101
|
Kim HJ, Ha S, Lee HY, Lee KJ. ROSics: chemistry and proteomics of cysteine modifications in redox biology. MASS SPECTROMETRY REVIEWS 2015; 34:184-208. [PMID: 24916017 PMCID: PMC4340047 DOI: 10.1002/mas.21430] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 04/30/2013] [Accepted: 11/20/2013] [Indexed: 05/29/2023]
Abstract
Post-translational modifications (PTMs) occurring in proteins determine their functions and regulations. Proteomic tools are available to identify PTMs and have proved invaluable to expanding the inventory of these tools of nature that hold the keys to biological processes. Cysteine (Cys), the least abundant (1-2%) of amino acid residues, are unique in that they play key roles in maintaining stability of protein structure, participating in active sites of enzymes, regulating protein function and binding to metals, among others. Cys residues are major targets of reactive oxygen species (ROS), which are important mediators and modulators of various biological processes. It is therefore necessary to identify the Cys-containing ROS target proteins, as well as the sites and species of their PTMs. Cutting edge proteomic tools which have helped identify the PTMs at reactive Cys residues, have also revealed that Cys residues are modified in numerous ways. These modifications include formation of disulfide, thiosulfinate and thiosulfonate, oxidation to sulfenic, sulfinic, sulfonic acids and thiosulfonic acid, transformation to dehydroalanine (DHA) and serine, palmitoylation and farnesylation, formation of chemical adducts with glutathione, 4-hydroxynonenal and 15-deoxy PGJ2, and various other chemicals. We present here, a review of relevant ROS biology, possible chemical reactions of Cys residues and details of the proteomic strategies employed for rapid, efficient and sensitive identification of diverse and novel PTMs involving reactive Cys residues of redox-sensitive proteins. We propose a new name, "ROSics," for the science which describes the principles of mode of action of ROS at molecular levels.
Collapse
Affiliation(s)
- Hee-Jung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans UniversitySeoul, 120-750, Korea
| | - Sura Ha
- Department of Chemistry, Korea Advanced Institute of Science & Technology (KAIST)Daejeon, 305-701, Korea
| | - Hee Yoon Lee
- Department of Chemistry, Korea Advanced Institute of Science & Technology (KAIST)Daejeon, 305-701, Korea
| | - Kong-Joo Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans UniversitySeoul, 120-750, Korea
| |
Collapse
|
102
|
Sun QA, Runge MS, Madamanchi NR. Oxidative stress, NADPH oxidases, and arteries. Hamostaseologie 2015; 36:77-88. [PMID: 25649240 DOI: 10.5482/hamo-14-11-0076] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis and its major complications - myocardial infarction and stroke - remain major causes of death and disability in the United States and world-wide. Indeed, with dramatic increases in obesity and diabetes mellitus, the prevalence and public health impact of cardiovascular diseases (CVD) will likely remain high. Major advances have been made in development of new therapies to reduce the incidence of atherosclerosis and CVD, in particular for treatment of hypercholesterolemia and hypertension. Oxidative stress is the common mechanistic link for many CVD risk factors. However, only recently have the tools existed to study the interface between oxidative stress and CVD in animal models. The most important source of reactive oxygen species (and hence oxidative stress) in vascular cells are the multiple forms of enzymes nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase). Recently published and emerging studies now clearly establish that: 1) NADPH oxidases are of critical importance in atherosclerosis and hypertension in animal models; 2) given the tissue-specific expression of key components of NADPH oxidase, it may be possible to target vascular oxidative stress for prevention of CVD.
Collapse
Affiliation(s)
| | - Marschall S Runge
- Marschall S. Runge, MD PhD, Department of Medicine, 125 MacNider Hall, University of North Carolina, Chapel Hill, NC 27599-7005, USA, E-mail:
| | | |
Collapse
|
103
|
Weaver JR, Grzesik W, Taylor-Fishwick DA. Inhibition of NADPH oxidase-1 preserves beta cell function. Diabetologia 2015; 58:113-21. [PMID: 25277953 DOI: 10.1007/s00125-014-3398-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/09/2014] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Upregulation of the reactive oxygen species (ROS)-producing enzyme NADPH oxidase (NOX)-1 in islets and beta cells follows acute exposure to inflammatory cytokines and is concomitant with beta cell dysfunction. NOX-1 is a candidate mediator of inflammation-induced beta cell dysfunction. This study aimed to determine whether selective inhibition of NADPH oxidase-1 presents a new strategy to preserve beta cell function. METHODS Induced beta cell dysfunction was studied in primary human donor islets, isolated mouse islets and murine beta cell lines. Islets and beta cells were stimulated with inflammatory cytokines (TNF-α, IL-1β, IFN-γ). NOX-1 activity was blocked by the selective inhibitor ML171. RESULTS Cytokine induction of intracellular ROS was reduced 80% with 1 μmol/l ML171 in murine beta cell lines (p < 0.01). Cytokine-induced apoptosis, measured by caspase-3 activation or quantified fluorescence microscopy, was prevented in islets and beta cell lines up to 100% with ML171 in a concentration-dependent manner (p < 0.05). Functionally, glucose-stimulated insulin secretion was abolished by cytokine exposure but preserved by ML171 in isolated mouse islets and murine beta cell lines. A feed-forward regulation of NOX-1 in islets and beta cell lines was disrupted by ML171. CONCLUSIONS/INTERPRETATION Stimulation of NOX-1 activity is a major component of inflammatory cytokine-induced beta cell dysfunction. Significant protection of beta cells is conferred with selective inhibition of NOX-1. Suppression of NOX-1 activity may present a new therapeutic strategy to preserve and protect beta cell function in diabetes.
Collapse
Affiliation(s)
- Jessica R Weaver
- Department of Microbiology and Molecular Cell Biology, LH 2128, Eastern Virginia Medical School, 700 W. Olney Road, Norfolk, VA, 23507, USA
| | | | | |
Collapse
|
104
|
Trocme C, Deffert C, Cachat J, Donati Y, Tissot C, Papacatzis S, Braunersreuther V, Pache JC, Krause KH, Holmdahl R, Barazzone-Argiroffo C, Carnesecchi S. Macrophage-specific NOX2 contributes to the development of lung emphysema through modulation of SIRT1/MMP-9 pathways. J Pathol 2014; 235:65-78. [PMID: 25116588 PMCID: PMC4280678 DOI: 10.1002/path.4423] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 08/05/2014] [Accepted: 08/05/2014] [Indexed: 12/19/2022]
Abstract
Reactive oxygen species (ROS) participate in the pathogenesis of emphysema. Among ROS-producing enzymes, NOX NADPH oxidases are thought to be responsible for tissue injury associated with several lung pathologies. To determine whether NOX2 and/or NOX1 participate in the development of emphysema, their expression patterns were first studied by immunohistochemistry in the lungs of emphysematous patients. Subsequently, we investigated their contribution to elastase-induced emphysema using NOX2- and NOX1-deficient mice. In human lung, NOX2 was mainly detected in macrophages of control and emphysematous lungs, while NOX1 was expressed in alveolar epithelium and bronchial cells. We observed an elevated number of NOX2-positive cells in human emphysematous lungs, as well as increased NOX2 and NOX1 mRNA expression in mouse lungs following elastase exposure. Elastase-induced alveolar airspace enlargement and elastin degradation were prevented in NOX2-deficient mice, but not in NOX1-deficient mice. This protection was independent of inflammation and correlated with reduced ROS production. Concomitantly, an elevation of sirtuin 1 (SIRT1) level and a decrease of matrix metalloproteinase-9 (MMP-9) expression and activity were observed in alveolar macrophages and neutrophils. We addressed the specific role of macrophage-restricted functional NOX2 in elastase-induced lung emphysema using Ncf1 mutant mice and Ncf1 macrophage rescue mice (Ncf1 mutant mice with transgenic expression of Ncf1 only in CD68-positive mononuclear phagocytes; the MN mouse). Compared to WT mice, the lack of functional NOX2 led to decreased elastase-induced ROS production and protected against emphysema. In contrast, ROS production was restored specifically in macrophages from Ncf1 rescue mice and contributes to emphysema. Taken together, our results demonstrate that NOX2 is involved in the pathogenesis of human emphysema and macrophage-specific NOX2 participates in elastase-induced emphysema through the involvement of SIRT1/MMP-9 pathways in mice.
Collapse
Affiliation(s)
- Candice Trocme
- Laboratory of Protein and Enzyme Biochemistry, University Hospital, Grenoble, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Zanetti F, Giacomello M, Donati Y, Carnesecchi S, Frieden M, Barazzone-Argiroffo C. Nicotine mediates oxidative stress and apoptosis through cross talk between NOX1 and Bcl-2 in lung epithelial cells. Free Radic Biol Med 2014; 76:173-84. [PMID: 25151121 DOI: 10.1016/j.freeradbiomed.2014.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/18/2014] [Accepted: 08/01/2014] [Indexed: 02/07/2023]
Abstract
Nicotine contributes to the onset and progression of several pulmonary diseases. Among the various pathophysiological mechanisms triggered by nicotine, oxidative stress and cell death are reported in several cell types. We found that chronic exposure to nicotine (48h) induced NOX1-dependent oxidative stress and apoptosis in primary pulmonary cells. In murine (MLE-12) and human (BEAS-2B) lung epithelial cell lines, nicotine acted as a sensitizer to cell death and synergistically enhanced apoptosis when cells were concomitantly exposed to hyperoxia. The precise signaling pathway was investigated in MLE-12 cells in which NOX1 was abrogated by a specific inhibitor or stably silenced by shRNA. In the early phase of exposure (1h), nicotine mediated intracellular Ca(2+) fluxes and activation of protein kinase C, which in its turn activated NOX1, leading to cellular and mitochondrial oxidative stress. The latter triggered the intrinsic apoptotic machinery by modulating the expression of Bcl-2 and Bax. Overexpression of Bcl-2 completely prevented nicotine's detrimental effects, suggesting Bcl-2as a downstream key regulator in nicotine/NOX1-induced cell damage. These results suggest that NOX1 is a major contributor to the generation of intracellular oxidative stress induced by nicotine and might be an important molecule to target in nicotine-related lung pathologies.
Collapse
Affiliation(s)
- Filippo Zanetti
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland
| | - Marta Giacomello
- Venetian Institute of Molecular Medicine, University of Padova, Padova, Italy
| | - Yves Donati
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland
| | - Stephanie Carnesecchi
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Constance Barazzone-Argiroffo
- Department of Pediatrics, Children's Hospital, 1211 Geneva, Switzerland; Department of Pathology and Immunology, Medical School, and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
106
|
Gomolak JR, Didion SP. Angiotensin II-induced endothelial dysfunction is temporally linked with increases in interleukin-6 and vascular macrophage accumulation. Front Physiol 2014; 5:396. [PMID: 25400581 PMCID: PMC4212611 DOI: 10.3389/fphys.2014.00396] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 09/24/2014] [Indexed: 01/18/2023] Open
Abstract
Angiotensin II (Ang II) is associated with vascular hypertrophy, endothelial dysfunction and activation of a number of inflammatory molecules, however the linear events involved in the development of hypertension and endothelial dysfunction produced in response to Ang II are not well defined. The goal of this study was to examine the dose- and temporal-dependent development of endothelial dysfunction in response to Ang II. Blood pressure and responses of carotid arteries were examined in control (C57Bl/6) mice and in mice infused with 50, 100, 200, 400, or 1000 ng/kg/min Ang II for either 14 or 28 Days. Infusion of Ang II was associated with graded and marked increases in systolic blood pressure and plasma Ang II concentrations. While low doses of Ang II (i.e., 50 and 100 ng/kg/min) had little to no effect on blood pressure or endothelial function, high doses of Ang II (e.g., 1000 ng/kg/min) were associated with large increases in arterial pressure and marked impairment of endothelial function. In contrast, intermediate doses of Ang II (200 and 400 ng/kg/min) while initially having no effect on systolic blood pressure were associated with significant increases in pressure over time. Despite increasing blood pressure, 200 ng/kg/min had no effect on endothelial function, whereas 400 ng/kg/min produced modest impairment on Day 14 and marked impairment of endothelial function on Day 28. The degree of endothelial dysfunction produced by 400 and 1000 ng/kg/min Ang II was reflective of parallel increases in plasma IL-6 levels and vascular macrophage content, suggesting that increases in arterial blood pressure precede the development of endothelial dysfunction. These findings are important as they demonstrate that along with increases in arterial pressure that increases in IL-6 and vascular macrophage accumulation correlate with the impairment of endothelial function produced by Ang II.
Collapse
Affiliation(s)
- Jessica R Gomolak
- Department of Pharmacology, The University of Mississippi Medical Center Jackson, MS, USA
| | - Sean P Didion
- Department of Pharmacology, The University of Mississippi Medical Center Jackson, MS, USA ; Department of Neurology, The University of Mississippi Medical Center Jackson, MS, USA
| |
Collapse
|
107
|
Wang Y, Chen F, Le B, Stepp DW, Fulton DJR. Impact of Nox5 polymorphisms on basal and stimulus-dependent ROS generation. PLoS One 2014; 9:e100102. [PMID: 24992705 PMCID: PMC4081039 DOI: 10.1371/journal.pone.0100102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 05/21/2014] [Indexed: 02/07/2023] Open
Abstract
Nox5 is an EF-hand containing, calcium-dependent isoform of the NADPH oxidase family of reactive oxygen species (ROS) generating enzymes. Altered expression and activity of Nox5 has been reported in cardiovascular diseases and cancers but the absence of Nox5 in rodents has precluded a greater understanding of its physiological and pathophysiological roles. Multiple polymorphisms have been identified within the coding sequence of human Nox5, but whether this translates into altered enzyme function is unknown. Herein, we have generated 15 novel mutants of Nox5β to evaluate the effect of exonic SNPs on basal and stimulated enzyme activity. Compared to the WT enzyme, ROS production was unchanged or slightly modified in the majority of mutants, but significantly decreased in 7. Focusing on M77K, Nox5 activity was dramatically reduced in unstimulated cells and following challenge with both calcium- and phosphorylation-dependent stimuli despite equivalent levels of expression. The M77K mutation did not influence the Nox5 phosphorylation or the ability to bind Hsp90, but in cell-free assays with excess co-factors and calcium, ROS production was dramatically reduced. A more conservative substitution M77V arising from another SNP yielded a different profile of enzyme activity and suggests a critical role of M77 in calcium-dependent ROS production. Two C-terminal mutants, R530H and G542R, were observed that had little to no activity and relatively high minor allele frequency (MAF). In conclusion, we have identified 7 missense SNPs in Nox5 that result in little or no enzyme activity. Whether humans with dysfunctional Nox5 variants have altered physiology or disease remains to be determined.
Collapse
Affiliation(s)
- Yusi Wang
- Vascular Biology Center, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Feng Chen
- Vascular Biology Center, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Brian Le
- Vascular Biology Center, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - David W. Stepp
- Vascular Biology Center, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- Department of Physiology, Georgia Regents University, Augusta, Georgia, United States of America
| | - David J. R. Fulton
- Vascular Biology Center, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- Department of Pharmacology, Georgia Regents University, Augusta, Georgia, United States of America
| |
Collapse
|
108
|
Chan EC, Dusting GJ, Liu GS, Jiang F. Redox mechanisms of the beneficial effects of heme oxygenase in hypertension. J Hypertens 2014; 32:1379-86; discussion 1387. [DOI: 10.1097/hjh.0000000000000179] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
109
|
González J, Valls N, Brito R, Rodrigo R. Essential hypertension and oxidative stress: New insights. World J Cardiol 2014; 6:353-366. [PMID: 24976907 PMCID: PMC4072825 DOI: 10.4330/wjc.v6.i6.353] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 03/01/2014] [Accepted: 05/08/2014] [Indexed: 02/06/2023] Open
Abstract
Essential hypertension is a highly prevalent pathological condition that is considered as one of the most relevant cardiovascular risk factors and is an important cause of morbidity and mortality around the world. Despite the fact that mechanisms underlying hypertension are not yet fully elucidated, a large amount of evidence shows that oxidative stress plays a central role in its pathophysiology. Oxidative stress can be defined as an imbalance between oxidant agents, such as superoxide anion, and antioxidant molecules, and leads to a decrease in nitric oxide bioavailability, which is the main factor responsible for maintaining the vascular tone. Several vasoconstrictor peptides, such as angiotensin II, endothelin-1 and urotensin II, act through their receptors to stimulate the production of reactive oxygen species, by activating enzymes like NADPH oxidase and xanthine oxidase. The knowledge of the mechanism described above has allowed generating new therapeutic strategies against hypertension based on the use of antioxidants agents, including vitamin C and E, N-Acetylcysteine, polyphenols and selenium, among others. These substances have different therapeutic targets, but all represent antioxidant reinforcement. Several clinical trials using antioxidants have been made. The aim of the present review is to provide new insights about the key role of oxidative stress in the pathophysiology of essential hypertension and new clinical attempts to demonstrate the usefulness of antioxidant therapy in the treatment of hypertension.
Collapse
|
110
|
Nox1 upregulates the function of vascular T-type calcium channels following chronic nitric oxide deficit. Pflugers Arch 2014; 467:727-35. [PMID: 24923576 DOI: 10.1007/s00424-014-1548-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is characterised by reduced nitric oxide bioavailability resulting from oxidative stress. Our previous studies have shown that nitric oxide deficit per se increases the contribution of T-type calcium channels to vascular tone through increased superoxide from NADPH oxidase (Nox). The aim of the present study was therefore to identify the Nox isoform responsible for modulating T-type channel function, as T-type channels are implicated in several pathophysiological conditions involving oxidative stress. We evaluated T-channel function in skeletal muscle arterioles in vivo, using a novel T-channel blocker, TTA-A2 (3 μmol/L), which demonstrated no cross reactivity with L-type channels. Wild-type and Nox2 knockout (Nox2ko) mice were treated with the nitric oxide synthase inhibitor L-NAME (40 mg/kg/day) for 2 weeks. L-NAME treatment significantly increased systolic blood pressure and the contribution of T-type calcium channels to arteriolar tone in wild-type mice, and this was not prevented by Nox2 deletion. In Nox2ko mice, pharmacological inhibition of Nox1 (10 μmol/L ML171), Nox4 (10 μmol/L VAS2870) and Nox4-derived hydrogen peroxide (500 U/mL catalase) significantly reduced the effect of chronic nitric oxide inhibition on T-type channel function. In contrast, in wild-type mice, ML171 and VAS2870, but not catalase, reduced the contribution of T-type channels to vascular tone, suggesting a role for Nox1 and non-selective actions of VAS2870. We conclude that Nox1, but not Nox2 or Nox4, is responsible for the upregulation of T-type calcium channels elicited by chronic nitric oxide deficit. These data point to an important role for this isoform in increasing T-type channel function during oxidative stress.
Collapse
|
111
|
Wilkinson-Berka JL, Deliyanti D, Rana I, Miller AG, Agrotis A, Armani R, Szyndralewiez C, Wingler K, Touyz RM, Cooper ME, Jandeleit-Dahm KA, Schmidt HHHW. NADPH oxidase, NOX1, mediates vascular injury in ischemic retinopathy. Antioxid Redox Signal 2014; 20:2726-40. [PMID: 24053718 PMCID: PMC4026404 DOI: 10.1089/ars.2013.5357] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIMS Ischemic retinal diseases such as retinopathy of prematurity are major causes of blindness due to damage to the retinal microvasculature. Despite this clinical situation, retinopathy of prematurity is mechanistically poorly understood. Therefore, effective preventative therapies are not available. However, hypoxic-induced increases in reactive oxygen species (ROS) have been suggested to be involved with NADPH oxidases (NOX), the only known dedicated enzymatic source of ROS. Our major aim was to determine the contribution of NOX isoforms (1, 2, and 4) to a rodent model of retinopathy of prematurity. RESULTS Using a genetic approach, we determined that only mice with a deletion of NOX1, but not NOX2 or NOX4, were protected from retinal neovascularization and vaso-obliteration, adhesion of leukocytes, microglial accumulation, and the increased generation of proangiogenic and proinflammatory factors and ROS. We complemented these studies by showing that the specific NOX inhibitor, GKT137831, reduced vasculopathy and ROS levels in retina. The source of NOX isoforms was evaluated in retinal vascular cells and neuro-glial elements. Microglia, the immune cells of the retina, expressed NOX1, 2, and 4 and responded to hypoxia with increased ROS formation, which was reduced by GKT137831. INNOVATION Our studies are the first to identify the NOX1 isoform as having an important role in the pathogenesis of retinopathy of prematurity. CONCLUSIONS Our findings suggest that strategies targeting NOX1 have the potential to be effective treatments for a range of ischemic retinopathies.
Collapse
|
112
|
Abstract
SIGNIFICANCE Reactive oxygen species (ROS) play a critical role in vascular disease. While there are many possible sources of ROS, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases play a central role. They are a source of "kindling radicals," which affect other enzymes, such as nitric oxide synthase endothelial nitric oxide synthase or xanthine oxidase. This is important, as risk factors for atherosclerosis (hypertension, diabetes, hypercholesterolemia, and smoking) regulate the expression and activity of NADPH oxidases in the vessel wall. RECENT ADVANCES There are seven isoforms in mammals: Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2. Nox1, Nox2, Nox4, and Nox5 are expressed in endothelium, vascular smooth muscle cells, fibroblasts, or perivascular adipocytes. Other homologues have not been found or are expressed at very low levels; their roles have not been established. Nox1/Nox2 promote the development of endothelial dysfunction, hypertension, and inflammation. Nox4 may have a role in protecting the vasculature during stress; however, when its activity is increased, it may be detrimental. Calcium-dependent Nox5 has been implicated in oxidative damage in human atherosclerosis. CRITICAL ISSUES NADPH oxidase-derived ROS play a role in vascular pathology as well as in the maintenance of normal physiological vascular function. We also discuss recently elucidated mechanisms such as the role of NADPH oxidases in vascular protection, vascular inflammation, pulmonary hypertension, tumor angiogenesis, and central nervous system regulation of vascular function and hypertension. FUTURE DIRECTIONS Understanding the role of individual oxidases and interactions between homologues in vascular disease is critical for efficient pharmacological regulation of vascular NADPH oxidases in both the laboratory and clinical practice.
Collapse
Affiliation(s)
- Anna Konior
- 1 Department of Internal Medicine, Jagiellonian University School of Medicine , Cracow, Poland
| | | | | | | |
Collapse
|
113
|
Gray SP, Jandeleit-Dahm K. The pathobiology of diabetic vascular complications--cardiovascular and kidney disease. J Mol Med (Berl) 2014; 92:441-52. [PMID: 24687627 DOI: 10.1007/s00109-014-1146-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/03/2014] [Accepted: 03/14/2014] [Indexed: 02/06/2023]
Abstract
With the increasing incidence of obesity and type 2 diabetes, it is predicted that more than half of Americans will have diabetes or pre-diabetes by 2020. Diabetic patients develop vascular complications at a much faster rate in comparison to non-diabetic individuals, and cardiovascular risk is increased up to tenfold. With the increasing incidence of diabetes across the world, the development of vascular complications will become an increasing medical burden. Diabetic vascular complications affect the micro- and macro-vasculature leading to kidney disease often requiring dialysis and transplantation or cardiovascular disease increasing the risk for myocardial infarction, stroke and amputations as well as leading to premature mortality. It has been suggested that many complex pathways contribute to the pathobiology of diabetic complications including hyperglycaemia itself, the production of advanced glycation end products (AGEs) and interaction with the receptors for AGEs such as the receptor for advanced glycation end products (RAGE), as well as the activation of vasoactive systems such as the renin-angiotensin aldosterone system (RAAS) and the endothelin system. More recently, it has been hypothesised that reactive oxygen species derived from NAD(P)H oxidases (Nox) may represent a common downstream mediator of vascular injury in diabetes. Current standard treatment of care includes the optimization of blood glucose and blood pressure usually including inhibitors of the renin-angiotensin system. Although these interventions are able to delay progression, they fail to prevent the development of complications. Thus, there is an urgent medical need to identify novel targets in diabetic vascular complications which may include the blockade of Nox-derived ROS formation, as well as blockade of AGE formation and inhibitors of RAGE activation. These strategies may provide superior protection against the deleterious effects of diabetes on the vasculature.
Collapse
Affiliation(s)
- Stephen P Gray
- Diabetes Complications Division, Baker IDI Heart & Diabetes Research Institute, PO Box 6492, St Kilda Rd, Melbourne, VIC, 8008, Australia,
| | | |
Collapse
|
114
|
Rodrigo R, González J. Role of Oxidative Stress in Hypertension. ROLE OF OXIDATIVE STRESS IN CHRONIC DISEASES 2014:199-245. [DOI: 10.1201/b16653-14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
115
|
Smith SME, Min J, Ganesh T, Diebold B, Kawahara T, Zhu Y, McCoy J, Sun A, Snyder JP, Fu H, Du Y, Lewis I, Lambeth JD. Ebselen and congeners inhibit NADPH oxidase 2-dependent superoxide generation by interrupting the binding of regulatory subunits. ACTA ACUST UNITED AC 2014; 19:752-63. [PMID: 22726689 DOI: 10.1016/j.chembiol.2012.04.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 04/18/2012] [Accepted: 04/27/2012] [Indexed: 02/07/2023]
Abstract
NADPH oxidases (Nox) are a primary source of reactive oxygen species (ROS), which function in normal physiology and, when overproduced, in pathophysiology. Recent studies using mice deficient in Nox2 identify this isoform as a novel target against Nox2-implicated inflammatory diseases. Nox2 activation depends on the binding of the proline-rich domain of its heterodimeric partner p22phox to p47phox. A high-throughput screen that monitored this interaction via fluorescence polarization identified ebselen and several of its analogs as inhibitors. Medicinal chemistry was performed to explore structure-activity relationships and to optimize potency. Ebselen and analogs potently inhibited Nox1 and Nox2 activity but were less effective against other isoforms. Ebselen also blocked translocation of p47phox to neutrophil membranes. Thus, ebselen and its analogs represent a class of compounds that inhibit ROS generation by interrupting the assembly of Nox2-activating regulatory subunits.
Collapse
Affiliation(s)
- Susan M E Smith
- Department of Pathology, Emory School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Gray SP, Jha JC, Di Marco E, Jandeleit-Dahm KA. NAD(P)H oxidase isoforms as therapeutic targets for diabetic complications. Expert Rev Endocrinol Metab 2014; 9:111-122. [PMID: 30743754 DOI: 10.1586/17446651.2014.887984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The development of macro- and microvascular complications is accelerated in diabetic patients. While some therapeutic regimes have helped in delaying progression of complications, none have yet been able to halt the progression and prevent vascular disease, highlighting the need to identify new therapeutic targets. Increased oxidative stress derived from the NADPH oxidase (Nox) family has recently been identified to play an important role in the pathophysiology of vascular disease. In recent years, specific Nox isoforms have been implicated in contributing to the development of atherosclerosis of major vessels, as well as damage of the small vessels within the kidney and the eye. With the use of novel Nox inhibitors, it has been demonstrated that these complications can be attenuated, indicating that targeting Nox derived oxidative stress holds potential as a new therapeutic strategy.
Collapse
Affiliation(s)
| | - Jay C Jha
- a Diabetic Complications Division, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Elyse Di Marco
- a Diabetic Complications Division, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Karin Am Jandeleit-Dahm
- a Diabetic Complications Division, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
117
|
Raaz U, Toh R, Maegdefessel L, Adam M, Nakagami F, Emrich FC, Spin JM, Tsao PS. Hemodynamic regulation of reactive oxygen species: implications for vascular diseases. Antioxid Redox Signal 2014; 20:914-28. [PMID: 23879326 PMCID: PMC3924901 DOI: 10.1089/ars.2013.5507] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Arterial blood vessels functionally and structurally adapt to altering hemodynamic forces in order to accommodate changing needs and to provide stress homeostasis. This ability is achieved at the cellular level by converting mechanical stimulation into biochemical signals (i.e., mechanotransduction). Physiological mechanical stress helps maintain vascular structure and function, whereas pathologic or aberrant stress may impair cellular mechano-signaling, and initiate or augment cellular processes that drive disease. RECENT ADVANCES Reactive oxygen species (ROS) may represent an intriguing class of mechanically regulated second messengers. Chronically enhanced ROS generation may be induced by adverse mechanical stresses, and is associated with a multitude of vascular diseases. Although a causal relationship has clearly been demonstrated in large numbers of animal studies, an effective ROS-modulating therapy still remains to be established by clinical studies. CRITICAL ISSUES AND FUTURE DIRECTIONS This review article focuses on the role of various mechanical forces (in the form of laminar shear stress, oscillatory shear stress, or cyclic stretch) as modulators of ROS-driven signaling, and their subsequent effects on vascular biology and homeostasis, as well as on specific diseases such as arteriosclerosis, hypertension, and abdominal aortic aneurysms. Specifically, it highlights the significance of the various NADPH oxidase (NOX) isoforms as critical ROS generators in the vasculature. Directed targeting of defined components in the complex network of ROS (mechano-)signaling may represent a key for successful translation of experimental findings into clinical practice.
Collapse
Affiliation(s)
- Uwe Raaz
- 1 Division of Cardiovascular Medicine, Stanford University School of Medicine , Stanford, California
| | | | | | | | | | | | | | | |
Collapse
|
118
|
Jha JC, Gray SP, Barit D, Okabe J, El-Osta A, Namikoshi T, Thallas-Bonke V, Wingler K, Szyndralewiez C, Heitz F, Touyz RM, Cooper ME, Schmidt HHHW, Jandeleit-Dahm KA. Genetic targeting or pharmacologic inhibition of NADPH oxidase nox4 provides renoprotection in long-term diabetic nephropathy. J Am Soc Nephrol 2014; 25:1237-54. [PMID: 24511132 DOI: 10.1681/asn.2013070810] [Citation(s) in RCA: 291] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Diabetic nephropathy may occur, in part, as a result of intrarenal oxidative stress. NADPH oxidases comprise the only known dedicated reactive oxygen species (ROS)-forming enzyme family. In the rodent kidney, three isoforms of the catalytic subunit of NADPH oxidase are expressed (Nox1, Nox2, and Nox4). Here we show that Nox4 is the main source of renal ROS in a mouse model of diabetic nephropathy induced by streptozotocin administration in ApoE(-/-) mice. Deletion of Nox4, but not of Nox1, resulted in renal protection from glomerular injury as evidenced by attenuated albuminuria, preserved structure, reduced glomerular accumulation of extracellular matrix proteins, attenuated glomerular macrophage infiltration, and reduced renal expression of monocyte chemoattractant protein-1 and NF-κB in streptozotocin-induced diabetic ApoE(-/-) mice. Importantly, administration of the most specific Nox1/4 inhibitor, GKT137831, replicated these renoprotective effects of Nox4 deletion. In human podocytes, silencing of the Nox4 gene resulted in reduced production of ROS and downregulation of proinflammatory and profibrotic markers that are implicated in diabetic nephropathy. Collectively, these results identify Nox4 as a key source of ROS responsible for kidney injury in diabetes and provide proof of principle for an innovative small molecule approach to treat and/or prevent chronic kidney failure.
Collapse
Affiliation(s)
- Jay C Jha
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Stephen P Gray
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - David Barit
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Jun Okabe
- Human Epigenetics Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Assam El-Osta
- Human Epigenetics Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Tamehachi Namikoshi
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Japan
| | - Vicki Thallas-Bonke
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Kirstin Wingler
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Faculty of Medicine, Health & Life Science, Maastricht University, Maastricht, The Netherlands
| | | | | | - Rhian M Touyz
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mark E Cooper
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Harald H H W Schmidt
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Faculty of Medicine, Health & Life Science, Maastricht University, Maastricht, The Netherlands
| | - Karin A Jandeleit-Dahm
- Diabetic Complications Division, Juvenile Diabetes Research Foundation Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, Victoria, Australia;
| |
Collapse
|
119
|
Montezano AC, Touyz RM. Reactive oxygen species, vascular Noxs, and hypertension: focus on translational and clinical research. Antioxid Redox Signal 2014; 20:164-82. [PMID: 23600794 PMCID: PMC3880913 DOI: 10.1089/ars.2013.5302] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 04/21/2013] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Reactive oxygen species (ROS) are signaling molecules that are important in physiological processes, including host defense, aging, and cellular homeostasis. Increased ROS bioavailability and altered redox signaling (oxidative stress) have been implicated in the onset and/or progression of chronic diseases, including hypertension. RECENT ADVANCES Although oxidative stress may not be the only cause of hypertension, it amplifies blood pressure elevation in the presence of other pro-hypertensive factors, such as salt loading, activation of the renin-angiotensin-aldosterone system, and sympathetic hyperactivity, at least in experimental models. A major source for ROS in the cardiovascular-renal system is a family of nicotinamide adenine dinucleotide phosphate oxidases (Noxs), including the prototypic Nox2-based Nox, and Nox family members: Nox1, Nox4, and Nox5. CRITICAL ISSUES Although extensive experimental data support a role for increased ROS levels and altered redox signaling in the pathogenesis of hypertension, the role in clinical hypertension is unclear, as a direct causative role of ROS in blood pressure elevation has yet to be demonstrated in humans. Nevertheless, what is becoming increasingly evident is that abnormal ROS regulation and aberrant signaling through redox-sensitive pathways are important in the pathophysiological processes which is associated with vascular injury and target-organ damage in hypertension. FUTURE DIRECTIONS There is a paucity of clinical information related to the mechanisms of oxidative stress and blood pressure elevation, and a few assays accurately measure ROS directly in patients. Such further ROS research is needed in humans and in the development of adequately validated analytical methods to accurately assess oxidative stress in the clinic.
Collapse
Affiliation(s)
- Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow , Glasgow, United Kingdom
| | | |
Collapse
|
120
|
Mercer KE, Sims CR, Yang CS, Wynne RA, Moutos C, Hogue WR, Lumpkin CK, Suva LJ, Chen JR, Badger TM, Ronis MJJ. Loss of functional NADPH oxidase 2 protects against alcohol-induced bone resorption in female p47phox-/- mice. Alcohol Clin Exp Res 2013; 38:672-82. [PMID: 24256560 DOI: 10.1111/acer.12305] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 09/13/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND In bone, NADPH oxidase (NOX)-derived reactive oxygen species (ROS) superoxide and/or hydrogen peroxide are an important stimulus for osteoclast differentiation and activity. Previously, we have demonstrated that chronic ethanol (EtOH) consumption generates excess NOX-dependent ROS in osteoblasts, which functions to stimulate nuclear factor kappa-β receptor ligand (RANKL)-RANK signaling, thus increasing osteoclastogenesis and activity. This activity can be blocked by co-administration of EtOH with the pan-NOX inhibitor diphenylene idonium (DPI). METHODS To test whether EtOH-induced bone loss is dependent on a functional NOX2 enzyme, 6-week-old female C57BL/6J-Ncf1/p47phox(-/-) (p47phox KO) and wild-type (WT) mice were pair-fed EtOH diets for 40 days. Bone loss was assessed by 3-point bending, micro-computed tomography and static histomorphometric analysis. Additionally, ST2 cultured cells were co-treated with EtOH and NOX inhibitors, DPI, gliotoxin, and plumbagin, after which changes in ROS production, and in RANKL and NOX mRNA expression were analyzed. RESULTS In WT mice, EtOH treatment significantly reduced bone density and mechanical strength, and increased total osteoclast number and activity. In EtOH-treated p47phox KO mice, bone density and mechanical strength were completely preserved. EtOH p47phox KO mice had no changes in osteoclast numbers or activity, and no elevations in serum CTX or RANKL gene expression (p < 0.05). In both WT and p47phox KO mice, EtOH feeding reduced biochemical markers of bone formation (p < 0.05). In vitro EtOH exposure of ST2 cells increased ROS, which was blocked by pretreating with DPI or the NOX2 inhibitor gliotoxin. EtOH-induced RANKL and NOX2 gene expression were inhibited by the NOX4-specific inhibitor plumbagin. CONCLUSIONS These data suggest that NOX2-derived ROS is necessary for EtOH-induced bone resorption. In osteoblasts, NOX2 and NOX4 appear to work in tandem to increase RANKL expression, whereas EtOH-mediated inhibition of bone formation occurs via a NOX2-independent mechanism.
Collapse
Affiliation(s)
- Kelly E Mercer
- Department of Pediatrics, Center for Orthopaedic Research at the University of Arkansas for Medical Sciences and Arkansas Children's Nutrition Center, Little Rock, Arkansas
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Chan SHH, Chan JYH. Angiotensin-generated reactive oxygen species in brain and pathogenesis of cardiovascular diseases. Antioxid Redox Signal 2013; 19:1074-84. [PMID: 22429119 DOI: 10.1089/ars.2012.4585] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE Overproduction of angiotensin II (Ang II) in brain contributes to the pathogenesis of cardiovascular diseases. One of the most promising theses that emerged during the last decade is that production of reactive oxygen species (ROS) and activation of redox-dependent signaling cascades underlie those Ang II actions. This review summarizes our status of understanding on the roles of ROS and redox-sensitive signaling in brain Ang II-dependent cardiovascular diseases, using hypertension and heart failure as illustrative examples. RECENT ADVANCES ROS generated by NADPH oxidase, mitochondrial electron transport chain, and proinflammatory cytokines activates mitogen-activated protein kinases and transcription factors, which in turn modulate ion channel functions and ultimately increase neuronal activity and sympathetic outflow in brain Ang II-dependent cardiovascular diseases. Antioxidants targeting ROS have been demonstrated to be beneficial to Ang II-induced hypertension and heart failure via protection from oxidative stress in brain regions that subserve cardiovascular regulation. CRITICAL ISSUES Intra-neuronal signaling and the downstream redox-sensitive proteins involved in controlling the neuronal discharge rate, the sympathetic outflow, and the pathogenesis of cardiovascular diseases need to be identified. The cross talk between Ang II-induced oxidative stress and neuroinflammation in neural mechanisms of cardiovascular diseases also warrants further elucidation. FUTURE DIRECTIONS Future studies are needed to identify new redox-based therapeutics that work not only in animal models, but also in patients suffering from the prevalent diseases. Upregulation of endogenous antioxidants in the regulation of ROS homeostasis is a potential therapeutic target, as are small molecule- or nanoformulated conjugate-based antioxidant therapy.
Collapse
Affiliation(s)
- Samuel H H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital , Kaohsiung, Taiwan, Republic of China
| | | |
Collapse
|
122
|
Braunersreuther V, Montecucco F, Asrih M, Ashri M, Pelli G, Galan K, Frias M, Burger F, Quinderé ALG, Montessuit C, Krause KH, Mach F, Jaquet V. Role of NADPH oxidase isoforms NOX1, NOX2 and NOX4 in myocardial ischemia/reperfusion injury. J Mol Cell Cardiol 2013; 64:99-107. [PMID: 24051369 DOI: 10.1016/j.yjmcc.2013.09.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 08/22/2013] [Accepted: 09/09/2013] [Indexed: 02/07/2023]
Abstract
Myocardial reperfusion injury is mediated by several processes including increase of reactive oxygen species (ROS). The aim of the study is to identify potential sources of ROS contributing to myocardial ischemia-reperfusion injury. For this purpose, we investigated myocardial ischemia/reperfusion pathology in mice deficient in various NADPH oxidase isoforms (Nox1, Nox2, Nox4, as well as Nox1/2 double knockout). Following 30min of ischemia and 24h of reperfusion, a significant decrease in the size of myocardial infarct was observed in Nox1-, Nox2- and Nox1/Nox2-, but not in Nox4-deficient mice. However, no protection was observed in a model of chronic ischemia, suggesting that NOX1 and NOX2-mediated oxidative damage occurs during reperfusion. Cardioprotective effect of Nox1 and Nox2 deficiencies was associated with decrease of neutrophil invasion, but, on the other hand an improved reperfusion injury was also observed in isolated perfused hearts (Langendorff model) suggesting that inflammatory cells were not the major source of oxidative damage. A decrease in global post-reperfusion oxidative stress was clearly detected in Nox2-, but not in Nox1-deficient hearts. Analysis of key signaling pathways during reperfusion suggests distinct cardioprotective patterns: increased phosphorylation was seen for Akt and Erk in Nox1-deficient mice and for Stat3 and Erk in Nox2-deficient mice. Consequently, NOX1 and NOX2 represent interesting drug targets for controlling reperfusion damage associated with revascularization in coronary disease.
Collapse
Affiliation(s)
- Vincent Braunersreuther
- Division of Cardiology, Department of Medicine, University Hospital, Foundation for Medical Researches, 64 Avenue Roseraie, 1211 Geneva, Switzerland; Division of Clinical Pathology, Department of Genetic Medicine and Laboratories, University Hospital, 1 rue Michel-Servet, 1211 Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Wang H, Albadawi H, Siddiquee Z, Stone JM, Panchenko MP, Watkins MT, Stone JR. Altered vascular activation due to deficiency of the NADPH oxidase component p22phox. Cardiovasc Pathol 2013; 23:35-42. [PMID: 24035466 DOI: 10.1016/j.carpath.2013.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/04/2013] [Accepted: 08/05/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reactive oxygen species generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase play important roles in vascular activation. The p22(phox) subunit is necessary for the activity of NADPH oxidase complexes utilizing Nox1, Nox2, Nox3, and Nox4 catalytic subunits. METHODS We assessed p22(phox)-deficient mice and human tissue for altered vascular activation. RESULTS Mice deficient in p22(phox) were smaller than their wild-type littermates but showed no alteration in basal blood pressure. The wild-type littermates were relatively resistant to forming intimal hyperplasia following carotid ligation, and the intimal hyperplasia that developed was not altered by p22(phox) deficiency. However, at the site of carotid artery ligation, the p22(phox)-deficient mice showed significantly less vascular elastic fiber loss compared with their wild-type littermates. This preservation of elastic fibers was associated with a reduced matrix metallopeptidase (MMP) 12/tissue inhibitor of metalloproteinase (TIMP) 1 expression ratio. A similar decrease in the relative MMP12/TIMP1 expression ratio occurred in human coronary artery smooth muscle cells upon knockdown of the hydrogen peroxide responsive kinase CK1αLS. In the ligated carotid arteries, the p22(phox)-deficient mice showed reduced expression of heterogeneous nuclear ribonucleoprotein C (hnRNP-C), suggesting reduced activity of CK1αLS. In a lung biopsy from a human patient with p22(phox) deficiency, there was also reduced vascular hnRNP-C expression. CONCLUSIONS These findings indicate that NADPH oxidase complexes modulate aspects of vascular activation including vascular elastic fiber loss, the MMP12/TIMP1 expression ratio, and the expression of hnRNP-C. Furthermore, these findings suggest that the effects of NADPH oxidase on vascular activation are mediated in part by protein kinase CK1αLS.
Collapse
MESH Headings
- Animals
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/pathology
- Carotid Artery, Common/enzymology
- Carotid Artery, Common/pathology
- Case-Control Studies
- Casein Kinase Ialpha/genetics
- Casein Kinase Ialpha/metabolism
- Cells, Cultured
- Coronary Vessels/enzymology
- Coronary Vessels/pathology
- Cytochrome b Group/deficiency
- Cytochrome b Group/genetics
- Elastic Tissue/enzymology
- Elastic Tissue/pathology
- Female
- Granulomatous Disease, Chronic/enzymology
- Granulomatous Disease, Chronic/genetics
- Granulomatous Disease, Chronic/pathology
- Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism
- Humans
- Hyperplasia
- Infant
- Male
- Matrix Metalloproteinase 12/metabolism
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- NADPH Oxidases/deficiency
- NADPH Oxidases/genetics
- Neointima
- RNA Interference
- Reactive Oxygen Species/metabolism
- Tissue Inhibitor of Metalloproteinase-1/metabolism
- Transfection
Collapse
Affiliation(s)
- He Wang
- Center for Systems Biology, Massachusetts General Hospital and Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
124
|
Shen H, Eguchi K, Kono N, Fujiu K, Matsumoto S, Shibata M, Oishi-Tanaka Y, Komuro I, Arai H, Nagai R, Manabe I. Saturated fatty acid palmitate aggravates neointima formation by promoting smooth muscle phenotypic modulation. Arterioscler Thromb Vasc Biol 2013; 33:2596-607. [PMID: 23968977 DOI: 10.1161/atvbaha.113.302099] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Obesity is a major risk factor of atherosclerotic cardiovascular disease. Circulating free fatty acid levels are known to be elevated in obese individuals and, along with dietary saturated fatty acids, are known to associate with cardiovascular events. However, little is known about the molecular mechanisms by which free fatty acids are linked to cardiovascular disease. APPROACH AND RESULTS We found that administration of palmitate, a major saturated free fatty acid, to mice markedly aggravated neointima formation induced by carotid artery ligation and that the neointima primarily consisted of phenotypically modulated smooth muscle cells (SMCs). In cultured SMCs, palmitate-induced phenotypic modulation was characterized by downregulation of SMC differentiation markers, such as SM α-actin and SM-myosin heavy chain, and upregulation of mediators involved in inflammation and remodeling of the vessel wall, such as platelet-derived growth factor B and matrix metalloproteinases. We also found that palmitate induced the expression of proinflammatory genes via a novel toll-like receptor 4/myeloid differentiation primary response 88/nuclear factor-κB/NADPH oxidase 1/reactive oxygen species signaling pathway: nuclear factor-κB was activated by palmitate via toll-like receptor 4 and its adapter, MyD88, and once active, it transactivated Nox1, encoding NADPH oxidase 1, a major reactive oxygen species generator in SMCs. Pharmacological inhibition and small interfering RNA-mediated knockdown of the components of this signaling pathway mitigated the palmitate-induced upregulation of proinflammatory genes. More importantly, Myd88 knockout mice were resistant to palmitate-induced exacerbation of neointima formation. CONCLUSIONS Palmitate seems to promote neointima formation by inducing inflammatory phenotypes in SMCs.
Collapse
Affiliation(s)
- Hua Shen
- From the Department of Cardiovascular Medicine (H.S., K.E., K.F., S.M., M.S., I.K., I.M.), Translational Systems Biology and Medicine Initiative (K.F.), Graduate School of Medicine, and Graduate School of Pharmaceutical Sciences (N.K., H.A.), The University of Tokyo, Tokyo, Japan; Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan (Y.O.-T.); and Jichi Medical University, Tochigi, Japan (R.N.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Hu X, Yang R, Zhang X, Chen L, Xiang X, Gong C, Wu X. Molecular cloning and functional characterization of the dual oxidase (BmDuox) gene from the silkworm Bombyx mori. PLoS One 2013; 8:e70118. [PMID: 23936382 PMCID: PMC3732266 DOI: 10.1371/journal.pone.0070118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 06/17/2013] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) from nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and their related dual oxidases are known to have significant roles in innate immunity and cell proliferation. In this study, the 5,545 bp cDNA of the silkworm Bombyx mori dual oxidase (BmDuox) gene containing a full-length open reading frame was cloned. It was shown to include an N-terminal signal peptide consisting of 28 amino acid residues, a 240 bp 5′-terminal untranslated region (5′-UTR), an 802 bp 3′-terminal region (3′-UTR), which contains nine ATTTA motifs, and a 4,503 bp open reading frame encoding a polypeptide of 1,500 amino acid residues. Structural analysis indicated that BmDuox contains a typical peroxidase domain at the N-terminus followed by a calcium-binding domain, a ferric-reducing domain, six transmembrane regions and binding domains for flavin adenine dinucleotide (FAD) and nicotinamide adenine dinucleotide (NAD). Transcriptional analysis revealed that BmDuox mRNA was expressed more highly in the head, testis and trachea compared to the midgut, hemocyte, Malpighian tube, ovary, fat bodies and silk glands. BmDuox mRNA was expressed during all the developmental stages of the silkworm. Subcellular localization revealed that BmDoux was present mainly in the periphery of the cells. Some cytoplasmic staining was detected, with rare signals in the nucleus. Expression of BmDuox was induced significantly in the larval midgut upon challenge by Escherichia coli and Bombyx mori nucleopolyhedrovirus (BmNPV). BmDuox-deleted larvae showed a marked increase in microbial proliferation in the midgut after ingestion of fluorescence-labeled bacteria compared to the control. We conclude that reducing BmDuox expression greatly increased the bacterial load, suggesting BmDuox has an important role in inhibiting microbial proliferation and the maintenance of homeostasis in the silkworm midgut.
Collapse
Affiliation(s)
- Xiaolong Hu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
126
|
Madamanchi NR, Runge MS. Redox signaling in cardiovascular health and disease. Free Radic Biol Med 2013; 61:473-501. [PMID: 23583330 PMCID: PMC3883979 DOI: 10.1016/j.freeradbiomed.2013.04.001] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 03/05/2013] [Accepted: 04/02/2013] [Indexed: 02/07/2023]
Abstract
Spatiotemporal regulation of the activity of a vast array of intracellular proteins and signaling pathways by reactive oxygen species (ROS) governs normal cardiovascular function. However, data from experimental and animal studies strongly support that dysregulated redox signaling, resulting from hyperactivation of various cellular oxidases or mitochondrial dysfunction, is integral to the pathogenesis and progression of cardiovascular disease (CVD). In this review, we address how redox signaling modulates the protein function, the various sources of increased oxidative stress in CVD, and the labyrinth of redox-sensitive molecular mechanisms involved in the development of atherosclerosis, hypertension, cardiac hypertrophy and heart failure, and ischemia-reperfusion injury. Advances in redox biology and pharmacology for inhibiting ROS production in specific cell types and subcellular organelles combined with the development of nanotechnology-based new in vivo imaging systems and targeted drug delivery mechanisms may enable fine-tuning of redox signaling for the treatment and prevention of CVD.
Collapse
Affiliation(s)
- Nageswara R Madamanchi
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Marschall S Runge
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
127
|
Rodiño-Janeiro BK, Paradela-Dobarro B, Castiñeiras-Landeira MI, Raposeiras-Roubín S, González-Juanatey JR, Álvarez E. Current status of NADPH oxidase research in cardiovascular pharmacology. Vasc Health Risk Manag 2013; 9:401-28. [PMID: 23983473 PMCID: PMC3750863 DOI: 10.2147/vhrm.s33053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The implications of reactive oxygen species in cardiovascular disease have been known for some decades. Rationally, therapeutic antioxidant strategies combating oxidative stress have been developed, but the results of clinical trials have not been as good as expected. Therefore, to move forward in the design of new therapeutic strategies for cardiovascular disease based on prevention of production of reactive oxygen species, steps must be taken on two fronts, ie, comprehension of reduction-oxidation signaling pathways and the pathophysiologic roles of reactive oxygen species, and development of new, less toxic, and more selective nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors, to clarify both the role of each NADPH oxidase isoform and their utility in clinical practice. In this review, we analyze the value of NADPH oxidase as a therapeutic target for cardiovascular disease and the old and new pharmacologic agents or strategies to prevent NADPH oxidase activity. Some inhibitors and different direct or indirect approaches are available. Regarding direct NADPH oxidase inhibition, the specificity of NADPH oxidase is the focus of current investigations, whereas the chemical structure-activity relationship studies of known inhibitors have provided pharmacophore models with which to search for new molecules. From a general point of view, small-molecule inhibitors are preferred because of their hydrosolubility and oral bioavailability. However, other possibilities are not closed, with peptide inhibitors or monoclonal antibodies against NADPH oxidase isoforms continuing to be under investigation as well as the ongoing search for naturally occurring compounds. Likewise, some different approaches include inhibition of assembly of the NADPH oxidase complex, subcellular translocation, post-transductional modifications, calcium entry/release, electron transfer, and genetic expression. High-throughput screens for any of these activities could provide new inhibitors. All this knowledge and the research presently underway will likely result in development of new drugs for inhibition of NADPH oxidase and application of therapeutic approaches based on their action, for the treatment of cardiovascular disease in the next few years.
Collapse
Affiliation(s)
- Bruno K Rodiño-Janeiro
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- European Molecular Biology Laboratory, Grenoble, France
| | | | | | - Sergio Raposeiras-Roubín
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Cardiology Department, University Clinic Hospital of Santiago de Compostela,
Santiago de Compostela, Spain
| | - José R González-Juanatey
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Cardiology Department, University Clinic Hospital of Santiago de Compostela,
Santiago de Compostela, Spain
- Medicine Department, University of Santiago de Compostela, Santiago de Compostela,
Spain
| | - Ezequiel Álvarez
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Medicine Department, University of Santiago de Compostela, Santiago de Compostela,
Spain
| |
Collapse
|
128
|
Lelli A, Gervais A, Colin C, Chéret C, de Almodovar CR, Carmeliet P, Krause KH, Boillée S, Mallat M. The NADPH oxidase Nox2 regulates VEGFR1/CSF-1R-mediated microglial chemotaxis and promotes early postnatal infiltration of phagocytes in the subventricular zone of the mouse cerebral cortex. Glia 2013; 61:1542-55. [DOI: 10.1002/glia.22540] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 05/08/2013] [Accepted: 05/10/2013] [Indexed: 11/09/2022]
Affiliation(s)
| | | | | | | | - Carmen Ruiz de Almodovar
- Laboratory of Angiogenesis and the Neurovascular Link, The Vesalius Research Center; VIB and K.U.Leuven, Campus Gasthuiberg; B3000-Leuven; Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and the Neurovascular Link, The Vesalius Research Center; VIB and K.U.Leuven, Campus Gasthuiberg; B3000-Leuven; Belgium
| | - Karl-Heinz Krause
- Department of Pathology and Immunology; University of Geneva, Centre Medical Universitaire; CH-1211 Geneva 4; Switzerland
| | | | | |
Collapse
|
129
|
Spencer NY, Zhou W, Li Q, Zhang Y, Luo M, Yan Z, Lynch TJ, Abbott D, Banfi B, Engelhardt JF. Hepatocytes produce TNF-α following hypoxia-reoxygenation and liver ischemia-reperfusion in a NADPH oxidase- and c-Src-dependent manner. Am J Physiol Gastrointest Liver Physiol 2013; 305:G84-94. [PMID: 23639811 PMCID: PMC3725690 DOI: 10.1152/ajpgi.00430.2012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell line studies have previously demonstrated that hypoxia-reoxygenation (H/R) leads to the production of NADPH oxidase 1 and 2 (NOX1 and NOX2)-dependent reactive oxygen species (ROS) required for the activation of c-Src and NF-κB. We now extend these studies into mouse models to evaluate the contribution of hepatocytes to the NOX- and c-Src-dependent TNF-α production that follows H/R in primary hepatocytes and liver ischemia-reperfusion (I/R). In vitro, c-Src-deficient primary hepatocytes produced less ROS and TNF-α following H/R compared with controls. In vivo, c-Src-KO mice also had impaired TNF-α and NF-κB responses following partial lobar liver I/R. Studies in NOX1 and p47phox knockout primary hepatocytes demonstrated that both NOX1 and p47phox are partially required for H/R-mediated TNF-α production. To further investigate the involvement of NADPH oxidases in the production of TNF-α following liver I/R, we performed additional in vivo experiments in knockout mice deficient for NOX1, NOX2, p47phox, Rac1, and/or Rac2. Cumulatively, these results demonstrate that NOX2 and its activator subunits (p47phox and Rac) control the secretion of TNF-α by the liver following I/R. Interestingly, in the absence of Kupffer cells and NOX2, NOX1 played a dominant role in TNF-α production following hepatic I/R. However, NOX1 deletion alone had little effect on I/R-induced TNF-α. Thus Kupffer cell-derived factors and NOX2 act to suppress hepatic NOX1-dependent TNF-α production. We conclude that c-Src and NADPH oxidase components are necessary for redox-mediated production of TNF-α following liver I/R and that hepatocytes play an important role in this process.
Collapse
Affiliation(s)
- Netanya Y. Spencer
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Weihong Zhou
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Qiang Li
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Yulong Zhang
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Meihui Luo
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Ziying Yan
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Thomas J. Lynch
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Duane Abbott
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Botond Banfi
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| |
Collapse
|
130
|
Chan SL, Baumbach GL. Deficiency of Nox2 prevents angiotensin II-induced inward remodeling in cerebral arterioles. Front Physiol 2013; 4:133. [PMID: 23805104 PMCID: PMC3693079 DOI: 10.3389/fphys.2013.00133] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 05/20/2013] [Indexed: 01/26/2023] Open
Abstract
Angiotensin II is an important determinant of inward remodeling in cerebral arterioles. Many of the vascular effects of angiotensin II are mediated by reactive oxygen species (ROS) generated from homologs of NADPH oxidase with Nox2 predominating in small arteries and arterioles. Therefore, we tested the hypothesis that superoxide generated by Nox2 plays a role in angiotensin II-induced cerebral arteriolar remodeling. We examined Nox2-deficient and wild-type (WT) mice in which a pressor or a non-pressor dose of angiotensin II (1000 or 200 ng/kg/day) or saline was infused for 4 weeks via osmotic minipumps. Systolic arterial pressure was measured by a tail-cuff method. Pressure and diameter of cerebral arterioles were measured through an open cranial window in anesthetized mice. Cross-sectional area (by histology) and superoxide level (by hydroethidine staining) of cerebral arterioles were determined ex vivo. The pressor, but not the non-pressor, dose of angiotensin II significantly increased systolic arterial pressure in both WT and Nox2-deficient mice. Both doses of angiotensin II increased superoxide levels and significantly reduced external diameter in maximally dilated cerebral arterioles in WT mice. Increased superoxide and inward remodeling were prevented in Nox2-deficient mice. Moreover, only the pressor dose of AngII increased cross-sectional area of arteriolar wall in WT mice and was prevented in Nox2-deficient mice. In conclusion, superoxide derived from Nox2-containing NADPH oxidase plays an important role in angiotensin II-mediated inward remodeling in cerebral arterioles. This effect appears to be independent of pressure and different from that of hypertrophy.
Collapse
Affiliation(s)
- Siu-Lung Chan
- Department of Pathology, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | | |
Collapse
|
131
|
Norton CE, Broughton BRS, Jernigan NL, Walker BR, Resta TC. Enhanced depolarization-induced pulmonary vasoconstriction following chronic hypoxia requires EGFR-dependent activation of NAD(P)H oxidase 2. Antioxid Redox Signal 2013; 18:1777-88. [PMID: 22966991 PMCID: PMC3619151 DOI: 10.1089/ars.2012.4836] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIMS Chronic hypoxia (CH) enhances depolarization-induced myofilament Ca(2+) sensitization and resultant pulmonary arterial constriction through superoxide (O(2)(-))-dependent stimulation of RhoA. Because NAD(P)H oxidase (NOX) has been implicated in the development of pulmonary hypertension, we hypothesized that vascular smooth muscle (VSM) depolarization increases NOX-derived O(2)(-) production leading to myofilament Ca(2+) sensitization and augmented vasoconstrictor reactivity following CH. As epidermal growth factor receptor (EGFR) mediates Rac1-dependent NOX activation in renal mesangial cells, we further sought to examine the role EGFR plays in this response. RESULTS Vasoconstrictor responses to depolarizing concentrations of KCl were greater in lungs isolated from CH (4 wk, 0.5 atm) rats compared to normoxic controls, and this effect of CH was abolished by the general NOX inhibitor, apocynin. CH similarly augmented KCl-induced vasoconstriction and O(2)(-) generation (assessed using the fluorescent indicator, dihydroethidium) in Ca(2+)-permeabilized, pressurized small pulmonary arteries. These latter responses to CH were prevented by general inhibition of NOX isoforms (apocynin, diphenylene iodonium), and by selective inhibition of NOX 2 (gp91ds-tat), Rac1 (NSC 23766), and EGFR (AG 1478). Consistent with these observations, CH increased KCl-induced EGFR phosphorylation, and augmented depolarization-induced Rac1 activation in an EGFR-dependent manner. INNOVATION This study establishes a novel signaling axis in VSM linking membrane depolarization to contraction that is independent of Ca(2+) influx, and which mediates myofilament Ca(2+) sensitization in the hypertensive pulmonary circulation. CONCLUSION CH augments membrane depolarization-induced pulmonary VSM Ca(2+) sensitization and vasoconstriction through EGFR-dependent stimulation of Rac1 and NOX 2.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | |
Collapse
|
132
|
Gray SP, Di Marco E, Okabe J, Szyndralewiez C, Heitz F, Montezano AC, de Haan JB, Koulis C, El-Osta A, Andrews KL, Chin-Dusting JPF, Touyz RM, Wingler K, Cooper ME, Schmidt HHHW, Jandeleit-Dahm KA. NADPH Oxidase 1 Plays a Key Role in Diabetes Mellitus–Accelerated Atherosclerosis. Circulation 2013; 127:1888-902. [PMID: 23564668 DOI: 10.1161/circulationaha.112.132159] [Citation(s) in RCA: 292] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Stephen P. Gray
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Elyse Di Marco
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Jun Okabe
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Cedric Szyndralewiez
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Freddy Heitz
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Augusto C. Montezano
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Judy B. de Haan
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Christine Koulis
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Assam El-Osta
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Karen L. Andrews
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Jaye P. F. Chin-Dusting
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Rhian M. Touyz
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Kirstin Wingler
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Mark E. Cooper
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Harald H. H. W. Schmidt
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| | - Karin A Jandeleit-Dahm
- From the Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (S.P.G., E.D.M., J.B.d.H., C.K., M.E.C., K.A.J.-D.); the Department of Medicine, Monash University, Monash, Australia (E.D.M., M.E.C., K.A.J.-D.); Epigenetics in Human Health and Disease, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (J.O., A.E.-O.); GenKyoTex SA, Geneva, Switzerland (C.S., F.H.); Ottawa Hospital Research Institute, Ottawa, Canada (A.C.M., R.M.T.); Institute of
| |
Collapse
|
133
|
Reactive oxygen species, Nox and angiotensin II in angiogenesis: implications for retinopathy. Clin Sci (Lond) 2013; 124:597-615. [PMID: 23379642 DOI: 10.1042/cs20120212] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pathological angiogenesis is a key feature of many diseases including retinopathies such as ROP (retinopathy of prematurity) and DR (diabetic retinopathy). There is considerable evidence that increased production of ROS (reactive oxygen species) in the retina participates in retinal angiogenesis, although the mechanisms by which this occurs are not fully understood. ROS is produced by a number of pathways, including the mitochondrial electron transport chain, cytochrome P450, xanthine oxidase and uncoupled nitric oxide synthase. The family of NADPH oxidase (Nox) enzymes are likely to be important given that their primary function is to produce ROS. Seven isoforms of Nox have been identified named Nox1-5, Duox (dual oxidase) 1 and Duox2. Nox1, Nox2 and Nox4 have been most extensively studied and are implicated in the development of conditions such as hypertension, cardiovascular disease and diabetic nephropathy. In recent years, evidence has accumulated to suggest that Nox1, Nox2 and Nox4 participate in pathological angiogenesis; however, there is no clear consensus about which Nox isoform is primarily responsible. In terms of retinopathy, there is growing evidence that Nox contribute to vascular injury. The RAAS (renin-angiotensin-aldosterone system), and particularly AngII (angiotensin II), is a key stimulator of Nox. It is known that a local RAAS exists in the retina and that blockade of AngII and aldosterone attenuate pathological angiogenesis in the retina. Whether the RAAS influences the production of ROS derived from Nox in retinopathy is yet to be fully determined. These topics will be reviewed with a particular emphasis on ROP and DR.
Collapse
|
134
|
Aoyama T, Paik YH, Watanabe S, Laleu B, Gaggini F, Fioraso-Cartier L, Molango S, Heitz F, Merlot C, Szyndralewiez C, Page P, Brenner DA. Nicotinamide adenine dinucleotide phosphate oxidase in experimental liver fibrosis: GKT137831 as a novel potential therapeutic agent. Hepatology 2012; 56:2316-27. [PMID: 22806357 PMCID: PMC3493679 DOI: 10.1002/hep.25938] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 06/13/2012] [Indexed: 02/06/2023]
Abstract
UNLABELLED Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) generates reactive oxygen species (ROS) in hepatic stellate cells (HSCs) during liver fibrosis. In response to fibrogenic agonists, such as angiotensin II (Ang II), the NOX1 components form an active complex, including Ras-related botulinum toxin substrate 1 (Rac1). Superoxide dismutase 1 (SOD1) interacts with the NOX-Rac1 complex to stimulate NOX activity. NOX4 is also induced in activated HSCs/myofibroblast by increased gene expression. Here, we investigate the role of an enhanced activity SOD1 G37R mutation (SODmu) and the effects of GKT137831, a dual NOX1/4 inhibitor, on HSCs and liver fibrosis. To induce liver fibrosis, wild-type (WT) and SOD1mu mice were treated with CCl(4) or bile duct ligation (BDL). Then, to address the role of NOX-SOD1-mediated ROS production in HSC activation and liver fibrosis, mice were treated with a NOX1/4 inhibitor. Fibrosis and ROS generation was assessed by histology and measurement of thiobarbituric acid reactive substances and NOX-related genes. Primary cultured HSCs isolated from WT, SODmu, and NOX1 knockout (KO) mice were assessed for ROS production, Rac1 activity, and NOX gene expression. Liver fibrosis was increased in SOD1mu mice, and ROS production and Rac1 activity were increased in SOD1mu HSCs. The NOX1/4 inhibitor, GKT137831, attenuated liver fibrosis and ROS production in both SOD1mu and WT mice as well as messenger RNA expression of fibrotic and NOX genes. Treatment with GKT137831 suppressed ROS production and NOX and fibrotic gene expression, but not Rac1 activity, in SOD1mut and WT HSCs. Both Ang II and tumor growth factor beta up-regulated NOX4, but Ang II required NOX1. CONCLUSIONS SOD1mu induces excessive NOX1 activation through Rac1 in HSCs, causing enhanced NOX4 up-regulation, ROS generation, and liver fibrosis. Treatment targeting NOX1/4 may be a new therapy for liver fibrosis.
Collapse
Affiliation(s)
- Tomonori Aoyama
- Department of Medicine, University of California San Diego, La Jolla, CA,Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yong-Han Paik
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Sumio Watanabe
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Benoît Laleu
- GenKyoTex SA, 16 Chemin des Aulx, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | - Francesca Gaggini
- GenKyoTex SA, 16 Chemin des Aulx, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | | | - Sophie Molango
- GenKyoTex SA, 16 Chemin des Aulx, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | - Freddy Heitz
- GenKyoTex SA, 16 Chemin des Aulx, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | - Cédric Merlot
- GenKyoTex SA, 16 Chemin des Aulx, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | | | - Patrick Page
- GenKyoTex SA, 16 Chemin des Aulx, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | - David A. Brenner
- Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
135
|
Li N, Li B, Brun T, Deffert-Delbouille C, Mahiout Z, Daali Y, Ma XJ, Krause KH, Maechler P. NADPH oxidase NOX2 defines a new antagonistic role for reactive oxygen species and cAMP/PKA in the regulation of insulin secretion. Diabetes 2012; 61:2842-50. [PMID: 22933115 PMCID: PMC3478534 DOI: 10.2337/db12-0009] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In insulin-secreting cells, expression of NADPH oxidase (NOX), a potent source of ROS, has been reported, along with controversial findings regarding its function. Here, the role of NOXs was investigated: first by expression and cellular localization in mouse and human pancreatic islets, and then by functional studies in islets isolated from Nox isoform-specific knockout mice. Both human and mouse β-cells express NOX, in particular NOX2. With use of Nox isoform-specific knockout mice, functional analysis revealed Nox2 as the predominant isoform. In human islets, NOX2 colocalized with both insulin granules and endosome/lysosome membranes. Nox2-deficient islets stimulated with 22.8 mmol/L glucose exhibited potentiation of insulin release compared with controls, an effect confirmed with in vitro knockdown of Nox2. The enhanced secretory function in Nox2-deficient islets was associated with both lower superoxide levels and elevated cAMP concentrations. In control islets, GLP-1 and other cAMP inducers suppressed glucose-induced ROS production similarly to Nox2 deficiency. Inhibiting cAMP-dependent protein kinase reduced the secretory response in Nox2-null islets, although not in control islets. This study ascribes a new role for NOX2 in pancreatic β-cells as negative modulator of the secretory response, reducing cAMP/PKA signaling secondary to ROS generation. Results also show reciprocal inhibition between the cAMP/PKA pathway and ROS.
Collapse
Affiliation(s)
- Ning Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
| | - Bin Li
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
| | - Thierry Brun
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
| | | | - Zahia Mahiout
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
| | - Youssef Daali
- Clinical Pharmacology and Toxicology, Geneva University Hospital, Geneva, Switzerland
| | - Xiao-Juan Ma
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
- Corresponding authors: Pierre Maechler, , and Karl-Heinz Krause,
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Corresponding authors: Pierre Maechler, , and Karl-Heinz Krause,
| |
Collapse
|
136
|
Csányi G, Yao M, Rodríguez AI, Al Ghouleh I, Sharifi-Sanjani M, Frazziano G, Huang X, Kelley EE, Isenberg JS, Pagano PJ. Thrombospondin-1 regulates blood flow via CD47 receptor-mediated activation of NADPH oxidase 1. Arterioscler Thromb Vasc Biol 2012; 32:2966-73. [PMID: 23087362 DOI: 10.1161/atvbaha.112.300031] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Although the matricellular protein thrombospondin-1 (TSP1) is highly expressed in the vessel wall in response to injury, its pathophysiological role in the development of vascular disease is poorly understood. This study was designed to test the hypothesis that TSP1 stimulates reactive oxygen species production in vascular smooth muscle cells and induces vascular dysfunction by promoting oxidative stress. METHODS AND RESULTS Nanomolar concentrations of TSP1 found in human vascular disease robustly stimulated superoxide (O(2)(•-)) levels in vascular smooth muscle cells at both cellular and tissue level as measured by cytochrome c and electron paramagnetic resonance. A peptide mimicking the C terminus of TSP1 known to specifically bind CD47 recapitulated this response. Transcriptional knockdown of CD47 and a monoclonal inhibitory CD47 antibody abrogated TSP1-triggered O(2)(•-) in vitro and ex vivo. TSP1 treatment of vascular smooth muscle cells activated phospholipase C and protein kinase C, resulting in phosphorylation of the NADPH oxidase organizer subunit p47(phox) and subsequent Nox1 activation, leading to impairment of arterial vasodilatation ex vivo. Further, we observed that blockade of CD47 and NADPH oxidase 1 gene silencing in vivo in rats improves TSP1-induced impairment of tissue blood flow after ischemia reperfusion. CONCLUSIONS Our data suggest a highly regulated process of reactive oxygen species stimulation and blood flow regulation promoted through a direct TSP1/CD47-mediated activation of Nox1. This is the first report, to our knowledge, of a matricellular protein acting as a ligand for NADPH oxidase activation and through specific engagement of integrin-associated protein CD47.
Collapse
Affiliation(s)
- Gábor Csányi
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Abhijit S, Bhaskaran R, Narayanasamy A, Chakroborty A, Manickam N, Dixit M, Mohan V, Balasubramanyam M. Hyperinsulinemia-induced vascular smooth muscle cell (VSMC) migration and proliferation is mediated by converging mechanisms of mitochondrial dysfunction and oxidative stress. Mol Cell Biochem 2012; 373:95-105. [PMID: 23073711 DOI: 10.1007/s11010-012-1478-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/26/2012] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is one of the major complications of diabetes and involves endothelial dysfunction, matrix alteration, and most importantly migration and proliferation of vascular smooth muscle cells (VSMCs). Although hyperglycemia and hyperinsulinemia are known to contribute to atherosclerosis, little is known about the specific cellular signaling pathways that mediate the detrimental hyperinsulinemic effects in VSMCs. Therefore, we investigated the cellular mechanisms of hyperinsulinemia-induced migration and proliferation of VSMCs. VSMCs were treated with insulin (100 nM) for 6 days and subjected to various physiological and molecular investigations. VSMCs subjected to hyperinsulinemia exhibited increased migration and proliferation, and this is paralleled by oxidative stress [increased NADPH oxidase activity, NADPH oxidase 1 mRNA expression, and reactive oxygen species (ROS) generation], alterations in mitochondrial physiology (membrane depolarization, decreased mitochondrial mass, and increased mitochondrial ROS), changes in mitochondrial biogenesis-related genes (mitofusin 1, mitofusin 2, dynamin-related protein 1, peroxisome proliferator-activated receptor gamma coactivator 1-alpha, peroxisome proliferator-activated receptor gamma coactivator 1-beta, nuclear respiratory factor 1, and uncoupling protein 2), and increased Akt phosphorylation. Diphenyleneiodonium, a known NADPH oxidase inhibitor significantly inhibited migration and proliferation of VSMCs and normalized all the above physiological and molecular perturbations. This study suggests a plausible crosstalk between mitochondrial dysfunction and oxidative stress under hyperinsulinemia and emphasizes counteracting mitochondrial dysfunction and oxidative stress as a novel therapeutic strategy for atherosclerosis.
Collapse
Affiliation(s)
- Shiny Abhijit
- Department of Cell and Molecular Biology, Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialities Centre, Gopalapuram, Chennai 600 086, India
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Stanic B, Pandey D, Fulton DJ, Miller FJ. Increased epidermal growth factor-like ligands are associated with elevated vascular nicotinamide adenine dinucleotide phosphate oxidase in a primate model of atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 32:2452-60. [PMID: 22879585 DOI: 10.1161/atvbaha.112.256107] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To characterize the relationship between the expression of epidermal growth factor (EGF)-like ligands and vascular nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression and activity in a primate model of atherosclerosis. METHODS AND RESULTS Adult male Cynomolgus monkeys were fed a normal or atherogenic (AS) diet for 45 months, after which animals from the AS group were placed on a normal diet for 8 months (regression). The expression of membrane-associated EGF-like ligands was increased in arteries from animals on the AS diet and normalized in the regression group. EGF-like ligands were distributed throughout atherosclerotic vessels but predominantly colocalized with macrophages. Consistent with ligand shedding, circulating heparin-bound EGF was elevated in the plasma of AS monkeys but not in those on regression diet. Atherosclerosis was associated with the activation of EGF receptor signaling. Expression of NADPH oxidase subunits Nox1 and Nox2 but not Nox4 or Nox5 was increased in arteries from monkeys on the AS diet and returned to normal with regression. Levels of Nox1 and Nox2 positively correlated with EGF-like ligands. In cultured monkey smooth muscle cells, treatment with EGF-like ligands increased Nox1 expression and activity. CONCLUSIONS These data identify EGF-like ligands as potential modulators of atherogenesis, resulting in part from increased vascular NADPH oxidase activity.
Collapse
Affiliation(s)
- Bojana Stanic
- Department of Internal Medicine The University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
139
|
Chan SHH, Chan JYH. Brain stem oxidative stress and its associated signaling in the regulation of sympathetic vasomotor tone. J Appl Physiol (1985) 2012; 113:1921-8. [PMID: 22837172 DOI: 10.1152/japplphysiol.00610.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is now compelling evidence from studies in humans and animals that overexcitation of the sympathetic nervous system plays an important role in the pathogenesis of cardiovascular diseases. An excellent example is neurogenic hypertension, in which central sympathetic overactivation is involved in the development, staging, and progression of the disease, and one of the underlying mechanisms involves oxidative stress in key brain stem sites that are engaged in the regulation of sympathetic vasomotor tone. Using the rostral ventrolateral medulla (RVLM) and nucleus tractus solitarii (NTS) as two illustrative brain stem neural substrates, this article provides an overview of the impact of reactive oxygen species and antioxidants on RVLM and NTS in the pathogenesis of neurogenic hypertension. This is followed by a discussion of the redox-sensitive signaling pathways, including several kinases, ion channels, and transcription factors that underpin the augmentation in sympathetic vasomotor tone. In addition, the emerging view that brain stem oxidative stress is also causally related to a reduction in sympathetic vasomotor tone and hypotension during brain stem death, methamphetamine intoxication, and temporal lobe status epilepticus will be presented, along with the causal contribution of the oxidant peroxynitrite formed by a reaction between nitric oxide synthase II (NOS II)-derived nitric oxide and superoxide. Also discussed as a reasonable future research direction is dissection of the cellular mechanisms and signaling cascades that may underlie the contributory role of nitric oxide generated by different NOS isoforms in the differential effects of oxidative stress in the RVLM or NTS on sympathetic vasomotor tone.
Collapse
Affiliation(s)
- Samuel H H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan, Republic of China
| | | |
Collapse
|
140
|
Youn JY, Gao L, Cai H. The p47phox- and NADPH oxidase organiser 1 (NOXO1)-dependent activation of NADPH oxidase 1 (NOX1) mediates endothelial nitric oxide synthase (eNOS) uncoupling and endothelial dysfunction in a streptozotocin-induced murine model of diabetes. Diabetologia 2012; 55:2069-79. [PMID: 22549734 PMCID: PMC3694990 DOI: 10.1007/s00125-012-2557-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 02/23/2012] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS We have previously shown that NADPH oxidase (NOX) lies upstream of uncoupled endothelial nitric oxide synthase (eNOS), which is known to occur in diabetic endothelium. However, it remains unclear which specific NOX isoform(s) is responsible for eNOS uncoupling and endothelial dysfunction in diabetic mouse models. The aim of the present study was to test the hypothesis that one or more NOX isoform(s) mediate(s) diabetic uncoupling of eNOS, which has been shown to occur in patients with diabetes to contribute to endothelial dysfunction. METHODS Diabetes was induced by streptozotocin administration. The N (ω)-nitro-L-arginine methyl ester (L-NAME)-sensitive superoxide production of aortic segments, reflective of eNOS uncoupling activity, was determined by electron spin resonance. RESULTS The L-NAME-sensitive superoxide production was more than doubled in wild-type diabetic mice, implicating uncoupling of eNOS. This was abolished in diabetic p47 ( phox-/-) (also known as Ncf1 (-/-)) mice, but preserved in Nox2 (-/y) (also known as Cybb (-/-)) mice made diabetic. The eNOS uncoupling activity was markedly attenuated in diabetic mice transfected with Nox1 or Nox1 organiser 1 (Noxo1) short interfering RNA (siRNA), and abolished in Nox1 (-/y) diabetic mice. Diabetes-induced impairment in endothelium-dependent vasorelaxation was also significantly attenuated in the Nox1 (-/y) mice made diabetic. By contrast, Nox4 siRNA, or inhibition of mitochondrial complex I or III with rotenone or siRNA, respectively, had no effect on diabetic uncoupling of eNOS. Overexpression of Dhfr, or oral administration of folic acid to improve dihydrofolate reductase (DHFR) function, recoupled eNOS in diabetes to improve endothelial function. CONCLUSIONS/INTERPRETATION Our data demonstrate for the first time that the p47(phox) and NOXO1-dependent activation of NOX1, but not that of NOX2, NOX4 or mitochondrion, mediates diabetic uncoupling of eNOS. NOX1-null mice are protected from diabetic endothelial dysfunction. Novel approaches to inhibit NOX1 and/or improve DHFR function, may prove to have therapeutic potential for diabetic endothelial dysfunction.
Collapse
Affiliation(s)
- J. Y. Youn
- Division of Molecular Medicine, Cardiovascular Research Laboratories, Departments of Anesthesiology and Medicine, David Geffen School of Medicine, University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA 90095, USA
| | - L. Gao
- Division of Molecular Medicine, Cardiovascular Research Laboratories, Departments of Anesthesiology and Medicine, David Geffen School of Medicine, University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA 90095, USA
| | - H. Cai
- Division of Molecular Medicine, Cardiovascular Research Laboratories, Departments of Anesthesiology and Medicine, David Geffen School of Medicine, University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA 90095, USA
| |
Collapse
|
141
|
Montezano AC, Touyz RM. Oxidative stress, Noxs, and hypertension: experimental evidence and clinical controversies. Ann Med 2012; 44 Suppl 1:S2-16. [PMID: 22713144 DOI: 10.3109/07853890.2011.653393] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are signaling molecules that influence many physiological processes. Increased ROS bioavailability and altered redox signaling (oxidative stress) have been implicated in chronic diseases including hypertension. Although oxidative stress may not be the sole cause of hypertension, it amplifies blood pressure elevation in the presence of other prohypertensive factors (salt, renin-angiotensin system, sympathetic hyperactivity). A major source for cardiovascular ROS is a family of non-phagocytic NADPH oxidases (Nox1, Nox2, Nox4, Nox5). Other sources of ROS involve mitochondrial electron transport enzymes, xanthine oxidase, and uncoupled nitric oxide synthase. Although evidence from experimental and animal studies supports a role for oxidative stress in the pathogenesis of hypertension, there is still no convincing proof that oxidative stress is a cause of human hypertension. However, what is clear is that oxidative stress is important in the molecular mechanisms associated with cardiovascular and renal injury in hypertension and that hypertension itself can contribute to oxidative stress. The present review addresses the putative function of ROS in the pathogenesis of hypertension and focuses on the role of Noxs in ROS generation in vessels and the kidney. Implications of oxidative stress in human hypertension are discussed, and clinical uncertainties are highlighted.
Collapse
Affiliation(s)
- Augusto C Montezano
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | |
Collapse
|
142
|
Altenhöfer S, Kleikers PWM, Radermacher KA, Scheurer P, Rob Hermans JJ, Schiffers P, Ho H, Wingler K, Schmidt HHHW. The NOX toolbox: validating the role of NADPH oxidases in physiology and disease. Cell Mol Life Sci 2012; 69:2327-43. [PMID: 22648375 PMCID: PMC3383958 DOI: 10.1007/s00018-012-1010-9] [Citation(s) in RCA: 288] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are cellular signals but also disease triggers; their relative excess (oxidative stress) or shortage (reductive stress) compared to reducing equivalents are potentially deleterious. This may explain why antioxidants fail to combat diseases that correlate with oxidative stress. Instead, targeting of disease-relevant enzymatic ROS sources that leaves physiological ROS signaling unaffected may be more beneficial. NADPH oxidases are the only known enzyme family with the sole function to produce ROS. Of the catalytic NADPH oxidase subunits (NOX), NOX4 is the most widely distributed isoform. We provide here a critical review of the currently available experimental tools to assess the role of NOX and especially NOX4, i.e. knock-out mice, siRNAs, antibodies, and pharmacological inhibitors. We then focus on the characterization of the small molecule NADPH oxidase inhibitor, VAS2870, in vitro and in vivo, its specificity, selectivity, and possible mechanism of action. Finally, we discuss the validation of NOX4 as a potential therapeutic target for indications including stroke, heart failure, and fibrosis.
Collapse
Affiliation(s)
- Sebastian Altenhöfer
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Pamela W. M. Kleikers
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Kim A. Radermacher
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | | | - J. J. Rob Hermans
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Paul Schiffers
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Heidi Ho
- National Stroke Research Institute, Melbourne, VIC Australia
| | - Kirstin Wingler
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Harald H. H. W. Schmidt
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
143
|
Chrissobolis S, Banfi B, Sobey CG, Faraci FM. Role of Nox isoforms in angiotensin II-induced oxidative stress and endothelial dysfunction in brain. J Appl Physiol (1985) 2012; 113:184-91. [PMID: 22628375 DOI: 10.1152/japplphysiol.00455.2012] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Angiotensin II (Ang II) promotes vascular disease through several mechanisms including by producing oxidative stress and endothelial dysfunction. Although multiple potential sources of reactive oxygen species exist, the relative importance of each is unclear, particularly in individual vascular beds. In these experiments, we examined the role of NADPH oxidase (Nox1 and Nox2) in Ang II-induced endothelial dysfunction in the cerebral circulation. Treatment with Ang II (1.4 mg·kg(-1)·day(-1) for 7 days), but not vehicle, increased blood pressure in all groups. In wild-type (WT; C57Bl/6) mice, Ang II reduced dilation of the basilar artery to the endothelium-dependent agonist acetylcholine compared with vehicle but had no effect on responses in Nox2-deficient (Nox2(-/y)) mice. Ang II impaired responses to acetylcholine in Nox1 WT (Nox1(+/y)) and caused a small reduction in responses to acetylcholine in Nox1-deficient (Nox1(-/y)) mice. Ang II did not impair responses to the endothelium-independent agonists nitroprusside or papaverine in either group. In WT mice, Ang II increased basal and phorbol-dibutyrate-stimulated superoxide production in the cerebrovasculature, and these increases were abolished in Nox2(-/y) mice. Overall, these data suggest that Nox2 plays a relatively prominent role in mediating Ang II-induced oxidative stress and cerebral endothelial dysfunction, with a minor role for Nox1.
Collapse
Affiliation(s)
- Sophocles Chrissobolis
- Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
144
|
The 1027th target candidate in stroke: Will NADPH oxidase hold up? EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2012; 4:11. [PMID: 22625431 PMCID: PMC3403875 DOI: 10.1186/2040-7378-4-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 05/24/2012] [Indexed: 02/07/2023]
Abstract
As recently reviewed, 1026 neuroprotective drug candidates in stroke research have all failed on their road towards validation and clinical translation, reasons being quality issues in preclinical research and publication bias. Quality control guidelines for preclinical stroke studies have now been established. However, sufficient understanding of the underlying mechanisms of neuronal death after stroke that could be possibly translated into new therapies is lacking. One exception is the hypothesis that cellular death is mediated by oxidative stress. Oxidative stress is defined as an excess of reactive oxygen species (ROS) derived from different possible enzymatic sources. Among these, NADPH oxidases (NOX1-5) stand out as they represent the only known enzyme family that has no other function than to produce ROS. Based on data from different NOX knockout mouse models in ischemic stroke, the most relevant isoform appears to be NOX4. Here we discuss the state-of-the-art of this target with respect to stroke and open questions that need to be addressed on the path towards clinical translation.
Collapse
|
145
|
Lassègue B, San Martín A, Griendling KK. Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system. Circ Res 2012; 110:1364-90. [PMID: 22581922 PMCID: PMC3365576 DOI: 10.1161/circresaha.111.243972] [Citation(s) in RCA: 611] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 03/09/2012] [Indexed: 02/07/2023]
Abstract
The NADPH oxidase (Nox) enzymes are critical mediators of cardiovascular physiology and pathophysiology. These proteins are expressed in virtually all cardiovascular cells, and regulate such diverse functions as differentiation, proliferation, apoptosis, senescence, inflammatory responses and oxygen sensing. They target a number of important signaling molecules, including kinases, phosphatases, transcription factors, ion channels, and proteins that regulate the cytoskeleton. Nox enzymes have been implicated in many different cardiovascular pathologies: atherosclerosis, hypertension, cardiac hypertrophy and remodeling, angiogenesis and collateral formation, stroke, and heart failure. In this review, we discuss in detail the biochemistry of Nox enzymes expressed in the cardiovascular system (Nox1, 2, 4, and 5), their roles in cardiovascular cell biology, and their contributions to disease development.
Collapse
Affiliation(s)
- Bernard Lassègue
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
146
|
Lee H, Ham SA, Kim MY, Kim JH, Paek KS, Kang ES, Kim HJ, Hwang JS, Yoo T, Park C, Kim JH, Lim DS, Han CW, Seo HG. Activation of PPARδ counteracts angiotensin II-induced ROS generation by inhibiting rac1 translocation in vascular smooth muscle cells. Free Radic Res 2012; 46:912-9. [PMID: 22519881 DOI: 10.3109/10715762.2012.687448] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Angiotensin II (Ang II)-mediated modification of the redox milieu of vascular smooth muscle cells (VSMCs) has been implicated in several pathophysiological processes, including cell proliferation, migration and differentiation. In this study, we demonstrate that the peroxisome proliferator-activated receptor (PPAR) δ counteracts Ang II-induced production of reactive oxygen species (ROS) in VSMCs. Activation of PPARδ by GW501516, a specific ligand for PPARδ, significantly reduced Ang II-induced ROS generation in VSMCs. This effect was, however, reversed in the presence of small interfering (si)RNA against PPARδ. The marked increase in ROS levels induced by Ang II was also eliminated by the inhibition of phosphatidylinositol 3-kinase (PI3K) but not of protein kinase C, suggesting the involvement of the PI3K/Akt signalling pathway in this process. Accordingly, ablation of Akt with siRNA further enhanced the inhibitory effects of GW501516 in Ang II-induced superoxide production. Ligand-activated PPARδ also blocked Ang II-induced translocation of Rac1 to the cell membrane, inhibiting the activation of NADPH oxidases and consequently ROS generation. These results indicate that ligand-activated PPARδ plays an important role in the cellular response to oxidative stress by decreasing ROS generated by Ang II in vascular cells.
Collapse
Affiliation(s)
- Hanna Lee
- Department of Animal Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangjin-Gu, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Takac I, Schröder K, Brandes RP. The Nox family of NADPH oxidases: friend or foe of the vascular system? Curr Hypertens Rep 2012; 14:70-8. [PMID: 22071588 DOI: 10.1007/s11906-011-0238-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NADPH (nicotinamide adenine dinucleotide phosphate) oxidases are important sources of reactive oxygen species (ROS). In the vascular system, ROS can have both beneficial and detrimental effects. Under physiologic conditions, ROS are involved in signaling pathways that regulate vascular tone as well as cellular processes like proliferation, migration and differentiation. However, high doses of ROS, which are produced after induction or activation of NADPH oxidases in response to cardiovascular risk factors and inflammation, contribute to the development of endothelial dysfunction and vascular disease. In vascular cells, the NADPH oxidase isoforms Nox1, Nox2, Nox4, and Nox5 are expressed, which differ in their activity, response to stimuli, and the type of ROS released. This review focuses on the specific role of different NADPH oxidase isoforms in vascular physiology and their potential contributions to vascular diseases.
Collapse
Affiliation(s)
- Ina Takac
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt am Main, Germany
| | | | | |
Collapse
|
148
|
Song P, Zou MH. Regulation of NAD(P)H oxidases by AMPK in cardiovascular systems. Free Radic Biol Med 2012; 52:1607-19. [PMID: 22357101 PMCID: PMC3341493 DOI: 10.1016/j.freeradbiomed.2012.01.025] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 01/25/2012] [Accepted: 01/27/2012] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are ubiquitously produced in cardiovascular systems. Under physiological conditions, ROS/RNS function as signaling molecules that are essential in maintaining cardiovascular function. Aberrant concentrations of ROS/RNS have been demonstrated in cardiovascular diseases owing to increased production or decreased scavenging, which have been considered common pathways for the initiation and progression of cardiovascular diseases such as atherosclerosis, hypertension, (re)stenosis, and congestive heart failure. NAD(P)H oxidases are primary sources of ROS and can be induced or activated by all known cardiovascular risk factors. Stresses, hormones, vasoactive agents, and cytokines via different signaling cascades control the expression and activity of these enzymes and of their regulatory subunits. But the molecular mechanisms by which NAD(P)H oxidase is regulated in cardiovascular systems remain poorly characterized. Investigations by us and others suggest that adenosine monophosphate-activated protein kinase (AMPK), as an energy sensor and modulator, is highly sensitive to ROS/RNS. We have also obtained convincing evidence that AMPK is a physiological suppressor of NAD(P)H oxidase in multiple cardiovascular cell systems. In this review, we summarize our current understanding of how AMPK functions as a physiological repressor of NAD(P)H oxidase.
Collapse
Affiliation(s)
| | - Ming-Hui Zou
- To whom correspondence should be addressed: Ming-Hui Zou, M.D., Ph.D., Department of Medicine, University of Oklahoma Health Science Center, 941 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA, Phone: 405-271-3974, Fax: 405-271-3973,
| |
Collapse
|
149
|
Wingler K, Hermans JJR, Schiffers P, Moens A, Paul M, Schmidt HHHW. NOX1, 2, 4, 5: counting out oxidative stress. Br J Pharmacol 2012; 164:866-83. [PMID: 21323893 PMCID: PMC3195911 DOI: 10.1111/j.1476-5381.2011.01249.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
For decades, oxidative stress has been discussed as a key mechanism of endothelial dysfunction and cardiovascular disease. However, attempts to validate and exploit this hypothesis clinically by supplementing antioxidants have failed. Nevertheless, this does not disprove the oxidative stress hypothesis. As a certain degree of reactive oxygen species (ROS) formation appears to be physiological and beneficial. To reduce oxidative stress therapeutically, two alternative approaches are being developed. One is the repair of key signalling components that are compromised by oxidative stress. These include uncoupled endothelial nitric oxide (NO) synthase and oxidized/heme-free NO receptor soluble guanylate cyclase. A second approach is to identify and effectively inhibit the relevant source(s) of ROS in a given disease condition. A highly likely target in this context is the family of NADPH oxidases. Animal models, including NOX knockout mice and new pharmacological inhibitors of NADPH oxidases have opened up a new era of oxidative stress research and have paved the way for new cardiovascular therapies.
Collapse
Affiliation(s)
- K Wingler
- Department of Pharmacology & Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | | | | | | | | | | |
Collapse
|
150
|
Bedard K, Jaquet V, Krause KH. NOX5: from basic biology to signaling and disease. Free Radic Biol Med 2012; 52:725-34. [PMID: 22182486 DOI: 10.1016/j.freeradbiomed.2011.11.023] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 11/10/2011] [Accepted: 11/12/2011] [Indexed: 02/07/2023]
Abstract
In mammals, the NADPH oxidase family of enzymes comprises seven members: NOXs 1-5, DUOX1, and DUOX2. All of these enzymes function to move an electron across cellular membranes, transferring it to oxygen to generate the superoxide anion. This generation of reactive oxygen species has important physiological and pathophysiological roles. NOX5 is perhaps the least well understood of these NOX isoforms, in part because the gene is not present in mice or rats. In recent years, however, there has been a rapid increase in our understanding of the NOX5 gene, the structural and biochemical aspects of the NOX5 enzyme, the role NOX5 plays in health and disease, and the development of novel NOX inhibitors. This review takes a look back at some historical aspects of the discovery of NOX5 and summarizes our current understanding of the enzyme.
Collapse
Affiliation(s)
- Karen Bedard
- Department of Pathology, Dalhousie University, Halifax, Canada
| | | | | |
Collapse
|