of Ciudad Real, Spain
| | | | | | - Manuel De la Mata
- Instituto Maimónides de Investigación Biomédica en Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Córdoba, Spain
| | - Milica Pesic
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Serbia
| | - Juan Ramón Peinado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - José M Villalba
- Department of Cell Biology, Physiology and Immunology, Faculty of Sciences, University of Córdoba, Agrifood Campus of International Excellence ceiA3, Córdoba, Spain
| | | | - Víctor M Pérez-García
- Laboratory of Mathematical Oncology, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Francisco J Alcaín
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Mario Durán-Prado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain.
| |
Collapse
|
191
|
Liu X, Long X, Liu W, Zhao Y, Hayashi T, Yamato M, Mizuno K, Fujisaki H, Hattori S, Tashiro SI, Ogura T, Atsuzawa Y, Ikejima T. Type I collagen induces mesenchymal cell differentiation into myofibroblasts through YAP-induced TGF-β1 activation. Biochimie 2018; 150:110-130. [PMID: 29777737 DOI: 10.1016/j.biochi.2018.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022]
Abstract
In organ fibrosis, mechanical stress and transforming growth factor beta-1 (TGF-β1) promote differentiation into myofibroblast from mesenchymal cells, leading to extracellular matrix (ECM) remodeling or active synthesis, deposition or degradation of ECM components. A major component of ECM, type I collagen (col I) triple helical molecules assemble into fibrils or are denatured to gelatin without triple-helicity in remodeling. However, whether changes of ECM components in remodeling have influence on mesenchymal cell differentiation remains elusive. This study adopted three states of collagen I existing in ECM remodeling: molecular collagen, fibrillar collagen and gelatin to see what are characteristics in the effects on two cell lines of mesenchymal origin, murine 3T3-L1 embryonic fibroblast and murine C2C12 myoblasts. The results showed that all three forms of collagen I were capable of inducing these two cells to differentiate into myofibroblasts characterized by increased expression of alpha-smooth muscle actin (α-SMA) mRNA. The expression of α-SMA is positively regulated by TGF-β1. Nuclear translocation of Yes-associated protein (YAP) is involved in this process. Focal adhesion kinase (FAK) is activated in the cells cultured on molecular collagen-coated plates, contributing to YAP activation. On the other hand, in the cells cultured on fibrillar collagen gel or gelatin-coated plates, oxidative stress but not FAK induce YAP activation. In conclusion, the three physicochemically distinct forms of col I induce the differentiation of mesenchymal cells into myofibroblasts through different pathways.
Collapse
Affiliation(s)
- Xiaoling Liu
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xinyu Long
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Weiwei Liu
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yeli Zhao
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Toshihiko Hayashi
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo, 192-0015, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Shin-Ichi Tashiro
- Department of Medical Education and Primary Care, Kyoto Prefectural University of Medicine, Kyoto, 603-8072, Japan
| | - Takaaki Ogura
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Yuji Atsuzawa
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Takashi Ikejima
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
192
|
Cobley JN, Fiorello ML, Bailey DM. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol 2018; 15:490-503. [PMID: 29413961 PMCID: PMC5881419 DOI: 10.1016/j.redox.2018.01.008] [Citation(s) in RCA: 745] [Impact Index Per Article: 106.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
The human brain consumes 20% of the total basal oxygen (O2) budget to support ATP intensive neuronal activity. Without sufficient O2 to support ATP demands, neuronal activity fails, such that, even transient ischemia is neurodegenerative. While the essentiality of O2 to brain function is clear, how oxidative stress causes neurodegeneration is ambiguous. Ambiguity exists because many of the reasons why the brain is susceptible to oxidative stress remain obscure. Many are erroneously understood as the deleterious result of adventitious O2 derived free radical and non-radical species generation. To understand how many reasons underpin oxidative stress, one must first re-cast free radical and non-radical species in a positive light because their deliberate generation enables the brain to achieve critical functions (e.g. synaptic plasticity) through redox signalling (i.e. positive functionality). Using free radicals and non-radical derivatives to signal sensitises the brain to oxidative stress when redox signalling goes awry (i.e. negative functionality). To advance mechanistic understanding, we rationalise 13 reasons why the brain is susceptible to oxidative stress. Key reasons include inter alia unsaturated lipid enrichment, mitochondria, calcium, glutamate, modest antioxidant defence, redox active transition metals and neurotransmitter auto-oxidation. We review RNA oxidation as an underappreciated cause of oxidative stress. The complex interplay between each reason dictates neuronal susceptibility to oxidative stress in a dynamic context and neural identity dependent manner. Our discourse sets the stage for investigators to interrogate the biochemical basis of oxidative stress in the brain in health and disease.
Collapse
Affiliation(s)
- James Nathan Cobley
- Free Radical Laboratory, Departments of Diabetes and Cardiovascular Sciences, Centre for Health Sciences, University of the Highlands and Islands, Inverness IV2 3HJ, UK.
| | - Maria Luisa Fiorello
- Free Radical Laboratory, Departments of Diabetes and Cardiovascular Sciences, Centre for Health Sciences, University of the Highlands and Islands, Inverness IV2 3HJ, UK
| | - Damian Miles Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Wales, CF37 4AT, UK
| |
Collapse
|
193
|
Hang D, Nan H, Kværner AS, De Vivo I, Chan AT, Hu Z, Shen H, Giovannucci E, Song M. Longitudinal associations of lifetime adiposity with leukocyte telomere length and mitochondrial DNA copy number. Eur J Epidemiol 2018; 33:485-495. [PMID: 29619669 PMCID: PMC8063494 DOI: 10.1007/s10654-018-0382-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/20/2018] [Indexed: 12/12/2022]
Abstract
Adiposity may cause adverse health outcomes by increasing oxidative stress and systemic inflammation, which can be reflected by altered telomere length (TL) and mitochondrial DNA copy number (mtCN) in peripheral blood leukocytes. However, little is known about the influence of lifetime adiposity on TL and mtCN in later life. This study was performed to investigate the associations of lifetime adiposity with leukocyte TL and mtCN in 9613 participants from the Nurses' Health Study. A group-based trajectory modelling approach was used to create trajectories of body shape from age 5 through 60 years, and a genetic risk score (GRS) was created based on 97 known adiposity susceptibility variants. Associations of body shape trajectories and GRS with dichotomized TL and mtCN were assessed by logistic regression models. After adjustment for lifestyle and dietary factors, compared with the lean-stable group, the lean-marked increase group had higher odds of having below-median TL (OR = 1.18, 95% CI 1.04, 1.35; P = 0.01), and the medium-marked increase group had higher odds of having below-median mtCN (OR = 1.28, 95% CI 1.00, 1.64; P = 0.047). There was a suggestive trend toward lower mtCN across the GRS quartiles (P for trend = 0.07). In conclusion, telomere attrition may be accelerated by marked weight gain in middle life, whereas mtCN is likely to be reduced persistently by adiposity over the life course. The findings indicate the importance of lifetime weight management to preserve functional telomeres and mitochondria.
Collapse
Affiliation(s)
- Dong Hang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Room 371, Bldg. 2, 665 Huntington Avenue, Boston, MA, 02115, USA
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongmei Nan
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Ane Sørlie Kværner
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Room 371, Bldg. 2, 665 Huntington Avenue, Boston, MA, 02115, USA
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew Tan Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Room 906, 55 Fruit Street, Boston, MA, 02114, USA
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Edward Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Room 371, Bldg. 2, 665 Huntington Avenue, Boston, MA, 02115, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Room 371, Bldg. 2, 665 Huntington Avenue, Boston, MA, 02115, USA.
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Room 906, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
194
|
Fisher KM, McLeish JA, Jamieson LE, Jiang J, Hopgood JR, McLaughlin S, Donaldson K, Campbell CJ. SERS as a tool for in vitro toxicology. Faraday Discuss 2018; 187:501-20. [PMID: 27032696 DOI: 10.1039/c5fd00216h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Measuring markers of stress such as pH and redox potential are important when studying toxicology in in vitro models because they are markers of oxidative stress, apoptosis and viability. While surface enhanced Raman spectroscopy is ideally suited to the measurement of redox potential and pH in live cells, the time-intensive nature and perceived difficulty in signal analysis and interpretation can be a barrier to its broad uptake by the biological community. In this paper we detail the development of signal processing and analysis algorithms that allow SERS spectra to be automatically processed so that the output of the processing is a pH or redox potential value. By automating signal processing we were able to carry out a comparative evaluation of the toxicology of silver and zinc oxide nanoparticles and correlate our findings with qPCR analysis. The combination of these two analytical techniques sheds light on the differences in toxicology between these two materials from the perspective of oxidative stress.
Collapse
Affiliation(s)
- Kate M Fisher
- EaStCHEM, School of Chemistry, University of Edinburgh, EH9 3FJ, UK.
| | - Jennifer A McLeish
- MRC Centre for Inflammation Research, ELEGI Colt Laboratory, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ, UK
| | - Lauren E Jamieson
- EaStCHEM, School of Chemistry, University of Edinburgh, EH9 3FJ, UK.
| | - Jing Jiang
- EaStCHEM, School of Chemistry, University of Edinburgh, EH9 3FJ, UK.
| | - James R Hopgood
- Institute for Digital Communications, Joint Research Institute for Signal and Image Processing, School of Engineering, University of Edinburgh, EH9 3JL, UK
| | - Stephen McLaughlin
- School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Ken Donaldson
- MRC Centre for Inflammation Research, ELEGI Colt Laboratory, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ, UK
| | - Colin J Campbell
- EaStCHEM, School of Chemistry, University of Edinburgh, EH9 3FJ, UK.
| |
Collapse
|
195
|
Zhang X, Liu P, Zhang C, Chiewchengchol D, Zhao F, Yu H, Li J, Kambara H, Luo KY, Venkataraman A, Zhou Z, Zhou W, Zhu H, Zhao L, Sakai J, Chen Y, Ho YS, Bajrami B, Xu B, Silberstein LE, Cheng T, Xu Y, Ke Y, Luo HR. Positive Regulation of Interleukin-1β Bioactivity by Physiological ROS-Mediated Cysteine S-Glutathionylation. Cell Rep 2018; 20:224-235. [PMID: 28683316 DOI: 10.1016/j.celrep.2017.05.070] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/18/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
Reactive oxygen species (ROS)-induced cysteine S-glutathionylation is an important posttranslational modification (PTM) that controls a wide range of intracellular protein activities. However, whether physiological ROS can modulate the function of extracellular components via S-glutathionylation is unknown. Using a screening approach, we identified ROS-mediated cysteine S-glutathionylation on several extracellular cytokines. Glutathionylation of the highly conserved Cys-188 in IL-1β positively regulates its bioactivity by preventing its ROS-induced irreversible oxidation, including sulfinic acid and sulfonic acid formation. We show this mechanism protects IL-1β from deactivation by ROS in an in vivo system of irradiation-induced bone marrow (BM) injury. Glutaredoxin 1 (Grx1), an enzyme that catalyzes deglutathionylation, was present and active in the extracellular space in serum and the BM, physiologically regulating IL-1β glutathionylation and bioactivity. Collectively, we identify cysteine S-glutathionylation as a cytokine regulatory mechanism that could be a therapeutic target in the treatment of various infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Pathology and Pathophysiology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA.
| | - Peng Liu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Christie Zhang
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Direkrit Chiewchengchol
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Fan Zhao
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Hongbo Yu
- Hematopathology, Flow Cytometry, Hematology, and Blood Bank Labs, VA Boston Healthcare System, West Roxbury, MA 02132, USA; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 01605, USA
| | - Jingyu Li
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Hiroto Kambara
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Kate Y Luo
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Arvind Venkataraman
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Ziling Zhou
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Haiyan Zhu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Li Zhao
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Jiro Sakai
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Yuanyuan Chen
- Department of Pathology and Pathophysiology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences and Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, MI 48201, USA
| | - Besnik Bajrami
- Mass Spectrometry Unit, Waters Corporation, Milford, MA 01757, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street MS015, Waltham, MA 02454, USA
| | - Leslie E Silberstein
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Tao Cheng
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Yuanfu Xu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Yuehai Ke
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Hongbo R Luo
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Department of Lab Medicine, Children's Hospital Boston, Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA.
| |
Collapse
|
196
|
Pietraforte D, Paulicelli E, Patrono C, Gambardella L, Scorza G, Testa A, Fattibene P. Protein oxidative damage and redox imbalance induced by ionising radiation in CHO cells. Free Radic Res 2018; 52:465-479. [DOI: 10.1080/10715762.2018.1446529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Donatella Pietraforte
- Core Facilities, EPR Area, Italian Institute of Health, Rome, Italy
- Center for Gender-Specific Medicine, Biomarkers Unit, Italian Institute of Health, Rome, Italy
| | | | - Clarice Patrono
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Lucrezia Gambardella
- Center for Gender-Specific Medicine, Biomarkers Unit, Italian Institute of Health, Rome, Italy
| | - Giuseppe Scorza
- Core Facilities, EPR Area, Italian Institute of Health, Rome, Italy
| | - Antonella Testa
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Rome, Italy
| | - Paola Fattibene
- Core Facilities, EPR Area, Italian Institute of Health, Rome, Italy
| |
Collapse
|
197
|
van der Vliet A, Danyal K, Heppner DE. Dual oxidase: a novel therapeutic target in allergic disease. Br J Pharmacol 2018; 175:1401-1418. [PMID: 29405261 DOI: 10.1111/bph.14158] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 12/11/2022] Open
Abstract
NADPH oxidases (NOXs) represent a family of enzymes that mediate regulated cellular production of reactive oxygen species (ROS) and play various functional roles in physiology. Among the NOX family, the dual oxidases DUOX1 and DUOX2 are prominently expressed in epithelial cell types at mucosal surfaces and have therefore been considered to have important roles in innate host defence pathways. Recent studies have revealed important insights into the host defence mechanisms of DUOX enzymes, which control innate immune response pathways in response to either microbial or allergic triggers. In this review, we discuss the current level of understanding with respect to the biological role(s) of DUOX enzymes and the unique role of DUOX1 in mediating innate immune responses to epithelial injury and allergens and in the development of allergic disease. These novel findings highlight DUOX1 as an attractive therapeutic target, and opportunities for the development of selective inhibitor strategies will be discussed.
Collapse
Affiliation(s)
- Albert van der Vliet
- Department of Pathology and Laboratory Medicine, The Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, USA.,Vermont Lung Center, University of Vermont, Burlington, VT, USA
| | - Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, The Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, USA.,Vermont Lung Center, 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
| |
Collapse
|
198
|
Chittiboyina S, Bai Y, Lelièvre SA. Microenvironment-Cell Nucleus Relationship in the Context of Oxidative Stress. Front Cell Dev Biol 2018; 6:23. [PMID: 29594114 PMCID: PMC5854663 DOI: 10.3389/fcell.2018.00023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/21/2018] [Indexed: 12/17/2022] Open
Abstract
The microenvironment is a source of reactive oxygen species (ROS) that influence cell phenotype and tissue homeostasis. The impact of ROS on redox pathways as well as directly on epigenetic mechanisms and the DNA illustrate communication with the cell nucleus. Changes in gene transcription related to redox conditions also influence the content and structure of the extracellular matrix. However, the importance of microenvironmental ROS for normal progression through life and disease development still needs to be thoroughly understood. We illustrate how different ROS concentration levels trigger various intracellular pathways linked to nuclear functions and determine processes necessary for the differentiation of stem cells. The abnormal predominance of ROS that leads to oxidative stress is emphasized in light of its impact on aging and diseases related to aging. These phenomena are discussed in the context of the possible contribution of extracellular ROS via direct diffusion into cells responsible for organ function, but also via an impact on stromal cells that triggers extracellular modifications and influences mechanotransduction. Finally, we argue that organs-on-a-chip with controlled microenvironmental conditions can help thoroughly grasp whether ROS production is readily a cause or a consequence of certain disorders, and better understand the concentration levels of extracellular ROS that are necessary to induce a switch in phenotype.
Collapse
Affiliation(s)
- Shirisha Chittiboyina
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
- 3D Cell Culture Core, Birck Nanotechnology Center, Purdue University Discovery Park, West Lafayette, IN, United States
| | - Yunfeng Bai
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Sophie A. Lelièvre
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
- 3D Cell Culture Core, Birck Nanotechnology Center, Purdue University Discovery Park, West Lafayette, IN, United States
- Center for Cancer Research, West Lafayette, IN, United States
| |
Collapse
|
199
|
Abstract
Hydrogen peroxide (H2O2) is produced on stimulation of many cell surface receptors and serves as an intracellular messenger in the regulation of diverse physiological events, mostly by oxidizing cysteine residues of effector proteins. Mammalian cells express multiple H2O2-eliminating enzymes, including catalase, glutathione peroxidase (GPx), and peroxiredoxin (Prx). A conserved cysteine in Prx family members is the site of oxidation by H2O2. Peroxiredoxins possess a high-affinity binding site for H2O2 that is lacking in catalase and GPx and which renders the catalytic cysteine highly susceptible to oxidation, with a rate constant several orders of magnitude greater than that for oxidation of cysteine in most H2O2 effector proteins. Moreover, Prxs are abundant and present in all subcellular compartments. The cysteines of most H2O2 effectors are therefore at a competitive disadvantage for reaction with H2O2. Recent Advances: Here we review intracellular sources of H2O2 as well as H2O2 target proteins classified according to biochemical and cellular function. We then highlight two strategies implemented by cells to overcome the kinetic disadvantage of most target proteins with regard to H2O2-mediated oxidation: transient inactivation of local Prx molecules via phosphorylation, and indirect oxidation of target cysteines via oxidized Prx. Critical Issues and Future Directions: Recent studies suggest that only a small fraction of the total pools of Prxs and H2O2 effector proteins localized in specific subcellular compartments participates in H2O2 signaling. Development of sensitive tools to selectively detect phosphorylated Prxs and oxidized effector proteins is needed to provide further insight into H2O2 signaling. Antioxid. Redox Signal. 28, 537-557.
Collapse
Affiliation(s)
- Sue Goo Rhee
- 1 Yonsei Biomedical Research Institute, Yonsei University College of Medicine , Seoul, Korea
| | - Hyun Ae Woo
- 2 College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul, Korea
| | - Dongmin Kang
- 3 Department of Life Science, Ewha Womans University , Seoul, Korea
| |
Collapse
|
200
|
A pivotal role for NF-κB in the macrophage inflammatory response to the myeloperoxidase oxidant hypothiocyanous acid. Arch Biochem Biophys 2018; 642:23-30. [DOI: 10.1016/j.abb.2018.01.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/04/2018] [Accepted: 01/29/2018] [Indexed: 01/04/2023]
|