1
|
Kong YX, Chiu J, Passam FH. "Sticki-ER": Functions of the Platelet Endoplasmic Reticulum. Antioxid Redox Signal 2024; 41:637-660. [PMID: 38284332 DOI: 10.1089/ars.2024.0566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Significance: The primary role of platelets is to generate a thrombus by platelet activation. Platelet activation relies on calcium mobilization from the endoplasmic reticulum (ER). ER resident proteins, which are externalized upon platelet activation, are essential for the function of platelet surface receptors and intercellular interactions. Recent Advances: The platelet ER is a conduit for changes in cellular function in response to the extracellular milieu. ER homeostasis is maintained by an appropriate redox balance, regulated calcium stores and normal protein folding. Alterations in ER function and ER stress results in ER proteins externalizing to the cell surface, including members of the protein disulfide isomerase family (PDIs) and chaperones. Critical Issues: The platelet ER is central to platelet function, but our understanding of its regulation is incomplete. Previous studies have focused on the function of PDIs in the extracellular space, and much less on their intracellular role. How platelets maintain ER homeostasis and how they direct ER chaperone proteins to facilitate intercellular signalling is unknown. Future Directions: An understanding of ER functions in the platelet is essential as these may determine critical platelet activities such as secretion and adhesion. Studies are necessary to understand the redox reactions of PDIs in the intracellular versus extracellular space, as these differentially affect platelet function. An unresolved question is how platelet ER proteins control calcium release. Regulation of protein folding in the platelet and downstream pathways of ER stress require further evaluation. Targeting the platelet ER may have therapeutic application in metabolic and neoplastic disease.
Collapse
Affiliation(s)
- Yvonne X Kong
- Haematology Research Group, Charles Perkins Centre; The University of Sydney, Camperdown, New South Wales, Australia
- Central Clinical School, Faculty of Medicine and Health; The University of Sydney, Camperdown, New South Wales, Australia
- Department of Haematology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Joyce Chiu
- ACRF Centenary Cancer Research Centre, The Centenary Institute; The University of Sydney, Camperdown, New South Wales, Australia
| | - Freda H Passam
- Haematology Research Group, Charles Perkins Centre; The University of Sydney, Camperdown, New South Wales, Australia
- Central Clinical School, Faculty of Medicine and Health; The University of Sydney, Camperdown, New South Wales, Australia
- Department of Haematology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
2
|
Porto FG, Tanaka LY, de Bessa TC, Oliveira PVS, Souza JMFD, Kajihara D, Fernandes CG, Santos PN, Laurindo FRM. Evidence for a protective role of Protein Disulfide Isomerase-A1 against aortic dissection. Atherosclerosis 2023; 382:117283. [PMID: 37774430 DOI: 10.1016/j.atherosclerosis.2023.117283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND AND AIMS Redox signaling is involved in the pathophysiology of aortic aneurysm/dissection. Protein Disulfide Isomerases and its prototype PDIA1 are thiol redox chaperones mainly from endoplasmic reticulum (ER), while PDIA1 cell surface pool redox-regulates thrombosis, cytoskeleton remodeling and integrin activation, which are mechanisms involved in aortic disease. Here we investigate the roles of PDIA1 in aortic dissection. METHODS Initially, we assessed the outcome of aortic aneurysm/dissection in transgenic PDIA1-overexpressing FVB mice using a model of 28-day exposure to lysyl oxidase inhibitor BAPN plus angiotensin-II infusion. In a second protocol, we assessed the effects of PDIA1 inhibitor isoquercetin (IQ) against aortic dissection in C57BL/6 mice exposed to BAPN for 28 days. RESULTS Transgenic PDIA1 overexpression associated with ca. 50% (p = 0.022) decrease (vs.wild-type) in mortality due to abdominal aortic rupture and protected against elastic fiber breaks in thoracic aorta. Conversely, exposure of mice to IQ increased thoracic aorta dissection-related mortality rates, from ca. 18%-50% within 28-days (p = 0.019); elastic fiber disruption and collagen deposition were also enhanced. The structurally-related compound diosmetin, which does not inhibit PDI, had negligible effects. In parallel, stretch-tension curves indicated that IQ amplified a ductile-type of biomechanical failure vs. control or BAPN-exposed mice aortas. IQ-induced effects seemed unassociated with nonspecific antioxidant effects or ER stress. In both models, echocardiographic analysis of surviving mice suggested that aortic rupture was dissociated from progressive dilatation. CONCLUSIONS Our data indicate a protective role of PDIA1 against aortic dissection/rupture and potentially uncovers a novel integrative mechanism coupling redox and biomechanical homeostasis in vascular remodeling.
Collapse
Affiliation(s)
- Fernando Garcez Porto
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Leonardo Yuji Tanaka
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Tiphany Coralie de Bessa
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Percillia Victoria Santos Oliveira
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Júlia Martins Felipe de Souza
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Daniela Kajihara
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Carolina Gonçalves Fernandes
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Patricia Nolasco Santos
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Francisco Rafael Martins Laurindo
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
3
|
Trostchansky A, Alarcon M. An Overview of Two Old Friends Associated with Platelet Redox Signaling, the Protein Disulfide Isomerase and NADPH Oxidase. Biomolecules 2023; 13:biom13050848. [PMID: 37238717 DOI: 10.3390/biom13050848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress participates at the baseline of different non-communicable pathologies such as cardiovascular diseases. Excessive formation of reactive oxygen species (ROS), above the signaling levels necessary for the correct function of organelles and cells, may contribute to the non-desired effects of oxidative stress. Platelets play a relevant role in arterial thrombosis, by aggregation triggered by different agonists, where excessive ROS formation induces mitochondrial dysfunction and stimulate platelet activation and aggregation. Platelet is both a source and a target of ROS, thus we aim to analyze both the platelet enzymes responsible for ROS generation and their involvement in intracellular signal transduction pathways. Among the proteins involved in these processes are Protein Disulphide Isomerase (PDI) and NADPH oxidase (NOX) isoforms. By using bioinformatic tools and information from available databases, a complete bioinformatic analysis of the role and interactions of PDI and NOX in platelets, as well as the signal transduction pathways involved in their effects was performed. We focused the study on analyzing whether these proteins collaborate to control platelet function. The data presented in the current manuscript support the role that PDI and NOX play on activation pathways necessary for platelet activation and aggregation, as well as on the platelet signaling imbalance produced by ROS production. Our data could be used to design specific enzyme inhibitors or a dual inhibition for these enzymes with an antiplatelet effect to design promising treatments for diseases involving platelet dysfunction.
Collapse
Affiliation(s)
- Andrés Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Marcelo Alarcon
- Thrombosis Research Center, Universidad de Talca, Talca 3460000, Chile
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
4
|
Miyano K, Okamoto S, Kajikawa M, Kiyohara T, Kawai C, Yamauchi A, Kuribayashi F. Regulation of Derlin-1-mediated degradation of NADPH oxidase partner p22 phox by thiol modification. Redox Biol 2022; 56:102479. [PMID: 36122532 PMCID: PMC9486109 DOI: 10.1016/j.redox.2022.102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
The transmembrane protein p22phox heterodimerizes with NADPH oxidase (Nox) 1–4 and is essential for the reactive oxygen species-producing capacity of oxidases. Missense mutations in the p22phox gene prevent the formation of phagocytic Nox2-based oxidase, which contributes to host defense. This results in chronic granulomatous disease (CGD), a severe primary immunodeficiency syndrome. In this study, we characterized missense mutations in p22phox (L51Q, L52P, E53V, and P55R) in the A22° type (wherein the p22phox protein is undetectable) of CGD. We demonstrated that these substitutions enhanced the degradation of the p22phox protein in the endoplasmic reticulum (ER) and the binding of p22phox to Derlin-1, a key component of ER-associated degradation (ERAD). Therefore, the L51-L52-E53-P55 sequence is responsible for protein stability in the ER. We observed that the oxidation of the thiol group of Cys-50, which is adjacent to the L51-L52-E53-P55 sequence, suppressed p22phox degradation. However, the suppression effect was markedly attenuated by the serine substitution of Cys-50. Blocking the free thiol of Cys-50 by alkylation or C50S substitution promoted the association of p22phox with Derlin-1. Derlin-1 depletion partially suppressed the degradation of p22phox mutant proteins. Furthermore, heterodimerization with p22phox (C50S) induced rapid degradation of not only Nox2 but also nonphagocytic Nox4 protein, which is responsible for redox signaling. Thus, the redox-sensitive Cys-50 appears to determine whether p22phox becomes a target for degradation by the ERAD system through its interaction with Derlin-1. Missense mutations in exon 3 of p22phox enhance the binding of p22phox to Derlin-1. Oxidation of the thiol group of p22phox Cys50 suppresses p22phox degradation. Serine substitution of Cys-50 increases the affinity of p22phox for Derlin-1. Stability of the p22phox protein is regulated by redox-sensitive Cys-50.
Collapse
Affiliation(s)
- Kei Miyano
- Department of Natural Sciences, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan; Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan.
| | - Shuichiro Okamoto
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| | - Mizuho Kajikawa
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Takuya Kiyohara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chikage Kawai
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| | - Akira Yamauchi
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| | - Futoshi Kuribayashi
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| |
Collapse
|
5
|
Brace N, Megson IL, Rossi AG, Doherty MK, Whitfield PD. SILAC-based quantitative proteomics to investigate the eicosanoid associated inflammatory response in activated macrophages. J Inflamm (Lond) 2022; 19:12. [PMID: 36050729 PMCID: PMC9438320 DOI: 10.1186/s12950-022-00309-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Macrophages play a central role in inflammation by phagocytosing invading pathogens, apoptotic cells and debris, as well as mediating repair of tissues damaged by trauma. In order to do this, these dynamic cells generate a variety of inflammatory mediators including eicosanoids such as prostaglandins, leukotrienes and hydroxyeicosatraenoic acids (HETEs) that are formed through the cyclooxygenase, lipoxygenase and cytochrome P450 pathways. The ability to examine the effects of eicosanoid production at the protein level is therefore critical to understanding the mechanisms associated with macrophage activation. RESULTS This study presents a stable isotope labelling with amino acids in cell culture (SILAC) -based proteomics strategy to quantify the changes in macrophage protein abundance following inflammatory stimulation with Kdo2-lipid A and ATP, with a focus on eicosanoid metabolism and regulation. Detailed gene ontology analysis, at the protein level, revealed several key pathways with a decrease in expression in response to macrophage activation, which included a promotion of macrophage polarisation and dynamic changes to energy requirements, transcription and translation. These findings suggest that, whilst there is evidence for the induction of a pro-inflammatory response in the form of prostaglandin secretion, there is also metabolic reprogramming along with a change in cell polarisation towards a reduced pro-inflammatory phenotype. CONCLUSIONS Advanced quantitative proteomics in conjunction with functional pathway network analysis is a useful tool to investigate the molecular pathways involved in inflammation.
Collapse
Affiliation(s)
- Nicole Brace
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Ian L Megson
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mary K Doherty
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Phillip D Whitfield
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK.
- Present Address: Glasgow Polyomics, Garscube Campus, University of Glasgow, Glasgow, G61 1BD, UK.
| |
Collapse
|
6
|
Zhang Y, Miao Q, Shi S, Hao H, Li X, Pu Z, Yang Y, An H, Zhang W, Kong Y, Pang X, Gu C, Gamper N, Wu Y, Zhang H, Du X. Protein disulfide isomerase modulation of TRPV1 controls heat hyperalgesia in chronic pain. Cell Rep 2022; 39:110625. [PMID: 35385753 DOI: 10.1016/j.celrep.2022.110625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
Protein disulfide isomerase (PDI) plays a key role in maintaining cellular homeostasis by mediating protein folding via catalyzing disulfide bond formation, breakage, and rearrangement in the endoplasmic reticulum. Increasing evidence suggests that PDI can be a potential treatment target for several diseases. However, the function of PDI in the peripheral sensory nervous system is unclear. Here we report the expression and secretion of PDI from primary sensory neurons is upregulated in inflammatory and neuropathic pain models. Deletion of PDI in nociceptive DRG neurons results in a reduction in inflammatory and neuropathic heat hyperalgesia. We demonstrate that secreted PDI activates TRPV1 channels through oxidative modification of extracellular cysteines of the channel, indicating that PDI acts as an unconventional positive modulator of TRPV1. These findings suggest that PDI in primary sensory neurons plays an important role in development of heat hyperalgesia and can be a potential therapeutic target for chronic pain.
Collapse
Affiliation(s)
- Yongxue Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Pharmacy, The First Hospital of Handan, Handan, Hebei, China
| | - Qi Miao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sai Shi
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, Hebei, China
| | - Han Hao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinmeng Li
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zeyao Pu
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yakun Yang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, Hebei, China
| | - Wei Zhang
- Department of Spinal Surgery of the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Youzhen Kong
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xu Pang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cunyang Gu
- Department of Pathology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China; Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, UK
| | - Yi Wu
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China.
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Abstract
Significance: Since protein disulfide isomerase (PDI) was first described in 1963, researchers have shown conclusively that PDI and sibling proteins are quintessential for thrombus formation. PDI, endoplasmic reticulum protein (ERp)5, ERp57, and ERp72 are released from platelets and vascular cells and interact with integrin αIIbβ3 on the outer surface of platelets. Recent Advances: At the cell surface they influence protein folding and function, propagating thrombosis and maintaining hemostasis. TMX1, which is a transmembrane thiol isomerase, is the first family member shown to negatively regulate platelets. Targets of thiol isomerases have been identified, including integrin α2β1, Von Willebrand Factor, GpIbα, nicotinamide adenine dinucleotide phosphate oxidase (Nox)-1, Nox-2, and tissue factor, all of which are pro-thrombotic, and several of which are on the cell surface. In spite of this, PDI can paradoxically catalyze the delivery of nitric oxide to platelets, which decrease thrombus formation. Critical Issues: Although the overall effect of PDI is to positively regulate platelet activation, it is still unclear how thiol isomerases function in pro-thrombotic states, such as obesity, diabetes, and cancer. In parallel, there has been a surge in the development of novel thiol isomerase inhibitors, which display selectivity, potency and modulate thrombosis and hemostasis. The availability of selective thiol isomerase inhibitors has culminated in clinical trials, with promising outcomes for the prevention of cancer-associated thrombosis. Future Directions: Altogether, thiol isomerases are perceived as an orchestrating force that regulates thrombus development. In the current review, we will explore the history of PDI in cardiovascular biology, detail known mechanisms of action, and summarize known thiol isomerase inhibitors.
Collapse
Affiliation(s)
- Renato Simões Gaspar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
8
|
Mustapha S, Mohammed M, Azemi AK, Yunusa I, Shehu A, Mustapha L, Wada Y, Ahmad MH, Ahmad WANW, Rasool AHG, Mokhtar SS. Potential Roles of Endoplasmic Reticulum Stress and Cellular Proteins Implicated in Diabesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8830880. [PMID: 33995826 PMCID: PMC8099518 DOI: 10.1155/2021/8830880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
The role of the endoplasmic reticulum (ER) has evolved from protein synthesis, processing, and other secretory pathways to forming a foundation for lipid biosynthesis and other metabolic functions. Maintaining ER homeostasis is essential for normal cellular function and survival. An imbalance in the ER implied stressful conditions such as metabolic distress, which activates a protective process called unfolded protein response (UPR). This response is activated through some canonical branches of ER stress, i.e., the protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1α (IRE1α), and activating transcription factor 6 (ATF6). Therefore, chronic hyperglycemia, hyperinsulinemia, increased proinflammatory cytokines, and free fatty acids (FFAs) found in diabesity (a pathophysiological link between obesity and diabetes) could lead to ER stress. However, limited data exist regarding ER stress and its association with diabesity, particularly the implicated proteins and molecular mechanisms. Thus, this review highlights the role of ER stress in relation to some proteins involved in diabesity pathogenesis and provides insight into possible pathways that could serve as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Pulau Pinang, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, College of Pharmacy, Columbia, SC, USA
| | - Aishatu Shehu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Lukman Mustapha
- Department of Pharmaceutical and Medicinal Chemistry, Kaduna State University, Kaduna, Nigeria
| | - Yusuf Wada
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mubarak Hussaini Ahmad
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
- School of Pharmacy Technician, Aminu Dabo College of Health Sciences and Technology, Kano, Nigeria
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
9
|
Karatas E, Raymond AA, Leon C, Dupuy JW, Di-Tommaso S, Senant N, Collardeau-Frachon S, Ruiz M, Lachaux A, Saltel F, Bouchecareilh M. Hepatocyte proteomes reveal the role of protein disulfide isomerase 4 in alpha 1-antitrypsin deficiency. JHEP Rep 2021; 3:100297. [PMID: 34151245 PMCID: PMC8192868 DOI: 10.1016/j.jhepr.2021.100297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 11/25/2022] Open
Abstract
Background & Aims A single point mutation in the Z-variant of alpha 1-antitrypsin (Z-AAT) alone can lead to both a protein folding and trafficking defect, preventing its exit from the endoplasmic reticulum (ER), and the formation of aggregates that are retained as inclusions within the ER of hepatocytes. These defects result in a systemic AAT deficiency (AATD) that causes lung disease, whereas the ER-retained aggregates can induce severe liver injury in patients with ZZ-AATD. Unfortunately, therapeutic approaches are still limited and liver transplantation represents the only curative treatment option. To overcome this limitation, a better understanding of the molecular basis of ER aggregate formation could provide new strategies for therapeutic intervention. Methods Our functional and omics approaches here based on human hepatocytes from patients with ZZ-AATD have enabled the identification and characterisation of the role of the protein disulfide isomerase (PDI) A4/ERP72 in features of AATD-mediated liver disease. Results We report that 4 members of the PDI family (PDIA4, PDIA3, P4HB, and TXNDC5) are specifically upregulated in ZZ-AATD liver samples from adult patients. Furthermore, we show that only PDIA4 knockdown or alteration of its activity by cysteamine treatment can promote Z-AAT secretion and lead to a marked decrease in Z aggregates. Finally, detailed analysis of the Z-AAT interactome shows that PDIA4 silencing provides a more conducive environment for folding of the Z mutant, accompanied by reduction of Z-AAT-mediated oxidative stress, a feature of AATD-mediated liver disease. Conclusions PDIA4 is involved in AATD-mediated liver disease and thus represents a therapeutic target for inhibition by drugs such as cysteamine. PDI inhibition therefore represents a potential therapeutic approach for treatment of AATD. Lay summary Protein disulfide isomerase (PDI) family members, and particularly PDIA4, are upregulated and involved in alpha 1-antitrypsin deficiency (AATD)-mediated liver disease in adults. PDI inhibition upon cysteamine treatment leads to improvements in features of AATD and hence represents a therapeutic approach for treatment of AATD-mediated liver disease. PDIA4 is upregulated and involved in alpha 1-antitrypsin deficiency (AATD)-mediated liver disease in adults. Knockdown of PDIA4 by siRNA or inhibition upon cysteamine treatment leads to improvements in features of AATD. RNA interference against PDIA4 or cysteamine represent approaches for treatment of AATD-mediated liver disease.
Collapse
Key Words
- AAT, alpha 1-antitrypsin
- AATD, alpha 1-antitrypsin deficiency
- Alpha 1-antitrypsin deficiency
- CF, cystic fibrosis
- CFTR, cystic fibrosis transmembrane conductance regulator
- Cysteamine
- ER, endoplasmic reticulum
- FFPE, formalin-fixed paraffin-embedded
- FKBP10, FK506-binding protein (FKBP) isoform 10
- HCC, hepatocellular carcinoma
- IHC, immunohistochemistry
- IP, immunoprecipitation
- Liver damage
- NHK, null Hong Kong variant of AAT
- P4HB, prolyl 4-hydroxylase subunit beta/PDIA1
- PDI, protein disulfide isomerase
- PDIA3, protein disulfide isomerase family A member 3/ERP57
- PDIA4
- PDIA4, protein disulfide isomerase family A member 4/ERP70/ERP72
- PDIi, PDI inhibitors
- Protein disulfide isomerase
- ROS, reactive oxygen species
- SURF4, proteins Surfeit 4
- Scr, scramble
- TRX, thioredoxin
- TXNDC5, thioredoxin domain containing 5/PDIA15
- Treatment
- WT, wild-type
- Z-AAT, alpha 1-antitrypsin Z variant
- ZZ, homozygosis for the Z mutant allele
- siRNA, small RNA interference
- ΔF508-CFTR, most common mutation of CFTR, which deletes phenylalanine508
Collapse
Affiliation(s)
- Esra Karatas
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France
| | - Anne-Aurélie Raymond
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France.,Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | - Céline Leon
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France
| | | | - Sylvaine Di-Tommaso
- Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | - Nathalie Senant
- Plateforme d'histopathologie, TBM-Core US 005, Bordeaux, France
| | - Sophie Collardeau-Frachon
- Department of Pathology, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Mathias Ruiz
- Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Alain Lachaux
- Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Frédéric Saltel
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France.,Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | | |
Collapse
|
10
|
Protein Disulphide Isomerase and NADPH Oxidase 1 Cooperate to Control Platelet Function and Are Associated with Cardiometabolic Disease Risk Factors. Antioxidants (Basel) 2021; 10:antiox10030497. [PMID: 33806982 PMCID: PMC8004975 DOI: 10.3390/antiox10030497] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Protein disulphide isomerase (PDI) and NADPH oxidase 1 (Nox-1) regulate platelet function and reactive oxygen species (ROS) generation, suggesting potentially interdependent roles. Increased platelet reactivity and ROS production have been correlated with cardiometabolic disease risk factors. Objectives: To establish whether PDI and Nox-1 cooperate to control platelet function. Methods: Immunofluorescence microscopy was utilised to determine expression and localisation of PDI and Nox-1. Platelet aggregation, fibrinogen binding, P-selectin exposure, spreading and calcium mobilization were measured as markers of platelet function. A cross-sectional population study (n = 136) was conducted to assess the relationship between platelet PDI and Nox-1 levels and cardiometabolic risk factors. Results: PDI and Nox-1 co-localized upon activation induced by the collagen receptor GPVI. Co-inhibition of PDI and Nox-1 led to additive inhibition of GPVI-mediated platelet aggregation, activation and calcium flux. This was confirmed in murine Nox-1−/− platelets treated with PDI inhibitor bepristat, without affecting bleeding. PDI and Nox-1 together contributed to GPVI signalling that involved the phosphorylation of p38 MAPK, p47phox, PKC and Akt. Platelet PDI and Nox-1 levels were upregulated in obesity, with platelet Nox-1 also elevated in hypertensive individuals. Conclusions: We show that PDI and Nox-1 cooperate to control platelet function and are associated with cardiometabolic risk factors.
Collapse
|
11
|
Victor P, Umapathy D, George L, Juttada U, Ganesh GV, Amin KN, Viswanathan V, Ramkumar KM. Crosstalk between endoplasmic reticulum stress and oxidative stress in the progression of diabetic nephropathy. Cell Stress Chaperones 2021; 26:311-321. [PMID: 33161510 PMCID: PMC7925747 DOI: 10.1007/s12192-020-01176-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence in substantiating the roles of endoplasmic reticulum stress, oxidative stress, and inflammatory responses and their interplay is evident in various diseases. However, an in-depth mechanistic understanding of the crosstalk between the intracellular stress signaling pathways and inflammatory responses and their participation in disease progression has not yet been explored. Progress has been made in our understanding of the cross talk and integrated stress signaling network between endoplasmic reticulum stress and oxidative stress towards the pathogenesis of diabetic nephropathy. In this present study, we studied the crosstalk between the endoplasmic reticulum stress and oxidative stress by understanding the role of protein disulfide isomerase and endoplasmic reticulum oxidase 1α, a key player in redox protein folding in the endoplasmic reticulum. We had recruited a total of 90 subjects and divided into three groups (control (n = 30), type 2 diabetes mellitus (n = 30), and diabetic nephropathy (n = 30)). We found that endoplasmic reticulum stress markers, activating transcription factor 6, inositol-requiring enzyme 1α, protein kinase RNA-like endoplasmic reticulum kinase, C/EBP homologous protein, and glucose-regulated protein-78; oxidative stress markers, thioredoxin-interacting protein and cytochrome b-245 light chain; and the crosstalk markers, protein disulfide isomerase and endoplasmic reticulum oxidase-1α, were progressively elevated in type 2 diabetes mellitus and diabetic nephropathy subjects. The association between the crosstalk markers showed a positive correlation with endoplasmic reticulum stress and oxidative stress markers. Further, the interplay between endoplasmic reticulum stress and oxidative stress was investigated in vitro using a human leukemic monocytic cell line under a hyperglycemic environment and examined the expression of protein disulfide isomerase and endoplasmic reticulum oxidase-1α. DCFH-DA assay and flow cytometry were performed to detect the production of free radicals. Further, phosphorylation of eIF2α in high glucose-exposed cells was studied using western blot. In conclusion, our results shed light on the crosstalk between endoplasmic reticulum stress and oxidative stress and significantly contribute to the onset and progression of diabetic nephropathy and therefore represent the major therapeutic targets for alleviating micro- and macrovascular complications associated with this metabolic disturbance. Graphical abstract.
Collapse
Affiliation(s)
- Paul Victor
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - Dhamodharan Umapathy
- Life Science Division, SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India
| | - Leema George
- Department of Biochemistry and Molecular Genetics, Prof. M. Viswanathan Diabetes Research Centre and M.V. Hospital for Diabetes (WHO Collaborating Centre for Research, Education & Training in Diabetes), Royapuram, Chennai, Tamil Nadu, 600013, India
| | - Udyama Juttada
- Department of Biochemistry and Molecular Genetics, Prof. M. Viswanathan Diabetes Research Centre and M.V. Hospital for Diabetes (WHO Collaborating Centre for Research, Education & Training in Diabetes), Royapuram, Chennai, Tamil Nadu, 600013, India
| | - Goutham V Ganesh
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - Karan Naresh Amin
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - Vijay Viswanathan
- Department of Biochemistry and Molecular Genetics, Prof. M. Viswanathan Diabetes Research Centre and M.V. Hospital for Diabetes (WHO Collaborating Centre for Research, Education & Training in Diabetes), Royapuram, Chennai, Tamil Nadu, 600013, India.
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603 203, India.
- Life Science Division, SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India.
| |
Collapse
|
12
|
Fernandes DC, Wosniak J, Gonçalves RC, Tanaka LY, Fernandes CG, Zanatta DB, de Mattos ABM, Strauss BE, Laurindo FRM. PDIA1 acts as master organizer of NOX1/NOX4 balance and phenotype response in vascular smooth muscle. Free Radic Biol Med 2021; 162:603-614. [PMID: 33227407 DOI: 10.1016/j.freeradbiomed.2020.11.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/29/2020] [Accepted: 11/17/2020] [Indexed: 02/05/2023]
Abstract
Changes in vascular smooth muscle cell (VSMC) phenotype underlie disease pathophysiology and are strongly regulated by NOX NADPH oxidases, with NOX1 favoring synthetic proliferative phenotype and NOX4 supporting differentiation. Growth factor-triggered NOX1 expression/activity strictly depends on the chaperone oxidoreductase protein disulfide isomerase-A1 (PDIA1). Intracellular PDIA1 is required for VSMC migration and cytoskeleton organization, while extracellular PDIA1 fine-tunes cytoskeletal mechanoadaptation and vascular remodeling. We hypothesized that PDIA1 orchestrates NOX1/NOX4 balance and VSMC phenotype. Using an inducible PDIA1 overexpression model in VSMC, we showed that early PDIA1 overexpression (for 24-48 h) increased NOX1 expression, hydrogen peroxide steady-state levels and spontaneous VSMC migration distances. Sustained PDIA1 overexpression for 72 h and 96 h supported high NOX1 levels while also increasing NOX4 expression and, remarkably, switched VSMC phenotype to differentiation. Differentiation was preceded by increased nuclear myocardin and serum response factor-response element activation, with no change in cell viability. Both NOX1 and hydrogen peroxide were necessary for later PDIA1-induced VSMC differentiation. In primary VSMC, PDIA1 knockdown decreased nuclear myocardin and increased the proliferating cell nuclear antigen expression. Newly-developed PDIA1-overexpressing mice (TgPDIA1) exhibited normal general and cardiovascular baseline phenotypes. However, in TgPDIA1 carotids, NOX1 was decreased while NOX4 and calponin expressions were enhanced, indicating overdifferentiation vs. normal carotids. Moreover, in a rabbit overdistension injury model during late vascular repair, PDIA1 silencing impaired VSMC redifferentiation and NOX1/NOX4 balance. Our results suggest a model in which PDIA1 acts as an upstream organizer of NOX1/NOX4 balance and related VSMC phenotype, accounting for baseline differentiation setpoint.
Collapse
Affiliation(s)
- Denise C Fernandes
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
| | - João Wosniak
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Renata C Gonçalves
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Carolina G Fernandes
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Daniela B Zanatta
- Viral Vector Laboratory, Center for Translational Research in Oncology/LIM24, Cancer Institute of Sao Paulo, School of Medicine, Sao Paulo, Brazil
| | - Ana Barbosa M de Mattos
- Laboratory of Genetic and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Bryan E Strauss
- Viral Vector Laboratory, Center for Translational Research in Oncology/LIM24, Cancer Institute of Sao Paulo, School of Medicine, Sao Paulo, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
| |
Collapse
|
13
|
PDI-Regulated Disulfide Bond Formation in Protein Folding and Biomolecular Assembly. Molecules 2020; 26:molecules26010171. [PMID: 33396541 PMCID: PMC7794689 DOI: 10.3390/molecules26010171] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
Disulfide bonds play a pivotal role in maintaining the natural structures of proteins to ensure their performance of normal biological functions. Moreover, biological molecular assembly, such as the gluten network, is also largely dependent on the intermolecular crosslinking via disulfide bonds. In eukaryotes, the formation and rearrangement of most intra- and intermolecular disulfide bonds in the endoplasmic reticulum (ER) are mediated by protein disulfide isomerases (PDIs), which consist of multiple thioredoxin-like domains. These domains assist correct folding of proteins, as well as effectively prevent the aggregation of misfolded ones. Protein misfolding often leads to the formation of pathological protein aggregations that cause many diseases. On the other hand, glutenin aggregation and subsequent crosslinking are required for the formation of a rheologically dominating gluten network. Herein, the mechanism of PDI-regulated disulfide bond formation is important for understanding not only protein folding and associated diseases, but also the formation of functional biomolecular assembly. This review systematically illustrated the process of human protein disulfide isomerase (hPDI) mediated disulfide bond formation and complemented this with the current mechanism of wheat protein disulfide isomerase (wPDI) catalyzed formation of gluten networks.
Collapse
|
14
|
Wang L, Deng L, Lin N, Shi Y, Chen J, Zhou Y, Chen D, Liu S, Li C. Berberine inhibits proliferation and apoptosis of vascular smooth muscle cells induced by mechanical stretch via the PDI/ERS and MAPK pathways. Life Sci 2020; 259:118253. [PMID: 32795536 DOI: 10.1016/j.lfs.2020.118253] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022]
Abstract
AIMS We recently demonstrated that mechanical stretch increases the proliferation and apoptosis of vascular smooth muscle cells (VSMCs) by activating the protein disulfide isomerase (PDI) redox system, thus accelerating atherosclerotic lesion formation in the transplanted vein. At present, there are no efficient intervention measures to prevent this phenomenon. Berberine inhibits pathological vascular remodeling caused by hypertension, but the underlying mechanism is controversial. Herein, we investigate the role of berberine and the underlying mechanism of its effects on mechanical stretch-induced VSMC proliferation and apoptosis. MAIN METHODS Mouse VSMCs cultivated on flexible membranes were pretreated for 1 h with one of the following substances: berberine, PDI inhibitor bacitracin, MAPK inhibitors, or ERS inhibitor 4-PBA. VSMCs were then subjected to mechanical stretch. Immunofluorescence and western blot were used to detect proliferation and apoptosis, as well as to analyze signaling pathways in VSMCs. KEY FINDINGS Our results showed that berberine inhibits the PDI-endoplasmic reticulum stress system, thereby attenuating the simultaneous increase of VSMC proliferation and apoptosis in response to mechanical stretch. Interestingly, MAPK inhibitors PD98059, SP600125, and SB202190 significantly reduced the activation of ERS signaling cascades, and their combination with berberine had additive effects. The ERS inhibitor 4-PBA reduced PDI activation and ERS signaling, but not MAPK phosphorylation. Moreover, caspase-3 and caspase-12 were downregulated by berberine. SIGNIFICANCE These results illustrate a novel mechanism of action of berberine that has practical implications. Our data provide important insights for the prevention and treatment of vascular remodeling and diseases caused by mechanical stretching during hypertension.
Collapse
Affiliation(s)
- Linli Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Lie Deng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Ning Lin
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Yi Shi
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, China
| | - Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Yan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Dadi Chen
- Experimental Center for Basic Medical Teaching, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China.
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China.
| |
Collapse
|
15
|
Tanaka LY, Oliveira PVS, Laurindo FRM. Peri/Epicellular Thiol Oxidoreductases as Mediators of Extracellular Redox Signaling. Antioxid Redox Signal 2020; 33:280-307. [PMID: 31910038 DOI: 10.1089/ars.2019.8012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Significance: Supracellular redox networks regulating cell-extracellular matrix (ECM) and organ system architecture merge with structural and functional (catalytic or allosteric) properties of disulfide bonds. This review addresses emerging evidence that exported thiol oxidoreductases (TORs), such as thioredoxin, protein disulfide isomerases (PDIs), quiescin sulfhydryl oxidases (QSOX)1, and peroxiredoxins, composing a peri/epicellular (pec)TOR pool, mediate relevant signaling. pecTOR functions depend mainly on kinetic and spatial regulation of thiol-disulfide exchange reactions governed by redox potentials, which are modulated by exported intracellular low-molecular-weight thiols, together conferring signal specificity. Recent Advances: pecTOR redox-modulates several targets including integrins, ECM proteins, surface molecules, and plasma components, although clear-cut documentation of direct effects is lacking in many cases. TOR catalytic pathways, displaying common patterns, culminate in substrate thiol reduction, oxidation, or isomerization. Peroxiredoxins act as redox/peroxide sensors, contrary to PDIs, which are likely substrate-targeted redox modulators. Emerging evidence suggests important pecTOR roles in patho(physio)logical processes, including blood coagulation, vascular remodeling, mechanosensing, endothelial function, immune responses, and inflammation. Critical Issues: Effects of pecPDIs supporting thrombosis/platelet activation have been well documented and reached the clinical arena. Roles of pecPDIA1 in vascular remodeling/mechanosensing are also emerging. Extracellular thioredoxin and pecPDIs redox-regulate immunoinflammation. Routes of TOR externalization remain elusive and appear to involve Golgi-independent routes. pecTORs are particularly accessible drug targets. Future Directions: Further understanding mechanisms of thiol redox reactions and developing assays for assessing pecTOR redox activities remain important research avenues. Also, addressing pecTORs as disease markers and achieving more efficient/specific drugs for pecTOR modulation are major perspectives for diagnostic/therapeutic improvements.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Percillia V S Oliveira
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
16
|
Beyond bacterial killing: NADPH oxidase 2 is an immunomodulator. Immunol Lett 2020; 221:39-48. [DOI: 10.1016/j.imlet.2020.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/09/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
|
17
|
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
|
18
|
Protein disulfide isomerase in cardiovascular disease. Exp Mol Med 2020; 52:390-399. [PMID: 32203104 PMCID: PMC7156431 DOI: 10.1038/s12276-020-0401-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 01/07/2023] Open
Abstract
Protein disulfide isomerase (PDI) participates in the pathogenesis of numerous diseases. Increasing evidence indicates that intravascular cell-derived PDI plays an important role in the initiation and progression of cardiovascular diseases, including thrombosis and vascular inflammation. Recent studies with PDI conditional knockout mice have advanced our understanding of the function of cell-specific PDI in disease processes. Furthermore, the identification and development of novel small-molecule PDI inhibitors has led into a new era of PDI research that transitioned from the bench to bedside. In this review, we will discuss recent findings on the regulatory role of PDI in cardiovascular disease. Efforts to untangle the functions of a large family of enzymes could lead researchers to new therapies for diverse cardiovascular diseases. Members of the protein disulfide isomerase (PDI) family chemically modify other proteins in ways that can alter both their structure and biological activity. Jaehyung Cho of the University of Illinois at Chicago, USA and coworkers have reviewed numerous studies linking PDI with cardiovascular diseases, including thrombosis, heart attack, vascular inflammation, and stroke. The authors also report progress in developing small-molecule PDI inhibitors that could yield the treatment for these conditions.
Collapse
|
19
|
Kim YM, Youn SW, Sudhahar V, Das A, Chandhri R, Cuervo Grajal H, Kweon J, Leanhart S, He L, Toth PT, Kitajewski J, Rehman J, Yoon Y, Cho J, Fukai T, Ushio-Fukai M. Redox Regulation of Mitochondrial Fission Protein Drp1 by Protein Disulfide Isomerase Limits Endothelial Senescence. Cell Rep 2019; 23:3565-3578. [PMID: 29924999 PMCID: PMC6324937 DOI: 10.1016/j.celrep.2018.05.054] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 02/28/2018] [Accepted: 05/16/2018] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial dynamics are tightly controlled by fusion and fission, and their dysregulation and excess reactive oxygen species (ROS) contribute to endothelial cell (EC) dysfunction. How redox signals regulate coupling between mitochondrial dynamics and endothelial (dys)function remains unknown. Here, we identify protein disulfide isomerase A1 (PDIA1) as a thiol reductase for the mitochondrial fission protein Drp1. A biotin-labeled Cys-OH trapping probe and rescue experiments reveal that PDIA1 depletion in ECs induces sulfenylation of Drp1 at Cys644, promoting mitochondrial fragmentation and ROS elevation without inducing ER stress, which drives EC senescence. Mechanistically, PDIA1 associates with Drp1 to reduce its redox status and activity. Defective wound healing and angiogenesis in diabetic or PDIA1+/- mice are restored by EC-targeted PDIA1 or the Cys oxidation-defective mutant Drp1. Thus, this study uncovers a molecular link between PDIA1 and Drp1 oxidoreduction, which maintains normal mitochondrial dynamics and limits endothelial senescence with potential translational implications for vascular diseases associated with diabetes or aging.
Collapse
Affiliation(s)
- Young-Mee Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Seock-Won Youn
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Varadarajan Sudhahar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA
| | - Archita Das
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Reyhaan Chandhri
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, 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
| | - Henar Cuervo Grajal
- 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
| | - Junghun Kweon
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Silvia Leanhart
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA
| | - Lianying He
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Peter T Toth
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Jan Kitajewski
- 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
| | - Jalees Rehman
- Departments of Medicine (Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA; Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
20
|
Gimenez M, Veríssimo-Filho S, Wittig I, Schickling BM, Hahner F, Schürmann C, Netto LES, Rosa JC, Brandes RP, Sartoretto S, De Lucca Camargo L, Abdulkader F, Miller FJ, Lopes LR. Redox Activation of Nox1 (NADPH Oxidase 1) Involves an Intermolecular Disulfide Bond Between Protein Disulfide Isomerase and p47 phox in Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2019; 39:224-236. [PMID: 30580571 DOI: 10.1161/atvbaha.118.311038] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- PDI (protein disulfide isomerase A1) was reported to support Nox1 (NADPH oxidase) activation mediated by growth factors in vascular smooth muscle cells. Our aim was to investigate the molecular mechanism by which PDI activates Nox1 and the functional implications of PDI in Nox1 activation in vascular disease. Approach and Results- Using recombinant proteins, we identified a redox interaction between PDI and the cytosolic subunit p47phox in vitro. Mass spectrometry of crosslinked peptides confirmed redox-dependent disulfide bonds between cysteines of p47phox and PDI and an intramolecular bond between Cys 196 and 378 in p47phox. PDI catalytic Cys 400 and p47phox Cys 196 were essential for the activation of Nox1 by PDI in vascular smooth muscle cells. Transfection of PDI resulted in the rapid oxidation of a redox-sensitive protein linked to p47phox, whereas PDI mutant did not promote this effect. Mutation of p47phox Cys 196, or the redox active cysteines of PDI, prevented Nox1 complex assembly and vascular smooth muscle cell migration. Proximity ligation assay confirmed the interaction of PDI and p47phox in murine carotid arteries after wire injury. Moreover, in human atheroma plaques, a positive correlation between the expression of PDI and p47phox occurred only in PDI family members with the a' redox active site. Conclusions- PDI redox cysteines facilitate Nox1 complex assembly, thus identifying a new mechanism through which PDI regulates Nox activity in vascular disease.
Collapse
Affiliation(s)
- Marcela Gimenez
- From the Department of Pharmacology (M.G., S.V.-F., S.S., L.D.L.C., L.R.L.), University of São Paulo, Brazil.,Department of Medicine, University of Iowa, Iowa City (M.G., B.M.S., F.J.M.)
| | - Sidney Veríssimo-Filho
- From the Department of Pharmacology (M.G., S.V.-F., S.S., L.D.L.C., L.R.L.), University of São Paulo, Brazil
| | - Ilka Wittig
- Functional Proteomics Core Unit (I.W.), Goethe-Universität, Frankfurt, Germany
| | - Brandon M Schickling
- Department of Medicine, University of Iowa, Iowa City (M.G., B.M.S., F.J.M.).,Department of Medicine, Duke University, Durham, NC (B.M.S., S.S., F.J.M.)
| | - Fabian Hahner
- Institute for Cardiovascular Physiology (F.H., C.S., R.P.B.), Goethe-Universität, Frankfurt, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology (F.H., C.S., R.P.B.), Goethe-Universität, Frankfurt, Germany
| | - Luis E S Netto
- Institute of Biomedical Sciences, Department of Genetics and Evolutionary Biology, Institute of Biosciences (L.E.S.N.), University of São Paulo, Brazil
| | - José César Rosa
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School (J.C.R.), University of São Paulo, Brazil
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology (F.H., C.S., R.P.B.), Goethe-Universität, Frankfurt, Germany
| | - Simone Sartoretto
- From the Department of Pharmacology (M.G., S.V.-F., S.S., L.D.L.C., L.R.L.), University of São Paulo, Brazil.,Department of Medicine, Duke University, Durham, NC (B.M.S., S.S., F.J.M.)
| | - Lívia De Lucca Camargo
- From the Department of Pharmacology (M.G., S.V.-F., S.S., L.D.L.C., L.R.L.), University of São Paulo, Brazil
| | - Fernando Abdulkader
- Department of Physiology and Biophysics (F.A.), University of São Paulo, Brazil
| | - Francis J Miller
- Department of Medicine, University of Iowa, Iowa City (M.G., B.M.S., F.J.M.).,Department of Medicine, Duke University, Durham, NC (B.M.S., S.S., F.J.M.).,Department of Medicine, Veterans Affairs Medical Center, Durham, NC (F.J.M.)
| | - Lucia Rossetti Lopes
- From the Department of Pharmacology (M.G., S.V.-F., S.S., L.D.L.C., L.R.L.), University of São Paulo, Brazil
| |
Collapse
|
21
|
Trostchansky A, Moore-Carrasco R, Fuentes E. Oxidative pathways of arachidonic acid as targets for regulation of platelet activation. Prostaglandins Other Lipid Mediat 2019; 145:106382. [PMID: 31634570 DOI: 10.1016/j.prostaglandins.2019.106382] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/12/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022]
Abstract
Platelet activation plays an important role in acute and chronic cardiovascular disease states. Multiple pathways contribute to platelet activation including those dependent upon arachidonic acid. Arachidonic acid is released from the platelet membrane by phospholipase A2 action and is then metabolized in the cytosol by specific arachidonic acid oxidation enzymes including prostaglandin H synthase, 12-lipoxygenase, and cytochrome P450 to produce pro- and anti-inflammatory eicosanoids. This review aims to analyze the role of arachidonic acid oxidation on platelet activation, the enzymes that use it as a substrate associated as novel therapeutics target for antiplatelet drugs.
Collapse
Affiliation(s)
- Andres Trostchansky
- Departamento de Bioquimica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - 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, Chile
| | - 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.
| |
Collapse
|
22
|
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
|
23
|
Borna H, Hosseini Qale Noe SH, Harchegani AB, Talatappe NR, Ghatrehsamani M, Ghanei M, Shahriary A. A review on proteomics analysis to reveal biological pathways and predictive proteins in sulfur mustard exposed patients: roles of inflammation and oxidative stress. Inhal Toxicol 2019; 31:3-11. [DOI: 10.1080/08958378.2018.1558316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Hojat Borna
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Hojjat Hosseini Qale Noe
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Asghar Beigi Harchegani
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nima Rahmani Talatappe
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdi Ghatrehsamani
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
De Bessa TC, Pagano A, Moretti AIS, Oliveira PVS, Mendonça SA, Kovacic H, Laurindo FRM. Subverted regulation of Nox1 NADPH oxidase-dependent oxidant generation by protein disulfide isomerase A1 in colon carcinoma cells with overactivated KRas. Cell Death Dis 2019; 10:143. [PMID: 30760703 PMCID: PMC6374413 DOI: 10.1038/s41419-019-1402-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 12/24/2018] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
Abstract
Protein disulfide isomerases including PDIA1 are implicated in cancer progression, but underlying mechanisms are unclear. PDIA1 is known to support vascular Nox1 NADPH oxidase expression/activation. Since deregulated reactive oxygen species (ROS) production underlies tumor growth, we proposed that PDIA1 is an upstream regulator of tumor-associated ROS. We focused on colorectal cancer (CRC) with distinct KRas activation levels. Analysis of RNAseq databanks and direct validation indicated enhanced PDIA1 expression in CRC with constitutive high (HCT116) vs. moderate (HKE3) and basal (Caco2) Ras activity. PDIA1 supported Nox1-dependent superoxide production in CRC; however, we first reported a dual effect correlated with Ras-level activity: in Caco2 and HKE3 cells, loss-of-function experiments indicate that PDIA1 sustains Nox1-dependent superoxide production, while in HCT116 cells PDIA1 restricted superoxide production, a behavior associated with increased Rac1 expression/activity. Transfection of Rac1G12V active mutant into HKE3 cells induced PDIA1 to become restrictive of Nox1-dependent superoxide, while in HCT116 cells treated with Rac1 inhibitor, PDIA1 became supportive of superoxide. PDIA1 silencing promoted diminished cell proliferation and migration in HKE3, not detectable in HCT116 cells. Screening of cell signaling routes affected by PDIA1 silencing highlighted GSK3β and Stat3. Also, E-cadherin expression after PDIA1 silencing was decreased in HCT116, consistent with PDIA1 support of epithelial-mesenchymal transition. Thus, Ras overactivation switches the pattern of PDIA1-dependent Rac1/Nox1 regulation, so that Ras-induced PDIA1 bypass can directly activate Rac1. PDIA1 may be a crucial regulator of redox-dependent adaptive processes related to cancer progression.
Collapse
Affiliation(s)
- Tiphany Coralie De Bessa
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France
| | - Alessandra Pagano
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France
| | - Ana Iochabel Soares Moretti
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Percillia Victoria Santos Oliveira
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Samir Andrade Mendonça
- Centro de Investigação Translacional em Oncologia do Instituto do Câncer do Estado de São Paulo (Icesp), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Herve Kovacic
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France.
| | - Francisco Rafael Martins Laurindo
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
25
|
Abstract
SIGNIFICANCE G protein-coupled receptors (GPCR) are the largest group of cell surface receptors, which link cells to their environment. Reactive oxygen species (ROS) can act as important cellular signaling molecules. The family of NADPH oxidases generates ROS in response to activated cell surface receptors. Recent Advances: Various signaling pathways linking GPCRs and activation of NADPH oxidases have been characterized. CRITICAL ISSUES Still, a more detailed analysis of G proteins involved in the GPCR-mediated activation of NADPH oxidases is needed. In addition, a more precise discrimination of NADPH oxidase activation due to either upregulation of subunit expression or post-translational subunit modifications is needed. Also, the role of noncanonical modulators of NADPH oxidase activation in the response to GPCRs awaits further analyses. FUTURE DIRECTIONS As GPCRs are one of the most popular classes of investigational drug targets, further detailing of G protein-coupled mechanisms in the activation mechanism of NADPH oxidases as well as better understanding of the link between newly identified NADPH oxidase interaction partners and GPCR signaling will provide new opportunities for improved efficiency and decreased off target effects of therapies targeting GPCRs.
Collapse
Affiliation(s)
- Andreas Petry
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany
| | - Agnes Görlach
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany .,2 DZHK (German Centre for Cardiovascular Research) , Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
26
|
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
|
27
|
Mennerich D, Kellokumpu S, Kietzmann T. Hypoxia and Reactive Oxygen Species as Modulators of Endoplasmic Reticulum and Golgi Homeostasis. Antioxid Redox Signal 2019; 30:113-137. [PMID: 29717631 DOI: 10.1089/ars.2018.7523] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE Eukaryotic cells execute various functions in subcellular compartments or organelles for which cellular redox homeostasis is of importance. Apart from mitochondria, hypoxia and stress-mediated formation of reactive oxygen species (ROS) were shown to modulate endoplasmic reticulum (ER) and Golgi apparatus (GA) functions. Recent Advances: Research during the last decade has improved our understanding of disulfide bond formation, protein glycosylation and secretion, as well as pH and redox homeostasis in the ER and GA. Thus, oxygen (O2) itself, NADPH oxidase (NOX) formed ROS, and pH changes appear to be of importance and indicate the intricate balance of intercompartmental communication. CRITICAL ISSUES Although the interplay between hypoxia, ER stress, and Golgi function is evident, the existence of more than 20 protein disulfide isomerase family members and the relative mild phenotypes of, for example, endoplasmic reticulum oxidoreductin 1 (ERO1)- and NOX4-knockout mice clearly suggest the existence of redundant and alternative pathways, which remain largely elusive. FUTURE DIRECTIONS The identification of these pathways and the key players involved in intercompartmental communication needs suitable animal models, genome-wide association, as well as proteomic studies in humans. The results of those studies will be beneficial for the understanding of the etiology of diseases such as type 2 diabetes, Alzheimer's disease, and cancer, which are associated with ROS, protein aggregation, and glycosylation defects.
Collapse
Affiliation(s)
- Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| | - Sakari Kellokumpu
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
Kehrel BE, Fender AC. Greetings from the endoplasmic reticulum (ER): escaping ER thiol isomerases regulate thrombosis. J Thromb Haemost 2018; 16:364-366. [PMID: 29194946 DOI: 10.1111/jth.13916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Indexed: 11/28/2022]
Affiliation(s)
- B E Kehrel
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Haemostasis, University Hospital Muenster, Muenster, Germany
| | - A C Fender
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Haemostasis, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
32
|
Moretti AIS, Pavanelli JC, Nolasco P, Leisegang MS, Tanaka LY, Fernandes CG, Wosniak J, Kajihara D, Dias MH, Fernandes DC, Jo H, Tran NV, Ebersberger I, Brandes RP, Bonatto D, Laurindo FRM. Conserved Gene Microsynteny Unveils Functional Interaction Between Protein Disulfide Isomerase and Rho Guanine-Dissociation Inhibitor Families. Sci Rep 2017; 7:17262. [PMID: 29222525 PMCID: PMC5722932 DOI: 10.1038/s41598-017-16947-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/21/2017] [Indexed: 02/07/2023] Open
Abstract
Protein disulfide isomerases (PDIs) support endoplasmic reticulum redox protein folding and cell-surface thiol-redox control of thrombosis and vascular remodeling. The family prototype PDIA1 regulates NADPH oxidase signaling and cytoskeleton organization, however the related underlying mechanisms are unclear. Here we show that genes encoding human PDIA1 and its two paralogs PDIA8 and PDIA2 are each flanked by genes encoding Rho guanine-dissociation inhibitors (GDI), known regulators of RhoGTPases/cytoskeleton. Evolutionary histories of these three microsyntenic regions reveal their emergence by two successive duplication events of a primordial gene pair in the last common vertebrate ancestor. The arrangement, however, is substantially older, detectable in echinoderms, nematodes, and cnidarians. Thus, PDI/RhoGDI pairing in the same transcription orientation emerged early in animal evolution and has been largely maintained. PDI/RhoGDI pairs are embedded into conserved genomic regions displaying common cis-regulatory elements. Analysis of gene expression datasets supports evidence for PDI/RhoGDI coexpression in developmental/inflammatory contexts. PDIA1/RhoGDIα were co-induced in endothelial cells upon CRISP-R-promoted transcription activation of each pair component, and also in mouse arterial intima during flow-induced remodeling. We provide evidence for physical interaction between both proteins. These data support strong functional links between PDI and RhoGDI families, which likely maintained PDI/RhoGDI microsynteny along > 800-million years of evolution.
Collapse
Affiliation(s)
- Ana I S Moretti
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Jessyca C Pavanelli
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Patrícia Nolasco
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Carolina G Fernandes
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - João Wosniak
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Daniela Kajihara
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Matheus H Dias
- Special Laboratory for Cell Cycle, Center of Toxins, Immune-Response and Cell Signaling - CeTICS-Cepid, Butantan Institute, São Paulo, Brazil
| | - Denise C Fernandes
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Hanjoong Jo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
| | - Ngoc-Vinh Tran
- Applied Bioinformatics Group, Institute of Cell Biology & Neuroscience, Goethe University, Frankfurt, Germany
| | - Ingo Ebersberger
- Applied Bioinformatics Group, Institute of Cell Biology & Neuroscience, Goethe University, Frankfurt, Germany
- Senckenberg Biodiversity and Climate Research Center (BiK-F), Frankfurt, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe University, Frankfurt, Germany
| | - Diego Bonatto
- Department of Molecular Biology and Biotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (Incor), University of São Paulo School of Medicine, São Paulo, Brazil.
| |
Collapse
|
33
|
Habash SS, Sobczak M, Siddique S, Voigt B, Elashry A, Grundler FMW. Identification and characterization of a putative protein disulfide isomerase (HsPDI) as an alleged effector of Heterodera schachtii. Sci Rep 2017; 7:13536. [PMID: 29051538 PMCID: PMC5648851 DOI: 10.1038/s41598-017-13418-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/21/2017] [Indexed: 12/27/2022] Open
Abstract
The plant-parasitic nematode Heterodera schachtii is an obligate biotroph that induces syncytial feeding sites in roots of its hosts. Nematodes produce effectors that are secreted into the host and facilitate infection process. Here we identified H. schachtii protein disulphide isomerase (HsPDI) as a putative effector that interferes with the host’s redox status. In situ hybridization showed that HsPdi is specifically localized within esophageal glands of pre-parasitic second stage juveniles (J2). HsPdi is up-regulated in the early parasitic J2s. Silencing of HsPdi by RNA interference in the J2s hampers their development and leads to structural malfunctions in associated feeding sites induced in Arabidopsis roots. Expression of HsPDI in Arabidopsis increases plant’s susceptibility towards H. schachtii. HsPdi expression is up-regulated in the presence of exogenous H2O2, whereas HsPdi silencing results in increased mortality under H2O2 stress. Stable expression of HsPDI in Arabidopsis plants decreases ROS burst induced by flg22. Transiently expressed HsPDI in N. benthamiana leaves is localized in the apoplast. HsPDI plays an important role in the interaction between nematode and plant, probably through inducing local changes in the redox status of infected host tissue. It also contributes to protect the nematode from exogenous H2O2 stress.
Collapse
Affiliation(s)
- Samer S Habash
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany
| | - Miroslaw Sobczak
- Department of Botany, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, PL-02787, Warsaw, Poland
| | - Shahid Siddique
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany
| | - Boris Voigt
- Rheinische Friedrich-Wilhelms-University of Bonn, Department of Plant Cell Biology, Institute of Cellular and Molecular Botany, Kirschallee 1, D-53115, Bonn, Germany
| | - Abdelnaser Elashry
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany.,Strube Research GmbH & Co. KG, Hauptstraße 1, 38387, Söllingen, Germany
| | - Florian M W Grundler
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany.
| |
Collapse
|
34
|
Thom SR, Bhopale VM, Yu K, Huang W, Kane MA, Margolis DJ. Neutrophil microparticle production and inflammasome activation by hyperglycemia due to cytoskeletal instability. J Biol Chem 2017; 292:18312-18324. [PMID: 28972154 DOI: 10.1074/jbc.m117.802629] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/11/2017] [Indexed: 12/23/2022] Open
Abstract
Microparticles are lipid bilayer-enclosed vesicles produced by cells under oxidative stress. MP production is elevated in patients with diabetes, but the underlying cellular mechanisms are poorly understood. We hypothesized that raising glucose above the physiological level of 5.5 mm would stimulate leukocytes to produce MPs and activate the nucleotide-binding domain, leucine-rich repeat pyrin domain-containing 3 (NLRP3) inflammasome. We found that when incubated in buffer with up to 20 mm glucose, human and murine neutrophils, but not monocytes, generate progressively more MPs with high interleukin (IL)-1β content. Enhanced MP production required generation of reactive chemical species by mitochondria, NADPH oxidase, and type 2 nitric-oxide synthase (NOS-2) and resulted in S-nitrosylation of actin. Depleting cells of capon (C-terminal PDZ ligand of neuronal nitric-oxide synthase protein), apoptosis-associated speck-like protein containing C-terminal caspase recruitment domain (ASC), or pro-IL-1β prevented the hyperglycemia-induced enhancement of reactive species production, MP generation, and IL-1β synthesis. Additional components required for these responses included inositol 1,3,5-triphosphate receptors, PKC, and enhancement of filamentous-actin turnover. Numerous proteins become localized to short filamentous actin in response to S-nitrosylation, including vasodilator-stimulated phosphoprotein, focal adhesion kinase, the membrane phospholipid translocation enzymes flippase and floppase, capon, NLRP3, and ASC. We conclude that an interdependent oxidative stress response to hyperglycemia perturbs neutrophil cytoskeletal stability leading to MP production and IL-1β synthesis.
Collapse
Affiliation(s)
- Stephen R Thom
- From the Department of Emergency Medicine, School of Medicine, and
| | - Veena M Bhopale
- From the Department of Emergency Medicine, School of Medicine, and
| | - Kevin Yu
- From the Department of Emergency Medicine, School of Medicine, and
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201 and
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201 and
| | - David J Margolis
- the Department of Dermatology and Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
35
|
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
|
36
|
Thom SR, Bhopale VM, Hu J, Yang M. Increased carbon dioxide levels stimulate neutrophils to produce microparticles and activate the nucleotide-binding domain-like receptor 3 inflammasome. Free Radic Biol Med 2017; 106:406-416. [PMID: 28288918 DOI: 10.1016/j.freeradbiomed.2017.03.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/22/2017] [Accepted: 03/08/2017] [Indexed: 11/22/2022]
Abstract
We hypothesized that elevations of carbon dioxide (CO2) commonly found in modern buildings will stimulate leukocytes to produce microparticles (MPs) and activate the nucleotide-binding domain-like receptor 3 (NLRP3) inflammasome due to mitochondrial oxidative stress. Human and murine neutrophils generate MPs with high interleukin-1β (IL-1β) content when incubated ex vivo in buffer equilibrated with 0.1-0.4% additional CO2. Enhanced MPs production requires mitochondrial reactive oxygen species production, which is mediated by activities of pyruvate carboxylase and phosphoenolpyruvate carboxykinase. Subsequent events leading to MPs generation include perturbation of inositol 1,3,5-triphosphate receptors, a transient elevation of intracellular calcium, activation of protein kinase C and NADPH oxidase (Nox). Concomitant activation of type-2 nitric oxide synthase yields secondary oxidants resulting in actin S-nitrosylation and enhanced filamentous actin turnover. Numerous proteins are linked to short filamentous actin including vasodilator-stimulated phosphoprotein, focal adhesion kinase, the membrane phospholipid translocation enzymes flippase and floppase, and the critical inflammasome protein ASC (Apoptosis-associated Speck protein with CARD domain). Elevations of CO2 cause oligomerization of the inflammasome components ASC, NLRP3, caspase 1, thioredoxin interacting protein, and calreticulin - a protein from endoplasmic reticulum, leading to IL-1β synthesis. An increased production rate of MPs containing elevated amounts of IL-1β persists for hours after short-term exposures to elevated CO2.
Collapse
Affiliation(s)
- Stephen R Thom
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Veena M Bhopale
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - JingPing Hu
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Ming Yang
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| |
Collapse
|
37
|
Wang L, Essex DW. A new antithrombotic strategy: inhibition of the C-terminal active site of protein disulfide isomerase. J Thromb Haemost 2017; 15:770-773. [PMID: 28109037 PMCID: PMC5546002 DOI: 10.1111/jth.13634] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 01/28/2023]
Affiliation(s)
- L Wang
- Sol Sherry Thrombosis Research Center, Division of Hematology, Department of Medicine, Temple University School of Medicine, Philadelphia, PA, USA
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Department of Medicine, Soochow University, Suzhou, China
| | - D W Essex
- Sol Sherry Thrombosis Research Center, Division of Hematology, Department of Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
38
|
Sousa HR, Gaspar RS, Sena EML, da Silva SA, Fontelles JL, AraUjo TLS, Mastrogiovanni M, Fries DM, Azevedo-Santos APS, Laurindo FRM, Trostchansky A, Paes AM. Novel antiplatelet role for a protein disulfide isomerase-targeted peptide: evidence of covalent binding to the C-terminal CGHC redox motif. J Thromb Haemost 2017; 15:774-784. [PMID: 28109047 DOI: 10.1111/jth.13633] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Indexed: 11/30/2022]
Abstract
Essentials Inhibitors of protein disulfide isomerase (PDI) have been considered a new antithrombotic class. CxxC is a PDI-targeted peptide that has been previously shown to inhibit its reductase activity. CxxC binds to surface PDI and inhibits ADP- and thrombin-evoked platelet activation and aggregation. CxxC binds to Cys400 on CGHC redox motif of PDI a' domain, a site for PDI prothrombotic activity. SUMMARY Background Protein disulfide isomerase (PDI) plays a major role in platelet aggregation, and its inhibitors have emerged as novel antithrombotic drugs. In previous work, we designed a peptide based on a PDI redox motif (CGHC) that inhibited both PDI reductase activity and PDI-modulated superoxide generation by neutrophil Nox2. Thus, we hypothesized that this peptide would also inhibit platelet aggregation by association with surface PDI. Methods Three peptides were used: CxxC, containing the PDI redox motif; Scr, presenting a scrambled sequence of the same residues and AxxA, with cysteines replaced by alanine. These peptides were tested under platelet aggregation and flow cytometry protocols to identify their possible antiplatelet activity. We labeled membrane free thiol and electrospray ionization liquid chromatography tandem mass spectrometry to test for an interaction. Results CxxC decreased platelet aggregation in a dose-dependent manner, being more potent at lower agonist concentrations, whereas neither AxxA nor Scr peptides exerted any effect. CxxC decreased aIIbb3 activation, but had no effect on the other markers. CxxC also decreased cell surface PDI pulldown without interfering with the total thiol protein content. Finally, we detected the addition of one CxxC molecule to reduced PDI through binding to Cys400 through mass spectrometry. Interestingly, CxxC did not react with oxidized PDI. Discussion CxxC has consistently shown its antiplatelet effects, both in PRP and washed platelets, corroborated by decreased aIIbb3 activation. The probable mechanism of action is through a mixed dissulphide bond with Cys400 of PDI, which has been shown to be essential for PDI's actions. Conclusion In summary, our data support antiplatelet activity for CxxC through binding to Cys400 in the PDI a0 domain, which can be further exploited as a model for sitedriven antithrombotic agent development.
Collapse
Affiliation(s)
- H R Sousa
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - R S Gaspar
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - E M L Sena
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - S A da Silva
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - J L Fontelles
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - T L S AraUjo
- Laboratory of Vascular Biology, Heart Institute, School of Medicine of the University of São Paulo, São Paulo, SP, Brazil
| | - M Mastrogiovanni
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - D M Fries
- Laboratory of Vascular Biology, Heart Institute, School of Medicine of the University of São Paulo, São Paulo, SP, Brazil
| | - A P S Azevedo-Santos
- Laboratory of Immunophysiology, Department of Pathology, Federal University of Maranhão, São Luís, MA, Brazil
| | - F R M Laurindo
- Laboratory of Vascular Biology, Heart Institute, School of Medicine of the University of São Paulo, São Paulo, SP, Brazil
| | - A Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - A M Paes
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Soares Moretti AI, Martins Laurindo FR. Protein disulfide isomerases: Redox connections in and out of the endoplasmic reticulum. Arch Biochem Biophys 2016; 617:106-119. [PMID: 27889386 DOI: 10.1016/j.abb.2016.11.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/08/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022]
Abstract
Protein disulfide isomerases are thiol oxidoreductase chaperones from thioredoxin superfamily. As redox folding catalysts from the endoplasmic reticulum (ER), their roles in ER-related redox homeostasis and signaling are well-studied. PDIA1 exerts thiol oxidation/reduction and isomerization, plus chaperone effects. Also, substantial evidence indicates that PDIs regulate thiol-disulfide switches in other cell locations such as cell surface and possibly cytosol. Subcellular PDI translocation routes remain unclear and seem Golgi-independent. The list of signaling and structural proteins reportedly regulated by PDIs keeps growing, via thiol switches involving oxidation, reduction and isomerization, S-(de)nytrosylation, (de)glutathyonylation and protein oligomerization. PDIA1 is required for agonist-triggered Nox NADPH oxidase activation and cell migration in vascular cells and macrophages, while PDIA1-dependent cytoskeletal regulation appears a converging pathway. Extracellularly, PDIs crucially regulate thiol redox signaling of thrombosis/platelet activation, e.g., integrins, and PDIA1 supports expansive caliber remodeling during injury repair via matrix/cytoskeletal organization. Some proteins display regulatory PDI-like motifs. PDI effects are orchestrated by expression levels or post-translational modifications. PDI is redox-sensitive, although probably not a mass-effect redox sensor due to kinetic constraints. Rather, the "all-in-one" organization of its peculiar redox/chaperone properties likely provide PDIs with precision and versatility in redox signaling, making them promising therapeutic targets.
Collapse
Affiliation(s)
- Ana Iochabel Soares Moretti
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | | |
Collapse
|
42
|
Trostchansky A, Rubbo H. Anti-inflammatory signaling actions of electrophilic nitro-arachidonic acid in vascular cells and astrocytes. Arch Biochem Biophys 2016; 617:155-161. [PMID: 27720684 DOI: 10.1016/j.abb.2016.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/28/2016] [Accepted: 10/03/2016] [Indexed: 10/20/2022]
Abstract
Nitrated derivatives of unsaturated fatty acids (nitro-fatty acids) are being formed and detected in human plasma, cell membranes and tissue, triggering signaling cascades via covalent and reversible post-translational modifications of nucleophilic amino acids in transcriptional regulatory proteins. Arachidonic acid (AA) represents a precursor of potent signaling molecules, i.e., prostaglandins and thromboxanes through enzymatic and non-enzymatic oxidative pathways. Arachidonic acid can be nitrated by reactive nitrogen species leading to the formation of nitro-arachidonic acid (NO2-AA). A critical issue is the influence of NO2-AA on prostaglandin endoperoxide H synthases, modulating inflammatory processes through redirection of AA metabolism and signaling. In this prospective article, we describe the key chemical and biochemical actions of NO2-AA in vascular and astrocytes. This includes the ability of NO2-AA to mediate unique redox signaling anti-inflammatory actions along with its therapeutic potential.
Collapse
Affiliation(s)
- Andrés Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Avda. General Flores 2125, Universidad de la República, Montevideo 11800, Uruguay
| | - Homero Rubbo
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Avda. General Flores 2125, Universidad de la República, Montevideo 11800, Uruguay.
| |
Collapse
|
43
|
Abstract
Since its discovery in 1999, a number of studies have evaluated the role of Nox1 NADPH oxidase in the cardiovascular system. Nox1 is activated in vascular cells in response to several different agonists, with its activity regulated at the transcriptional level as well as by NADPH oxidase complex formation, protein stabilization and post-translational modification. Nox1 has been shown to decrease the bioavailability of nitric oxide, transactivate the epidermal growth factor receptor, induce pro-inflammatory signalling, and promote cell migration and proliferation. Enhanced expression and activity of Nox1 under pathologic conditions results in excessive production of reactive oxygen species and dysregulated cellular function. Indeed, studies using genetic models of Nox1 deficiency or overexpression have revealed roles for Nox1 in the pathogenesis of cardiovascular diseases ranging from atherosclerosis to hypertension, restenosis and ischaemia/reperfusion injury. These data suggest that Nox1 is a potential therapeutic target for vascular disease, and drug development efforts are ongoing to identify a specific bioavailable inhibitor of Nox1.
Collapse
|
44
|
Perri ER, Thomas CJ, Parakh S, Spencer DM, Atkin JD. The Unfolded Protein Response and the Role of Protein Disulfide Isomerase in Neurodegeneration. Front Cell Dev Biol 2016; 3:80. [PMID: 26779479 PMCID: PMC4705227 DOI: 10.3389/fcell.2015.00080] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022] Open
Abstract
The maintenance and regulation of proteostasis is a critical function for post-mitotic neurons and its dysregulation is increasingly implicated in neurodegenerative diseases. Despite having different clinical manifestations, these disorders share similar pathology; an accumulation of misfolded proteins in neurons and subsequent disruption to cellular proteostasis. The endoplasmic reticulum (ER) is an important component of proteostasis, and when the accumulation of misfolded proteins occurs within the ER, this disturbs ER homeostasis, giving rise to ER stress. This triggers the unfolded protein response (UPR), distinct signaling pathways that whilst initially protective, are pro-apoptotic if ER stress is prolonged. ER stress is increasingly implicated in neurodegenerative diseases, and emerging evidence highlights the complexity of the UPR in these disorders, with both protective and detrimental components being described. Protein Disulfide Isomerase (PDI) is an ER chaperone induced during ER stress that is responsible for the formation of disulfide bonds in proteins. Whilst initially considered to be protective, recent studies have revealed unconventional roles for PDI in neurodegenerative diseases, distinct from its normal function in the UPR and the ER, although these mechanisms remain poorly defined. However, specific aspects of PDI function may offer the potential to be exploited therapeutically in the future. This review will focus on the evidence linking ER stress and the UPR to neurodegenerative diseases, with particular emphasis on the emerging functions ascribed to PDI in these conditions.
Collapse
Affiliation(s)
- Emma R Perri
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne, VIC, Australia
| | - Colleen J Thomas
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University Melbourne, VIC, Australia
| | - Sonam Parakh
- Department of Biomedical Sciences, Faculty of Medicine and Human Science, Macquarie University Sydney, NSW, Australia
| | - Damian M Spencer
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne, VIC, Australia
| | - Julie D Atkin
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe UniversityMelbourne, VIC, Australia; Department of Biomedical Sciences, Faculty of Medicine and Human Science, Macquarie UniversitySydney, NSW, Australia
| |
Collapse
|
45
|
Heckler EJ, Kholodovych V, Jain M, Liu T, Li H, Beuve A. Mapping Soluble Guanylyl Cyclase and Protein Disulfide Isomerase Regions of Interaction. PLoS One 2015; 10:e0143523. [PMID: 26618351 PMCID: PMC4664405 DOI: 10.1371/journal.pone.0143523] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 11/05/2015] [Indexed: 11/19/2022] Open
Abstract
Soluble guanylyl cyclase (sGC) is a heterodimeric nitric oxide (NO) receptor that produces cyclic GMP. This signaling mechanism is a key component in the cardiovascular system. NO binds to heme in the β subunit and stimulates the catalytic conversion of GTP to cGMP several hundred fold. Several endogenous factors have been identified that modulate sGC function in vitro and in vivo. In previous work, we determined that protein disulfide isomerase (PDI) interacts with sGC in a redox-dependent manner in vitro and that PDI inhibited NO-stimulated activity in cells. To our knowledge, this was the first report of a physical interaction between sGC and a thiol-redox protein. To characterize this interaction between sGC and PDI, we first identified peptide linkages between sGC and PDI, using a lysine cross-linking reagent and recently developed mass spectrometry analysis. Together with Flag-immunoprecipitation using sGC domain deletions, wild-type (WT) and mutated PDI, regions of sGC involved in this interaction were identified. The observed data were further explored with computational modeling to gain insight into the interaction mechanism between sGC and oxidized PDI. Our results indicate that PDI interacts preferentially with the catalytic domain of sGC, thus providing a mechanism for PDI inhibition of sGC. A model in which PDI interacts with either the α or the β catalytic domain is proposed.
Collapse
Affiliation(s)
- Erin J. Heckler
- Department of Pharmacology and Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States of America
| | - Vladyslav Kholodovych
- High Performance and Research Computing, OIRT, Rutgers University, New Brunswick, NJ, United States of America
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States of America
| | - Mohit Jain
- Proteomics Core, New Jersey Medical School, Rutgers University, Newark, NJ, United States of America
| | - Tong Liu
- Proteomics Core, New Jersey Medical School, Rutgers University, Newark, NJ, United States of America
| | - Hong Li
- Proteomics Core, New Jersey Medical School, Rutgers University, Newark, NJ, United States of America
| | - Annie Beuve
- Department of Pharmacology and Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, United States of America
- * E-mail:
| |
Collapse
|
46
|
Bhopale VM, Yang M, Yu K, Thom SR. Factors Associated with Nitric Oxide-mediated β2 Integrin Inhibition of Neutrophils. J Biol Chem 2015; 290:17474-84. [PMID: 26032418 DOI: 10.1074/jbc.m115.651620] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Indexed: 11/06/2022] Open
Abstract
This investigation explored the mechanism for inhibition of β2 integrin adhesion molecules when neutrophils are exposed to nitric oxide ((•)NO). Roles for specific proteins were elucidated using chemical inhibitors, depletion with small inhibitory RNA, and cells from knock-out mice. Optimal inhibition occurs with exposures to a (•)NO flux of ∼ 28 nmol/min for 2 min or more, which sets up an autocatalytic cascade triggered by activating type 2 nitric-oxide synthase (NOS-2) and NADPH oxidase (NOX). Integrin inhibition does not occur with neutrophils exposed to a NOX inhibitor (Nox2ds), a NOS-2 inhibitor (1400 W), or with cells from mice lacking NOS-2 or the gp91(phox) component of NOX. Reactive species cause S-nitrosylation of cytosolic actin that enhances actin polymerization. Protein cross-linking and actin filament formation assays indicate that increased polymerization occurs because of associations involving vasodilator-stimulated phosphoprotein, focal adhesion kinase, and protein-disulfide isomerase in proximity to actin filaments. These effects were inhibited in cells exposed to ultraviolet light which photo-reverses S-nitrosylated cysteine residues and by co-incubations with cytochalasin D. The autocatalytic cycle can be arrested by protein kinase G activated with 8-bromo-cyclic GMP and by a high (•)NO flux (∼ 112 nmol/min) that inactivates NOX.
Collapse
Affiliation(s)
- Veena M Bhopale
- From the Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Ming Yang
- From the Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Kevin Yu
- From the Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Stephen R Thom
- From the Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
47
|
Parakh S, Atkin JD. Novel roles for protein disulphide isomerase in disease states: a double edged sword? Front Cell Dev Biol 2015; 3:30. [PMID: 26052512 PMCID: PMC4439577 DOI: 10.3389/fcell.2015.00030] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/28/2015] [Indexed: 12/14/2022] Open
Abstract
Protein disulphide isomerase (PDI) is a multifunctional redox chaperone of the endoplasmic reticulum (ER). Since it was first discovered 40 years ago the functions ascribed to PDI have evolved significantly and recent studies have recognized its distinct functions, with adverse as well as protective effects in disease. Furthermore, post translational modifications of PDI abrogate its normal functional roles in specific disease states. This review focusses on recent studies that have identified novel functions for PDI relevant to specific diseases.
Collapse
Affiliation(s)
- Sonam Parakh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Julie D Atkin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia ; Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University Bundoora, VIC, Australia
| |
Collapse
|
48
|
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: 19] [Impact Index Per Article: 1.9] [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
|
49
|
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
|
50
|
Nakao LS, Everley RA, Marino SM, Lo SM, de Souza LE, Gygi SP, Gladyshev VN. Mechanism-based proteomic screening identifies targets of thioredoxin-like proteins. J Biol Chem 2015; 290:5685-95. [PMID: 25561728 DOI: 10.1074/jbc.m114.597245] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Thioredoxin (Trx)-fold proteins are protagonists of numerous cellular pathways that are subject to thiol-based redox control. The best characterized regulator of thiols in proteins is Trx1 itself, which together with thioredoxin reductase 1 (TR1) and peroxiredoxins (Prxs) comprises a key redox regulatory system in mammalian cells. However, there are numerous other Trx-like proteins, whose functions and redox interactors are unknown. It is also unclear if the principles of Trx1-based redox control apply to these proteins. Here, we employed a proteomic strategy to four Trx-like proteins containing CXXC motifs, namely Trx1, Rdx12, Trx-like protein 1 (Txnl1) and nucleoredoxin 1 (Nrx1), whose cellular targets were trapped in vivo using mutant Trx-like proteins, under conditions of low endogenous expression of these proteins. Prxs were detected as key redox targets of Trx1, but this approach also supported the detection of TR1, which is the Trx1 reductant, as well as mitochondrial intermembrane proteins AIF and Mia40. In addition, glutathione peroxidase 4 was found to be a Rdx12 redox target. In contrast, no redox targets of Txnl1 and Nrx1 could be detected, suggesting that their CXXC motifs do not engage in mixed disulfides with cellular proteins. For some Trx-like proteins, the method allowed distinguishing redox and non-redox interactions. Parallel, comparative analyses of multiple thiol oxidoreductases revealed differences in the functions of their CXXC motifs, providing important insights into thiol-based redox control of cellular processes.
Collapse
Affiliation(s)
- Lia S Nakao
- From the Division of Genetics, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, the Universidade Federal do Paraná, Departamento de Patologia Básica, 81531-980, Curitiba, PR, Brazil, and
| | - Robert A Everley
- the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Stefano M Marino
- From the Division of Genetics, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Sze M Lo
- the Universidade Federal do Paraná, Departamento de Patologia Básica, 81531-980, Curitiba, PR, Brazil, and
| | - Luiz E de Souza
- the Universidade Federal do Paraná, Departamento de Patologia Básica, 81531-980, Curitiba, PR, Brazil, and
| | - Steven P Gygi
- the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Vadim N Gladyshev
- From the Division of Genetics, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115,
| |
Collapse
|