1
|
Nagarkoti S, Kim YM, Ash D, Das A, Vitriol E, Read TA, Youn SW, Sudhahar V, McMenamin M, Hou Y, Boatwright H, Caldwell R, Essex DW, Cho J, Fukai T, Ushio-Fukai M. Protein disulfide isomerase A1 as a novel redox sensor in VEGFR2 signaling and angiogenesis. Angiogenesis 2023; 26:77-96. [PMID: 35984546 PMCID: PMC9918675 DOI: 10.1007/s10456-022-09852-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 07/26/2022] [Indexed: 02/04/2023]
Abstract
VEGFR2 signaling in endothelial cells (ECs) is regulated by reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria, which plays an important role in postnatal angiogenesis. However, it remains unclear how highly diffusible ROS signal enhances VEGFR2 signaling and reparative angiogenesis. Protein disulfide isomerase A1 (PDIA1) functions as an oxidoreductase depending on the redox environment. We hypothesized that PDIA1 functions as a redox sensor to enhance angiogenesis. Here we showed that PDIA1 co-immunoprecipitated with VEGFR2 or colocalized with either VEGFR2 or an early endosome marker Rab5 at the perinuclear region upon stimulation of human ECs with VEGF. PDIA1 silencing significantly reduced VEGF-induced EC migration, proliferation and spheroid sprouting via inhibiting VEGFR2 signaling. Mechanistically, VEGF stimulation rapidly increased Cys-OH formation of PDIA1 via the NOX4-mitochondrial ROS axis. Overexpression of "redox-dead" mutant PDIA1 with replacement of the active four Cys residues with Ser significantly inhibited VEGF-induced PDIA1-CysOH formation and angiogenic responses via reducing VEGFR2 phosphorylation. Pdia1+/- mice showed impaired angiogenesis in developmental retina and Matrigel plug models as well as ex vivo aortic ring sprouting model. Study using hindlimb ischemia model revealed that PDIA1 expression was markedly increased in angiogenic ECs of ischemic muscles, and that ischemia-induced limb perfusion recovery and neovascularization were impaired in EC-specific Pdia1 conditional knockout mice. These results suggest that PDIA1 can sense VEGF-induced H2O2 signal via CysOH formation to promote VEGFR2 signaling and angiogenesis in ECs, thereby enhancing postnatal angiogenesis. The oxidized PDIA1 is a potential therapeutic target for treatment of ischemic vascular diseases.
Collapse
Affiliation(s)
- Sheela Nagarkoti
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Young-Mee Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Department of Medicine (Cardiology), University of Illinois at Chicago, Chicago, IL, USA
| | - Dipankar Ash
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Archita Das
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Eric Vitriol
- Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Tracy-Ann Read
- Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Seock-Won Youn
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Varadarajan Sudhahar
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Malgorzata McMenamin
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Yali Hou
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Harriet Boatwright
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
| | - Ruth Caldwell
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Vision Discovery Institute, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - David W Essex
- Department of Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA
- Departments of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney-Walker Blvd, Augusta, GA, 30912, USA.
- Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
2
|
Mahmood F, Xu R, Awan MUN, Song Y, Han Q, Xia X, Zhang J. PDIA3: Structure, functions and its potential role in viral infections. Biomed Pharmacother 2021; 143:112110. [PMID: 34474345 DOI: 10.1016/j.biopha.2021.112110] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
The catalysis of disulphide (SS) bonds is the most important characteristic of protein disulphide isomerase (PDI) family. Catalysis occurs in the endoplasmic reticulum, which contains many proteins, most of which are secretory in nature and that have at least one s-s bond. Protein disulphide isomerase A3 (PDIA3) is a member of the PDI family that acts as a chaperone. PDIA3 is highly expressed in response to cellular stress, and also intercept the apoptotic cellular death related to endoplasmic reticulum (ER) stress, and protein misfolding. PDIA3 expression is elevated in almost 70% of cancers and its expression has been linked with overall low cell invasiveness, survival and metastasis. Viral diseases present a significant public health threat. The presence of PDIA3 on the cell surface helps different viruses to enter the cells and also helps in replication. Therefore, inhibitors of PDIA3 have great potential to interfere with viral infections. In this review, we summarize what is known about the basic structure, functions and role of PDIA3 in viral infections. The review will inspire studies of pathogenic mechanisms and drug targeting to counter viral diseases.
Collapse
Affiliation(s)
- Faisal Mahmood
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Ruixian Xu
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Maher Un Nisa Awan
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Yuzhu Song
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Qinqin Han
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China
| | - Xueshan Xia
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China.
| | - Jinyang Zhang
- Molecular Medicine Research Centre of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 Jingming South Road, Kunming 650500, China.
| |
Collapse
|
3
|
Daiber A, Chlopicki S. Revisiting pharmacology of oxidative stress and endothelial dysfunction in cardiovascular disease: Evidence for redox-based therapies. Free Radic Biol Med 2020; 157:15-37. [PMID: 32131026 DOI: 10.1016/j.freeradbiomed.2020.02.026] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/05/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
According to the latest Global Burden of Disease Study data, non-communicable diseases in general and cardiovascular disease (CVD) in particular are the leading cause of premature death and reduced quality of life. Demographic shifts, unhealthy lifestyles and a higher burden of adverse environmental factors provide an explanation for these findings. The expected growing prevalence of CVD requires enhanced research efforts for identification and characterisation of novel therapeutic targets and strategies. Cardiovascular risk factors including classical (e.g. hypertension, diabetes, hypercholesterolaemia) and non-classical (e.g. environmental stress) factors induce the development of endothelial dysfunction, which is closely associated with oxidant stress and vascular inflammation and results in CVD, particularly in older adults. Most classically successful therapies for CVD display vasoprotective, antioxidant and anti-inflammatory effects, but were originally designed with other therapeutic aims. So far, only a few 'redox drugs' are in clinical use and many antioxidant strategies have not met expectations. With the present review, we summarise the actual knowledge on CVD pathomechanisms, with special emphasis on endothelial dysfunction, adverse redox signalling and oxidative stress, highlighting the preclinical and clinical evidence. In addition, we provide a brief overview of established CVD therapies and their relation to endothelial dysfunction and oxidative stress. Finally, we discuss novel strategies for redox-based CVD therapies trying to explain why, despite a clear link between endothelial dysfunction and adverse redox signalling and oxidative stress, redox- and oxidative stress-based therapies have not yet provided a breakthrough in the treatment of endothelial dysfunction and CVD.
Collapse
Affiliation(s)
- Andreas Daiber
- The Center for Cardiology, Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; The Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Stefan Chlopicki
- The Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland; Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland.
| |
Collapse
|
4
|
Shergalis AG, Hu S, Bankhead A, Neamati N. Role of the ERO1-PDI interaction in oxidative protein folding and disease. Pharmacol Ther 2020; 210:107525. [PMID: 32201313 DOI: 10.1016/j.pharmthera.2020.107525] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/04/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023]
Abstract
Protein folding in the endoplasmic reticulum is an oxidative process that relies on protein disulfide isomerase (PDI) and endoplasmic reticulum oxidase 1 (ERO1). Over 30% of proteins require the chaperone PDI to promote disulfide bond formation. PDI oxidizes cysteines in nascent polypeptides to form disulfide bonds and can also reduce and isomerize disulfide bonds. ERO1 recycles reduced PDI family member PDIA1 using a FAD cofactor to transfer electrons to oxygen. ERO1 dysfunction critically affects several diseases states. Both ERO1 and PDIA1 are overexpressed in cancers and implicated in diabetes and neurodegenerative diseases. Cancer-associated ERO1 promotes cell migration and invasion. Furthermore, the ERO1-PDIA1 interaction is critical for epithelial-to-mesenchymal transition. Co-expression analysis of ERO1A gene expression in cancer patients demonstrated that ERO1A is significantly upregulated in lung adenocarcinoma (LUAD), glioblastoma and low-grade glioma (GBMLGG), pancreatic ductal adenocarcinoma (PAAD), and kidney renal papillary cell carcinoma (KIRP) cancers. ERO1Α knockdown gene signature correlates with knockdown of cancer signaling proteins including IGF1R, supporting the search for novel, selective ERO1 inhibitors for the treatment of cancer. In this review, we explore the functions of ERO1 and PDI to support inhibition of this interaction in cancer and other diseases.
Collapse
Affiliation(s)
- Andrea G Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, United States
| | - Shuai Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, United States; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Armand Bankhead
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, United States.
| |
Collapse
|
5
|
Fuentes E, Moore-Carrasco R, de Andrade Paes AM, Trostchansky A. Role of Platelet Activation and Oxidative Stress in the Evolution of Myocardial Infarction. J Cardiovasc Pharmacol Ther 2019; 24:509-520. [DOI: 10.1177/1074248419861437] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myocardial infarction, commonly known as heart attack, evolves from the rupture of unstable atherosclerotic plaques to coronary thrombosis and myocardial ischemia–reperfusion injury. A body of evidence supports a close relationship between the alterations following an ischemia–reperfusion injury-induced oxidative stress and platelet activity. Through their critical role in thrombogenesis and inflammatory responses, platelets are fully (totally) implicated from atherothrombotic plaque formation to myocardial infarction onset and expansion. However, mere platelet aggregation prevention does not offer full protection, suggesting that other antiplatelet therapy mechanisms may also be involved. Thus, the present review discusses the integrative role of platelets, oxidative stress, and antiplatelet therapy in triggering myocardial infarction pathophysiology.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca, Chile
| | - Rodrigo Moore-Carrasco
- Departamento de Bioquímica Clínica e Inmunohematología, Facultad de Ciencias de la Salud, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca, Chile
| | - Antonio Marcus de Andrade Paes
- Laboratory of Experimental Physiology, Health Sciences Graduate Program and Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Andres Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
6
|
Gonzalez-Perilli L, Prolo C, Álvarez MN. Arachidonic Acid and Nitroarachidonic: Effects on NADPH Oxidase Activity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1127:85-95. [PMID: 31140173 DOI: 10.1007/978-3-030-11488-6_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Arachidonic acid (AA) is a polyunsaturated fatty acid that participates in the inflammatory response mainly through bioactive-lipids formation in macrophages and also in the phagocytic NADPH oxidase 2 (NOX2) activation. NOX2 is the enzyme responsible for a huge superoxide formation in macrophages, essential to eliminate pathogens inside the phagosome. The oxidase is an enzymatic complex comprised of a membrane-bound flavocytochrome b 558 (gp91phox/p22phox), three cytosolic subunits (p47phox, p40phox and p67phox) and a Rac-GTPase. The enzyme becomes active when macrophages are exposed to appropriate stimuli that trigger the phosphorylation of cytosolic subunits and its migration to plasmatic membrane to form the active complex. It is proposed that AA stimulates NOX2 activity through AA interaction with different components of the NADPH oxidase complex. In inflammatory conditions, there is an increase in reactive oxygen and nitrogen species that results in the production of nitrated derivatives of AA, such as nitroarachidonic acid (NO2-AA). NO2-AA is capable to inhibit NOX2 activity by interfering with p47phox migration to the membrane without affecting phosphorylation of cytosolic proteins. Also, NO2-AA is capable to interact with protein disulfide isomerase (PDI), which is involved on NOX2 active complex formation. It has been demonstrated that NO2-AA forms a covalent adduct with PDI that could prevent the interaction with NOX2 and it would explain the inhibitory effects of the fatty acid upon NOX2. Together, current data indicate that AA is an important activator of NOX2 formed in the early events of the inflammatory response, leading to a massive production of oxidants that may, in turn, promote NO2-AA formation and shutting down the oxidative burst. Hence, AA and its derivatives could have antagonistic roles on NOX2 activity regulation.
Collapse
Affiliation(s)
- Lucía Gonzalez-Perilli
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay
| | - Carolina Prolo
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay
| | - María Noel Álvarez
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
7
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 PMCID: PMC6442925 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/30/2018] [Accepted: 05/06/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
8
|
Fradin T, Bechor E, Berdichevsky Y, Dahan I, Pick E. Binding of p67phoxto Nox2 is stabilized by disulfide bonds between cysteines in the369Cys-Gly-Cys371triad in Nox2 and in p67phox. J Leukoc Biol 2018; 104:1023-1039. [DOI: 10.1002/jlb.4a0418-173r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/09/2018] [Accepted: 06/23/2018] [Indexed: 01/05/2023] Open
Affiliation(s)
- Tanya Fradin
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Edna Bechor
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Yevgeny Berdichevsky
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Iris Dahan
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Edgar Pick
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| |
Collapse
|
9
|
Elmasry K, Ibrahim AS, Saleh H, Elsherbiny N, Elshafey S, Hussein KA, Al-Shabrawey M. Role of endoplasmic reticulum stress in 12/15-lipoxygenase-induced retinal microvascular dysfunction in a mouse model of diabetic retinopathy. Diabetologia 2018; 61:1220-1232. [PMID: 29468369 PMCID: PMC5878142 DOI: 10.1007/s00125-018-4560-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 12/20/2017] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Our earlier studies have established the role of 12/15-lipoxygenase (LO) in mediating the inflammatory reaction in diabetic retinopathy. However, the exact mechanism is still unclear. The goal of the current study was to identify the potential role of endoplasmic reticulum (ER) stress as a major cellular stress response in the 12/15-LO-induced retinal changes in diabetic retinopathy. METHODS We used in vivo and in vitro approaches. For in vivo studies, experimental diabetes was induced in wild-type (WT) mice and 12/15-Lo (also known as Alox15) knockout mice (12/15-Lo-/-); ER stress was then evaluated after 12-14 weeks of diabetes. We also tested the effect of intravitreal injection of 12-hydroxyeicosatetraenoic acid (HETE) on retinal ER stress in WT mice and in mice lacking the catalytic subunit of NADPH oxidase, encoded by Nox2 (also known as Cybb) (Nox2-/- mice). In vitro studies were performed using human retinal endothelial cells (HRECs) treated with 15-HETE (0.1 μmol/l) or vehicle, with or without ER stress or NADPH oxidase inhibitors. This was followed by evaluation of ER stress response, NADPH oxidase expression/activity and the levels of phosphorylated vascular endothelial growth factor receptor-2 (p-VEGFR2) by western blotting and immunoprecipitation assays. Moreover, real-time imaging of intracellular calcium (Ca2+) release in HRECs treated with or without 15-HETE was performed using confocal microscopy. RESULTS Deletion of 12/15-Lo significantly attenuated diabetes-induced ER stress in mouse retina. In vitro, 15-HETE upregulated ER stress markers such as phosphorylated RNA-dependent protein kinase-like ER-regulated kinase (p-PERK), activating transcription factor 6 (ATF6) and protein disulfide isomerase (PDI) in HRECs. Inhibition of ER stress reduced 15-HETE-induced-leucocyte adhesion, VEGFR2 phosphorylation and NADPH oxidase expression/activity. However, inhibition of NADPH oxidase or deletion of Nox2 had no effect on ER stress induced by the 12/15-LO-derived metabolites both in vitro and in vivo. We also found that 15-HETE increases the intracellular calcium in HRECs. CONCLUSIONS/INTERPRETATION ER stress contributes to 12/15-LO-induced retinal inflammation in diabetic retinopathy via activation of NADPH oxidase and VEGFR2. Perturbation of calcium homeostasis in the retina might also play a role in linking 12/15-LO to retinal ER stress and subsequent microvascular dysfunction in diabetic retinopathy.
Collapse
Affiliation(s)
- Khaled Elmasry
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB 2602, Augusta, GA, 30912, USA
- Department of Ophthalmology and Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed S Ibrahim
- Department of Ophthalmology and Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Heba Saleh
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Nehal Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Sally Elshafey
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Khaled A Hussein
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB 2602, Augusta, GA, 30912, USA
- Oral Medicine and Surgery Research Division, National Research Centre, Dokki, Egypt
| | - Mohamed Al-Shabrawey
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB 2602, Augusta, GA, 30912, USA.
- Department of Ophthalmology and Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
10
|
Morris G, Puri BK, Walder K, Berk M, Stubbs B, Maes M, Carvalho AF. The Endoplasmic Reticulum Stress Response in Neuroprogressive Diseases: Emerging Pathophysiological Role and Translational Implications. Mol Neurobiol 2018; 55:8765-8787. [PMID: 29594942 PMCID: PMC6208857 DOI: 10.1007/s12035-018-1028-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/20/2018] [Indexed: 02/07/2023]
Abstract
The endoplasmic reticulum (ER) is the main cellular organelle involved in protein synthesis, assembly and secretion. Accumulating evidence shows that across several neurodegenerative and neuroprogressive diseases, ER stress ensues, which is accompanied by over-activation of the unfolded protein response (UPR). Although the UPR could initially serve adaptive purposes in conditions associated with higher cellular demands and after exposure to a range of pathophysiological insults, over time the UPR may become detrimental, thus contributing to neuroprogression. Herein, we propose that immune-inflammatory, neuro-oxidative, neuro-nitrosative, as well as mitochondrial pathways may reciprocally interact with aberrations in UPR pathways. Furthermore, ER stress may contribute to a deregulation in calcium homoeostasis. The common denominator of these pathways is a decrease in neuronal resilience, synaptic dysfunction and even cell death. This review also discusses how mechanisms related to ER stress could be explored as a source for novel therapeutic targets for neurodegenerative and neuroprogressive diseases. The design of randomised controlled trials testing compounds that target aberrant UPR-related pathways within the emerging framework of precision psychiatry is warranted.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, Wales, SA15 2LW, UK
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, England, W12 0HS, UK.
| | - Ken Walder
- The Centre for Molecular and Medical Research, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
- Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Florey Institute for Neuroscience and Mental Health, Melbourne, Australia
| | - Brendon Stubbs
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, UK
- Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Faculty of Health, Social Care and Education, Anglia Ruskin University, Chelmsford, UK
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - André F Carvalho
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| |
Collapse
|
11
|
Cole KS, Grandjean JMD, Chen K, Witt CH, O'Day J, Shoulders MD, Wiseman RL, Weerapana E. Characterization of an A-Site Selective Protein Disulfide Isomerase A1 Inhibitor. Biochemistry 2018. [PMID: 29521097 DOI: 10.1021/acs.biochem.8b00178] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Protein disulfide isomerase A1 (PDIA1) is an endoplasmic reticulum (ER)-localized thiol-disulfide oxidoreductase that is an important folding catalyst for secretory pathway proteins. PDIA1 contains two active-site domains (a and a'), each containing a Cys-Gly-His-Cys (CGHC) active-site motif. The two active-site domains share 37% sequence identity and function independently to perform disulfide-bond reduction, oxidation, and isomerization. Numerous inhibitors for PDIA1 have been reported, yet the selectivity of these inhibitors toward the a and a' sites is poorly characterized. Here, we identify a potent and selective PDIA1 inhibitor, KSC-34, with 30-fold selectivity for the a site over the a' site. KSC-34 displays time-dependent inhibition of PDIA1 reductase activity in vitro with a kinact/ KI of 9.66 × 103 M-1 s-1 and is selective for PDIA1 over other members of the PDI family, and other cellular cysteine-containing proteins. We provide the first cellular characterization of an a-site selective PDIA1 inhibitor and demonstrate that KSC-34 has minimal sustained effects on the cellular unfolded protein response, indicating that a-site inhibition does not induce global protein folding-associated ER stress. KSC-34 treatment significantly decreases the rate of secretion of a destabilized, amyloidogenic antibody light chain, thereby minimizing pathogenic amyloidogenic extracellular proteins that rely on high PDIA1 activity for proper folding and secretion. Given the poor understanding of the contribution of each PDIA1 active site to the (patho)physiological functions of PDIA1, site selective inhibitors like KSC-34 provide useful tools for delineating the pathological role and therapeutic potential of PDIA1.
Collapse
Affiliation(s)
- Kyle S Cole
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| | - Julia M D Grandjean
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Kenny Chen
- Department of Chemistry , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Collin H Witt
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| | - Johanna O'Day
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| | - Matthew D Shoulders
- Department of Chemistry , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - R Luke Wiseman
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Eranthie Weerapana
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| |
Collapse
|
12
|
Shih YC, Chen CL, Zhang Y, Mellor RL, Kanter EM, Fang Y, Wang HC, Hung CT, Nong JY, Chen HJ, Lee TH, Tseng YS, Chen CN, Wu CC, Lin SL, Yamada KA, Nerbonne JM, Yang KC. Endoplasmic Reticulum Protein TXNDC5 Augments Myocardial Fibrosis by Facilitating Extracellular Matrix Protein Folding and Redox-Sensitive Cardiac Fibroblast Activation. Circ Res 2018. [PMID: 29535165 DOI: 10.1161/circresaha.117.312130] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
RATIONALE Cardiac fibrosis plays a critical role in the pathogenesis of heart failure. Excessive accumulation of extracellular matrix (ECM) resulting from cardiac fibrosis impairs cardiac contractile function and increases arrhythmogenicity. Current treatment options for cardiac fibrosis, however, are limited, and there is a clear need to identify novel mediators of cardiac fibrosis to facilitate the development of better therapeutics. Exploiting coexpression gene network analysis on RNA sequencing data from failing human heart, we identified TXNDC5 (thioredoxin domain containing 5), a cardiac fibroblast (CF)-enriched endoplasmic reticulum protein, as a potential novel mediator of cardiac fibrosis, and we completed experiments to test this hypothesis directly. OBJECTIVE The objective of this study was to determine the functional role of TXNDC5 in the pathogenesis of cardiac fibrosis. METHODS AND RESULTS RNA sequencing and Western blot analyses revealed that TXNDC5 mRNA and protein were highly upregulated in failing human left ventricles and in hypertrophied/failing mouse left ventricle. In addition, cardiac TXNDC5 mRNA expression levels were positively correlated with those of transcripts encoding transforming growth factor β1 and ECM proteins in vivo. TXNDC5 mRNA and protein were increased in human CF (hCF) under transforming growth factor β1 stimulation in vitro. Knockdown of TXNDC5 attenuated transforming growth factor β1-induced hCF activation and ECM protein upregulation independent of SMAD3 (SMAD family member 3), whereas increasing expression of TXNDC5 triggered hCF activation and proliferation and increased ECM protein production. Further experiments showed that TXNDC5, a protein disulfide isomerase, facilitated ECM protein folding and that depletion of TXNDC5 led to ECM protein misfolding and degradation in CF. In addition, TXNDC5 promotes hCF activation and proliferation by enhancing c-Jun N-terminal kinase activity via increased reactive oxygen species, derived from NAD(P)H oxidase 4. Transforming growth factor β1-induced TXNDC5 upregulation in hCF was dependent on endoplasmic reticulum stress and activating transcription factor 6-mediated transcriptional control. Targeted disruption of Txndc5 in mice (Txndc5-/-) revealed protective effects against isoproterenol-induced cardiac hypertrophy, reduced fibrosis (by ≈70%), and markedly improved left ventricle function; post-isoproterenol left ventricular ejection fraction was 59.1±1.5 versus 40.1±2.5 (P<0.001) in Txndc5-/- versus wild-type mice, respectively. CONCLUSIONS The endoplasmic reticulum protein TXNDC5 promotes cardiac fibrosis by facilitating ECM protein folding and CF activation via redox-sensitive c-Jun N-terminal kinase signaling. Loss of TXNDC5 protects against β agonist-induced cardiac fibrosis and contractile dysfunction. Targeting TXNDC5, therefore, could be a powerful new therapeutic approach to mitigate excessive cardiac fibrosis, thereby improving cardiac function and outcomes in patients with heart failure.
Collapse
Affiliation(s)
- Ying-Chun Shih
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chao-Ling Chen
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Yan Zhang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Rebecca L Mellor
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Evelyn M Kanter
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Yun Fang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Hua-Chi Wang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chen-Ting Hung
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Jing-Yi Nong
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Hui-Ju Chen
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Tzu-Han Lee
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Yi-Shuan Tseng
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chiung-Nien Chen
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chau-Chung Wu
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Shuei-Liong Lin
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Kathryn A Yamada
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Jeanne M Nerbonne
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Kai-Chien Yang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei.
| |
Collapse
|
13
|
Wong KHK, Cai Y, Ying F, Chen X, Vanhoutte PM, Tang EHC. Deletion of Rap1 disrupts redox balance and impairs endothelium-dependent relaxations. J Mol Cell Cardiol 2018; 115:1-9. [PMID: 29277598 DOI: 10.1016/j.yjmcc.2017.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 01/29/2023]
Abstract
AIMS Repressor activator protein 1 (Rap1) is conventionally known as a static structural component of the telomere, but recent evidence indicates that it exerts functions within and outside the nucleus taking part in metabolic regulation and promoting inflammatory responses. The present study investigated whether or not Rap1 deletion affects oxidative stress and nitric oxide (NO) bioavailability in the vascular wall, thus modulating endothelial function. METHODS AND RESULTS Vascular responsiveness was studied in wire myographs in aortae from Rap1 wildtype and knockout mice. Deletion of Rap1 impaired endothelium-dependent relaxations elicited by acetylcholine. Rap1 deficiency did not affect the activation of endothelial NO synthase or the sensitivity of vascular smooth muscle to NO donors. The blunted acetylcholine-mediated relaxations in Rap1 deficient aortae were restored with nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors, apocynin or VAS2870. Rap1 deletion lowered cellular thiol-redox status and diminished activities of thiol-redox enzymes, thioredoxin 1 and glutaredoxin 1. CONCLUSIONS The capacity of thioredoxin 1 and glutaredoxin 1 to reduce intra-protein disulfide bridges is weakened in Rap1 deficient mice, resulting in hyper-activation of NADPH oxidase and greater reactive oxygen species generation. The high oxidative stress in Rap1 deficient mice is implicated with greater oxidative breakdown of NO, explaining the blunted acetylcholine-mediated relaxations in this animal. These findings imply that Rap1 plays an unanticipated role in regulating the fate of NO (a pivotal determinant of vascular homeostasis) and thus identify a new physiological importance of the telomere-associated protein.
Collapse
Affiliation(s)
- Kenneth H K Wong
- Department of Pharmacology and Pharmacy, State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yin Cai
- Department of Anaethesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Fan Ying
- Department of Pharmacology and Pharmacy, State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xinyi Chen
- Department of Pharmacology and Pharmacy, State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Paul M Vanhoutte
- Department of Pharmacology and Pharmacy, State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Eva H C Tang
- Department of Pharmacology and Pharmacy, State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; School of Biomedical Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Barra NG, Lisyansky M, Vanduzer TA, Raha S, Holloway AC, Hardy DB. Maternal nicotine exposure leads to decreased cardiac protein disulfide isomerase and impaired mitochondrial function in male rat offspring. J Appl Toxicol 2017; 37:1517-1526. [PMID: 28681937 DOI: 10.1002/jat.3503] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 11/08/2022]
Abstract
Smoking throughout pregnancy can lead to complications during gestation, parturition and neonatal development. Thus, nicotine replacement therapies are a popular alternative thought to be safer than cigarettes. However, recent studies in rodents suggest that fetal and neonatal nicotine exposure alone results in cardiac dysfunction and high blood pressure. While it is well known that perinatal nicotine exposure causes increased congenital abnormalities, the mechanisms underlying longer-term deficits in cardiac function are not completely understood. Recently, our laboratory demonstrated that nicotine impairs placental protein disulfide isomerase (PDI) triggering an increase in endoplasmic reticulum stress, leading us to hypothesize that this may also occur in the heart. At 3 months of age, nicotine-exposed offspring had 45% decreased PDI levels in the absence of endoplasmic reticulum stress. Given the association of PDI and superoxide dismutase enzymes, we further observed that antioxidant superoxide dismutase-2 levels were reduced by 32% in these offspring concomitant with a 26-49% decrease in mitochondrial complex proteins (I, II, IV and V) and tissue inhibitor of metalloproteinase-4, a critical matrix metalloprotease for cardiac contractility and health. Collectively, this study suggests that perinatal nicotine exposure decreases PDI, which can promote oxidative damage and mitochondrial damage, associated with a premature decline in cardiac function.
Collapse
Affiliation(s)
- Nicole G Barra
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Maria Lisyansky
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Taylor A Vanduzer
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Sandeep Raha
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Alison C Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Daniel B Hardy
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.,Departments of Obstetrics and Gynecology, Children's Health Research Institute, Lawson, Health Research Institute, Western University, London, Ontario, Canada
| |
Collapse
|
15
|
Daiber A, Di Lisa F, Oelze M, Kröller‐Schön S, Steven S, Schulz E, Münzel T. Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol 2017; 174:1670-1689. [PMID: 26660451 PMCID: PMC5446573 DOI: 10.1111/bph.13403] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are associated with and/or caused by oxidative stress. This concept has been proven by using the approach of genetic deletion of reactive species producing (pro-oxidant) enzymes as well as by the overexpression of reactive species detoxifying (antioxidant) enzymes leading to a marked reduction of reactive oxygen and nitrogen species (RONS) and in parallel to an amelioration of the severity of diseases. Likewise, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of antioxidant RONS detoxifying enzymes. Thus, the consequences of the interaction (redox crosstalk) of superoxide/hydrogen peroxide produced by mitochondria with other ROS producing enzymes such as NADPH oxidases (Nox) are of outstanding importance and will be discussed including the consequences for endothelial nitric oxide synthase (eNOS) uncoupling as well as the redox regulation of the vascular function/tone in general (soluble guanylyl cyclase, endothelin-1, prostanoid synthesis). Pathways and potential mechanisms leading to this crosstalk will be analysed in detail and highlighted by selected examples from the current literature including hypoxia, angiotensin II-induced hypertension, nitrate tolerance, aging and others. The general concept of redox-based activation of RONS sources via "kindling radicals" and enzyme-specific "redox switches" will be discussed providing evidence that mitochondria represent key players and amplifiers of the burden of oxidative stress. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Fabio Di Lisa
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Matthias Oelze
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Swenja Kröller‐Schön
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Sebastian Steven
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
- Center of Thrombosis and HemostasisMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Eberhard Schulz
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| | - Thomas Münzel
- Center for Cardiology, Laboratory of Molecular CardiologyMedical Center of the Johannes Gutenberg UniversityMainzGermany
| |
Collapse
|
16
|
Marschall R, Tudzynski P. The Protein Disulfide Isomerase of Botrytis cinerea: An ER Protein Involved in Protein Folding and Redox Homeostasis Influences NADPH Oxidase Signaling Processes. Front Microbiol 2017; 8:960. [PMID: 28611757 PMCID: PMC5447010 DOI: 10.3389/fmicb.2017.00960] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/15/2017] [Indexed: 11/13/2022] Open
Abstract
Botrytis cinerea is a filamentous plant pathogen, which infects hundreds of plant species; within its lifestyle, the production of reactive oxygen species (ROS) and a balanced redox homeostasis are essential parameters. The pathogen is capable of coping with the plant’s oxidative burst and even produces its own ROS to enhance the plant’s oxidative burst. Highly conserved NADPH oxidase (Nox) complexes produce the reactive molecules. The membrane-associated complexes regulate a large variety of vegetative and pathogenic processes. Besides their commonly accepted function at the plasma membrane, recent studies reveal that Nox complexes are also active at the membrane of the endoplasmic reticulum. In this study, we identified the essential ER protein BcPdi1 as new interaction partner of the NoxA complex in B. cinerea. Mutants that lack this ER chaperone display overlapping phenotypes to mutants of the NoxA signaling pathway. The protein appears to be involved in all major developmental processes, such as the formation of sclerotia, conidial anastomosis tubes and infection cushions (IC’s) and is needed for full virulence. Moreover, expression analyses and reporter gene studies indicate that BcPdi1 affects the redox homeostasis and unfolded protein response (UPR)-related genes. Besides the close association between BcPdi1 and BcNoxA, interaction studies provide evidence that the ER protein might likewise be involved in Ca2+ regulated processes. Finally, we were able to show that the potential key functions of the protein BcPdi1 might be affected by its phosphorylation state.
Collapse
Affiliation(s)
- Robert Marschall
- Institut für Biologie und Biotechnologie der Pflanzen, Westfälische Wilhelms-Universität MünsterMünster, Germany
| | - Paul Tudzynski
- Institut für Biologie und Biotechnologie der Pflanzen, Westfälische Wilhelms-Universität MünsterMünster, Germany
| |
Collapse
|
17
|
Santos GB, Gonzalez-Perilli L, Mastrogiovanni M, Aicardo A, Cerdeira CD, Trostchansky A, Brigagão MRPL. Nitroxide 4-hydroxy-2,2',6,6'-tetramethylpiperidine 1-oxyl (Tempol) inhibits the reductase activity of protein disulfide isomerase via covalent binding to the Cys 400 residue on CXXC redox motif at the a'active site. Chem Biol Interact 2017; 272:117-124. [PMID: 28532685 DOI: 10.1016/j.cbi.2017.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/17/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIM Oxidative stress arising from inflammatory processes is a serious cause of cell and tissue damage. Tempol is an efficient antioxidant with superoxide dismutase-like activity. The purpose of this paper is to address the inhibition of protein disulfide isomerase (PDI), an essential redox chaperone whose active sites contain the Cys-Gly-His-Cys (CXXC) motif, by the nitroxide Tempol. RESULTS In the presence of Tempol (5-120 μM), the reductase activity of PDI was reversibly affected both in vitro and in activated mice neutrophils, with an IC50 of 22.9 ± 10.8 μM. Inhibitory activity was confirmed by using both the insulin method and fluorescent formation of eosin-glutathione (E-GSH). The capacity of Tempol to bind the enzyme was determined by EPR and mass spectrometry. EPR Tempol signal decreased in the presence of PDI while remained unaffected when PDI thiols were previously blocked with NEM. When total protein was analyzed, 1 and 4 molecules of Tempol were bound to the protein. However, only one was found to be covalently bound to PDI at the a'active site. More specifically, Cys400 was modified by Tempol. CONCLUSION We have shown that the nitroxide Tempol acts as an inhibitor of PDI through covalent binding to the Cys400 of the protein structure. Since PDI is coupled with the assembly of the NADPH oxidase complex of phagocytes, these findings reveal a novel action of Tempol that presents potential clinical applications for therapeutic intervention to target PDI knockdown in pathological processes in which this protein is engaged.
Collapse
Affiliation(s)
- Gérsika Bitencourt Santos
- Department of Biochemistry (DBq), Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, Brazil
| | - Lucia Gonzalez-Perilli
- Biochemistry Department, Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mauricio Mastrogiovanni
- Biochemistry Department, Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Adrián Aicardo
- Biochemistry Department, Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Cláudio Daniel Cerdeira
- Department of Biochemistry (DBq), Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, Brazil
| | - Andrés Trostchansky
- Biochemistry Department, Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | | |
Collapse
|
18
|
Nitroarachidonic acid (NO 2AA) inhibits protein disulfide isomerase (PDI) through reversible covalent adduct formation with critical cysteines. Biochim Biophys Acta Gen Subj 2017; 1861:1131-1139. [PMID: 28215702 DOI: 10.1016/j.bbagen.2017.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 02/04/2017] [Accepted: 02/08/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Nitroarachidonic acid (NO2AA) exhibits pleiotropic anti-inflammatory actions in a variety of cell types. We have recently shown that NO2AA inhibits phagocytic NADPH oxidase 2 (NOX2) by preventing the formation of the active complex. Recent work indicates the participation of protein disulfide isomerase (PDI) activity in NOX2 activation. Cysteine (Cys) residues at PDI active sites could be targets for NO2AA- nitroalkylation regulating PDI activity which could explain our previous observation. METHODS PDI reductase and chaperone activities were assessed using the insulin and GFP renaturation methods in the presence or absence of NO2AA. To determine the covalent reaction with PDI as well as the site of reaction, the PEG-switch assay and LC-MS/MS studies were performed. RESULTS AND CONCLUSIONS We determined that both activities of PDI were inhibited by NO2AA in a dose- and time- dependent manner and independent from release of nitric oxide. Since nitroalkenes are potent electrophiles and PDI has critical Cys residues for its activity, then formation of a covalent adduct between NO2AA and PDI is feasible. To this end we demonstrated the reversible covalent modification of PDI by NO2AA. Trypsinization of modified PDI confirmed that the Cys residues present in the active site a' of PDI were key targets accounting for nitroalkene modification. GENERAL SIGNIFICANCE PDI may contribute to NOX2 activation. As such, inhibition of PDI by NO2AA might be involved in preventing NOX2 activation. Future work will be directed to determine if the covalent modifications observed play a role in the reported NO2AA inhibition of NOX2 activity.
Collapse
|
19
|
Potential Role of Protein Disulfide Isomerase in Metabolic Syndrome-Derived Platelet Hyperactivity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2423547. [PMID: 28053690 PMCID: PMC5174184 DOI: 10.1155/2016/2423547] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/17/2016] [Accepted: 11/01/2016] [Indexed: 02/08/2023]
Abstract
Metabolic Syndrome (MetS) has become a worldwide epidemic, alongside with a high socioeconomic cost, and its diagnostic criteria must include at least three out of the five features: visceral obesity, hypertension, dyslipidemia, insulin resistance, and high fasting glucose levels. MetS shows an increased oxidative stress associated with platelet hyperactivation, an essential component for thrombus formation and ischemic events in MetS patients. Platelet aggregation is governed by the peroxide tone and the activity of Protein Disulfide Isomerase (PDI) at the cell membrane. PDI redox active sites present active cysteine residues that can be susceptible to changes in plasma oxidative state, as observed in MetS. However, there is a lack of knowledge about the relationship between PDI and platelet hyperactivation under MetS and its metabolic features, in spite of PDI being a mediator of important pathways implicated in MetS-induced platelet hyperactivation, such as insulin resistance and nitric oxide dysfunction. Thus, the aim of this review is to analyze data available in the literature as an attempt to support a possible role for PDI in MetS-induced platelet hyperactivation.
Collapse
|
20
|
Yoshioka J. Thioredoxin superfamily and its effects on cardiac physiology and pathology. Compr Physiol 2016; 5:513-30. [PMID: 25880503 DOI: 10.1002/cphy.c140042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A precise control of oxidation/reduction of protein thiols is essential for intact cardiac physiology. Irreversible oxidative modifications have been proposed to play a role in the pathogenesis of cardiovascular diseases. An imbalance of redox homeostasis with diminution of antioxidant capacities predisposes the heart to oxidant injury. There is growing interest in endoplasmic reticulum (ER) stress in the cardiovascular field, since perturbation of redox homeostasis in the ER is sufficient to cause ER stress. Because a number of human diseases are related to altered redox homeostasis and defects in protein folding, many research efforts have been devoted in recent years to understanding the structure and enzymatic properties of the thioredoxin superfamily. The thioredoxin superfamily has been well documented as thiol oxidoreductases to exert a role in various cell signaling pathways. The redox properties of the thioredoxin motif account for the different functions of several members of the thioredoxin superfamily. While thioredoxin and glutaredoxin primarily act as antioxidants by reducing protein disulfides and mixed disulfide, another member of the superfamily, protein disulfide isomerase (PDI), can act as an oxidant by forming intrachain disulfide bonds that contribute to proper protein folding. Increasing evidence suggests a pivotal role of PDI in the survival pathway that promotes cardiomyocyte survival and leads to more favorable cardiac remodeling. Thus, the thiol redox state is important for cellular redox signaling and survival pathway in the heart. This review summarizes the key features of major members of the thioredoxin superfamily directly involved in cardiac physiology and pathology.
Collapse
Affiliation(s)
- Jun Yoshioka
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| |
Collapse
|
21
|
Manfredi G, Kawamata H. Mitochondria and endoplasmic reticulum crosstalk in amyotrophic lateral sclerosis. Neurobiol Dis 2015; 90:35-42. [PMID: 26282323 DOI: 10.1016/j.nbd.2015.08.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/31/2015] [Accepted: 08/12/2015] [Indexed: 12/13/2022] Open
Abstract
Physical and functional interactions between mitochondria and the endoplasmic reticulum (ER) are crucial for cell life. These two organelles are intimately connected and collaborate to essential processes, such as calcium homeostasis and phospholipid biosynthesis. The connections between mitochondria and endoplasmic reticulum occur through structures named mitochondria associated membranes (MAMs), which contain lipid rafts and a large number of proteins, many of which serve multiple functions at different cellular sites. Growing evidence strongly suggests that alterations of ER-mitochondria interactions are involved in neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), a devastating and rapidly fatal motor neuron disease. Mutations in proteins that participate in ER-mitochondria interactions and MAM functions are increasingly being associated with genetic forms of ALS and other neurodegenerative diseases. This evidence strongly suggests that, rather than considering the two organelles separately, a better understanding of the disease process can derive from studying the alterations in their crosstalk. In this review we discuss normal and pathological ER-mitochondria interactions and the evidence that link them to ALS.
Collapse
Affiliation(s)
- Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 401 East 61st Street, New York, NY 10065, United States.
| | - Hibiki Kawamata
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 401 East 61st Street, New York, NY 10065, United States.
| |
Collapse
|
22
|
Klafke JZ, Porto FG, Batista R, Bochi GV, Moresco RN, da Luz PL, Viecili PRN. Association between hypertriglyceridemia and protein oxidation and proinflammatory markers in normocholesterolemic and hypercholesterolemic individuals. Clin Chim Acta 2015; 448:50-7. [PMID: 26115893 DOI: 10.1016/j.cca.2015.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/16/2015] [Accepted: 06/16/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND Although hypercholesterolemia is a well-established risk factor for coronary heart disease, evidence suggests that increased triglyceride (TG) concentrations are also an independent risk factor. TG concentrations >150mg/dl are observed nearly twice as often in subjects with atherosclerosis. We assessed the association between hypertriglyceridemia and protein oxidation and proinflammatory markers in normocholesterolemic and hypercholesterolemic individuals. METHODS We included 127 volunteers enrolled in Cruz Alta, RS, Brazil. The patients were stratified based on total cholesterol and TG concentrations for analysis of associations with inflammation (high-sensitivity C-reactive protein - hs-CRP), endothelial dysfunction (nitric oxide - NOx) and oxidative stress (advanced oxidation protein products - AOPPs; ischemia-modified albumin - IMA). Correlations between variables were determined and multiple regression analysis was employed to investigate whether some variables correlate with TG concentrations. RESULTS Hypertriglyceridemia was related to oxidative stress and proinflammatory markers in individuals independent of total cholesterol concentrations. Moreover, the results indicate a stronger association of tested biomarkers with TG concentrations than with total cholesterol. The results indicate a positive correlation between oxidative stress and TG concentrations in the sera of hypercholesterolemia subjects. AOPPs and IMA concentrations were associated with the presence of hypertriglyceridemia in a manner that was independent of age, gender, hypertension and diabetes mellitus disease, smoking habits, sedentary lifestyle, BMI, waist circumference, LDL, HDL and total cholesterol concentrations. CONCLUSIONS We speculate that TG concentrations can reflect the enhancement of protein oxidation and proinflammation.
Collapse
Affiliation(s)
- Jonatas Zeni Klafke
- Programa de Pós-Graduação em Atenção Integral à Saúde, Universidade de Cruz Alta, 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa, Instituto de Cardiologia de Cruz Alta, 98010-110 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde, Universidade de Cruz Alta, 98020-290 Cruz Alta, RS, Brazil.
| | - Fernando Garcez Porto
- Programa de Pós-Graduação em Atenção Integral à Saúde, Universidade de Cruz Alta, 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa, Instituto de Cardiologia de Cruz Alta, 98010-110 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde, Universidade de Cruz Alta, 98020-290 Cruz Alta, RS, Brazil
| | - Roselaine Batista
- Centro de Ensino e Pesquisa, Instituto de Cardiologia de Cruz Alta, 98010-110 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde, Universidade de Cruz Alta, 98020-290 Cruz Alta, RS, Brazil
| | - Guilherme Vargas Bochi
- Laboratório de Bioquímica Clínica, Departamento de Análises Clínicas e Toxicológicas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Rafael Noal Moresco
- Laboratório de Bioquímica Clínica, Departamento de Análises Clínicas e Toxicológicas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Protásio Lemos da Luz
- Instituto de Cardiologia-InCor-Hospital da Clínicas da Faculdade de Medicina da Universidade de São Paulo-HCFM-USP, São Paulo, SP, Brazil
| | - Paulo Ricardo Nazário Viecili
- Programa de Pós-Graduação em Atenção Integral à Saúde, Universidade de Cruz Alta, 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa, Instituto de Cardiologia de Cruz Alta, 98010-110 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde, Universidade de Cruz Alta, 98020-290 Cruz Alta, RS, Brazil
| |
Collapse
|
23
|
Guo S, Chen X. The human Nox4: gene, structure, physiological function and pathological significance. J Drug Target 2015; 23:888-96. [PMID: 25950600 DOI: 10.3109/1061186x.2015.1036276] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increased generation of reactive oxygen species (ROS) has been implicated in the pathogenesis of a variety of diseases such as cardiovascular diseases and cancer. NADPH oxidase (Nox), a multicomponent enzyme, has been identified as one of the key sources of ROS. Nox4, one of the seven members of Nox family (Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2), has been extensively investigated in recent years. Its unique structures result in the constitutive generation of hydrogen peroxide (H2O2) as the main product. As a key oxygen sensor, Nox4-derived H2O2 plays diverse roles in cell proliferation, migration and death. Increased expression of Nox4 in cancer has been observed, which participates in metastasis, angiogenesis and apoptosis. Expression of Nox4 in endothelial cells actively mediated endothelial activation, dysfunction and injury, which contributes to the development of atherosclerosis, hypertension, cardiac hypertrophy and among others. This article explores the experimental studies related to the gene, structure, physiological function and pathological significance of Nox4. As Nox4 might serve as a potential target for the therapy of cardiovascular diseases and cancer, the Nox4 inhibitor is also discussed in this article.
Collapse
Affiliation(s)
- Shuhui Guo
- a State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao , China
| | - Xiuping Chen
- a State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao , China
| |
Collapse
|
24
|
Androwiki ACD, Camargo LDL, Sartoretto S, Couto GK, Ribeiro IMR, Veríssimo-Filho S, Rossoni LV, Lopes LR. Protein disulfide isomerase expression increases in resistance arteries during hypertension development. Effects on Nox1 NADPH oxidase signaling. Front Chem 2015; 3:24. [PMID: 25870854 PMCID: PMC4375999 DOI: 10.3389/fchem.2015.00024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/11/2015] [Indexed: 11/30/2022] Open
Abstract
NADPH oxidases derived reactive oxygen species (ROS) play an important role in vascular function and remodeling in hypertension through redox signaling processes. Previous studies demonstrated that protein disulfide isomerase (PDI) regulates Nox1 expression and ROS generation in cultured vascular smooth muscle cells. However, the role of PDI in conductance and resistance arteries during hypertension development remains unknown. The aim of the present study was to investigate PDI expression and NADPH oxidase dependent ROS generation during hypertension development. Mesenteric resistance arteries (MRA) and thoracic aorta were isolated from 6, 8, and 12 week-old spontaneously hypertensive (SHR) and Wistar rats. ROS production (dihydroethidium fluorescence), PDI (WB, imunofluorescence), Nox1 and NOX4 (RT-PCR) expression were evaluated. Results show a progressive increase in ROS generation in MRA and aorta from 8 to 12 week-old SHR. This effect was associated with a concomitant increase in PDI and Nox1 expression only in MRA. Therefore, suggesting a positive correlation between PDI and Nox1 expression during the development of hypertension in MRA. In order to investigate if this effect was due to an increase in arterial blood pressure, pre hypertensive SHR were treated with losartan (20 mg/kg/day for 30 days), an AT1 receptor antagonist. Losartan decreased blood pressure and ROS generation in both vascular beds. However, only in SHR MRA losartan treatment lowered PDI and Nox1 expression to control levels. In MRA PDI inhibition (bacitracin, 0.5 mM) decreased Ang II redox signaling (p-ERK 1/2). Altogether, our results suggest that PDI plays a role in triggering oxidative stress and vascular dysfunction in resistance but not in conductance arteries, increasing Nox1 expression and activity. Therefore, PDI could be a new player in oxidative stress and functional alterations in resistance arteries during the establishment of hypertension.
Collapse
Affiliation(s)
- Aline C D Androwiki
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Lívia de Lucca Camargo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Simone Sartoretto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Gisele K Couto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Izabela M R Ribeiro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Sidney Veríssimo-Filho
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Luciana V Rossoni
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| | - Lucia R Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo São Paulo, Brazil
| |
Collapse
|
25
|
Bechor E, Dahan I, Fradin T, Berdichevsky Y, Zahavi A, Federman Gross A, Rafalowski M, Pick E. The dehydrogenase region of the NADPH oxidase component Nox2 acts as a protein disulfide isomerase (PDI) resembling PDIA3 with a role in the binding of the activator protein p67 (phox.). Front Chem 2015; 3:3. [PMID: 25699251 PMCID: PMC4316792 DOI: 10.3389/fchem.2015.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/09/2015] [Indexed: 11/28/2022] Open
Abstract
The superoxide (O(·-) 2)-generating NADPH oxidase of phagocytes consists of a membrane component, cytochrome b 558 (a heterodimer of Nox2 and p22 (phox) ), and four cytosolic components, p47 (phox) , p67 (phox) , p40 (phox) , and Rac. The catalytic component, responsible for O(·-) 2 generation, is Nox2. It is activated by the interaction of the dehydrogenase region (DHR) of Nox2 with the cytosolic components, principally with p67 (phox) . Using a peptide-protein binding assay, we found that Nox2 peptides containing a (369)CysGlyCys(371) triad (CGC) bound p67 (phox) with high affinity, dependent upon the establishment of a disulfide bond between the two cysteines. Serially truncated recombinant Nox2 DHR proteins bound p67 (phox) only when they comprised the CGC triad. CGC resembles the catalytic motif (CGHC) of protein disulfide isomerases (PDIs). This led to the hypothesis that Nox2 establishes disulfide bonds with p67 (phox) via a thiol-dilsulfide exchange reaction and, thus, functions as a PDI. Evidence for this was provided by the following: (1) Recombinant Nox2 protein, which contained the CGC triad, exhibited PDI-like disulfide reductase activity; (2) Truncation of Nox2 C-terminal to the CGC triad or mutating C369 and C371 to R, resulted in loss of PDI activity; (3) Comparison of the sequence of the DHR of Nox2 with PDI family members revealed three small regions of homology with PDIA3; (4) Two monoclonal anti-Nox2 antibodies, with epitopes corresponding to regions of Nox2/PDIA3 homology, reacted with PDIA3 but not with PDIA1; (5) A polyclonal anti-PDIA3 (but not an anti-PDIA1) antibody reacted with Nox2; (6) p67 (phox) , in which all cysteines were mutated to serines, lost its ability to bind to a Nox2 peptide containing the CGC triad and had an impaired capacity to support oxidase activity in vitro. We propose a model of oxidase assembly in which binding of p67 (phox) to Nox2 via disulfide bonds, by virtue of the intrinsic PDI activity of Nox2, stabilizes the primary interaction between the two components.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Edgar Pick
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv UniversityTel Aviv, Israel
| |
Collapse
|
26
|
Jaronen M, Goldsteins G, Koistinaho J. ER stress and unfolded protein response in amyotrophic lateral sclerosis-a controversial role of protein disulphide isomerase. Front Cell Neurosci 2014; 8:402. [PMID: 25520620 PMCID: PMC4251436 DOI: 10.3389/fncel.2014.00402] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 11/07/2014] [Indexed: 01/20/2023] Open
Abstract
Accumulation of proteins in aberrant conformation occurs in many neurodegenerative diseases. Furthermore, dysfunctions in protein handling in endoplasmic reticulum (ER) and the following ER stress have been implicated in a vast number of diseases, such as amyotrophic lateral sclerosis (ALS). During excessive ER stress unfolded protein response (UPR) is activated to return ER to its normal physiological balance. The exact mechanisms of protein misfolding, accumulation and the following ER stress, which could lead to neurodegeneration, and the question whether UPR is a beneficial compensatory mechanism slowing down the neurodegenerative processes, are of interest. Protein disulphide isomerase (PDI) is a disulphide bond-modulating ER chaperone, which can also facilitate the ER-associated degradation (ERAD) of misfolded proteins. In this review we discuss the recent findings of ER stress, UPR and especially the role of PDI in ALS.
Collapse
Affiliation(s)
- Merja Jaronen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland ; Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School Boston, MA, USA
| | - Gundars Goldsteins
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland
| | - Jari Koistinaho
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland
| |
Collapse
|
27
|
Drummond GR, Sobey CG. Endothelial NADPH oxidases: which NOX to target in vascular disease? Trends Endocrinol Metab 2014; 25:452-63. [PMID: 25066192 DOI: 10.1016/j.tem.2014.06.012] [Citation(s) in RCA: 246] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/29/2014] [Accepted: 06/30/2014] [Indexed: 02/03/2023]
Abstract
NADPH oxidases (NOXs) are reactive oxygen species (ROS)-generating enzymes implicated in the pathophysiology of vascular diseases such as hypertension and stroke. Endothelial cells express four NOX isoforms including the superoxide-generating enzymes NOX1, NOX2, and NOX5 and the hydrogen peroxide-generating enzyme NOX4. Studies on arteries from patients with coronary artery disease, and in animals with experimentally induced hypertension, diabetes, or atherosclerosis, suggest that NOX1, NOX2, and NOX5 promote endothelial dysfunction, inflammation, and apoptosis in the vessel wall, whereas NOX4 is by contrast vasoprotective in increasing nitric oxide bioavailability and suppressing cell death pathways. Based on these findings and promising preclinical studies with the NOX1/NOX2 antagonist, apocynin, we suggest that the field is poised for clinical evaluation of NOX inhibitors as therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Grant R Drummond
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Surgery, Monash Medical Centre, Southern Clinical School, Monash University, Clayton, Victoria, Australia.
| | - Christopher G Sobey
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Surgery, Monash Medical Centre, Southern Clinical School, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
28
|
Ali Khan H, Mutus B. Protein disulfide isomerase a multifunctional protein with multiple physiological roles. Front Chem 2014; 2:70. [PMID: 25207270 PMCID: PMC4144422 DOI: 10.3389/fchem.2014.00070] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/07/2014] [Indexed: 11/19/2022] Open
Abstract
Protein disulfide isomerase (PDI), is a member of the thioredoxin superfamily of redox proteins. PDI has three catalytic activities including, thiol-disulfide oxireductase, disulfide isomerase and redox-dependent chaperone. Originally, PDI was identified in the lumen of the endoplasmic reticulum and subsequently detected at additional locations, such as cell surfaces and the cytosol. This review will provide an overview of the recent advances in relating the structural features of PDI to its multiple catalytic roles as well as its physiological and pathophysiological functions related to redox regulation and protein folding.
Collapse
Affiliation(s)
- Hyder Ali Khan
- Chemistry and Biochemistry Department, University of Windsor Windsor, ON, Canada
| | - Bulent Mutus
- Chemistry and Biochemistry Department, University of Windsor Windsor, ON, Canada
| |
Collapse
|
29
|
Szumiel I. Ionizing radiation-induced oxidative stress, epigenetic changes and genomic instability: The pivotal role of mitochondria. Int J Radiat Biol 2014; 91:1-12. [DOI: 10.3109/09553002.2014.934929] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
30
|
Abstract
Protein disulfide isomerase (PDI), ERp5, and ERp57, among perhaps other thiol isomerases, are important for the initiation of thrombus formation. Using the laser injury thrombosis model in mice to induce in vivo arterial thrombus formation, it was shown that thrombus formation is associated with PDI secretion by platelets, that inhibition of PDI blocked platelet thrombus formation and fibrin generation, and that endothelial cell activation leads to PDI secretion. Similar results using this and other thrombosis models in mice have demonstrated the importance of ERp5 and ERp57 in the initiation of thrombus formation. The integrins, αIIbβ3 and αVβ3, play a key role in this process and interact directly with PDI, ERp5, and ERp57. The mechanism by which thiol isomerases participate in thrombus generation is being evaluated using trapping mutant forms to identify substrates of thiol isomerases that participate in the network pathways linking thiol isomerases, platelet receptor activation, and fibrin generation. PDI as an antithrombotic target is being explored using isoquercetin and quercetin 3-rutinoside, inhibitors of PDI identified by high throughput screening. Regulation of thiol isomerase expression, analysis of the storage, and secretion of thiol isomerases and determination of the electron transfer pathway are key issues to understanding this newly discovered mechanism of regulation of the initiation of thrombus formation.
Collapse
Affiliation(s)
- Bruce Furie
- From the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | | |
Collapse
|
31
|
Abstract
SIGNIFICANCE Understanding isoform- and context-specific subcellular Nox reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase compartmentalization allows relevant functional inferences. This review addresses the interplay between Nox NADPH oxidases and the endoplasmic reticulum (ER), an increasingly evident player in redox pathophysiology given its role in redox protein folding and stress responses. RECENT ADVANCES Catalytic/regulatory transmembrane subunits are synthesized in the ER and their processing includes folding, N-glycosylation, heme insertion, p22phox heterodimerization, as shown for phagocyte Nox2. Dual oxidase (Duox) maturation also involves the regulation by ER-resident Duoxa2. The ER is the activation site for some isoforms, typically Nox4, but potentially other isoforms. Such location influences redox/Nox-mediated calcium signaling regulation via ER targets, such as sarcoendoplasmic reticulum calcium ATPase (SERCA). Growing evidence suggests that Noxes are integral signaling elements of the unfolded protein response during ER stress, with Nox4 playing a dual prosurvival/proapoptotic role in this setting, whereas Nox2 enhances proapoptotic signaling. ER chaperones such as protein disulfide isomerase (PDI) closely interact with Noxes. PDI supports growth factor-dependent Nox1 activation and mRNA expression, as well as migration in smooth muscle cells, and PDI overexpression induces acute spontaneous Nox activation. CRITICAL ISSUES Mechanisms of PDI effects include possible support of complex formation and RhoGTPase activation. In phagocytes, PDI supports phagocytosis, Nox activation, and redox-dependent interactions with p47phox. Together, the results implicate PDI as possible Nox organizer. FUTURE DIRECTIONS We propose that convergence between Noxes and ER may have evolutive roots given ER-related functional contexts, which paved Nox evolution, namely calcium signaling and pathogen killing. Overall, the interplay between Noxes and the ER may provide relevant insights in Nox-related (patho)physiology.
Collapse
Affiliation(s)
- Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine , São Paulo, Brazil
| | | | | |
Collapse
|
32
|
Wittenberg G, Levitan A, Klein T, Dangoor I, Keren N, Danon A. Knockdown of the Arabidopsis thaliana chloroplast protein disulfide isomerase 6 results in reduced levels of photoinhibition and increased D1 synthesis in high light. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2014; 78:1003-13. [PMID: 24684167 DOI: 10.1111/tpj.12525] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 03/18/2014] [Accepted: 03/27/2014] [Indexed: 05/09/2023]
Abstract
A chloroplast protein disulfide isomerase (PDI) was previously proposed to regulate translation of the unicellular green alga Chlamydomonas reinhardtii chloroplast psbA mRNA, encoding the D1 protein, in response to light. Here we show that AtPDI6, one of 13 Arabidopsis thaliana PDI genes, also plays a role in the chloroplast. We found that AtPDI6 is targeted and localized to the chloroplast. Interestingly, AtPDI6 knockdown plants displayed higher resistance to photoinhibition than wild-type plants when exposed to a tenfold increase in light intensity. The AtPDI6 knockdown plants also displayed a higher rate of D1 synthesis under a similar light intensity. The increased resistance to photoinhibition may not be rationalized by changes in antenna or non-photochemical quenching. Thus, the increased D1 synthesis rate, which may result in a larger proportion of active D1 under light stress, may led to the decrease in photoinhibition. These results suggest that, although the D1 synthesis rates observed in wild-type plants under high light intensities are elevated, repair can potentially occur faster. The findings implicate AtPDI6 as an attenuator of D1 synthesis, modulating photoinhibition in a light-regulated manner.
Collapse
Affiliation(s)
- Gal Wittenberg
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | | | | | | | | |
Collapse
|
33
|
Dias FF, Amaral KB, Carmo LAS, Shamri R, Dvorak AM, Weller PF, Melo RCN. Human Eosinophil Leukocytes Express Protein Disulfide Isomerase in Secretory Granules and Vesicles: Ultrastructural Studies. J Histochem Cytochem 2014; 62:450-459. [PMID: 24670795 DOI: 10.1369/0022155414531437] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Protein disulfide isomerase (PDI) has fundamental roles in the oxidative folding of proteins in the endoplasmic reticulum (ER) of eukaryotic cells. The study of this molecule has been attracting considerable attention due to its association with other cell functions and human diseases. In leukocytes, such as neutrophils, PDI is involved with cell adhesion, signaling and inflammation. However, the expression of PDI in other leukocytes, such as eosinophils, important cells in inflammatory, allergic and immunomodulatory responses, remains to be defined. Here we used different approaches to investigate PDI expression within human eosinophils. Western blotting and flow cytometry demonstrated high PDI expression in both unstimulated and CCL11/eotaxin-1-stimulated eosinophils, with similar levels in both conditions. By using an immunogold electron microscopy technique that combines better epitope preservation and secondary Fab-fragments of antibodies linked to 1.4-nm gold particles for optimal access to microdomains, we identified different intracellular sites for PDI. In addition to predictable strong PDI labeling at the nuclear envelope, other unanticipated sites, such as secretory granules, lipid bodies and vesicles, including large transport vesicles (eosinophil sombrero vesicles), were also labeled. Thus, we provide the first identification of PDI in human eosinophils, suggesting that this molecule may have additional/specific functions in these leukocytes.
Collapse
Affiliation(s)
- Felipe F Dias
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, MG, Brazil (FFD,KBA,LASC,RCNM)Department of Pathology (AMD)Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (RS,PFW,RCNM)
| | - Kátia B Amaral
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, MG, Brazil (FFD,KBA,LASC,RCNM)Department of Pathology (AMD)Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (RS,PFW,RCNM)
| | - Lívia A S Carmo
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, MG, Brazil (FFD,KBA,LASC,RCNM)Department of Pathology (AMD)Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (RS,PFW,RCNM)
| | - Revital Shamri
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, MG, Brazil (FFD,KBA,LASC,RCNM)Department of Pathology (AMD)Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (RS,PFW,RCNM)
| | - Ann M Dvorak
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, MG, Brazil (FFD,KBA,LASC,RCNM)Department of Pathology (AMD)Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (RS,PFW,RCNM)
| | - Peter F Weller
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, MG, Brazil (FFD,KBA,LASC,RCNM)Department of Pathology (AMD)Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (RS,PFW,RCNM)
| | - Rossana C N Melo
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, MG, Brazil (FFD,KBA,LASC,RCNM)Department of Pathology (AMD)Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (RS,PFW,RCNM)
| |
Collapse
|
34
|
Protein disulfide isomerase: a promising target for cancer therapy. Drug Discov Today 2014; 19:222-40. [DOI: 10.1016/j.drudis.2013.10.017] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 09/12/2013] [Accepted: 10/22/2013] [Indexed: 12/15/2022]
|
35
|
Griffiths HR, Dias IHK, Willetts RS, Devitt A. Redox regulation of protein damage in plasma. Redox Biol 2014; 2:430-5. [PMID: 24624332 PMCID: PMC3949090 DOI: 10.1016/j.redox.2014.01.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 01/11/2014] [Indexed: 01/12/2023] Open
Abstract
The presence and concentrations of modified proteins circulating in plasma depend on rates of protein synthesis, modification and clearance. In early studies, the proteins most frequently analysed for damage were those which were more abundant in plasma (e.g. albumin and immunoglobulins) which exist at up to 10 orders of magnitude higher concentrations than other plasma proteins e.g. cytokines. However, advances in analytical techniques using mass spectrometry and immuno-affinity purification methods, have facilitated analysis of less abundant, modified proteins and the nature of modifications at specific sites is now being characterised. The damaging reactive species that cause protein modifications in plasma principally arise from reactive oxygen species (ROS) produced by NADPH oxidases (NOX), nitric oxide synthases (NOS) and oxygenase activities; reactive nitrogen species (RNS) from myeloperoxidase (MPO) and NOS activities; and hypochlorous acid from MPO. Secondary damage to proteins may be caused by oxidized lipids and glucose autooxidation. In this review, we focus on redox regulatory control of those enzymes and processes which control protein maturation during synthesis, produce reactive species, repair and remove damaged plasma proteins. We have highlighted the potential for alterations in the extracellular redox compartment to regulate intracellular redox state and, conversely, for intracellular oxidative stress to alter the cellular secretome and composition of extracellular vesicles. Through secreted, redox-active regulatory molecules, changes in redox state may be transmitted to distant sites. Loss of redox homeostasis may affect the secretome content and protein concentration, transmitting redox signals to distant cells through extracellular vesicles. Damaged glycoforms may arise from oxidants or aberrant biosynthetic regulation. Reactive species generation by NOX and NOS is controlled through redox regulation. Cell surface and plasma thiol-oxidised proteins can be reduced and their activity modulated by thioredoxin, protein disulphide isomerase and reductases.
Collapse
Key Words
- Ageing
- BH4, tetrahydrobiopterin
- COX, cyclo-oxygenase
- CRP, C-reactive protein
- ER, endoplasmic reticulum
- ERO1, endoplasmic reticulum oxidoreductin 1
- EV, extracellular vesicles
- FX1, factor XI
- GPI, glycoprotein 1
- GPX, glutathione peroxidase
- GRX, glutaredoxin
- GSH, glutathione
- Glycosylation
- MIRNA, microRNA
- MPO, myeloperoxidase
- NO, nitric oxide
- NOS, nitric oxide synthase
- NOX, NADPH oxidase
- Nitration
- O2•−, superoxide anion radical
- ONOO-, peroxynitrite
- Oxidation
- PDI, protein disulphide isomerase
- Peroxiredoxin
- Prx, peroxiredoxin
- RNS, reactive nitrogen species
- ROS, reactive nitrogen species
- Thioredoxin
- Trx, thioredoxin
- VWF, von Willebrand factor
- XO, xanthine oxidase
Collapse
Affiliation(s)
- Helen R Griffiths
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Irundika H K Dias
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Rachel S Willetts
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Andrew Devitt
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| |
Collapse
|
36
|
Schulz E, Wenzel P, Münzel T, Daiber A. Mitochondrial redox signaling: Interaction of mitochondrial reactive oxygen species with other sources of oxidative stress. Antioxid Redox Signal 2014; 20:308-24. [PMID: 22657349 PMCID: PMC3887453 DOI: 10.1089/ars.2012.4609] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Oxidative stress is a well established hallmark of cardiovascular disease and there is strong evidence for a causal role of reactive oxygen and nitrogen species (RONS) therein. RECENT ADVANCES Improvement of cardiovascular complications by genetic deletion of RONS producing enzymes and overexpression of RONS degrading enzymes proved the involvement of these species in cardiovascular disease at a molecular level. Vice versa, overexpression of RONS producing enzymes as well as deletion of antioxidant enzymes was demonstrated to aggravate cardiovascular complications. CRITICAL ISSUES With the present overview we present and discuss different pathways how mitochondrial RONS interact (crosstalk) with other sources of oxidative stress, namely NADPH oxidases, xanthine oxidase and an uncoupled nitric oxide synthase. The potential mechanisms of how this crosstalk proceeds are discussed in detail. Several examples from the literature are summarized (including hypoxia, angiotensin II mediated vascular dysfunction, cellular starvation, nitrate tolerance, aging, hyperglycemia, β-amyloid stress and others) and the underlying mechanisms are put together to a more general concept of redox-based activation of different sources of RONS via enzyme-specific "redox switches". Mitochondria play a key role in this concept providing redox triggers for oxidative damage in the cardiovascular system but also act as amplifiers to increase the burden of oxidative stress. FUTURE DIRECTIONS Based on these considerations, the characterization of the role of mitochondrial RONS formation in cardiac disease as well as inflammatory processes but also the role of mitochondria as potential therapeutic targets in these pathophysiological states should be addressed in more detail in the future.
Collapse
Affiliation(s)
- Eberhard Schulz
- 1 2nd Medical Clinic, Molecular Cardiology, Medical Center of the Johannes Gutenberg University , Mainz, Germany
| | | | | | | |
Collapse
|
37
|
Camargo LDL, Babelova A, Mieth A, Weigert A, Mooz J, Rajalingam K, Heide H, Wittig I, Lopes LR, Brandes RP. Endo-PDI is required for TNFα-induced angiogenesis. Free Radic Biol Med 2013; 65:1398-1407. [PMID: 24103565 DOI: 10.1016/j.freeradbiomed.2013.09.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/08/2013] [Accepted: 09/29/2013] [Indexed: 12/20/2022]
Abstract
Protein disulfide isomerase (PDI) and its homologs are oxidoreductases facilitating protein folding in the ER. Endo-PDI (also termed ERp46) is highly expressed in endothelial cells. It belongs to the PDI family but its physiological function is largely unknown. We studied the role of Endo-PDI in endothelial angiogenic responses. Stimulation of human umbilical vein endothelial cells (with TNFα (10ng/ml) increased ERK1/2 phosphorylation. This effect was largely attenuated by Endo-PDI siRNA, whereas JNK and p38 MAP kinase phosphorylation was Endo-PDI independent. Similarly, TNFα-stimulated NF-κB signaling determined by IκBα degradation as well as TNFα-induced ICAM expression was unaffected by Endo-PDI siRNA. The action of Endo-PDI was not mediated by extracellular thiol exchange or cell surface PDI as demonstrated by nonpermeative inhibitors and PDI-neutralizing antibody. Moreover, exogenously added PDI failed to restore ERK1/2 activation after Endo-PDI knockdown. This suggests that Endo-PDI acts intracellularly potentially by maintaining the Ras/Raf/MEK/ERK pathway. Indeed, knockdown of Endo-PDI attenuated Ras activation measured by G-LISA and Raf phosphorylation. ERK activation influences gene expression by the transcriptional factor AP-1, which controls MMP-9 and cathepsin B, two proteases required for angiogenesis. TNFα-stimulated MMP-9 and cathepsin B induction was reduced by silencing of Endo-PDI. Accordingly, inhibition of cathepsin B or Endo-PDI siRNA blocked the TNFα-stimulated angiogenic response in the spheroid outgrowth assays. Moreover ex vivo tube formation and in vivo Matrigel angiogenesis in response to TNFα were attenuated by Endo-PDI siRNA. In conclusion, our study establishes Endo-PDI as a novel, important mediator of AP-1-driven gene expression and endothelial angiogenic function.
Collapse
Affiliation(s)
- Livia de Lucca Camargo
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany; Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andrea Babelova
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Anja Mieth
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute for Biochemistry I, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Juliane Mooz
- Institute for Biochemistry II, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | | | - Heinrich Heide
- Functional Proteomics, SFB815 Core Unit, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Ilka Wittig
- Functional Proteomics, SFB815 Core Unit, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Lucia Rossetti Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
38
|
Prado GN, Romero JR, Rivera A. Endothelin-1 receptor antagonists regulate cell surface-associated protein disulfide isomerase in sickle cell disease. FASEB J 2013; 27:4619-29. [PMID: 23913858 PMCID: PMC3804753 DOI: 10.1096/fj.13-228577] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/09/2013] [Indexed: 01/29/2023]
Abstract
Increased endothelin-1 (ET-1) levels, disordered thiol protein status, and erythrocyte hydration status play important roles in sickle cell disease (SCD) through unresolved mechanisms. Protein disulfide isomerase (PDI) is an oxidoreductase that mediates thiol/disulfide interchange reactions. We provide evidence that PDI is present in human and mouse erythrocyte membranes and that selective blockade with monoclonal antibodies against PDI leads to reduced Gardos channel activity (1.6±0.03 to 0.56±0.02 mmol·10(13) cell(-1)·min(-1), P<0.001) and density of sickle erythrocytes (D50: 1.115±0.001 to 1.104±0.001 g/ml, P=0.012) with an IC50 of 4 ng/ml. We observed that erythrocyte associated-PDI activity was increased in the presence of ET-1 (3.1±0.2 to 5.6±0.4%, P<0.0001) through a mechanism that includes casein kinase II. Consistent with these results, in vivo treatment of BERK sickle transgenic mice with ET-1 receptor antagonists lowered circulating and erythrocyte associated-PDI activity (7.1±0.3 to 5.2±0.2%, P<0.0001) while improving hematological parameters and Gardos channel activity. Thus, our results suggest that PDI is a novel target in SCD that regulates erythrocyte volume and oxidative stress and may contribute to cellular adhesion and endothelial activation leading to vasoocclusion as observed in SCD.
Collapse
Affiliation(s)
- Gregory N Prado
- 1Department of Laboratory Medicine, Bader 7, Boston Children's Hospital, 300 Longwood Ave., Boston, MA 02115, USA.
| | | | | |
Collapse
|
39
|
Microglial cells are involved in the susceptibility of NADPH oxidase knockout mice to 6-hydroxy-dopamine-induced neurodegeneration. PLoS One 2013; 8:e75532. [PMID: 24086556 PMCID: PMC3781051 DOI: 10.1371/journal.pone.0075532] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/15/2013] [Indexed: 01/22/2023] Open
Abstract
We explored the impact of Nox-2 in modulating inflammatory-mediated microglial responses in the 6-hydroxydopamine (6-OHDA)-induced Parkinson’s disease (PD) model. Nox1 and Nox2 gene expression were found to increase in striatum, whereas a marked increase of Nox2 expression was observed in substantia nigra (SN) of wild-type (wt) mice after PD induction. Gp91phox-/- 6-OHDA-lesioned mice exhibited a significant reduction in the apomorphine-induced rotational behavior, when compared to wt mice. Immunolabeling assays indicated that striatal 6-OHDA injections reduced the number of dopaminergic (DA) neurons in the SN of wt mice. In gp91phox-/- 6-OHDA-lesioned mice the DA degeneration was negligible, suggesting an involvement of Nox in 6-OHDA-mediated SN degeneration. Gp91phox-/- 6-OHDA-lesioned mice treated with minocycline, a tetracycline derivative that exerts multiple anti-inflammatory effects, including microglial inhibition, exhibited increased apomorphine-induced rotational behavior and degeneration of DA neurons after 6-OHDA injections. The same treatment also increased TNF-α release and potentiated NF-κB activation in the SN of gp91phox-/--lesioned mice. Our results demonstrate for the first time that inhibition of microglial cells increases the susceptibility of gp91phox-/- 6-OHDA lesioned mice to develop PD. Blockade of microglia leads to NF-κB activation and TNF-α release into the SN of gp91phox-/- 6-OHDA lesioned mice, a likely mechanism whereby gp91phox-/- 6-OHDA lesioned mice may be more susceptible to develop PD after microglial cell inhibition. Nox2 adds an essential level of regulation to signaling pathways underlying the inflammatory response after PD induction.
Collapse
|
40
|
Hernandes MS, Britto LRG. NADPH oxidase and neurodegeneration. Curr Neuropharmacol 2013; 10:321-7. [PMID: 23730256 PMCID: PMC3520042 DOI: 10.2174/157015912804143540] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 07/22/2012] [Accepted: 08/08/2012] [Indexed: 12/21/2022] Open
Abstract
NADPH oxidase (Nox) is a unique, multi-protein, electron transport system that produces large amounts of superoxide via the reduction of molecular oxygen. Nox-derived reactive oxygen species (ROS) are known to be involved in a variety of physiological processes, including host defense and signal transduction. However, over the past decade, the involvement of (Nox)-dependent oxidative stress in the pathophysiology of several neurodegenerative diseases has been increasingly recognized. ROS produced by Nox proteins contribute to neurodegenerative diseases through distinct mechanisms, such as oxidation of DNA, proteins, lipids, amino acids and metals, in addition to activation of redox-sensitive signaling pathways. In this review, we discuss the recent literature on Nox involvement in neurodegeneration, focusing on Parkinson and Alzheimer diseases.
Collapse
Affiliation(s)
- Marina S Hernandes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas and Núcleo de Apoio à Pesquisa em Neurociência Aplicada, Universidade de São Paulo, SP, Brasil
| | | |
Collapse
|
41
|
Immenschuh S, Rahayu P, Bayat B, Saragih H, Rachman A, Santoso S. Antibodies against dengue virus nonstructural protein-1 induce heme oxygenase-1 via a redox-dependent pathway in human endothelial cells. Free Radic Biol Med 2013; 54:85-92. [PMID: 23103292 DOI: 10.1016/j.freeradbiomed.2012.10.551] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 10/11/2012] [Accepted: 10/18/2012] [Indexed: 11/24/2022]
Abstract
Heme oxygenase (HO)-1, the inducible isoform of the first and rate-limiting enzyme of heme degradation, affords anti-inflammatory protection via its cell-type-specific effects in endothelial cells (ECs). In dengue hemorrhagic fever (DHF), which is the life-threatening form of dengue virus (DV) infection, endothelial interactions of cross-reactive antibodies against the DV nonstructural glycoprotein-1 (NS1) are associated with endothelial dysfunction. In this study, we investigated whether anti-NS1 antibodies might regulate HO-1 gene expression in human ECs. Serum from DHF patients with high anti-NS1 titers and a monoclonal anti-NS1 antibody upregulated HO-1 gene expression in human umbilical vein ECs, which was blocked by purified NS1 antigen. Immunoprecipitation studies showed that anti-NS1 antibodies specifically bound to the oxidoreductase protein disulfide isomerase (PDI) on ECs. Moreover, anti-NS1-mediated HO-1 induction was reduced by inhibition of PDI enzyme activity. Reactive oxygen species, which were generated by NADPH oxidase and in turn activated the phosphatidylinositol 3-kinase (PI3K)/Akt cascade, were involved in this upregulation of HO-1 gene expression. Finally, apoptosis of ECs caused by anti-NS1 antibodies was increased by pharmacological inhibition of HO-1 enzyme activity. In conclusion, HO-1 gene expression is upregulated by anti-NS1 antibodies via activation of a redox-dependent PDI/PI3K/Akt-mediated pathway in human ECs.
Collapse
Affiliation(s)
- Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany.
| | | | | | | | | | | |
Collapse
|
42
|
Jaronen M, Vehviläinen P, Malm T, Keksa-Goldsteine V, Pollari E, Valonen P, Koistinaho J, Goldsteins G. Protein disulfide isomerase in ALS mouse glia links protein misfolding with NADPH oxidase-catalyzed superoxide production. Hum Mol Genet 2012; 22:646-55. [PMID: 23118353 DOI: 10.1093/hmg/dds472] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Protein disulfide isomerase (PDI) is an oxidoreductase assisting oxidative protein folding in the endoplasmic reticulum of all types of cells, including neurons and glia. In neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS), up-regulation of PDI is an important part of unfolded protein response (UPR) that is thought to represent an adaption reaction and thereby protect the neurons. Importantly, studies on animal models of familial ALS with mutant Cu/Zn superoxide dismutase 1 (SOD1) have shown that the mutant SOD1 in astrocytes or microglia strongly regulates the progression of the disease. Here, we found an early up-regulation of PDI in microglia of transgenic (tg) mutant SOD1 mice, indicating that in addition to neurons, UPR takes place in glial cells in ALS. The observation was supported by the finding that also the expression of a UPR marker GADD34 (growth arrest and DNA damage-inducible protein) was induced in the spinal cord glia of tg mutant SOD1 mice. Because mutant SOD1 can cause sustained activation of NADPH oxidase (NOX), we investigated the role of PDI in UPR-induced NOX activation in microglia. In BV-2 microglia, UPR resulted in NOX activation with increased production of superoxide and increased release of tumor necrosis factor-α. The phenomenon was recapitulated in primary rat microglia, murine macrophages and human monocytes. Importantly, pharmacological inhibition of PDI or its down-regulation by short interfering RNAs prevented NOX activation in microglia and subsequent production of superoxide. Thus, results strongly demonstrate that UPR, caused by protein misfolding, may lead to PDI-dependent NOX activation and contribute to neurotoxicity in neurodegenerative diseases including ALS.
Collapse
Affiliation(s)
- Merja Jaronen
- Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Laurindo FRM, Pescatore LA, Fernandes DDC. Protein disulfide isomerase in redox cell signaling and homeostasis. Free Radic Biol Med 2012; 52:1954-69. [PMID: 22401853 DOI: 10.1016/j.freeradbiomed.2012.02.037] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 02/23/2012] [Accepted: 02/24/2012] [Indexed: 12/16/2022]
Abstract
Thiol proteins may potentially act as redox signaling adaptor proteins, adjusting reactive oxygen species intermediates to specific signals and redox signals to cell homeostasis. In this review, we discuss redox effects of protein disulfide isomerase (PDI), a thioredoxin superfamily oxidoreductase from the endoplasmic reticulum (ER). Abundantly expressed PDI displays ubiquity, interactions with redox and nonredox proteins, versatile effects, and several posttranslational modifications. The PDI family contains >20 members with at least some apparent complementary actions. PDI has oxidoreductase, isomerase, and chaperone effects, the last not directly dependent on its thiols. PDI is a converging hub for pathways of disulfide bond introduction into ER-processed proteins, via hydrogen peroxide-generating mechanisms involving the oxidase Ero1α, as well as hydrogen peroxide-consuming reactions involving peroxiredoxin IV and the novel peroxidases Gpx7/8. PDI is a candidate pathway for coupling ER stress to oxidant generation. Emerging information suggests a convergence between PDI and Nox family NADPH oxidases. PDI silencing prevents Nox responses to angiotensin II and inhibits Akt phosphorylation in vascular cells and parasite phagocytosis in macrophages. PDI overexpression spontaneously enhances Nox activation and expression. In neutrophils, PDI redox-dependently associates with p47phox and supports the respiratory burst. At the cell surface, PDI exerts transnitrosation, thiol reductase, and apparent isomerase activities toward targets including adhesion and matrix proteins and proteases. Such effects mediate redox-dependent adhesion, coagulation/thrombosis, immune functions, and virus internalization. The route of PDI externalization remains elusive. Such multiple redox effects of PDI may contribute to its conspicuous expression and functional role in disease, rendering PDI family members putative redox cell signaling adaptors.
Collapse
Affiliation(s)
- Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, 05403-000 São Paulo, Brazil.
| | | | | |
Collapse
|
44
|
Amanso AM, Griendling KK. Differential roles of NADPH oxidases in vascular physiology and pathophysiology. Front Biosci (Schol Ed) 2012; 4:1044-64. [PMID: 22202108 DOI: 10.2741/s317] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) are produced by all vascular cells and regulate the major physiological functions of the vasculature. Production and removal of ROS are tightly controlled and occur in discrete subcellular locations, allowing for specific, compartmentalized signaling. Among the many sources of ROS in the vessel wall, NADPH oxidases are implicated in physiological functions such as control of vasomotor tone, regulation of extracellular matrix and phenotypic modulation of vascular smooth muscle cells. They are involved in the response to injury, whether as an oxygen sensor during hypoxia, as a regulator of protein processing, as an angiogenic stimulus, or as a mechanism of wound healing. These enzymes have also been linked to processes leading to disease development, including migration, proliferation, hypertrophy, apoptosis and autophagy. As a result, NADPH oxidases participate in atherogenesis, systemic and pulmonary hypertension and diabetic vascular disease. The role of ROS in each of these processes and diseases is complex, and a more full understanding of the sources, targets, cell-specific responses and counterbalancing mechanisms is critical for the rational development of future therapeutics.
Collapse
Affiliation(s)
- Angelica M Amanso
- Department of Medicine, Division of Cardiology, Emory University, Division of Cardiology, Atlanta, GA 30322, USA
| | | |
Collapse
|
45
|
de Andrade CR, Stolf BS, Debbas V, Rosa DS, Kalil J, Coelho V, Laurindo FRM. Quiescin sulfhydryl oxidase (QSOX) is expressed in the human atheroma core: possible role in apoptosis. In Vitro Cell Dev Biol Anim 2011; 47:716-27. [PMID: 22069028 DOI: 10.1007/s11626-011-9461-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 09/26/2011] [Indexed: 12/30/2022]
Abstract
Quiescin sulfhydryl oxidases (QSOXs) catalyze the formation of disulfide bonds in peptides and proteins, and in vertebrates comprise two proteins: QSOX1 and QSOX2. QSOX1, the most extensively studied type, has been implicated in protein folding, production of extracellular matrix, redox regulation, protection from apoptosis, angiogenesis, and cell differentiation. Atherosclerosis is an immunopathological condition in which redox processes, apoptosis, cell differentiation, and matrix secretion/maturation have critical roles. Considering these data, we hypothesized that QSOX1 could be involved in this disease, possibly reducing apoptosis and angiogenesis inside the plaque. QSOX1 labeling in normal human carotid vessels showed predominant expression by endothelium, subendothelium, and adventitia. In atherosclerotic plaques, however, QSOX1 was highly expressed in macrophages at the lipid core. QSOX1 expression was also studied in terms of mRNA and protein in cell types present in plaques under apoptotic or activating stimuli, emulating conditions found in the atherosclerotic process. QSOX1 mRNA increased in endothelial cells and macrophages after the induction of apoptosis. At the protein level, the correlation between apoptosis and QSOX1 expression was not evident in all cell types, possibly because of a rapid secretion of QSOX1. Our results propose for the first time possible roles for QSOX1 in atherosclerosis, being upregulated in endothelial cells and macrophages by apoptosis and cell activation, and possibly controlling these processes, as well as angiogenesis. The quantitative differences in QSOX1 induction may depend on the cell type and also on local factors.
Collapse
Affiliation(s)
- Claudia R de Andrade
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
46
|
Stolf BS, Smyrnias I, Lopes LR, Vendramin A, Goto H, Laurindo FRM, Shah AM, Santos CXC. Protein disulfide isomerase and host-pathogen interaction. ScientificWorldJournal 2011; 11:1749-61. [PMID: 22125433 PMCID: PMC3201685 DOI: 10.1100/2011/289182] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 09/07/2011] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) production by immunological cells is known to cause damage to pathogens. Increasing evidence accumulated in the last decade has shown, however, that ROS (and redox signals) functionally regulate different cellular pathways in the host-pathogen interaction. These especially affect (i) pathogen entry through protein redox switches and redox modification (i.e., intra- and interdisulfide and cysteine oxidation) and (ii) phagocytic ROS production via Nox family NADPH oxidase enzyme and the control of phagolysosome function with key implications for antigen processing. The protein disulfide isomerase (PDI) family of redox chaperones is closely involved in both processes and is also implicated in protein unfolding and trafficking across the endoplasmic reticulum (ER) and towards the cytosol, a thiol-based redox locus for antigen processing. Here, we summarise examples of the cellular association of host PDI with different pathogens and explore the possible roles of pathogen PDIs in infection. A better understanding of these complex regulatory steps will provide insightful information on the redox role and coevolutional biological process, and assist the development of more specific therapeutic strategies in pathogen-mediated infections.
Collapse
Affiliation(s)
- Beatriz S Stolf
- Department of Parasitology, University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
47
|
de A. Paes AM, Veríssimo-Filho S, Guimarães LL, Silva ACB, Takiuti JT, Santos CXC, Janiszewski M, Laurindo FRM, Lopes LR. Protein disulfide isomerase redox-dependent association with p47phox: evidence for an organizer role in leukocyte NADPH oxidase activation. J Leukoc Biol 2011; 90:799-810. [DOI: 10.1189/jlb.0610324] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
48
|
Civelek M, Manduchi E, Riley RJ, Stoeckert CJ, Davies PF. Coronary artery endothelial transcriptome in vivo: identification of endoplasmic reticulum stress and enhanced reactive oxygen species by gene connectivity network analysis. ACTA ACUST UNITED AC 2011; 4:243-52. [PMID: 21493819 DOI: 10.1161/circgenetics.110.958926] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial function is central to the localization of atherosclerosis. The in vivo endothelial phenotypic footprints of arterial bed identity and site-specific atherosusceptibility are addressed. METHODS AND RESULTS Ninety-eight endothelial cell samples from 13 discrete coronary and noncoronary arterial regions of varying susceptibilities to atherosclerosis were isolated from 76 normal swine. Transcript profiles were analyzed to determine the steady-state in vivo endothelial phenotypes. An unsupervised systems biology approach using weighted gene coexpression networks showed highly correlated endothelial genes. Connectivity network analysis identified 19 gene modules, 12 of which showed significant association with circulatory bed classification. Differential expression of 1300 genes between coronary and noncoronary artery endothelium suggested distinct coronary endothelial phenotypes, with highest significance expressed in gene modules enriched for biological functions related to endoplasmic reticulum (ER) stress and unfolded protein binding, regulation of transcription and translation, and redox homeostasis. Furthermore, within coronary arteries, comparison of endothelial transcript profiles of susceptible proximal regions to protected distal regions suggested the presence of ER stress conditions in susceptible sites. Accumulation of reactive oxygen species throughout coronary endothelium was greater than in noncoronary endothelium consistent with coronary artery ER stress and lower endothelial expression of antioxidant genes in coronary arteries. CONCLUSIONS Gene connectivity analyses discriminated between coronary and noncoronary endothelial transcript profiles and identified differential transcript levels associated with increased ER and oxidative stress in coronary arteries consistent with enhanced susceptibility to atherosclerosis.
Collapse
Affiliation(s)
- Mete Civelek
- Department of Bioengineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
49
|
Amanso AM, Debbas V, Laurindo FRM. Proteasome inhibition represses unfolded protein response and Nox4, sensitizing vascular cells to endoplasmic reticulum stress-induced death. PLoS One 2011; 6:e14591. [PMID: 21297867 PMCID: PMC3027620 DOI: 10.1371/journal.pone.0014591] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Accepted: 12/06/2010] [Indexed: 12/21/2022] Open
Abstract
Background Endoplasmic reticulum (ER) stress has pathophysiological relevance in vascular diseases and merges with proteasome function. Proteasome inhibition induces cell stress and may have therapeutic implications. However, whether proteasome inhibition potentiates ER stress-induced apoptosis and the possible mechanisms involved in this process are unclear. Methodology/Principal Findings Here we show that proteasome inhibition with MG132, per se at non-lethal levels, sensitized vascular smooth muscle cells to caspase-3 activation and cell death during ER stress induced by tunicamycin (Tn). This effect was accompanied by suppression of both proadaptive (KDEL chaperones) and proapoptotic (CHOP/GADD153) unfolded protein response markers, although, intriguingly, the splicing of XBP1 was markedly enhanced and sustained. In parallel, proteasome inhibition completely prevented ER stress-induced increase in NADPH oxidase activity, as well as increases in Nox4 isoform and protein disulfide isomerase mRNA expression. Increased Akt phosphorylation due to proteasome inhibition partially offset the proapoptotic effect of Tn or MG132. Although proteasome inhibition enhanced oxidative stress, reactive oxygen species scavenging had no net effect on sensitization to Tn or MG132-induced cell death. Conclusion/Relevance These data indicate unfolded protein response-independent pathways whereby proteasome inhibition sensitizes vascular smooth muscle to ER stress-mediated cell death. This may be relevant to understand the therapeutic potential of such compounds in vascular disease associated with increased neointimal hyperplasia.
Collapse
Affiliation(s)
- Angélica M. Amanso
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Victor Debbas
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Francisco R. M. Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
50
|
Abstract
The paper reviews the rapidly expanding pool of information on cellular and molecular mechanisms of autophagy, including autophagy types, macroautophagy induction, formation of autophagosomes and cross-talk between autophagy and apoptosis. Special attention is given to generation of reactive oxygen species (ROS) in various cellular compartments of cells under stress conditions inducing autophagy. The roles of hydrogen peroxide and superoxide in autophagy are analysed based on the recent experimental work. The relation between ROS and life span prolongation is briefly discussed, with the final conclusion that the paradox of dual role of ROS in life and death may be solved to a considerable extent due to research on autophagy.
Collapse
Affiliation(s)
- Irena Szumiel
- Center of Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland.
| |
Collapse
|