1
|
Ahmed R, Augustine R, Chaudhry M, Akhtar UA, Zahid AA, Tariq M, Falahati M, Ahmad IS, Hasan A. Nitric oxide-releasing biomaterials for promoting wound healing in impaired diabetic wounds: State of the art and recent trends. Pharmacotherapy 2022; 149:112707. [PMID: 35303565 DOI: 10.1016/j.biopha.2022.112707] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
Impaired diabetic wounds are serious pathophysiological complications associated with persistent microbial infections including failure in the closure of wounds, and the cause of a high frequency of lower limb amputations. The healing of diabetic wounds is attenuated due to the lack of secretion of growth factors, prolonged inflammation, and/or inhibition of angiogenic activity. Diabetic wound healing can be enhanced by supplying nitric oxide (NO) endogenously or exogenously. NO produced inside the cells by endothelial nitric oxide synthase (eNOS) naturally aids wound healing through its beneficial vasculogenic effects. However, during hyperglycemia, the activity of eNOS is affected, and thus there becomes an utmost need for the topical supply of NO from exogenous sources. Thus, NO-donors that can release NO are loaded into wound healing patches or wound coverage matrices to treat diabetic wounds. The burst release of NO from its donors is prevented by encapsulating them in polymeric hydrogels or nanoparticles for supplying NO for an extended duration of time to the diabetic wounds. In this article, we review the etiology of diabetic wounds, wound healing strategies, and the role of NO in the wound healing process. We further discuss the challenges faced in translating NO-donors as a clinically viable nanomedicine strategy for the treatment of diabetic wounds with a focus on the use of biomaterials for the encapsulation and in vivo controlled delivery of NO-donors.
Collapse
Affiliation(s)
- Rashid Ahmed
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar; Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, Mirpur 10250, AJK, Pakistan; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA
| | - Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar
| | - Maryam Chaudhry
- Department of Continuing Education, University of Oxford, OX1 2JD Oxford, United Kingdom
| | - Usman A Akhtar
- Department of Chemical Engineering, College of Engineering, Qatar University, Doha 2713, Qatar
| | - Alap Ali Zahid
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar
| | - Muhammad Tariq
- Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, Mirpur 10250, AJK, Pakistan
| | - Mojtaba Falahati
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Irfan S Ahmad
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA; Department of Agricultural and Biological Engineering, University of Illinois at Urbana Champaign, IL, USA; Carle Illinois College of Medicine, University of Illinois at Urbana Champaign, IL, USA
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
2
|
Caballano-Infantes E, Cahuana GM, Bedoya FJ, Salguero-Aranda C, Tejedo JR. The Role of Nitric Oxide in Stem Cell Biology. Antioxidants (Basel) 2022; 11:497. [PMID: 35326146 PMCID: PMC8944807 DOI: 10.3390/antiox11030497] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a gaseous biomolecule endogenously synthesized with an essential role in embryonic development and several physiological functions, such as regulating mitochondrial respiration and modulation of the immune response. The dual role of NO in embryonic stem cells (ESCs) has been previously reported, preserving pluripotency and cell survival or inducing differentiation with a dose-dependent pattern. In this line, high doses of NO have been used in vitro cultures to induce focused differentiation toward different cell lineages being a key molecule in the regenerative medicine field. Moreover, optimal conditions to promote pluripotency in vitro are essential for their use in advanced therapies. In this sense, the molecular mechanisms underlying stemness regulation by NO have been studied intensively over the current years. Recently, we have reported the role of low NO as a hypoxia-like inducer in pluripotent stem cells (PSCs), which supports using this molecule to maintain pluripotency under normoxic conditions. In this review, we stress the role of NO levels on stem cells (SCs) fate as a new approach for potential cell therapy strategies. Furthermore, we highlight the recent uses of NO in regenerative medicine due to their properties regulating SCs biology.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Javier Bedoya
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Salguero-Aranda
- Department of Pathology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, CSIC-University of Seville, 41013 Seville, Spain;
- Spanish Biomedical Research Network Centre in Oncology-CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41004 Seville, Spain
| | - Juan R. Tejedo
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3
|
Caballano-Infantes E, Díaz I, Hitos AB, Cahuana GM, Martínez-Ruiz A, Soria-Juan B, Rodríguez-Griñolo R, Hmadcha A, Martín F, Soria B, Tejedo JR, Bedoya FJ. Stemness of Human Pluripotent Cells: Hypoxia-Like Response Induced by Low Nitric Oxide. Antioxidants (Basel) 2021; 10:antiox10091408. [PMID: 34573040 PMCID: PMC8472328 DOI: 10.3390/antiox10091408] [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] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022] Open
Abstract
The optimization of conditions to promote the stemness of pluripotent cells in vitro is instrumental for their use in advanced therapies. We show here that exposure of human iPSCs and human ESCs to low concentrations of the chemical NO donor DETA/NO leads to stabilization of hypoxia-inducible factors (HIF-1α and HIF-2α) under normoxia, with this effect being dependent on diminished Pro 402 hydroxylation and decreased degradation by the proteasome. Moreover, the master genes of pluripotency, NANOG and OCT-4, were upregulated. NO also induces a shift in the metabolic profile of PSCs, with an increased expression of hypoxia response genes in glycolysis. Furthermore, a reduction in the mitochondrial membrane potential with lower oxygen consumption and increased expression of mitochondrial fusion regulators, such as DRP1, was observed. The results reported here indicate that NO mimics hypoxia response in human PSCs and enhances their stemness properties when cultured under normoxic conditions.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Correspondence: (E.C.-I.); (F.J.B.)
| | - Irene Díaz
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Ana Belén Hitos
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28009 Madrid, Spain;
| | | | - Rosario Rodríguez-Griñolo
- Departamento de Economía, Métodos Cuantitativo e Historia Económica, Universidad Pablo de Olavide, 41013 Seville, Spain;
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Franz Martín
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Bernat Soria
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
- ISABIAL and Institute of Bioengineering, University Miguel Hernández de Elche, 03010 Alicante, Spain
| | - Juan R. Tejedo
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Francisco Javier Bedoya
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
- Correspondence: (E.C.-I.); (F.J.B.)
| |
Collapse
|
4
|
The Role of Protein S-Nitrosylation in Protein Misfolding-Associated Diseases. Life (Basel) 2021; 11:life11070705. [PMID: 34357077 PMCID: PMC8304259 DOI: 10.3390/life11070705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
Abnormal and excessive nitrosative stress contributes to neurodegenerative disease associated with the production of pathological levels of misfolded proteins. The accumulated findings strongly suggest that excessive NO production can induce and deepen these pathological processes, particularly by the S-nitrosylation of target proteins. Therefore, the relationship between S-nitrosylated proteins and the accumulation of misfolded proteins was reviewed. We particularly focused on the S-nitrosylation of E3-ubiquitin-protein ligase, parkin, and endoplasmic reticulum chaperone, PDI, which contribute to the accumulation of misfolded proteins. In addition to the target proteins being S-nitrosylated, NOS, which produces NO, and GSNOR, which inhibits S-nitrosylation, were also suggested as potential therapeutic targets for protein misfolding-associated diseases.
Collapse
|
5
|
Wang X, Meul T, Meiners S. Exploring the proteasome system: A novel concept of proteasome inhibition and regulation. Pharmacol Ther 2020; 211:107526. [PMID: 32173559 DOI: 10.1016/j.pharmthera.2020.107526] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/08/2020] [Indexed: 12/13/2022]
Abstract
The proteasome is a well-identified therapeutic target for cancer treatment. It acts as the main protein degradation system in the cell and degrades key mediators of cell growth, survival and function. The term "proteasome" embraces a whole family of distinct complexes, which share a common proteolytic core, the 20S proteasome, but differ by their attached proteasome activators. Each of these proteasome complexes plays specific roles in the control of cellular function. In addition, distinct proteasome interacting proteins regulate proteasome activity in subcellular compartments and in response to cellular signals. Proteasome activators and regulators may thus serve as building blocks to fine-tune proteasome function in the cell according to cellular needs. Inhibitors of the proteasome, e.g. the FDA approved drugs Velcade™, Kyprolis™, Ninlaro™, inactivate the catalytic 20S core and effectively block protein degradation of all proteasome complexes in the cell resulting in inhibition of cell growth and induction of apoptosis. Efficacy of these inhibitors, however, is hampered by their pronounced cytotoxic side-effects as well as by the emerging development of resistance to catalytic proteasome inhibitors. Targeted inhibition of distinct buiding blocks of the proteasome system, i.e. proteasome activators or regulators, represents an alternative strategy to overcome these limitations. In this review, we stress the importance of the diversity of the proteasome complexes constituting an entire proteasome system. Our building block concept provides a rationale for the defined targeting of distinct proteasome super-complexes in disease. We thereby aim to stimulate the development of innovative therapeutic approaches beyond broad catalytic proteasome inhibition.
Collapse
Affiliation(s)
- Xinyuan Wang
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Thomas Meul
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany.
| |
Collapse
|
6
|
Nitric oxide mediated redox regulation of protein homeostasis. Cell Signal 2018; 53:348-356. [PMID: 30408515 DOI: 10.1016/j.cellsig.2018.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Nitric oxide is a versatile diffusible signaling molecule, whose biosynthesis by three NO synthases (NOS) is tightly regulated at transcriptional and posttranslational levels, availability of co-factors, and calcium binding. Above normal levels of NO have beneficial protective effects for example in the cardiovascular system, but also contribute to the pathophysiology in the context of inflammatory diseases, and to aging and neurodegeneration in the nervous system. The effect specificity relies on the functional and spatial specificity of the NOS isoenzymes, and on the duality of two major signaling mechanisms (i) activation of soluble guanylycylase (sGC)-dependent cGMP production and (ii) direct S-nitrosylation of redox sensitive cysteines of susceptible proteins. The present review summarizes the functional implications of S-nitrosylation in the context of proteostasis, and focuses on two NO target proteins, heat shock cognate of 70 kDa (Hsc70/HSPA8) and the ubiquitin 2 ligase (UBE2D), because both are modified on functionally critical cysteines and are key regulators of chaperone mediated and assisted autophagy and proteasomal protein degradation. SNO modifications of these candidates are associated with protein accumulations and adoption of a senescent phenotype of neuronal cells suggesting that S-nitrosylations of protein homeostatic machineries contribute to aging phenomena.
Collapse
|
7
|
Bailey J, Davis S, Shaw A, Diotallevi M, Fischer R, Benson MA, Zhu H, Brown J, Bhattacharya S, Kessler BM, Channon KM, Crabtree MJ. Tetrahydrobiopterin modulates ubiquitin conjugation to UBC13/UBE2N and proteasome activity by S-nitrosation. Sci Rep 2018; 8:14310. [PMID: 30254268 PMCID: PMC6156325 DOI: 10.1038/s41598-018-32481-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/06/2018] [Indexed: 12/16/2022] Open
Abstract
Nitric Oxide (NO) is an intracellular signalling mediator, which affects many biological processes via the posttranslational modification of proteins through S-nitrosation. The availability of NO and NOS-derived reactive oxygen species (ROS) from enzymatic uncoupling are determined by the NO synthase cofactor Tetrahydrobiopterin (BH4). Here, using a global proteomics "biotin-switch" approach, we identified components of the ubiquitin-proteasome system to be altered via BH4-dependent NO signalling by protein S-nitrosation. We show S-nitrosation of ubiquitin conjugating E2 enzymes, in particular the catalytic residue C87 of UBC13/UBE2N, leading to impaired polyubiquitylation by interfering with the formation of UBC13~Ub thioester intermediates. In addition, proteasome cleavage activity in cells also seems to be altered by S-nitrosation, correlating with the modification of cysteine residues within the 19S regulatory particle and catalytic subunits of the 20S complex. Our results highlight the widespread impact of BH4 on downstream cellular signalling as evidenced by the effect of a perturbed BH4-dependent NO-Redox balance on critical processes within the ubiquitin-proteasome system (UPS). These studies thereby uncover a novel aspect of NO associated modulation of cellular homeostasis.
Collapse
Affiliation(s)
- Jade Bailey
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Simon Davis
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Andrew Shaw
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Marina Diotallevi
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Matthew A Benson
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Hanneng Zhu
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - James Brown
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Shoumo Bhattacharya
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Keith M Channon
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Mark J Crabtree
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| |
Collapse
|
8
|
Nitric oxide differentially affects proteasome activator 28 after arterial injury in type 1 and type 2 diabetic rats. J Surg Res 2016; 202:413-21. [PMID: 27229117 DOI: 10.1016/j.jss.2016.01.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND Diabetic patients display aggressive restenosis after vascular interventions, likely because of proproliferative influences of hyperglycemia and hyperinsulinemia. We have shown that nitric oxide (NO) inhibits neointimal hyperplasia in type 2, but not in type 1, diabetic rats. Here, we examined proteasome activator 28 (PA28) after arterial injury in different diabetic environments, with or without NO. We hypothesize that NO differentially affects PA28 levels based on metabolic environment. MATERIALS AND METHODS Vascular smooth muscle cell (VSMC) lysates from male, nondiabetic Lean Zucker (LZ) and Zucker Diabetic Fatty (ZDF) rats were assayed for 26S proteasome activity with or without PA28 and S-nitroso-N-acetylpenicillamine. LZ and ZDF VSMCs were treated with (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate for 24 h. Balloon-injured carotid arteries from LZ, streptozotocin-injected LZ (STZ, type 1), and ZDF (type 2) rats treated with disodium 1-[2-(carboxylato)pyrrolidin-1-iyl]diazen-1-ium-1,2-diolate were harvested at 3 or 14 d. PA28α was assessed by Western blotting and immunofluorescent staining. RESULTS S-nitroso-N-acetylpenicillamine reversed PA28-stimulated increases in 26S proteasome activity in LZ and ZDF VSMCs. Increased (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate lowered PA28α in LZ VSMCs but increased PA28α in ZDF VSMCs. At 3 d after injury, disodium 1-[2-(carboxylato)pyrrolidin-1-iyl]diazen-1-ium-1,2-diolate potentiated injury-induced PA28α decreases in LZ, STZ, and ZDF rats, suggesting VSMCs, depleted at this early time point, are major sources of PA28α. At 14 d after injury, total PA28α staining returned to baseline. However, although intimal and medial PA28α staining increased in injured STZ rats, adventitial PA28α staining increased in injured ZDF rats. CONCLUSIONS PA28 dysregulation may explain the differential ability of NO to inhibit neointimal hyperplasia in type 1 versus type 2 diabetes.
Collapse
|
9
|
Feng X, Jiang Y, Xie L, Jiang L, Li J, Sun C, Xu H, Wang R, Zhou M, Zhou Y, Dan H, Wang Z, Ji N, Deng P, Liao G, Geng N, Wang Y, Zhang D, Lin Y, Ye L, Liang X, Li L, Luo G, Feng M, Fang J, Zeng X, Wang Z, Chen Q. Overexpression of proteasomal activator PA28α serves as a prognostic factor in oral squamous cell carcinoma. J Exp Clin Cancer Res 2016; 35:35. [PMID: 26892607 PMCID: PMC4759779 DOI: 10.1186/s13046-016-0309-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 02/11/2016] [Indexed: 02/05/2023] Open
Abstract
Background Despite recent advances in oral squamous cell carcinoma (OSCC) diagnosis and therapy, disease recurrence remains common and is strongly associated with mortality. It is therefore critical to identify new targets for both treatment and diagnostic purposes. We aimed at investigating the role of PA28α, a proteasomal activator in OSCC. Methods The expression of PA28α was examined in a panel of OSCC cell lines and tissues, associated with oncomine analysis. In a large OSCC patient cohort, the prognostic value of PA28α expression was evaluated. Primary clinical end points were recurrence-free and overall survival rate. Functional involvement of PA28α in OSCC was examined in both in vitro and in vivo models upon specific siRNA knockdown. Results PA28α was found to be overexpressed in OSCC cell lines and tumor tissues. High expression of PA28α was significantly associated with recurrence and poorer overall survival. Specific knockdown of PA28α inhibited OSCC cell proliferation, migration, invasion in vitro and reduced the growth of OSCC xenografts in vivo. Multivariate Cox regression analyses revealed PA28α as independent prognostic predictors. Conclusions These results suggest that PA28α is involved in OSCC oncogenesis and may serve as a potential prognostic factor. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0309-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaodong Feng
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Yuchen Jiang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Liang Xie
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Lu Jiang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Jing Li
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Chongkui Sun
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Hao Xu
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China.
| | - Ruinan Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Min Zhou
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Yu Zhou
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Hongxia Dan
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Zhiyong Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Ning Ji
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Peng Deng
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Ga Liao
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Ning Geng
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Yun Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Dunfang Zhang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Xinhua Liang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Gang Luo
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Mingye Feng
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Juan Fang
- Guangdong Provincial Key Laboratory of Oral Diseases, Stomatological Hospital, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| | - Zhi Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China. .,Guangdong Provincial Key Laboratory of Oral Diseases, Stomatological Hospital, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, No. 14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
10
|
Wilck N, Ludwig A. Targeting the ubiquitin-proteasome system in atherosclerosis: status quo, challenges, and perspectives. Antioxid Redox Signal 2014; 21:2344-63. [PMID: 24506455 DOI: 10.1089/ars.2013.5805] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Atherosclerosis is a vascular disease of worldwide significance with fatal complications such as myocardial infarction, stroke, and peripheral artery disease. Atherosclerosis is recognized as a chronic inflammatory disease leading to arterial plaque formation and vessel narrowing in different vascular beds. Besides the strong inflammatory nature of atherosclerosis, it is also characterized by proliferation, apoptosis, and enhanced oxidative stress. The ubiquitin-proteasome system (UPS) is the major intracellular degradation system in eukaryotic cells. Besides its essential role in the degradation of dysfunctional and oxidatively damaged proteins, it is involved in many processes that influence disease progression in atherosclerosis. Hence, it is logical to ask whether targeting the proteasome is a reasonable and feasible option for the treatment of atherosclerosis. RECENT ADVANCES Several lines of evidence suggest stage-specific dysfunction of the UPS in atherogenesis. Regulation of key processes by the proteasome in atherosclerosis, as well as the modulation of these processes by proteasome inhibitors in vascular cells, is outlined in this review. The treatment of atherosclerotic animal models with proteasome inhibitors yielded partly opposing results, the potentially underlying reasons of which are discussed here. CRITICAL ISSUES AND FUTURE DIRECTIONS Targeting UPS function in atherosclerosis is a promising but challenging option. Limitations of current proteasome inhibitors, dose dependency, and the cell specificity of effects, as well as the potential of future therapeutics are discussed. A stage-specific in-depth exploration of UPS function in atherosclerosis in the future will help identify targets and windows for beneficial intervention.
Collapse
Affiliation(s)
- Nicola Wilck
- 1 Medizinische Klinik für Kardiologie und Angiologie, Charité-Universitätsmedizin Berlin , Campus Mitte, Berlin, Germany
| | | |
Collapse
|
11
|
Lavin B, Gómez M, Pello OM, Castejon B, Piedras MJ, Saura M, Zaragoza C. Nitric oxide prevents aortic neointimal hyperplasia by controlling macrophage polarization. Arterioscler Thromb Vasc Biol 2014; 34:1739-46. [PMID: 24925976 DOI: 10.1161/atvbaha.114.303866] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Nitric oxide synthase 3 (NOS3) prevents neointima hyperplasia by still unknown mechanisms. To demonstrate the significance of endothelial nitric oxide in the polarization of infiltrated macrophages through the expression of matrix metalloproteinase (MMP)-13 in neointima formation. APPROACH AND RESULTS After aortic endothelial denudation, NOS3 null mice show elevated neointima formation, detecting increased mobilization of LSK (lineage-negative [Lin]-stem-cell antigen 1 [SCA1]+KIT+) progenitor cells, and high ratios of M1 (proinflammatory) to M2 (resolving) macrophages, accompanied by high expression of interleukin-5, interleukin-6, MCP-1 (monocyte chemoattractant protein), VEGF (vascular endothelial growth factor), GM-CSF (granulocyte-macrophage colony stimulating factor), interleukin-1β, and interferon-γ. In conditional c-Myc knockout mice, in which M2 polarization is defective, denuded aortas showed extensive wall thickening as well. Conditioned medium from NOS3-deficient endothelium induced extensive repolarization of M2 macrophages to an M1 phenotype, and vascular smooth muscle cells proliferated and migrated faster in conditioned medium from M1 macrophages. Among the different proteins participating in cell migration, MMP-13 was preferentially expressed by M1 macrophages. M1-mediated vascular smooth muscle cell migration was inhibited when macrophages were isolated from MMP-13-deficient mice, whereas exogenous administration of MMP-13 to vascular smooth muscle cell fully restored migration. Excess vessel wall thickening in mice lacking NOS3 was partially reversed by simultaneous deletion of MMP-13, indicating that NOS3 prevents neointimal hyperplasia by preventing MMP-13 activity. An excess of M1-polarized macrophages that coexpress MMP-13 was also detected in human carotid samples from endarterectomized patients. CONCLUSIONS These findings indicate that at least M1 macrophage-mediated expression of MMP-13 in NOS3 null mice induces neointima formation after vascular injury, suggesting that MMP-13 may represent a new promising target in vascular disease.
Collapse
Affiliation(s)
- Begoña Lavin
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Monica Gómez
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Oscar M Pello
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Borja Castejon
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Maria J Piedras
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Marta Saura
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Carlos Zaragoza
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.).
| |
Collapse
|
12
|
Liu H, Yu S, Zhang H, Xu J. Identification of nitric oxide as an endogenous inhibitor of 26S proteasomes in vascular endothelial cells. PLoS One 2014; 9:e98486. [PMID: 24853093 PMCID: PMC4031199 DOI: 10.1371/journal.pone.0098486] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 05/02/2014] [Indexed: 01/22/2023] Open
Abstract
The 26S proteasome plays a fundamental role in almost all eukaryotic cells, including vascular endothelial cells. However, it remains largely unknown how proteasome functionality is regulated in the vasculature. Endothelial nitric oxide (NO) synthase (eNOS)-derived NO is known to be essential to maintain endothelial homeostasis. The aim of the present study was to establish the connection between endothelial NO and 26S proteasome functionality in vascular endothelial cells. The 26S proteasome reporter protein levels, 26S proteasome activity, and the O-GlcNAcylation of Rpt2, a key subunit of the proteasome regulatory complex, were assayed in 26S proteasome reporter cells, human umbilical vein endothelial cells (HUVEC), and mouse aortic tissues isolated from 26S proteasome reporter and eNOS knockout mice. Like the other selective NO donors, NO derived from activated eNOS (by pharmacological and genetic approach) increased O-GlcNAc modification of Rpt2, reduced proteasome chymotrypsin-like activity, and caused 26S proteasome reporter protein accumulation. Conversely, inactivation of eNOS reversed all the effects. SiRNA knockdown of O-GlcNAc transferase (OGT), the key enzyme that catalyzes protein O-GlcNAcylation, abolished NO-induced effects. Consistently, adenoviral overexpression of O-GlcNAcase (OGA), the enzyme catalyzing the removal of the O-GlcNAc group, mimicked the effects of OGT knockdown. Finally, compared to eNOS wild type aortic tissues, 26S proteasome reporter mice lacking eNOS exhibited elevated 26S proteasome functionality in parallel with decreased Rpt2 O-GlcNAcylation, without changing the levels of Rpt2 protein. In conclusion, the eNOS-derived NO functions as a physiological suppressor of the 26S proteasome in vascular endothelial cells.
Collapse
Affiliation(s)
- Hongtao Liu
- Section of Endocrinology, Department of Medicine and Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Shujie Yu
- Section of Endocrinology, Department of Medicine and Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Hua Zhang
- Section of Endocrinology, Department of Medicine and Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jian Xu
- Section of Endocrinology, Department of Medicine and Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
13
|
Vogt C, Xing Q, He W, Li B, Frost MC, Zhao F. Fabrication and Characterization of a Nitric Oxide-Releasing Nanofibrous Gelatin Matrix. Biomacromolecules 2013; 14:2521-30. [DOI: 10.1021/bm301984w] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Caleb Vogt
- Stem Cell and Tissue Engineering
Lab, Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Qi Xing
- Stem Cell and Tissue Engineering
Lab, Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Weilue He
- Polymer and Biomaterial Lab,
Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Bowen Li
- Department of Material Science
and Engineering Michigan Technological University Houghton, Michigan
49931, United States
| | - Megan C. Frost
- Polymer and Biomaterial Lab,
Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Feng Zhao
- Stem Cell and Tissue Engineering
Lab, Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
14
|
Tsihlis ND, Kapadia MR, Vavra AK, Flannery WD, Oustwani CS, Jiang Q, Kibbe MR. Nitric oxide may inhibit neointimal hyperplasia by decreasing isopeptidase T levels and activity in the vasculature. J Vasc Surg 2013; 58:179-86. [PMID: 23375434 DOI: 10.1016/j.jvs.2012.10.066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 09/27/2012] [Accepted: 10/01/2012] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Isopeptidase T is a cysteine protease deubiquitinating enzyme that hydrolyzes unanchored polyubiquitin chains to free monoubiquitin. Nitric oxide (NO) decreases 26S proteasome activity in vascular smooth muscle cells (VSMCs) and inhibits neointimal hyperplasia in animal models. As NO can cause S-nitrosylation of active-site cysteines, we hypothesize that NO inhibits isopeptidase T activity through S-nitrosylation. Because accumulation of polyubiquitin chains inhibits the 26S proteasome, this may be one mechanism through which NO prevents neointimal hyperplasia. METHODS To investigate our hypothesis, we examined the effect of NO on isopeptidase T activity, levels, and localization in VSMCs in vitro and in a rat carotid balloon injury model in vivo. RESULTS NO inhibited recombinant isopeptidase T activity by 82.8% (t = 60 minutes, P < .001 vs control). Dithiothreitol and glutathione (5 mmol/L) both significantly reversed NO-mediated inhibition of isopeptidase T activity (P < .001). NO caused a time-dependent increase in S-nitrosylated isopeptidase T levels in VSMCs, which was reversible with dithiothreitol, indicating that isopeptidase T undergoes reversible S-nitrosylation on exposure to NO in vitro. Although NO did not affect isopeptidase T levels or subcellular localization in VSMCs in vitro, it decreased isopeptidase T levels and increased ubiquitinated proteins after balloon injury in vivo. CONCLUSIONS Local administration of NO may prevent neointimal hyperplasia by inhibiting isopeptidase T levels and activity in the vasculature, thereby inhibiting the 26S proteasome in VSMCs. These data provide additional mechanistic insights into the ability of NO to prevent neointimal hyperplasia after vascular interventions.
Collapse
Affiliation(s)
- Nick D Tsihlis
- Division of Vascular Surgery, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | | | |
Collapse
|