1
|
Batori RK, Bordan Z, Padgett CA, Huo Y, Chen F, Atawia RT, Lucas R, Ushio-Fukai M, Fukai T, Belin de Chantemele EJ, Stepp DW, Fulton DJR. PFKFB3 Connects Glycolytic Metabolism with Endothelial Dysfunction in Human and Rodent Obesity. Antioxidants (Basel) 2025; 14:172. [PMID: 40002359 PMCID: PMC11851787 DOI: 10.3390/antiox14020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity and type 2 diabetes (T2D) increase cardiovascular risk, largely due to altered metabolic state. An early consequence of T2D/obesity is the loss of endothelial function and impaired nitric oxide (NO) signaling. In blood vessels, endothelial nitric oxide synthase (eNOS) synthesizes NO to maintain vessel homeostasis. The biological actions of NO are compromised by superoxide that is generated by NADPH oxidases (NOXs). Herein we investigated how altered metabolism affects superoxide/NO balance in obesity. We found that eNOS expression and NO bioavailability are significantly decreased in endothelial cells (ECs) from T2D patients and animal models of obesity. In parallel, PFKFB3, a key glycolytic regulatory enzyme, is significantly increased in ECs of obese animals. EC overexpression of wild-type and a cytosol-restricted mutant PFKFB3 decreased NO production due to increased eNOS-T495 phosphorylation. PFKFB3 also blunted Akt-S473 phosphorylation, reducing stimulus-dependent phosphorylation of S1177 and the activation of eNOS. Furthermore, PFKFB3 enhanced the activities of NOX1 and NOX5, which are major contributors to endothelial dysfunction. Prolonged exposure of ECs to high glucose or TNFα, which are hallmarks of T2D, leads to increased PFKFB3 expression. These results demonstrate a novel functional relationship between endothelial metabolism, ROS, and NO balance that may contribute to endothelial dysfunction in obesity.
Collapse
Affiliation(s)
- Robert K. Batori
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
| | - Zsuzsanna Bordan
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
| | - Caleb A. Padgett
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
| | - Yuqing Huo
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 210029, China;
| | - Reem T. Atawia
- Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK 73096, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Medicine (Cardiology), Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Eric J. Belin de Chantemele
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Medicine (Cardiology), Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David W. Stepp
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David J. R. Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.B.); (Z.B.); (C.A.P.); (R.L.); (M.U.-F.); (T.F.); (E.J.B.d.C.); (D.W.S.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
2
|
Zhang W, Yu P, Liu W, Wang L, Song X, Yao Y, Liu X, Meng X. Mechanism of sodium nitroprusside regulating ginseng quality. Sci Rep 2025; 15:1562. [PMID: 39794439 PMCID: PMC11724116 DOI: 10.1038/s41598-025-85905-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025] Open
Abstract
The roots of Panax ginseng C. A. Meyer (ginseng) are one of the traditional medicinal herbs in Asian countries and is known as the "king of all herbs". The most important active components of ginseng are the secondary metabolite saponins, which are closely related to ecological stress. Unsuitable ecological stress can generate a large amount of reactive oxygen species (ROS), by which the secondary metabolism is regulated, and the quality of herbs can be significantly improved. The purpose of this study was to investigate the effect of sodium nitroprusside (SNP) treatment on the quality of fresh ginseng roots. In this study, 5-year-old fresh ginseng was exposed to 0.1, 0.5, and 2 mmol/L SNP, a nitric oxide (NO) donor for five consecutive days. SNP significantly increased the levels of O2·-, H2O2, malondialdehyde (MDA), NADPH oxidase (NOX), superoxide dismutase (SOD), catalase (CAT), peroxides (POD), ascorbate peroxidase (APX), glutathione reductase (GR), ascorbate (AsA) and GSH/GSSG. The main root treated by 0.5 mmol/L SNP for three days was the best, with the activities of the key enzymes of the ginsenoside synthesis pathway, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR), farnesyl pyrophosphate synthase (FPS), squalene synthase (SS), squalene epoxidase (SE), and dammarane diol-II synthase (DS) activities increased markedly; the ginsenosides Rg1 + Re, Rb1, Rf, Rc, Rg2 + Rh1 and the total ginsenoside contents increased by 51.0%, 77.7%, 44.6%, 26.8%, 63.2% and 48.2%, respectively, but only a trace amount of the ginsenoside monomer Rb2 decreased 23.4%. The fibrous roots treated by 0.1 mmol/L SNP for four days showed the best effect, HMGCR, FPS, SS, SE, and DS also increased significantly; ginsenosides Rg1 + Re, Rb1, Ro, Rc, Rf, Rb3, Rb2, and total saponin contents increased 37.6%, 47.8%, 34.2%, 75.1%, 51.0%, 49.4%, 28.3%, and 20.4%, respectively. The 1,3-diphosphoglycerate (1,3-DPG) and phosphoenolpyruvate carboxylase (PEPC), related to primary metabolism, were also significantly elevated. The Morris water maze (MWM), histopathological analysis and oxidative stress indexes in brain tissues were used to evaluate the anti-aging effect, indicating that the SNP-treated ginseng further ameliorated D-gal-induced the impaired memory function and oxidative stress in mice, implying the efficacy of SNP-treated ginseng was better than untreated ginseng's. SNP can build the physiological state of ginseng under ecological stress, stimulate the antioxidant protection mechanism, increase the secondary metabolites, and improve the quality of ginseng.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, 150040, Hei-longjiang, China
| | - Pengcheng Yu
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, 150040, Hei-longjiang, China
| | - Wenfei Liu
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, 150040, Hei-longjiang, China
| | - Liyang Wang
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, 150040, Hei-longjiang, China
| | - Xiaowen Song
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, 150040, Hei-longjiang, China
| | - Yao Yao
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, 150040, Hei-longjiang, China
| | - Xiubo Liu
- Jiamusi College, Heilongjiang University of Chinese Medicine, Jiamusi, 154007, Heilongjiang, China
| | - Xiangcai Meng
- Department of Pharmacognosy, Heilongjiang University of Chinese Medicine, Harbin, 150040, Hei-longjiang, China.
| |
Collapse
|
3
|
Singh I, Kim J, Touhidul Islam SM, Fei Q, Singh AK, Won J. The role of S-nitrosoglutathione reductase (GSNOR) in T cell-mediated immunopathology of experimental autoimmune encephalomyelitis (EAE). Neuroscience 2025; 564:1-12. [PMID: 39532197 DOI: 10.1016/j.neuroscience.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/14/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Previously, we reported that both S-nitrosoglutathione (GSNO), a carrier of cellular nitric oxide, and N6022, an injectable form of GSNO reductase (GSNOR) inhibitor that increases endogenous GSNO levels, alleviate experimental autoimmune encephalomyelitis (EAE) in mice by suppressing Th1 and Th17 immune responses. Building on these findings, we explored the role of GSNOR in EAE pathogenesis and evaluated the efficacy of an orally active GSNOR inhibitor (N91115) in treating the EAE disease. EAE mice exhibited heightened expression/activity of GSNOR in the spinal cord, and the knockout of the GSNOR gene resulted in much milder clinical manifestations of EAE, with lower degrees of demyelination and axonal loss, reduced microglial and astrocyte activations, as well as suppressed Th1 and Th17 cell responses, alongside bolstered Treg immune responses. Next, we evaluated the efficacy of N91115 against EAE immunopathology. Consistent with our findings in GSNOR deficient EAE mice, daily N91115 administration reduced clinical EAE severity, with less spinal cord demyelination and axonal loss compared to untreated EAE mice. Furthermore, N91115 treated EAE mice showed diminished Th1 and Th17 immune responses and enhanced Treg responses. This observation underscores the potential of increased GSNOR expression and activity as a risk factor exacerbating EAE immunopathology, while simultaneously highlighting its potential as a target for modifying the disease. Furthermore, the balanced immune regulation provided by orally active N91115 (IL-6/IL-17a vs. IL-10) presents a promising alternative to immunosuppressive drugs, reducing the risk of opportunistic infections.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spinal Cord/metabolism
- Spinal Cord/immunology
- Mice, Inbred C57BL
- Female
- Mice, Knockout
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Mice
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Aldehyde Oxidoreductases/metabolism
- Aldehyde Oxidoreductases/antagonists & inhibitors
- Microglia/drug effects
- Microglia/metabolism
- Microglia/pathology
- Alcohol Dehydrogenase
Collapse
Affiliation(s)
- Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| | - Judong Kim
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - S M Touhidul Islam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Qiao Fei
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
4
|
Zhang X, Zhang D, Zhong C, Li W, Dinesh-Kumar SP, Zhang Y. Orchestrating ROS regulation: coordinated post-translational modification switches in NADPH oxidases. THE NEW PHYTOLOGIST 2025; 245:510-522. [PMID: 39468860 DOI: 10.1111/nph.20231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024]
Abstract
Reactive oxygen species (ROS) are among the most important signaling molecules, playing a significant role in plant growth, development, and responses to various environmental stresses. Respiratory burst oxidase homologs (RBOHs) are key enzymes in ROS production. Plants tightly regulate the activation and deactivation of RBOHs through various post-translational modifications (PTMs), including phosphorylation, ubiquitination, S-nitrosylation, and persulfidation. These PTMs fine-tune ROS production, ensuring normal plant growth and development while facilitating rapid responses to abiotic and biotic stresses. This review discusses the effects of different PTMs on RBOH function and their biological relevance. Additionally, we examine the evolutionary conservation of PTM sites and emphasize the complex interplay between multiple PTMs regulating RBOHs.
Collapse
Affiliation(s)
- Xinyu Zhang
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Dingliang Zhang
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Chenchen Zhong
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Wenli Li
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Savithramma P Dinesh-Kumar
- Department of Plant Biology and The Genome Center, College of Biological Sciences, University of California, Davis, Davis, CA, 95616, USA
| | - Yongliang Zhang
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
5
|
Barbosa PO, Tanus-Santos JE, Cavalli RDC, Bengtsson T, Montenegro MF, Sandrim VC. The Nitrate-Nitrite-Nitric Oxide Pathway: Potential Role in Mitigating Oxidative Stress in Hypertensive Disorders of Pregnancy. Nutrients 2024; 16:1475. [PMID: 38794713 PMCID: PMC11124146 DOI: 10.3390/nu16101475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Hypertensive diseases of pregnancy (HDPs) represent a global clinical challenge, affecting 5-10% of women and leading to complications for both maternal well-being and fetal development. At the heart of these complications is endothelial dysfunction, with oxidative stress emerging as a pivotal causative factor. The reduction in nitric oxide (NO) bioavailability is a vital indicator of this dysfunction, culminating in blood pressure dysregulation. In the therapeutic context, although antihypertensive medications are commonly used, they come with inherent concerns related to maternal-fetal safety, and a percentage of women do not respond to these therapies. Therefore, alternative strategies that directly address the pathophysiology of HDPs are required. This article focuses on the potential of the nitrate-nitrite-NO pathway, abundantly present in dark leafy greens and beetroot, as an alternative approach to treating HDPs. The objective of this review is to discuss the prospective antioxidant role of nitrate. We hope our discussion paves the way for using nitrate to improve endothelial dysfunction and control oxidative stress, offering a potential therapy for managing HDPs.
Collapse
Affiliation(s)
- Priscila Oliveira Barbosa
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, SP, Brazil; (P.O.B.)
| | - José E. Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo—USP, Ribeirão Preto 14049-900, SP, Brazil;
| | - Ricardo de Carvalho Cavalli
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, SP, Brazil; (P.O.B.)
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Marcelo F. Montenegro
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | | |
Collapse
|
6
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
7
|
Luo S, Ye D, Wang Y, Liu X, Wang X, Xie L, Ji Y. Roles of Protein S-Nitrosylation in Endothelial Homeostasis and Dysfunction. Antioxid Redox Signal 2024; 40:186-205. [PMID: 37742108 DOI: 10.1089/ars.2023.0406] [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: 09/25/2023]
Abstract
Significance: Nitric oxide (NO) plays several distinct roles in endothelial homeostasis. Except for activating the guanylyl cyclase enzyme-dependent cyclic guanosine monophosphate signaling pathway, NO can bind reactive cysteine residues in target proteins, a process known as S-nitrosylation (SNO). SNO is proposed to explain the multiple biological functions of NO in the endothelium. Investigating the targets and mechanism of protein SNO in endothelial cells (ECs) can provide new strategies for treating endothelial dysfunction-related diseases. Recent Advances: In response to different environments, proteomics has identified multiple SNO targets in ECs. Functional studies confirm that SNO regulates NO bioavailability, inflammation, permeability, oxidative stress, mitochondrial function, and insulin sensitivity in ECs. It also influences EC proliferation, migration, apoptosis, and transdifferentiation. Critical Issues: Single-cell transcriptomic analysis of ECs isolated from different mouse tissues showed heterogeneous gene signatures. However, litter research focuses on the heterogeneous properties of SNO proteins in ECs derived from different tissues. Although metabolism reprogramming plays a vital role in endothelial functions, little is known about how protein SNO regulates metabolism reprogramming in ECs. Future Directions: Precisely deciphering the effects of protein SNO in ECs isolated from different tissues under different conditions is necessary to further characterize the relationship between protein SNO and endothelial dysfunction-related diseases. In addition, identifying SNO targets that can influence endothelial metabolic reprogramming and the underlying mechanism can offer new views on the crosstalk between metabolism and post-translational protein modification. Antioxid. Redox Signal. 40, 186-205.
Collapse
Affiliation(s)
- Shanshan Luo
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Danyu Ye
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Yu Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xingeng Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xiaoqian Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| |
Collapse
|
8
|
Dent MR, DeMartino AW. Nitric oxide and thiols: Chemical biology, signalling paradigms and vascular therapeutic potential. Br J Pharmacol 2023:10.1111/bph.16274. [PMID: 37908126 PMCID: PMC11058123 DOI: 10.1111/bph.16274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023] Open
Abstract
Nitric oxide (• NO) interactions with biological thiols play crucial, but incompletely determined, roles in vascular signalling and other biological processes. Here, we highlight two recently proposed signalling paradigms: (1) the formation of a vasodilating labile nitrosyl ferrous haem (NO-ferrohaem) facilitated by thiols via thiyl radical generation and (2) polysulfides/persulfides and their interaction with • NO. We also describe the specific (bio)chemical routes in which • NO and thiols react to form S-nitrosothiols, a broad class of small molecules, and protein post-translational modifications that can influence protein function through catalytic site or allosteric structural changes. S-Nitrosothiol formation depends upon cellular conditions, but critically, an appropriate oxidant for either the thiol (yielding a thiyl radical) or • NO (yielding a nitrosonium [NO+ ]-donating species) is required. We examine the roles of these collective • NO/thiol species in vascular signalling and their cardiovascular therapeutic potential.
Collapse
Affiliation(s)
- Matthew R. Dent
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony W. DeMartino
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Cipriano A, Viviano M, Feoli A, Milite C, Sarno G, Castellano S, Sbardella G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J Med Chem 2023; 66:11632-11655. [PMID: 37650225 PMCID: PMC10510401 DOI: 10.1021/acs.jmedchem.3c00770] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Indexed: 09/01/2023]
Abstract
NADPH oxidases (NOXs) form a family of electron-transporting membrane enzymes whose main function is reactive oxygen species (ROS) generation. Strong evidence suggests that ROS produced by NOX enzymes are major contributors to oxidative damage under pathologic conditions. Therefore, blocking the undesirable actions of these enzymes is a therapeutic strategy for treating various pathological disorders, such as cardiovascular diseases, inflammation, and cancer. To date, identification of selective NOX inhibitors is quite challenging, precluding a pharmacologic demonstration of NOX as therapeutic targets in vivo. The aim of this Perspective is to furnish an updated outlook about the small-molecule NOX inhibitors described over the last two decades. Structures, activities, and in vitro/in vivo specificity are discussed, as well as the main biological assays used.
Collapse
Affiliation(s)
- Alessandra Cipriano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Monica Viviano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Alessandra Feoli
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Ciro Milite
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Giuliana Sarno
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Sabrina Castellano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Gianluca Sbardella
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| |
Collapse
|
10
|
Haigh S, Brown ZL, Shivers MA, Sellers HG, West MA, Barman SA, Stepp DW, Csanyi G, Fulton DJR. A Reappraisal of the Utility of L-012 to Measure Superoxide from Biologically Relevant Sources. Antioxidants (Basel) 2023; 12:1689. [PMID: 37759992 PMCID: PMC10525458 DOI: 10.3390/antiox12091689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The detection of superoxide anion (O2●-) in biological tissues remains challenging. Barriers to convenient and reproducible measurements include expensive equipment, custom probes, and the need for high sensitivity and specificity. The luminol derivative, L-012, has been used to measure O2●- since 1993 with mixed results and concerns over specificity. The goal of this study was to better define the conditions for use and their specificity. We found that L-012 coupled with depolymerized orthovanadate, a relatively impermeable tyrosine phosphatase inhibitor, yielded a highly sensitive approach to detect extracellular O2●-. In O2●- producing HEK-NOX5 cells, orthovanadate increased L-012 luminescence 100-fold. The combination of L-012 and orthovanadate was highly sensitive, stable, scalable, completely reversed by superoxide dismutase, and selective for O2●- generating NOXes versus NOX4, which produces H2O2. Moreover, there was no signal from cells transfected with NOS3 (NO●) and NOS2(ONOO-). To exclude the effects of altered tyrosine phosphorylation, O2●- was detected using non-enzymatic synthesis with phenazine methosulfate and via novel coupling of L-012 with niobium oxalate, which was less active in inducing tyrosine phosphorylation. Overall, our data shows that L-012 coupled with orthovanadate or other periodic group 5 salts yields a reliable, sensitive, and specific approach to measuring extracellular O2●- in biological systems.
Collapse
Affiliation(s)
- Stephen Haigh
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - Zach L. Brown
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - Mitch A. Shivers
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - Hunter G. Sellers
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - Madison A. West
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - Scott A. Barman
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - David W. Stepp
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - Gabor Csanyi
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| | - David J. R. Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
- David Fulton Vascular Biology Center, Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, CB 3316, Augusta, GA 30909, USA
| |
Collapse
|
11
|
Richter SM, Massman LC, Stuehr DJ, Sweeny EA. Functional interactions between NADPH oxidase 5 and actin. Front Cell Dev Biol 2023; 11:1116833. [PMID: 36776559 PMCID: PMC9909703 DOI: 10.3389/fcell.2023.1116833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
NADPH oxidase 5 (NOX5) is a transmembrane oxidative signaling enzyme which produces superoxide in response to intracellular calcium flux. Increasing evidence indicates that NOX5 is involved in a variety of physiological processes as well as human disease, however, details of NOX5 signaling pathways and targets of NOX5 mediated oxidative modifications remain poorly resolved. Actin dynamics have previously been shown to be modulated by oxidative modification, however, a direct connection to NOX5 expression and activity has not been fully explored. Here we show that NOX5 and actin interact in the cell, and each modulate the activity of the other. Using actin effector molecules jasplakinolide, cytochalasin D and latrunculin A, we show that changes in actin dynamics affect NOX5 superoxide production. In tandem, NOX5 oxidatively modifies actin, and shifts the ratio of filamentous to monomeric actin. Finally, we show that knockdown of NOX5 in the pancreatic cancer cell line PSN-1 impairs cell migration. Together our findings indicate an important link between actin dynamics and oxidative signaling through NOX5.
Collapse
Affiliation(s)
- Samantha M Richter
- Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lilyanna C Massman
- Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, United States
| | - Elizabeth A Sweeny
- Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
12
|
Ouyang M, Ouyang X, Peng Z, Liu M, Xu G, Zou Z, Zhang M, Shang Q. Heart-targeted amelioration of sepsis-induced myocardial dysfunction by microenvironment responsive nitric oxide nanogenerators in situ. J Nanobiotechnology 2022; 20:263. [PMID: 35672697 PMCID: PMC9171488 DOI: 10.1186/s12951-022-01457-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/07/2022] [Indexed: 12/28/2022] Open
Abstract
Background A balanced endogenous level of bioavailable nitric oxide (NO) plays a key role in maintaining cardiovascular homeostasis. The bioactive NO level in the cardiomyocytes was much reduced during sepsis. However, it is clinically challenging for the NO gas therapy due to the lack of spatial and temporal release system with precise control. The purpose of this study is to design a NO-releasing biomaterial with heart-targeted capability responsive to the infectious microenvironment, thus ameliorating lipopolysaccharide (LPS)-induced cardiac dysfunction. Results The heart-targeted NO delivery and in situ releasing system, PCM-MSN@LA, was synthesized using hollow mesoporous silica nanoparticles (MSN) as the carrier, and L-arginine (LA) as the NO donor. The myocardial delivery was successfully directed to heart by specific peptide (PCM) combined with low-intensity focused ultrasound (LIFU) guidance. The myocardial system synthesized NO from the LA released from PCM-MSN@LA in the presence of increased endogenous nitric oxide synthase (NOS) activity induced by LPS. This targeted NO release in situ achieved extraordinary protective effects against LPS-challenged myocardial injury by reducing the recruitment of inflammatory cells, inhibiting oxidative stress and maintaining the mitochondria integrity. In particular, this protection was not compromised by simultaneous circulation collapse as an adverse event in the context. Conclusions PCM-MSN@LA + LIFU exhibited extraordinary cardioprotective effects against severe sepsis in the hearts of LPS-treated animals without the side effect of NO diffusion. This technology has great potential to be served as a novel therapeutic strategy for sepsis-induced myocardial injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01457-y.
Collapse
|
13
|
Jung P, Ha E, Zhang M, Fall C, Hwang M, Taylor E, Stetkevich S, Bhanot A, Wilson CG, Figueroa JD, Obenaus A, Bragg S, Tone B, Eliamani S, Holshouser B, Blood AB, Liu T. Neuroprotective role of nitric oxide inhalation and nitrite in a Neonatal Rat Model of Hypoxic-Ischemic Injury. PLoS One 2022; 17:e0268282. [PMID: 35544542 PMCID: PMC9094545 DOI: 10.1371/journal.pone.0268282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 04/26/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND There is evidence from various models of hypoxic-ischemic injury (HII) that nitric oxide (NO) is protective. We hypothesized that either inhaled NO (iNO) or nitrite would alleviate brain injury in neonatal HII via modulation of mitochondrial function. METHODS We tested the effects of iNO and nitrite on the Rice-Vannucci model of HII in 7-day-old rats. Brain mitochondria were isolated for flow cytometry, aconitase activity, electron paramagnetic resonance, and Seahorse assays. RESULTS Pretreatment of pups with iNO decreased survival in the Rice-Vannucci model of HII, while iNO administered post-insult did not. MRI analysis demonstrated that pre-HII iNO at 40 ppm and post-HII iNO at 20 ppm decreased the brain lesion sizes from 6.3±1.3% to 1.0±0.4% and 1.8±0.8%, respectively. Intraperitoneal nitrite at 0.165 μg/g improved neurobehavioral performance but was harmful at higher doses and had no effect on brain infarct size. NO reacted with complex IV at the heme a3 site, decreased the oxidative stress of mitochondria challenged with anoxia and reoxygenation, and suppressed mitochondrial oxygen respiration. CONCLUSIONS This study suggests that iNO administered following neonatal HII may be neuroprotective, possibly via its modulation of mitochondrial function.
Collapse
Affiliation(s)
- Peter Jung
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Euntaik Ha
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Meijuan Zhang
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Carolyn Fall
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Mindy Hwang
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Emily Taylor
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Samuel Stetkevich
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Aditi Bhanot
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Christopher G. Wilson
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Johnny D. Figueroa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California, Irvine, CA, United States of America
| | - Shannon Bragg
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Beatriz Tone
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Saburi Eliamani
- Center for Imaging Research, Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Barbara Holshouser
- Center for Imaging Research, Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Arlin B. Blood
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Taiming Liu
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| |
Collapse
|
14
|
NO rapidly mobilizes cellular heme to trigger assembly of its own receptor. Proc Natl Acad Sci U S A 2022; 119:2115774119. [PMID: 35046034 PMCID: PMC8795550 DOI: 10.1073/pnas.2115774119] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/11/2022] Open
Abstract
Nitric oxide (NO) performs many biological functions, but how it operates at the molecular and cellular levels is not fully understood. We discovered that cell NO generation at physiologic levels triggers a rapid redeployment of intracellular heme, an iron-containing cofactor, and we show that this drives the assembly of the natural NO receptor protein, soluble guanylyl cyclase, which is needed for NO to perform its biological signaling functions. Our study uncovers a way that NO can shape biological signaling processes and a way that cells may use NO to control their hemeprotein activities through deployment of the heme cofactor. These concepts broaden our understanding of NO function in biology and medicine. Nitric oxide (NO) signaling in biology relies on its activating cyclic guanosine monophosphate (cGMP) production by the NO receptor soluble guanylyl cyclase (sGC). sGC must obtain heme and form a heterodimer to become functional, but paradoxically often exists as an immature heme-free form in cells and tissues. Based on our previous finding that NO can drive sGC maturation, we investigated its basis by utilizing a fluorescent sGC construct whose heme level can be monitored in living cells. We found that NO generated at physiologic levels quickly triggered cells to mobilize heme to immature sGC. This occurred when NO was generated within cells or by neighboring cells, began within seconds of NO exposure, and led cells to construct sGC heterodimers and thus increase their active sGC level by several-fold. The NO-triggered heme deployment involved cellular glyceraldehyde-3-phosphate dehydrogenase (GAPDH)–heme complexes and required the chaperone hsp90, and the newly formed sGC heterodimers remained functional long after NO generation had ceased. We conclude that NO at physiologic levels triggers assembly of its own receptor by causing a rapid deployment of cellular heme. Redirecting cellular heme in response to NO is a way for cells and tissues to modulate their cGMP signaling and to more generally tune their hemeprotein activities wherever NO biosynthesis takes place.
Collapse
|
15
|
Lairion F, Saporito-Magriñá C, Musacco-Sebio R, Fuda J, Torti H, Repetto MG. Nitric oxide, chronic iron and copper overloads and regulation of redox homeostasis in rat liver. J Biol Inorg Chem 2021; 27:23-36. [PMID: 34791544 DOI: 10.1007/s00775-021-01908-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/27/2021] [Indexed: 01/21/2023]
Abstract
Iron [Fe(II)] and copper [Cu(II)] ions produced liver oxidative stress and damage, and as a consequence, changes in the antioxidant protection. The objective of this work is to evaluate whether control of redox homeostasis in chronic overload of Fe(II) and Cu(II) is associated with nitric oxide (NO) and antioxidant enzymes protection in liver. Male Sprague-Dawley rats of 80-90 g received the standard diet ad libitum and drinking water supplemented with either 1.0 g/L of ferrous chloride (0.1% w/v, n = 24) or 0.5 g/L cupric sulfate (0.05% w/v, n = 24) for 42 days. The activities of the enzymes involved in the control of cellular redox homeostasis, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx), were determined by spectrophotometric methods, and NO production was determined by the determination of nitrite levels in liver. Chronic overload with Fe(II) and Cu(II) led to a significant increase of NO production while hampering the activity of NADPH oxidase. Meanwhile, the animals supplemented with Fe(II) showed a decrease in SOD and Gpx activities in liver homogenates with respect to baseline activity after 7 days of treatment, whereas the rats which received Cu(II) showed an increased SOD and catalase activity after 28 and 7 days of chronic overload. Further research is required to understand whether the modulation of the activity of these enzymes upon Cu and Fe overload is involved in a common toxic pathway or may serve to control the steady state of oxidant species related to redox signaling pathways.
Collapse
Affiliation(s)
- Fabiana Lairion
- Departamento de Ciencias Químicas, Cátedra de Química General e Inorgánica, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junin 956, 1113AAD, Buenos Aires, Argentina
| | - Christian Saporito-Magriñá
- Departamento de Ciencias Químicas, Cátedra de Química General e Inorgánica, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junin 956, 1113AAD, Buenos Aires, Argentina
| | - Rosario Musacco-Sebio
- Departamento de Ciencias Químicas, Cátedra de Química General e Inorgánica, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junin 956, 1113AAD, Buenos Aires, Argentina
| | - Julian Fuda
- Departamento de Físicomatemática, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Horacio Torti
- Departamento de Físicomatemática, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Marisa Gabriela Repetto
- Departamento de Ciencias Químicas, Cátedra de Química General e Inorgánica, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junin 956, 1113AAD, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Bioquímica y Medicina Molecular (IBIMOL, UBA-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
16
|
Sweeny EA, Hunt AP, Batka AE, Schlanger S, Lehnert N, Stuehr DJ. Nitric oxide and heme-NO stimulate superoxide production by NADPH oxidase 5. Free Radic Biol Med 2021; 172:252-263. [PMID: 34139309 PMCID: PMC8355125 DOI: 10.1016/j.freeradbiomed.2021.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 01/05/2023]
Abstract
Nitric oxide (NO) is a ubiquitous cell signaling molecule which mediates widespread and diverse processes in the cell. These NO dependent effects often involve activation (e.g. NO binding to the heme group of soluble guanylyl cyclase for cGMP production) or inactivation (e.g. S-nitrosation) of protein targets. We studied the effect of NO and heme-NO on the transmembrane signaling enzyme NADPH oxidase 5 (NOX5), a heme protein which produces superoxide in response to increases in intracellular calcium. We found that treatment with NO donors increases NOX5 activity through heme-dependent effects, and that this effect could be recapitulated by the addition of heme-NO. This work adds to our understanding of NOX5 regulation in the cell but also provides a framework for understanding how NO could cause widespread changes in hemeprotein activity based on different affinities for heme v. heme-NO, and helps explain the opposing roles NO plays in activation and inactivation of hemeprotein targets.
Collapse
Affiliation(s)
- Elizabeth A Sweeny
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrew P Hunt
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Allison E Batka
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Simon Schlanger
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nicolai Lehnert
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
17
|
Meng J, Lv Z, Zhang Y, Wang Y, Qiao X, Sun C, Chen Y, Guo M, Han W, Ye A, Xie T, Chu B, Shi C, Yang S, Chen C. Precision Redox: The Key for Antioxidant Pharmacology. Antioxid Redox Signal 2021; 34:1069-1082. [PMID: 33270507 PMCID: PMC8080931 DOI: 10.1089/ars.2020.8212] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
Significance: The redox balance of cells provides a stable microenvironment for biological macromolecules to perform their physiological functions. As redox imbalance is closely related to the occurrence and development of a variety of diseases, antioxidant therapies are an attractive option. However, redox-based therapeutic strategies have not yet shown satisfactory results. To find the key reason is of great significance. Recent Advances: We emphasize the precise nature of redox regulation and elucidate the importance and necessity of precision redox strategies from three aspects: differences in redox status, differences in redox function, and differences in the effects of redox therapy. We then propose the "5R" principle of precision redox in antioxidant pharmacology: "Right species, Right place, Right time, Right level, and Right target." Critical Issues: Redox status must be considered in the context of species, time, place, level, and target. The function of a biomacromolecule and its cellular signaling role are closely dependent on redox status. Accurate evaluation of redox status and specific interventions are critical for the success of redox treatments. Precision redox is the key for antioxidant pharmacology. The precise application of antioxidants as nutritional supplements is also key to the general health of the population. Future Directions: Future studies to develop more accurate methods for detecting redox status and accurately evaluating the redox state of different physiological and pathological processes are needed. Antioxidant pharmacology should consider the "5R" principle rather than continuing to apply global nonspecific antioxidant treatments. Antioxid. Redox Signal. 34, 1069-1082.
Collapse
Affiliation(s)
- Jiao Meng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Lv
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yingmin Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuanyuan Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chuanxin Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yuzhe Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Miaomiao Guo
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wensheng Han
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Aojun Ye
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ting Xie
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Boyu Chu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chang Shi
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shangpo Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Castro B, Citterico M, Kimura S, Stevens DM, Wrzaczek M, Coaker G. Stress-induced reactive oxygen species compartmentalization, perception and signalling. NATURE PLANTS 2021; 7:403-412. [PMID: 33846592 PMCID: PMC8751180 DOI: 10.1038/s41477-021-00887-0] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 02/24/2021] [Indexed: 05/19/2023]
Abstract
Reactive oxygen species (ROS) are essential for life and are involved in the regulation of almost all biological processes. ROS production is critical for plant development, response to abiotic stresses and immune responses. Here, we focus on recent discoveries in ROS biology emphasizing abiotic and biotic stress responses. Recent advancements have resulted in the identification of one of the first sensors for extracellular ROS and highlighted waves of ROS production during stress signalling in Arabidopsis. Enzymes that produce ROS, including NADPH oxidases, exhibit precise regulation through diverse post-translational modifications. Discoveries highlight the importance of both amino- and carboxy-terminal regulation of NADPH oxidases through protein phosphorylation and cysteine oxidation. Here, we discuss advancements in ROS compartmentalization, systemic ROS waves, ROS sensing and post-translational modification of ROS-producing enzymes and identify areas where foundational gaps remain.
Collapse
Affiliation(s)
- Bardo Castro
- Department of Plant Pathology, University of California, Davis, Davis, CA, USA
| | - Matteo Citterico
- Organismal and Evolutionary Biology Research Programme, Viikki Plant Science Centre, University of Helsinki, Helsinki, Finland
| | - Sachie Kimura
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan
| | - Danielle M Stevens
- Department of Plant Pathology, University of California, Davis, Davis, CA, USA
| | - Michael Wrzaczek
- Organismal and Evolutionary Biology Research Programme, Viikki Plant Science Centre, University of Helsinki, Helsinki, Finland.
- Institute of Plant Molecular Biology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic.
| | - Gitta Coaker
- Department of Plant Pathology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
19
|
Horimatsu T, Blomkalns AL, Ogbi M, Moses M, Kim D, Patel S, Gilreath N, Reid L, Benson TW, Pye J, Ahmadieh S, Thompson A, Robbins N, Mann A, Edgell A, Benjamin S, Stansfield BK, Huo Y, Fulton DJ, Agarwal G, Singh N, Offermanns S, Weintraub NL, Kim HW. Niacin protects against abdominal aortic aneurysm formation via GPR109A independent mechanisms: role of NAD+/nicotinamide. Cardiovasc Res 2020; 116:2226-2238. [PMID: 31710686 PMCID: PMC7695356 DOI: 10.1093/cvr/cvz303] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/23/2019] [Accepted: 11/07/2019] [Indexed: 01/07/2023] Open
Abstract
AIMS Chronic adventitial and medial infiltration of immune cells play an important role in the pathogenesis of abdominal aortic aneurysms (AAAs). Nicotinic acid (niacin) was shown to inhibit atherosclerosis by activating the anti-inflammatory G protein-coupled receptor GPR109A [also known as hydroxycarboxylic acid receptor 2 (HCA2)] expressed on immune cells, blunting immune activation and adventitial inflammatory cell infiltration. Here, we investigated the role of niacin and GPR109A in regulating AAA formation. METHODS AND RESULTS Mice were supplemented with niacin or nicotinamide, and AAA was induced by angiotensin II (AngII) infusion or calcium chloride (CaCl2) application. Niacin markedly reduced AAA formation in both AngII and CaCl2 models, diminishing adventitial immune cell infiltration, concomitant inflammatory responses, and matrix degradation. Unexpectedly, GPR109A gene deletion did not abrogate the protective effects of niacin against AAA formation, suggesting GPR109A-independent mechanisms. Interestingly, nicotinamide, which does not activate GPR109A, also inhibited AAA formation and phenocopied the effects of niacin. Mechanistically, both niacin and nicotinamide supplementation increased nicotinamide adenine dinucleotide (NAD+) levels and NAD+-dependent Sirt1 activity, which were reduced in AAA tissues. Furthermore, pharmacological inhibition of Sirt1 abrogated the protective effect of nicotinamide against AAA formation. CONCLUSION Niacin protects against AAA formation independent of GPR109A, most likely by serving as an NAD+ precursor. Supplementation of NAD+ using nicotinamide-related biomolecules may represent an effective and well-tolerated approach to preventing or treating AAA.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Calcium Chloride
- Cells, Cultured
- Dilatation, Pathologic
- Disease Models, Animal
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- NAD/metabolism
- Niacin/pharmacology
- Niacinamide/pharmacology
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Signal Transduction
- Sirtuin 1/metabolism
Collapse
Affiliation(s)
- Tetsuo Horimatsu
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Andra L Blomkalns
- Department of Emergency Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mourad Ogbi
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Mary Moses
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - David Kim
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Sagar Patel
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Nicole Gilreath
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Lauren Reid
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Tyler W Benson
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Jonathan Pye
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Samah Ahmadieh
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Allie Thompson
- Department of Emergency Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Robbins
- Department of Emergency Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Adrien Mann
- Department of Emergency Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ashlee Edgell
- Department of Emergency Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Stephanie Benjamin
- Department of Emergency Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brian K Stansfield
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yuqing Huo
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David J Fulton
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Gautam Agarwal
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nagendra Singh
- Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Ludwigstraße, Bad Nauheim, Germany
| | - Neal L Weintraub
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Ha Won Kim
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Vascular Biology Center, Medical College of Georgia, Augusta University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| |
Collapse
|
20
|
Oral nitrite treatment increases S-nitrosylation of vascular protein kinase C and attenuates the responses to angiotensin II. Redox Biol 2020; 38:101769. [PMID: 33126056 PMCID: PMC7596338 DOI: 10.1016/j.redox.2020.101769] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023] Open
Abstract
Nitrate and nitrite supplement deficient endogenous nitric oxide (NO) formation. While these anions may generate NO, recent studies have shown that circulating nitrite levels do not necessarily correlate with the antihypertensive effect of oral nitrite administration and that formation of nitrosylated species (RXNO) in the stomach is critically involved in this effect. This study examined the possibility that RXNO formed in the stomach after oral nitrite administration promotes target protein nitrosylation in the vasculature, inhibits vasoconstriction and the hypertensive responses to angiotensin II. Our results show that oral nitrite treatment enhances circulating RXNO concentrations (measured by ozone-based chemiluminescence methods), increases aortic protein kinase C (PKC) nitrosylation (measured by resin-assisted capture SNO-RAC method), and reduces both angiotensin II-induced vasoconstriction (isolated aortic ring preparation) and hypertensive (in vivo invasive blood pressure measurements) effects implicating PKC nitrosylation as a key mechanism for the responses to oral nitrite. Treatment of rats with the nitrosylating compound S-nitrosoglutathione (GSNO) resulted in the same effects described for oral nitrite. Moreover, partial depletion of thiols with buthionine sulfoximine prevented PKC nitrosylation and the blood pressure effects of oral nitrite. Further confirming a role for PKC nitrosylation, preincubation of aortas with GSNO attenuated the responses to both angiotensin II and to a direct PKC activator, and this effect was attenuated by ascorbate (reverses GSNO-induced nitrosylation). GSNO-induced nitrosylation also inhibited the increases in Ca2+ mobilization in angiotensin II-stimulated HEK293T cells expressing angiotensin type 1 receptor. Together, these results are consistent with the idea that PKC nitrosylation in the vasculature may underlie oral nitrite treatment-induced reduction in the vascular and hypertensive responses to angiotensin II. Oral nitrite treatment exerts antihypertensive effects. The mechanisms explaining such effects are not entirely known. Oral nitrite treatment increases circulating concentrations of nitrosylating species. Vascular PKC nitrosylation attenuates the vascular responses to angiotensin II.
Collapse
|
21
|
Abstract
Significance: The oxidative stress, resulting from an imbalance in the production and scavenging of reactive oxygen species (ROS), is known to be involved in the development and progression of several pathologies. The excess of ROS production is often due to an overactivation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) and for this reason these enzymes became promising therapeutic targets. However, even if NOX are now well characterized, the development of new therapies is limited by the lack of highly isoform-specific inhibitors. Recent Advances: In the past decade, several groups and laboratories have screened thousands of molecules to identify new specific inhibitors with low off-target effects. These works have led to the characterization of several new potent NOX inhibitors; however, their specificity varies a lot depending on the molecules. Critical Issues: Here, we are reviewing more than 25 known NOX inhibitors, focusing mainly on the newly identified ones such as APX-115, NOS31, Phox-I1 and 2, GLX7013114, and GSK2795039. To have a better overall view of these molecules, the inhibitors were classified according to their specificity, from pan-NOX inhibitors to highly isoform-specific ones. We are also presenting the use of these compounds both in vitro and in vivo. Future Directions: Several of these new molecules are potent and very specific inhibitors that could be good candidates for the development of new drugs. Even if the results are very promising, most of these compounds were only validated in vitro or in mice models and further investigations will be required before using them as potential therapies.
Collapse
Affiliation(s)
- Mathieu Chocry
- Aix-Marseille Université, Institut de Neurophysiopathologie (INP), CNRS, Marseille, France
| | - Ludovic Leloup
- Aix-Marseille Université, Institut de Neurophysiopathologie (INP), CNRS, Marseille, France
| |
Collapse
|
22
|
Sweeny EA, Schlanger S, Stuehr DJ. Dynamic regulation of NADPH oxidase 5 by intracellular heme levels and cellular chaperones. Redox Biol 2020; 36:101656. [PMID: 32738790 PMCID: PMC7394750 DOI: 10.1016/j.redox.2020.101656] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
NADPH oxidase 5 (NOX5) is a transmembrane signaling enzyme that produces superoxide in response to elevated cytosolic calcium. In addition to its association with numerous human diseases, NOX5 has recently been discovered to play crucial roles in the immune response and cardiovascular system. Details of NOX5 maturation, and specifically its response to changes in intracellular heme levels have remained unclear. Here we establish an experimental system in mammalian cells that allows us to probe the influence of heme availability on ROS production by NOX5. We identified a mode of dynamic regulatory control over NOX5 activity through modulation of its heme saturation and oligomeric state by intracellular heme levels and Hsp90 binding. This regulatory mechanism allows for fine-tuning and reversible modulation of NOX5 activity in response to stimuli.
Collapse
Affiliation(s)
- Elizabeth A Sweeny
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Simon Schlanger
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
23
|
Wei L, Zhang J, Wang C, Liao W. Recent progress in the knowledge on the alleviating effect of nitric oxide on heavy metal stress in plants. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2020; 147:161-171. [PMID: 31865162 DOI: 10.1016/j.plaphy.2019.12.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 11/03/2019] [Accepted: 12/16/2019] [Indexed: 06/10/2023]
Abstract
Recently, nitric oxide (NO), a redox-related signaling molecule, is considered to be a key regulator in plant growth and development as well as response to abiotic stresses. Heavy metal (HM) stress is one of the most serious threats to affect crop growth and production. HM stress attributes to the production of reactive oxygen species (ROS), leading to oxidative stress in plants. Thus, to minimize the toxic effects of HM stress, plants directly or indirectly activate different ROS-scavenging mechanisms comprised antioxidative enzymes and non-enzymatic antioxidants. Understanding the roles of NO is essential to elucidate how NO activates the appropriate set of responses to HM stress. Moreover, the regulation of key genes or proteins is very important in response to stress stimuli. Therefore, here we focus on the recent knowledge concerning the alleviating effect of NO on HM stress, covering from HM iron accumulation to antioxidant system to related gene and protein expression.
Collapse
Affiliation(s)
- Lijuan Wei
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District, Lanzhou, 730070, PR China
| | - Jing Zhang
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District, Lanzhou, 730070, PR China
| | - Chunlei Wang
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District, Lanzhou, 730070, PR China
| | - Weibiao Liao
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District, Lanzhou, 730070, PR China.
| |
Collapse
|
24
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
25
|
Bouressam ML, Lecat S, Raoul A, Gaucher C, Perrin-Sarrado C, Lartaud I, Dupuis F. S-nitrosoglutathione inhibits cerebrovascular angiotensin II-dependent and -independent AT 1 receptor responses: A possible role of S-nitrosation. Br J Pharmacol 2019; 176:2049-2062. [PMID: 30822355 DOI: 10.1111/bph.14644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 01/21/2019] [Accepted: 02/13/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Angiotensin II (AngII) and NO regulate the cerebral circulation. AngII AT1 receptors exert ligand-dependent and ligand-independent (myogenic tone [MT]) vasoconstriction of cerebral vessels. NO induces post-translational modifications of proteins such as S-nitrosation (redox modification of cysteine residues). In cultured cells, S-nitrosation decreases AngII's affinity for the AT1 receptor. The present work evaluated the functional consequences of S-nitrosation on both AngII-dependent and AngII-independent cerebrovascular responses. EXPERIMENTAL APPROACH S-Nitrosation was induced in rat isolated middle cerebral arteries by pretreatment with the NO donors, S-nitrosoglutathione (GSNO) or sodium nitroprusside (SNP). Agonist-dependent activation of AT1 receptors was evaluated by obtaining concentration-response curves to AngII. Ligand-independent activation of AT1 receptors was evaluated by calculating MT (active vs. passive diameter) at pressures ranging from 20 to 200 mmHg in the presence or not of a selective AT1 receptor inverse agonist. KEY RESULTS GSNO or SNP completely abolished the AngII-dependent AT1 receptor-mediated vasoconstriction of cerebral arteries. GSNO had no impact on responses to other vasoconstrictors sharing (phenylephrine, U46619) or not (5-HT) the same signalling pathway. MT was reduced by GSNO, and the addition of losartan did not further decrease MT, suggesting that GSNO blocks AT1 receptor-dependent MT. Ascorbate (which reduces S-nitrosated compounds) restored the response to AngII but not the soluble GC inhibitor ODQ, suggesting that these effects are mediated by S-nitrosation rather than by S-nitrosylation. CONCLUSIONS AND IMPLICATIONS In rat middle cerebral arteries, GSNO pretreatment specifically affects the AT1 receptor and reduces both AngII-dependent and AngII-independent activation, most likely through AT1 receptor S-nitrosation.
Collapse
Affiliation(s)
| | - Sandra Lecat
- BSC UMR7242 "GPCRs, pain and inflammation" team, CNRS, Université de Strasbourg Labex Medalis, Illkirch, France
| | | | | | | | | | | |
Collapse
|
26
|
Touyz RM, Anagnostopoulou A, Rios F, Montezano AC, Camargo LL. NOX5: Molecular biology and pathophysiology. Exp Physiol 2019; 104:605-616. [PMID: 30801870 PMCID: PMC6519284 DOI: 10.1113/ep086204] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review provides a comprehensive overview of Nox5 from basic biology to human disease and highlights unique features of this Nox isoform What advances does it highlight? Major advances in Nox5 biology relate to crystallization of the molecule and new insights into the pathophysiological role of Nox5. Recent discoveries have unravelled the crystal structure of Nox5, the first Nox isoform to be crystalized. This provides new opportunities to develop drugs or small molecules targeted to Nox5 in an isoform-specific manner, possibly for therapeutic use. Moreover genome wide association studies (GWAS) identified Nox5 as a new blood pressure-associated gene and studies in mice expressing human Nox5 in a cell-specific manner have provided new information about the (patho) physiological role of Nox5 in the cardiovascular system and kidneys. Nox5 seems to be important in the regulation of vascular contraction and kidney function. In cardiovascular disease and diabetic nephropathy, Nox5 activity is increased and this is associated with increased production of reactive oxygen species and oxidative stress implicated in tissue damage. ABSTRACT Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox), comprise seven family members (Nox1-Nox5 and dual oxidase 1 and 2) and are major producers of reactive oxygen species in mammalian cells. Reactive oxygen species are crucially involved in cell signalling and function. All Noxs share structural homology comprising six transmembrane domains with two haem-binding regions and an NADPH-binding region on the intracellular C-terminus, whereas their regulatory systems, mechanisms of activation and tissue distribution differ. This explains the diverse function of Noxs. Of the Noxs, NOX5 is unique in that rodents lack the gene, it is regulated by Ca2+ , it does not require NADPH oxidase subunits for its activation, and it is not glycosylated. NOX5 localizes in the perinuclear and endoplasmic reticulum regions of cells and traffics to the cell membrane upon activation. It is tightly regulated through numerous post-translational modifications and is activated by vasoactive agents, growth factors and pro-inflammatory cytokines. The exact pathophysiological significance of NOX5 remains unclear, but it seems to be important in the physiological regulation of sperm motility, vascular contraction and lymphocyte differentiation, and NOX5 hyperactivation has been implicated in cardiovascular disease, kidney injury and cancer. The field of NOX5 biology is still in its infancy, but with new insights into its biochemistry and cellular regulation, discovery of the NOX5 crystal structure and genome-wide association studies implicating NOX5 in disease, the time is now ripe to advance NOX5 research. This review provides a comprehensive overview of our current understanding of NOX5, from basic biology to human disease, and highlights the unique characteristics of this enigmatic Nox isoform.
Collapse
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Aikaterini Anagnostopoulou
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Francisco Rios
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Livia L. Camargo
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| |
Collapse
|
27
|
Luo M, Tian R, Lu N. Nitric oxide protected against NADPH oxidase-derived superoxide generation in vascular endothelium: Critical role for heme oxygenase-1. Int J Biol Macromol 2019; 126:549-554. [DOI: 10.1016/j.ijbiomac.2018.12.252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/25/2018] [Accepted: 12/26/2018] [Indexed: 12/26/2022]
|
28
|
Abstract
NOX (NADPH oxidases) are a family of NADPH-dependent transmembrane enzymes that synthesize superoxide and other reactive oxygen species. There are seven isoforms (NOX1-5 and DUOX1-2) which derive from a common ancestral NOX. NOX enzymes are distinguished by different modes of activation, the types of ROS that are produced, the cell types where they are expressed, and distinct functional roles. NOX5 was one of the earliest eukaryotic Nox enzymes to evolve and ironically the last isoform to be discovered in humans. In the time since its discovery, our knowledge of the regulation of NOX5 has expanded tremendously, and we now have a more comprehensive understanding of the molecular mechanisms underlying NOX5-dependent ROS production. In contrast, the cell types where NOX5 is robustly expressed and its functional significance in health and disease remain an underdeveloped area. The goal of this chapter is to provide an up-to-date overview of the mechanisms regulating NOX5 function and its importance in human physiology and pathophysiology.
Collapse
Affiliation(s)
- David J R Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
29
|
NADPH Oxidase (Rboh) Activity is Up Regulated during Sweet Pepper ( Capsicum annuum L.) Fruit Ripening. Antioxidants (Basel) 2019; 8:antiox8010009. [PMID: 30609654 PMCID: PMC6356770 DOI: 10.3390/antiox8010009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/06/2018] [Accepted: 12/25/2018] [Indexed: 11/21/2022] Open
Abstract
In plants, NADPH oxidase (NOX) is also known as a respiratory burst oxidase homolog (Rboh). This highly important enzyme, one of the main enzymatic sources of superoxide radicals (O2•−), is involved in the metabolism of reactive oxygen and nitrogen species (ROS and RNS), which is active in the non-climacteric pepper (Capsicum annuum L.) fruit. We used sweet pepper fruits at two ripening stages (green and red) to biochemically analyze the O2•−-generating Rboh activity and the number of isozymes during this physiological process. Malondialdehyde (MDA) content, an oxidative stress marker, was also assayed as an index of lipid peroxidation. In red fruits, MDA was observed to increase 2-fold accompanied by a 5.3-fold increase in total Rboh activity. Using in-gel assays of Rboh activity, we identified a total of seven CaRboh isozymes (I–VII) which were differentially modulated during ripening. CaRboh-III and CaRboh-I were the most prominent isozymes in green and red fruits, respectively. An in vitro assay showed that CaRboh activity is inhibited in the presence of nitric oxide (NO) donors, peroxynitrite (ONOO−) and glutathione (GSH), suggesting that CaRboh can undergo S-nitrosation, Tyr-nitration, and glutathionylation, respectively. In summary, this study provides a basic biochemical characterization of CaRboh activity in pepper fruits and indicates that this O2•−-generating Rboh is involved in nitro-oxidative stress associated with sweet pepper fruit ripening.
Collapse
|
30
|
Saxena N, Won J, Choi S, Singh AK, Singh I. S-nitrosoglutathione reductase (GSNOR) inhibitor as an immune modulator in experimental autoimmune encephalomyelitis. Free Radic Biol Med 2018; 121:57-68. [PMID: 29694854 PMCID: PMC6083447 DOI: 10.1016/j.freeradbiomed.2018.04.558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/27/2022]
Abstract
We previously reported that S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier, attenuated TH17-mediated immune responses in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). Cellular GSNO homeostasis is regulated via its synthesis by reaction between nitric oxide and glutathione and its enzymatic catabolism by GSNO reductase (GSNOR). In this study, we evaluated potential of reversible inhibitor of GSNOR (N6022) in comparison with exogenous GSNO in immunopathogenesis of EAE. Daily treatment of EAE mice with N6022 or exogenous GSNO significantly attenuated the clinical disease of EAE, but N6022 treatment showed greater efficacy than GSNO. Both N6022 and exogenous GSNO treatments increased the spleen levels of GSNO, as documented by increased protein-associated S-nitrosothiols, and inhibited polarization and CNS effector function of proinflammatory TH17 cells while inducing the polarization and CNS effector function of anti-inflammatory CD4+ CD25+ FOXP3- regulatory T (Treg) cells. Moreover, N6022 further attenuated TH1 while inducing TH2 and CD4+ CD25+ FOXP3+ Treg in their polarization and CNS effector functions. Similar to GSNO, the N6022 treatment protected against the EAE disease induced demyelination. However, neither exogenous GSNO nor N6022 treatment did not cause significant systemic lymphopenic effect as compared to FTY720. Taken together, these data document that optimization of cellular GSNO homeostasis by GSNOR inhibitor (N6022) in NO metabolizing cells attenuates EAE disease via selective inhibition of pro-inflammatory subsets of CD4+ cells (TH1/TH17) while upregulating anti-inflammatory subsets of CD4+ cells (TH2/Treg) without causing lymphopenic effects and thus offers a potential treatment option for MS/EAE.
Collapse
MESH Headings
- Alcohol Dehydrogenase/antagonists & inhibitors
- Animals
- Benzamides/pharmacology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/enzymology
- CD4-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Mice
- Mice, Inbred C57BL
- Protein S/metabolism
- Pyrroles/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/enzymology
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/drug effects
- Th1 Cells/enzymology
- Th1 Cells/immunology
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Nishant Saxena
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Seungho Choi
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| |
Collapse
|
31
|
Calabró V, Litterio MC, Fraga CG, Galleano M, Piotrkowski B. Effects of quercetin on heart nitric oxide metabolism in l-NAME treated rats. Arch Biochem Biophys 2018; 647:47-53. [PMID: 29621523 DOI: 10.1016/j.abb.2018.03.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/30/2018] [Accepted: 03/31/2018] [Indexed: 02/07/2023]
Abstract
This study investigated the effects of a quercetin-supplemented diet on the biochemical changes installed in the heart of NO-deficient rats in terms of oxidants production and NO bioavailability determinants. Sprague-Dawley rats were subjected to Nω-nitro-l-arginine methyl ester (l-NAME) treatment (360 mg/L l-NAME in the drinking water, 4 d) with or without supplementation with quercetin (4 g/kg diet). l-NAME administration led to increased blood pressure (BP) (30%), decreased nitric oxide synthase (NOS) activity (50%), and increases in NADPH oxidase (NOX)-dependent superoxide anion production (60%) and p47phox protein level (65%). The co-administration of quercetin prevented the increase in BP and the activation of NOX but did not modify the decrease in NOS activity caused by l-NAME. In addition, quercetin affected oxidative stress parameters as glutathione oxidation, and the activities of oxidant detoxifying enzymes superoxide dismutase, glutathione peroxidase, and catalase. Thus, quercetin administration counteracts l-NAME effects on NO bioavailability determinants in vivo, essentially through controlling NOX-mediated superoxide anion production.
Collapse
Affiliation(s)
- Valeria Calabró
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina
| | - María C Litterio
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina
| | - Cesar G Fraga
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina; Department of Nutrition, University of California, Davis, USA
| | - Monica Galleano
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina.
| | - Barbara Piotrkowski
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Bioquímica y Medicina Molecular (IBIMOL), UBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
32
|
Rouaud F, Boucher JL, Slama-Schwok A, Rocchi S. Mechanism of melanoma cells selective apoptosis induced by a photoactive NADPH analogue. Oncotarget 2018; 7:82804-82819. [PMID: 27756874 PMCID: PMC5347734 DOI: 10.18632/oncotarget.12651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/02/2016] [Indexed: 02/07/2023] Open
Abstract
Melanoma is one of the most lethal cancers when it reaches a metastatic stage. Despite the spectacular achievements of targeted therapies (BRAF inhibitors) or immuno-therapies (anti-CTLA4 or anti-PD1), most patients with melanoma will need additional treatments. Here we used a photoactive NADPH analogue called NS1 to induce cell death by inhibition of NADPH oxidases NOX in melanoma cells, including melanoma cells isolated from patients. In contrast, healthy melanocytes growth was unaffected by NS1 treatment. NS1 established an early Endoplasmic Reticulum stress by the early release of calcium mediated by (a) calcium-dependent redox-sensitive ion channel(s). These events initiated autophagy and apoptosis in all tested melanoma cells independently of their mutational status. The autophagy promoted by NS1 was incomplete. The autophagic flux was blocked at late stage events, consistent with the accumulation of p62, and a close localization of LC3 with NS1 associated with NS1 inhibition of NOX1 in autophagosomes. This hypothesis of a specific incomplete autophagy and apoptosis driven by NS1 was comforted by the use of siRNAs and pharmacological inhibitors blocking different processes. This study highlights the potential therapeutic interest of NS1 inducing cell death by triggering a selective ER stress and incomplete autophagy in melanoma cells harbouring wt and BRAF mutation.
Collapse
Affiliation(s)
- Florian Rouaud
- INSERM U1065 Team 1, Université de Nice Sophia Antipolis et Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | | | | | - Stéphane Rocchi
- INSERM U1065 Team 1, Université de Nice Sophia Antipolis et Centre Méditerranéen de Médecine Moléculaire, Nice, France
| |
Collapse
|
33
|
Lucena SV, Moura GEDD, Rodrigues T, Watashi CM, Melo FH, Icimoto MY, Viana GM, Nader HB, Monteiro HP, Tersariol ILS, Ogata FT. Heparan sulfate proteoglycan deficiency up-regulates the intracellular production of nitric oxide in Chinese hamster ovary cell lines. J Cell Physiol 2017; 233:3176-3194. [PMID: 28833096 DOI: 10.1002/jcp.26160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/17/2017] [Indexed: 01/01/2023]
Affiliation(s)
| | | | - Tiago Rodrigues
- Centro de Ciências Naturais e Humanas (CCNH)-UFABC; Santo André São Paulo Brazil
| | - Carolina M. Watashi
- Centro de Ciências Naturais e Humanas (CCNH)-UFABC; Santo André São Paulo Brazil
| | - Fabiana H. Melo
- Faculdade de Ciências Médicas da Santa Casa de São Paulo; São Paulo São Paulo Brazil
| | | | | | - Helena B. Nader
- Departamento de Bioquímica-UNIFESP; São Paulo São Paulo Brazil
| | | | - Ivarne L. S. Tersariol
- Departamento de Bioquímica-UNIFESP; São Paulo São Paulo Brazil
- Centro Interdisciplinar de Investigação Bioquímica UMC; Mogi das Cruzes São PauloSão Paulo Brazil
| | - Fernando T. Ogata
- Departamento de Bioquímica-UNIFESP; São Paulo São Paulo Brazil
- Division of Biochemistry, Medical Biochemistry & Biophysics, Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
34
|
The emerging role of NADPH oxidase NOX5 in vascular disease. Clin Sci (Lond) 2017; 131:981-990. [PMID: 28473473 DOI: 10.1042/cs20160846] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/14/2017] [Accepted: 03/19/2017] [Indexed: 01/11/2023]
Abstract
Oxidative stress is a consequence of up-regulation of pro-oxidant enzyme-induced reactive oxygen species (ROS) production and concomitant depletion of antioxidants. Elevated levels of ROS act as an intermediate and are the common denominator for various diseases including diabetes-associated macro-/micro-vascular complications and hypertension. A range of enzymes are capable of generating ROS, but the pro-oxidant enzyme family, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs), are the only enzymes known to be solely dedicated to ROS generation in the vascular tissues, kidney, aortas and eyes. While there is convincing evidence for a role of NOX1 in vascular and eye disease and for NOX4 in renal injury, the role of NOX5 in disease is less clear. Although NOX5 is highly up-regulated in humans in disease, it is absent in rodents. Thus, so far it has not been possible to study NOX5 in traditional mouse or rat models of disease. In the present review, we summarize and critically analyse the emerging evidence for a pathophysiological role of NOX5 in disease including the expression, regulation and molecular and cellular mechanisms which have been demonstrated to be involved in NOX5 activation.
Collapse
|
35
|
Fulton DJR, Li X, Bordan Z, Haigh S, Bentley A, Chen F, Barman SA. Reactive Oxygen and Nitrogen Species in the Development of Pulmonary Hypertension. Antioxidants (Basel) 2017; 6:antiox6030054. [PMID: 28684719 PMCID: PMC5618082 DOI: 10.3390/antiox6030054] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/29/2017] [Accepted: 07/01/2017] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease of the lung vasculature that involves the loss of endothelial function together with inappropriate smooth muscle cell growth, inflammation, and fibrosis. These changes underlie a progressive remodeling of blood vessels that alters flow and increases pulmonary blood pressure. Elevated pressures in the pulmonary artery imparts a chronic stress on the right ventricle which undergoes compensatory hypertrophy but eventually fails. How PAH develops remains incompletely understood and evidence for the altered production of reactive oxygen and nitrogen species (ROS, RNS respectively) in the pulmonary circulation has been well documented. There are many different types of ROS and RNS, multiple sources, and collective actions and interactions. This review summarizes past and current knowledge of the sources of ROS and RNS and how they may contribute to the loss of endothelial function and changes in smooth muscle proliferation in the pulmonary circulation.
Collapse
Affiliation(s)
- David J R Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Xueyi Li
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Zsuzsanna Bordan
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Stephen Haigh
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Austin Bentley
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China.
| | - Scott A Barman
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
36
|
Rojas M, Lemtalsi T, Toque HA, Xu Z, Fulton D, Caldwell RW, Caldwell RB. NOX2-Induced Activation of Arginase and Diabetes-Induced Retinal Endothelial Cell Senescence. Antioxidants (Basel) 2017; 6:antiox6020043. [PMID: 28617308 PMCID: PMC5488023 DOI: 10.3390/antiox6020043] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 05/30/2017] [Accepted: 06/09/2017] [Indexed: 12/19/2022] Open
Abstract
Increases in reactive oxygen species (ROS) and decreases in nitric oxide (NO) have been linked to vascular dysfunction during diabetic retinopathy (DR). Diabetes can reduce NO by increasing ROS and by increasing activity of arginase, which competes with nitric oxide synthase (NOS) for their commons substrate l-arginine. Increased ROS and decreased NO can cause premature endothelial cell (EC) senescence leading to defective vascular repair. We have previously demonstrated the involvement of NADPH oxidase 2 (NOX2)-derived ROS, decreased NO and overactive arginase in DR. Here, we investigated their impact on diabetes-induced EC senescence. Studies using diabetic mice and retinal ECs treated with high glucose or H2O2 showed that increases in ROS formation, elevated arginase expression and activity, and decreased NO formation led to premature EC senescence. NOX2 blockade or arginase inhibition prevented these effects. EC senescence was also increased by inhibition of NOS activity and this was prevented by treatment with a NO donor. These results indicate that diabetes/high glucose-induced activation of arginase and decreases in NO bioavailability accelerate EC senescence. NOX2-generated ROS contribute importantly to this process. Blockade of NOX2 or arginase represents a strategy to prevent diabetes-induced premature EC senescence by preserving NO bioavailability.
Collapse
Affiliation(s)
- Modesto Rojas
- Vascular Biology Center, Augusta University, 1459 Laney Walker Boulevard, Augusta, GA 30912-2500, USA.
- VA Medical Center, One Freedom Way, Augusta, GA 30904-6285, USA.
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, 1459 Laney Walker Boulevard, Augusta, GA 30912-2500, USA.
- VA Medical Center, One Freedom Way, Augusta, GA 30904-6285, USA.
| | - Haroldo A Toque
- Department of Pharmacology & Toxicology, Augusta University, 1459 Laney Walker, Boulevard, Augusta, GA 30912-2500, USA.
| | - Zhimin Xu
- Vascular Biology Center, Augusta University, 1459 Laney Walker Boulevard, Augusta, GA 30912-2500, USA.
- VA Medical Center, One Freedom Way, Augusta, GA 30904-6285, USA.
| | - David Fulton
- Vascular Biology Center, Augusta University, 1459 Laney Walker Boulevard, Augusta, GA 30912-2500, USA.
- Department of Pharmacology & Toxicology, Augusta University, 1459 Laney Walker, Boulevard, Augusta, GA 30912-2500, USA.
| | - Robert William Caldwell
- Department of Pharmacology & Toxicology, Augusta University, 1459 Laney Walker, Boulevard, Augusta, GA 30912-2500, USA.
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, 1459 Laney Walker Boulevard, Augusta, GA 30912-2500, USA.
- VA Medical Center, One Freedom Way, Augusta, GA 30904-6285, USA.
| |
Collapse
|
37
|
Lee HJ, Lee DY, Mariappan MM, Feliers D, Ghosh-Choudhury G, Abboud HE, Gorin Y, Kasinath BS. Hydrogen sulfide inhibits high glucose-induced NADPH oxidase 4 expression and matrix increase by recruiting inducible nitric oxide synthase in kidney proximal tubular epithelial cells. J Biol Chem 2017; 292:5665-5675. [PMID: 28188286 DOI: 10.1074/jbc.m116.766758] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/07/2017] [Indexed: 12/19/2022] Open
Abstract
High-glucose increases NADPH oxidase 4 (NOX4) expression, reactive oxygen species generation, and matrix protein synthesis by inhibiting AMP-activated protein kinase (AMPK) in renal cells. Because hydrogen sulfide (H2S) inhibits high glucose-induced matrix protein increase by activating AMPK in renal cells, we examined whether H2S inhibits high glucose-induced expression of NOX4 and matrix protein and whether H2S and NO pathways are integrated. High glucose increased NOX4 expression and activity at 24 h in renal proximal tubular epithelial cells, which was inhibited by sodium hydrosulfide (NaHS), a source of H2S. High glucose decreased AMPK phosphorylation and activity, which was restored by NaHS. Compound C, an AMPK inhibitor, prevented NaHS inhibition of high glucose-induced NOX4 expression. NaHS inhibition of high glucose-induced NOX4 expression was abrogated by N(ω)-nitro-l-arginine methyl ester, an inhibitor of NOS. NaHS unexpectedly augmented the expression of inducible NOS (iNOS) but not endothelial NOS. iNOS siRNA and 1400W, a selective iNOS inhibitor, abolished the ameliorative effects of NaHS on high glucose-induced NOX4 expression, reactive oxygen species generation, and, matrix laminin expression. Thus, H2S recruits iNOS to generate NO to inhibit high glucose-induced NOX4 expression, oxidative stress, and matrix protein accumulation in renal epithelial cells; the two gasotransmitters H2S and NO and their interaction may serve as therapeutic targets in diabetic kidney disease.
Collapse
Affiliation(s)
- Hak Joo Lee
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and.,the South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Doug Yoon Lee
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and
| | - Meenalakshmi M Mariappan
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and.,the South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Denis Feliers
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and.,the South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Goutam Ghosh-Choudhury
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and.,the South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Hanna E Abboud
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and.,the South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Yves Gorin
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and
| | - Balakuntalam S Kasinath
- From the University of Texas Health Science Center, San Antonio, Texas 78229-3900 and .,the South Texas Veterans Health Care System, San Antonio, Texas 78229
| |
Collapse
|
38
|
RhoA S-nitrosylation as a regulatory mechanism influencing endothelial barrier function in response to G +-bacterial toxins. Biochem Pharmacol 2016; 127:34-45. [PMID: 28017778 DOI: 10.1016/j.bcp.2016.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/19/2016] [Indexed: 01/05/2023]
Abstract
Disruption of the endothelial barrier in response to Gram positive (G+) bacterial toxins is a major complication of acute lung injury (ALI) and can be further aggravated by antibiotics which stimulate toxin release. The integrity of the pulmonary endothelial barrier is mediated by the balance of disruptive forces such as the small GTPase RhoA, and protective forces including endothelium-derived nitric oxide (NO). How NO protects against the barrier dysfunction is incompletely understood and our goal was to determine whether NO and S-nitrosylation can modulate RhoA activity and whether this mechanism is important for G+ toxin-induced microvascular permeability. We found that the G+ toxin listeriolysin-O (LLO) increased RhoA activity and that NO and S-NO donors inhibit RhoA activity. RhoA was robustly S-nitrosylated as determined by biotin-switch and mercury column analysis. MS revealed that three primary cysteine residues are S-nitrosylated including cys16, cys20 and cys159. Mutation of these residues to serine diminished S-nitrosylation to endogenous NO and mutant RhoA was less sensitive to inhibition by S-NO. G+-toxins stimulated the denitrosylation of RhoA which was not mediated by S-nitrosoglutathione reductase (GSNOR), thioredoxin (TRX) or thiol-dependent enzyme activity but was instead stimulated directly by elevated calcium levels. Calcium-promoted the direct denitrosylation of WT but not mutant RhoA and mutant RhoA adenovirus was more effective than WT in disrupting the barrier integrity of human lung microvascular endothelial cells. In conclusion, we reveal a novel mechanism by which NO and S-nitrosylation reduces RhoA activity which may be of significance in the management of pulmonary endothelial permeability induced by G+-toxins.
Collapse
|
39
|
Fuseler JW, Valarmathi MT. Nitric Oxide Modulates Postnatal Bone Marrow-Derived Mesenchymal Stem Cell Migration. Front Cell Dev Biol 2016; 4:133. [PMID: 27933292 PMCID: PMC5122209 DOI: 10.3389/fcell.2016.00133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/01/2016] [Indexed: 01/06/2023] Open
Abstract
Nitric oxide (NO) is a small free-radical gas molecule, which is highly diffusible and can activate a wide range of downstream effectors, with rapid and widespread cellular effects. NO is a versatile signaling mediator with a plethora of cellular functions. For example, NO has been shown to regulate actin, the microfilament, dependent cellular functions, and also acts as a putative stem cell differentiation-inducing agent. In this study, using a wound-healing model of cellular migration, we have explored the effect of exogenous NO on the kinetics of movement and morphological changes in postnatal bone marrow-derived mesenchymal stem cells (MSCs). Cellular migration kinetics and morphological changes of the migrating MSCs were measured in the presence of an NO donor (S-Nitroso-N-Acetyl-D,L-Penicillamine, SNAP), especially, to track the dynamics of single-cell responses. Two experimental conditions were assessed, in which SNAP (200 μM) was applied to the MSCs. In the first experimental group (SN-1), SNAP was applied immediately following wound formation, and migration kinetics were determined for 24 h. In the second experimental group (SN-2), MSCs were pretreated for 7 days with SNAP prior to wound formation and the determination of migration kinetics. The generated displacement curves were further analyzed by non-linear regression analysis. The migration displacement of the controls and NO treated MSCs (SN-1 and SN-2) was best described by a two parameter exponential functions expressing difference constant coefficients. Additionally, changes in the fractal dimension (D) of migrating MSCs were correlated with their displacement kinetics for all the three groups. Overall, these data suggest that NO may evidently function as a stop migration signal by disordering the cytoskeletal elements required for cell movement and proliferation of MSCs.
Collapse
Affiliation(s)
- John W Fuseler
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Mani T Valarmathi
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign Urbana, IL, USA
| |
Collapse
|
40
|
Chen F, Li X, Aquadro E, Haigh S, Zhou J, Stepp DW, Weintraub NL, Barman SA, Fulton DJR. Inhibition of histone deacetylase reduces transcription of NADPH oxidases and ROS production and ameliorates pulmonary arterial hypertension. Free Radic Biol Med 2016; 99:167-178. [PMID: 27498117 PMCID: PMC5240036 DOI: 10.1016/j.freeradbiomed.2016.08.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/30/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Excessive levels of reactive oxygen species (ROS) and increased expression of NADPH oxidases (Nox) have been proposed to contribute to pulmonary artery hypertension (PAH) and other cardiovascular diseases (CVD). Nox enzymes are major sources of ROS but the mechanisms regulating changes in Nox expression in disease states remain poorly understood. Epigenetics encompasses a number of mechanisms that cells employ to regulate the ability to read and transcribe DNA. Histone acetylation is a prominent example of an epigenetic mechanism regulating the expression of numerous genes by altering chromatin accessibility. The goal of this study was to determine whether inhibition of histone deacetylases (HDAC) affects the expression of Nox isoforms and reduces pulmonary hypertension. In immune cells, we found that multiple HDAC inhibitors robustly decreased Nox2 mRNA and protein expression in a dose-dependent manner concomitant with reduced superoxide production. This effect was not restricted to Nox2 as expression of Nox1, Nox4 and Nox5 was also reduced by HDAC inhibition. Surprisingly, Nox promoter-luciferase activity was unchanged in the presence of HDAC inhibitors. In macrophages and lung fibroblasts, ChIP experiments revealed that HDAC inhibitors block the binding of RNA polymerase II and the histone acetyltransferase p300 to the Nox2, Nox4 and Nox5 promoter regions and decrease histones activation marks (H3K4me3 and H3K9ac) at these promoter sites. We further show that the ability of CRISPR-ON to drive transcription of Nox1, Nox2, Nox4 and Nox5 genes is blocked by HDAC inhibitors. In a monocrotaline (MCT) rat model of PAH, multiple HDAC isoforms are upregulated in isolated pulmonary arteries, and HDAC inhibitors attenuate Nox expression in isolated pulmonary arteries and reduce indices of PAH. In conclusion, HDAC inhibitors potently suppress Nox gene expression both in vitro and in vivo via epigenetically regulating chromatin accessibility.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029 China; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA.
| | - Xueyi Li
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Emily Aquadro
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Stephen Haigh
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Jiliang Zhou
- Department of Pharmacology, Augusta University, Augusta, GA 30912, USA
| | - David W Stepp
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Medicine, Augusta University, Augusta, GA 30912, USA
| | - Scott A Barman
- Department of Pharmacology, Augusta University, Augusta, GA 30912, USA
| | - David J R Fulton
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Pharmacology, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
41
|
Zollbrecht C, Persson AEG, Lundberg JO, Weitzberg E, Carlström M. Nitrite-mediated reduction of macrophage NADPH oxidase activity is dependent on xanthine oxidoreductase-derived nitric oxide but independent of S-nitrosation. Redox Biol 2016; 10:119-127. [PMID: 27744114 PMCID: PMC5065649 DOI: 10.1016/j.redox.2016.09.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Inorganic nitrite has shown beneficial effects in cardiovascular and metabolic diseases partly via attenuation of NADPH-oxidase (NOX)-mediated oxidative stress. However, the exact mechanisms are still unclear. Here we investigated the role of S-nitrosation or altered expression of NOX subunits, and the role of xanthine oxidoreductase (XOR) in nitrite-derived nitric oxide (NO) production. METHODS Mouse macrophages were activated with LPS in the presence or absence of nitrite. NOX activity was measured by lucigenin-dependent chemiluminescence. Gene and protein expression of NOX2 subunits and XOR were investigated using qPCR and Western Blot. S-nitrosation of Nox2 and p22phox was studied with a Biotin Switch assay. Uric acid levels in cell culture medium were analyzed as a measure of XOR activity, and NO production was assessed by DAF-FM fluorescence. RESULTS NOX activity in activated macrophages was significantly reduced by nitrite. Reduced NOX activity was not attributed to decreased NOX gene expression. However, protein levels of p47phox and p67phox subunits were reduced by nitrite in activated macrophages. Protein expression of Nox2 and p22phox was not influenced by this treatment and neither was their S-nitrosation status. Increased uric acid levels after nitrite and diminished NO production during XOR-inhibition with febuxostat suggest that XOR is more active during nitrite-treatment of activated macrophages and plays an important role in the bioactivation of nitrite. CONCLUSIONS Our findings contribute to the mechanistic understanding about the therapeutic effects associated with nitrite supplementation in many diseases. We show that nitrite-mediated inhibition of NOX activity cannot be explained by S-nitrosation of the NOX enzyme, but that changes in NOX2 expression and XOR function may contribute.
Collapse
Affiliation(s)
- Christa Zollbrecht
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - A Erik G Persson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
42
|
Samuvel DJ, Shunmugavel A, Singh AK, Singh I, Khan M. S-Nitrosoglutathione ameliorates acute renal dysfunction in a rat model of lipopolysaccharide-induced sepsis. ACTA ACUST UNITED AC 2016; 68:1310-9. [PMID: 27484743 DOI: 10.1111/jphp.12608] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/05/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Sepsis induces an inflammatory response that results in acute renal failure (ARF). The current study is to evaluate the role of S-Nitrosoglutathione (GSNO) in renoprotection from lipopolysaccharide (LPS)-induced sepsis. METHODS Rats were divided to three groups. First group received LPS (5 mg/kg body weight), second group was treated with LPS + GSNO (50 μg/kg body weight), and third group was administered with vehicle (saline). They were sacrificed on day 1 and 3 post-LPS injection. Serum levels of nitric oxide (NO), creatinine and blood urea nitrogen (BUN) were analysed. Tissue morphology, T lymphocyte infiltrations, and the expression of inflammatory (TNF-α, iNOS) and anti-inflammatory (IL-10) mediators as well as glutathione (GSH) levels were determined. KEY FINDING Lipopolysaccharide significantly decreased body weight and increased cellular T lymphocyte infiltration, caspase-3 and iNOS and decreased PPAR-γ in renal tissue. NO, creatinine and BUN were significantly elevated after LPS challenge, and they significantly decreased after GSNO treatment. TNF-α level was found significantly increased in LPS-treated serum and kidney. GSNO treatment of LPS-challenged rats decreased caspase-3, iNOS, TNF-α, T lymphocyte infiltration and remarkably increased levels of IL-10, PPAR-γ and GSH. CONCLUSION GSNO can be used as a renoprotective agent for the treatment of sepsis-induced acute kidney injury.
Collapse
Affiliation(s)
- Devadoss J Samuvel
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
43
|
Chen F, Yin C, Dimitropoulou C, Fulton DJR. Cloning, Characteristics, and Functional Analysis of Rabbit NADPH Oxidase 5. Front Physiol 2016; 7:284. [PMID: 27486403 PMCID: PMC4950256 DOI: 10.3389/fphys.2016.00284] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/23/2016] [Indexed: 12/31/2022] Open
Abstract
Background: Nox5 was the last member of the Nox enzyme family to be identified. Functionally distinct from the other Nox isoforms, our understanding of its physiological significance has been hampered by the absence of Nox5 in mouse and rat genomes. Nox5 is present in the genomes of other species such as the rabbit that have broad utility as models of cardiovascular disease. However, the mRNA sequence, characteristics, and functional analysis of rabbit Nox5 has not been fully defined and were the goals of the current study. Methods: Rabbit Nox5 was amplified from rabbit tissue, cloned, and sequenced. COS-7 cells were employed for expression and functional analysis via Western blotting and measurements of superoxide. We designed and synthesized miRNAs selectively targeting rabbit Nox5. The nucleotide and amino acid sequences of rabbit Nox5 were aligned with those of putative rabbit isoforms (X1, X2, X3, and X4). A phylogenetic tree was generated based on the mRNA sequence for Nox5 from rabbit and other species. Results: Sequence alignment revealed that the identified rabbit Nox5 was highly conserved with the predicted sequence of rabbit Nox5. Cell based experiments reveal that rabbit Nox5 was robustly expressed and produced superoxide at rest and in a calcium and PMA-dependent manner that was susceptible to superoxide dismutase and the flavoprotein inhibitor, DPI. miRNA-1 was shown to be most effective in down-regulating the expression of rabbit Nox5. Phylogenetic analysis revealed a close relationship between rabbit and armadillo Nox5. Rabbit Nox5 was relatively closely related to human Nox5, but lies in a distinct cluster. Conclusion: Our study establishes the suitability of the rabbit as a model organism to further our understanding of the role of Nox5 in cardiovascular and other diseases and provides new information on the genetic relationship of Nox5 genes in different species.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical UniversityNanjing, Jiangsu, China; Vascular Biology Center, Medical College of Georgia at Augusta UniversityAugusta, GA, USA
| | - Caiyong Yin
- Department of Forensic Medicine, Nanjing Medical University Nanjing, Jiangsu, China
| | | | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University Augusta, GA, USA
| |
Collapse
|
44
|
Porpino SKP, Zollbrecht C, Peleli M, Montenegro MF, Brandão MCR, Athayde-Filho PF, França-Silva MS, Larsson E, Lundberg JO, Weitzberg E, Persson EG, Braga VA, Carlström M. Nitric oxide generation by the organic nitrate NDBP attenuates oxidative stress and angiotensin II-mediated hypertension. Br J Pharmacol 2016; 173:2290-302. [PMID: 27160064 DOI: 10.1111/bph.13511] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/11/2016] [Accepted: 05/02/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE NO deficiency and oxidative stress are crucially involved in the development or progression of cardiovascular disease, including hypertension and stroke. We have previously demonstrated that acute treatment with the newly discovered organic nitrate, 2-nitrate-1,3-dibuthoxypropan (NDBP), is associated with NO-like effects in the vasculature. This study aimed to further characterize the mechanism(s) and to elucidate the therapeutic potential in a model of hypertension and oxidative stress. EXPERIMENTAL APPROACH A combination of ex vivo, in vitro and in vivo approaches was used to assess the effects of NDBP on vascular reactivity, NO release, NADPH oxidase activity and in a model of hypertension. KEY RESULTS Ex vivo vascular studies demonstrated NDBP-mediated vasorelaxation in mesenteric resistance arteries, which was devoid of tolerance. In vitro studies using liver and kidney homogenates revealed dose-dependent and sustained NO generation by NDBP, which was attenuated by the xanthine oxidase inhibitor febuxostat. In addition, NDBP reduced NADPH oxidase activity in the liver and prevented angiotensin II-induced activation of NADPH oxidase in the kidney. In vivo studies showed that NDBP halted the progression of hypertension in mice with chronic angiotensin II infusion. This was associated with attenuated cardiac hypertrophy, and reduced NADPH oxidase-derived oxidative stress and fibrosis in the kidney and heart. CONCLUSION AND IMPLICATIONS The novel organic nitrate NDBP halts the progression of angiotensin II-mediated hypertension. Mechanistically, our findings suggest that NDBP treatment is associated with sustained NO release and attenuated activity of NADPH oxidase, which to some extent requires functional xanthine oxidase.
Collapse
Affiliation(s)
- Suênia K P Porpino
- Dept. of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Dept. of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Christa Zollbrecht
- Dept. of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Peleli
- Dept. of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Maria C R Brandão
- Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | | | - Erik Larsson
- Dept. of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jon O Lundberg
- Dept. of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Dept. of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Erik G Persson
- Dept. of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Valdir A Braga
- Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Mattias Carlström
- Dept. of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Abstract
Purpose of review Extensive data indicate a role for reactive oxygen species (ROS) and redox signaling in vascular damage in hypertension. However, molecular mechanisms underlying these processes remain unclear, but oxidative post-translational modification of vascular proteins is critical. This review discusses how proteins are oxidatively modified and how redox signaling influences vascular smooth muscle cell growth and vascular remodeling in hypertension. We also highlight Nox5 as a novel vascular ROS-generating oxidase. Recent findings Oxidative stress in hypertension leads to oxidative imbalance that affects vascular cell function through redox signaling. Many Nox isoforms produce ROS in the vascular wall, and recent findings show that Nox5 may be important in humans. ROS regulate signaling by numerous processes including cysteine oxidative post-translational modification such as S-nitrosylation, S-glutathionylation and sulfydration. In vascular smooth muscle cells, this influences cellular responses to oxidative stimuli promoting changes from a contractile to a proliferative phenotype. Summary In hypertension, Nox-induced ROS production is increased, leading to perturbed redox signaling through oxidative modifications of vascular proteins. This influences mitogenic signaling and cell cycle regulation, leading to altered cell growth and vascular remodeling in hypertension.
Collapse
|
46
|
Farnese FS, Menezes-Silva PE, Gusman GS, Oliveira JA. When Bad Guys Become Good Ones: The Key Role of Reactive Oxygen Species and Nitric Oxide in the Plant Responses to Abiotic Stress. FRONTIERS IN PLANT SCIENCE 2016; 7:471. [PMID: 27148300 PMCID: PMC4828662 DOI: 10.3389/fpls.2016.00471] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/24/2016] [Indexed: 05/18/2023]
Abstract
The natural environment of plants is composed of a complex set of abiotic stresses and their ability to respond to these stresses is highly flexible and finely balanced through the interaction between signaling molecules. In this review, we highlight the integrated action between reactive oxygen species (ROS) and reactive nitrogen species (RNS), particularly nitric oxide (NO), involved in the acclimation to different abiotic stresses. Under stressful conditions, the biosynthesis transport and the metabolism of ROS and NO influence plant response mechanisms. The enzymes involved in ROS and NO synthesis and scavenging can be found in different cells compartments and their temporal and spatial locations are determinant for signaling mechanisms. Both ROS and NO are involved in long distances signaling (ROS wave and GSNO transport), promoting an acquired systemic acclimation to abiotic stresses. The mechanisms of abiotic stresses response triggered by ROS and NO involve some general steps, as the enhancement of antioxidant systems, but also stress-specific mechanisms, according to the stress type (drought, hypoxia, heavy metals, etc.), and demand the interaction with other signaling molecules, such as MAPK, plant hormones, and calcium. The transduction of ROS and NO bioactivity involves post-translational modifications of proteins, particularly S-glutathionylation for ROS, and S-nitrosylation for NO. These changes may alter the activity, stability, and interaction with other molecules or subcellular location of proteins, changing the entire cell dynamics and contributing to the maintenance of homeostasis. However, despite the recent advances about the roles of ROS and NO in signaling cascades, many challenges remain, and future studies focusing on the signaling of these molecules in planta are still necessary.
Collapse
Affiliation(s)
- Fernanda S. Farnese
- Laboratory of Plant Ecophysiology, Instituto Federal Goiano – Campus Rio VerdeGoiás, Brazil
| | - Paulo E. Menezes-Silva
- Laboratory of Plant Ecophysiology, Instituto Federal Goiano – Campus Rio VerdeGoiás, Brazil
| | - Grasielle S. Gusman
- Laboratory of Plant Chemistry, Univiçosa – Faculdade de Ciências Biológicas e da SaúdeViçosa, Brazil
| | - Juraci A. Oliveira
- Department of General Biology, Universidade Federal de ViçosaViçosa, Brazil
| |
Collapse
|
47
|
Basudhar D, Ridnour LA, Cheng R, Kesarwala AH, Heinecke J, Wink DA. Biological signaling by small inorganic molecules. Coord Chem Rev 2016; 306:708-723. [PMID: 26688591 PMCID: PMC4680994 DOI: 10.1016/j.ccr.2015.06.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Small redox active molecules such as reactive nitrogen and oxygen species and hydrogen sulfide have emerged as important biological mediators that are involved in various physiological and pathophysiological processes. Advancement in understanding of cellular mechanisms that tightly regulate both generation and reactivity of these molecules is central to improved management of various disease states including cancer and cardiovascular dysfunction. Imbalance in the production of redox active molecules can lead to damage of critical cellular components such as cell membranes, proteins and DNA and thus may trigger the onset of disease. These small inorganic molecules react independently as well as in a concerted manner to mediate physiological responses. This review provides a general overview of the redox biology of these key molecules, their diverse chemistry relevant to physiological processes and their interrelated nature in cellular signaling.
Collapse
Affiliation(s)
- Debashree Basudhar
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Lisa A. Ridnour
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Robert Cheng
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Aparna H. Kesarwala
- Radiation Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Julie Heinecke
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - David A. Wink
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| |
Collapse
|
48
|
|
49
|
Chen F, Haigh S, Yu Y, Benson T, Wang Y, Li X, Dou H, Bagi Z, Verin AD, Stepp DW, Csanyi G, Chadli A, Weintraub NL, Smith SME, Fulton DJR. Nox5 stability and superoxide production is regulated by C-terminal binding of Hsp90 and CO-chaperones. Free Radic Biol Med 2015; 89:793-805. [PMID: 26456056 PMCID: PMC4751585 DOI: 10.1016/j.freeradbiomed.2015.09.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 08/15/2015] [Accepted: 09/03/2015] [Indexed: 10/22/2022]
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone that orchestrates the folding and stability of proteins that regulate cellular signaling, proliferation and inflammation. We have previously shown that Hsp90 controls the production of reactive oxygen species by modulating the activity of Noxes1-3 and 5, but not Nox4. The goal of the current study was to define the regions on Nox5 that bind Hsp90 and determine how Hsp90 regulates enzyme activity. In isolated enzyme activity assays, we found that Hsp90 inhibitors selectively decrease superoxide, but not hydrogen peroxide, production. The addition of Hsp90 alone only modestly increases Nox5 enzyme activity but in combination with the co-chaperones, Hsp70, HOP, Hsp40, and p23 it robustly stimulated superoxide, but not hydrogen peroxide, production. Proximity ligation assays reveal that Nox5 and Hsp90 interact in intact cells. In cell lysates using a co-IP approach, Hsp90 binds to Nox5 but not Nox4, and the degree of binding can be influenced by calcium-dependent stimuli. Inhibition of Hsp90 induced the degradation of full length, catalytically inactive and a C-terminal fragment (aa398-719) of Nox5. In contrast, inhibition of Hsp90 did not affect the expression levels of N-terminal fragments (aa1-550) suggesting that Hsp90 binding maintains the stability of C-terminal regions. In Co-IP assays, Hsp90 was bound only to the C-terminal region of Nox5. Further refinement using deletion analysis revealed that the region between aa490-550 mediates Hsp90 binding. Converse mapping experiments show that the C-terminal region of Nox5 bound to the M domain of Hsp90 (aa310-529). In addition to Hsp90, Nox5 bound other components of the foldosome including co-chaperones Hsp70, HOP, p23 and Hsp40. Silencing of HOP, Hsp40 and p23 reduced Nox5-dependent superoxide. In contrast, increased expression of Hsp70 decreased Nox5 activity whereas a mutant of Hsp70 failed to do so. Inhibition of Hsp90 results in the loss of higher molecular weight complexes of Nox5 and decreased interaction between monomers. Collectively these results show that the C-terminal region of Nox5 binds to the M domain of Hsp90 and that the binding of Hsp90 and select co-chaperones facilitate oligomerization and the efficient production of superoxide.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029 China; Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA.
| | - Steven Haigh
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Yanfang Yu
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Tyler Benson
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Yusi Wang
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Xueyi Li
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Huijuan Dou
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Zsolt Bagi
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Alexander D Verin
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - David W Stepp
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Gabor Csanyi
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Ahmed Chadli
- Cancer Research Center, Molecular Chaperones Program, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Susan M E Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw GA 30152, USA
| | - David J R Fulton
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA.
| |
Collapse
|
50
|
Qian J, Tian W, Jiang X, Tamosiuniene R, Sung YK, Shuffle EM, Tu AB, Valenzuela A, Jiang S, Zamanian RT, Fiorentino DF, Voelkel NF, Peters-Golden M, Stenmark KR, Chung L, Rabinovitch M, Nicolls MR. Leukotriene B4 Activates Pulmonary Artery Adventitial Fibroblasts in Pulmonary Hypertension. Hypertension 2015; 66:1227-1239. [PMID: 26558820 DOI: 10.1161/hypertensionaha.115.06370] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 09/10/2015] [Indexed: 12/14/2022]
Abstract
A recent study demonstrated a significant role for leukotriene B4 (LTB4) causing pulmonary vascular remodeling in pulmonary arterial hypertension. LTB4 was found to directly injure luminal endothelial cells and promote growth of the smooth muscle cell layer of pulmonary arterioles. The purpose of this study was to determine the effects of LTB4 on the pulmonary adventitial layer, largely composed of fibroblasts. Here, we demonstrate that LTB4 enhanced human pulmonary artery adventitial fibroblast proliferation, migration, and differentiation in a dose-dependent manner through its cognate G-protein-coupled receptor, BLT1. LTB4 activated human pulmonary artery adventitial fibroblast by upregulating p38 mitogen-activated protein kinase as well as Nox4-signaling pathways. In an autoimmune model of pulmonary hypertension, inhibition of these pathways blocked perivascular inflammation, decreased Nox4 expression, reduced reactive oxygen species production, reversed arteriolar adventitial fibroblast activation, and attenuated pulmonary hypertension development. This study uncovers a novel mechanism by which LTB4 further promotes pulmonary arterial hypertension pathogenesis, beyond its established effects on endothelial and smooth muscle cells, by activating adventitial fibroblasts.
Collapse
Affiliation(s)
- Jin Qian
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| | - Rasa Tamosiuniene
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| | - Yon K Sung
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| | - Eric M Shuffle
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| | - Allen B Tu
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| | | | - Shirley Jiang
- Stanford University, School of Medicine, Stanford, CA 94305
| | | | | | | | | | - Kurt R Stenmark
- University of Colorado Denver, School of Medicine, Aurora, CO 80045
| | - Lorinda Chung
- Stanford University, School of Medicine, Stanford, CA 94305
| | | | - Mark R Nicolls
- VA Palo Alto Health Care System, Palo Alto, CA 94304.,Stanford University, School of Medicine, Stanford, CA 94305
| |
Collapse
|