401
|
Tang X, Luo YX, Chen HZ, Liu DP. Mitochondria, endothelial cell function, and vascular diseases. Front Physiol 2014; 5:175. [PMID: 24834056 PMCID: PMC4018556 DOI: 10.3389/fphys.2014.00175] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/16/2014] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are perhaps the most sophisticated and dynamic responsive sensing systems in eukaryotic cells. The role of mitochondria goes beyond their capacity to create molecular fuel and includes the generation of reactive oxygen species, the regulation of calcium, and the activation of cell death. In endothelial cells, mitochondria have a profound impact on cellular function under both healthy and diseased conditions. In this review, we summarize the basic functions of mitochondria in endothelial cells and discuss the roles of mitochondria in endothelial dysfunction and vascular diseases, including atherosclerosis, diabetic vascular dysfunction, pulmonary artery hypertension, and hypertension. Finally, the potential therapeutic strategies to improve mitochondrial function in endothelial cells and vascular diseases are also discussed, with a focus on mitochondrial-targeted antioxidants and calorie restriction.
Collapse
Affiliation(s)
- Xiaoqiang Tang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| | - Yu-Xuan Luo
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| |
Collapse
|
402
|
Garrido-Urbani S, Jaquet V, Imhof BA. [ROS and NADPH oxidase: key regulators of tumor vascularisation]. Med Sci (Paris) 2014; 30:415-21. [PMID: 24801037 DOI: 10.1051/medsci/20143004016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress is the result of an imbalance between the production of reactive oxygen species (ROS) and antioxidant mechanisms. It is characterized by damage of all cellular components, DNA, proteins, lipids. ROS are nevertheless important for the physiology of an organism, as they are involved in the innate immune defense and several intracellular signaling pathways. They play an important role in tumorigenesis by promoting tumor vasculature, which is essential to their growth and metastatic processes. There are many sources of ROS in the cells, but the NOX enzymes (NADPH oxidase-dependent) are now recognized to have a major role in the oxidative stress process. Indeed, they are present in many tissues where their only function is to produce ROS. This article discusses the NOX in endothelial cells and their role in the tumor angiogenesis.
Collapse
Affiliation(s)
- Sarah Garrido-Urbani
- Centre médical universitaire, département pathologie et immunologie, faculté de médecine, université de Genève, 1, rue Michel Servet, 1211 Genève, Suisse
| | - Vincent Jaquet
- Centre médical universitaire, département pathologie et immunologie, faculté de médecine, université de Genève, 1, rue Michel Servet, 1211 Genève, Suisse
| | - Beat A Imhof
- Centre médical universitaire, département pathologie et immunologie, faculté de médecine, université de Genève, 1, rue Michel Servet, 1211 Genève, Suisse
| |
Collapse
|
403
|
Abstract
SIGNIFICANCE Adhesion and migration induced by cytokines or growth factors are well-organized processes in cellular motility. Reactive oxygen species (ROS) are specifically produced by the Nox family of NADPH oxidases. RECENT ADVANCES The signal transduction of migration and adhesion depends on ROS produced by Nox enzymes and factors that initiate migration and adhesion and stimulate cellular ROS formation. CRITICAL ISSUES The identification of molecular targets of ROS formation in the signal transduction of adhesion and migration is still in its beginnings, but a site and isoform-specific contribution of Nox enzymes to this process becomes apparent. Nox-derived ROS, therefore, act as second messengers that are specifically modifying signaling proteins involved in adhesion and migration. FUTURE DIRECTIONS Individual protein targets of Nox-mediated redox signaling in different cell types and tissues will be identified. Isoform-specific Nox inhibitors will be developed to modulate the ROS-dependent component of migration and adhesion. These compounds might be suited to elicit differential effects between pathophysiologic and physiologic adhesion and migration.
Collapse
Affiliation(s)
- Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität , Frankfurt am Main, Germany
| |
Collapse
|
404
|
Mitochondrial regulation of NADPH oxidase in hindlimb unweighting rat cerebral arteries. PLoS One 2014; 9:e95916. [PMID: 24759683 PMCID: PMC3997512 DOI: 10.1371/journal.pone.0095916] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/01/2014] [Indexed: 02/07/2023] Open
Abstract
Exposure to microgravity results in post-flight cardiovascular deconditioning and orthostatic intolerance in astronauts. Vascular oxidative stress injury and mitochondrial dysfunction have been indicated in this process. To elucidate the mechanism for this condition, we investigated whether mitochondria regulated NADPH oxidase in hindlimb unweighting (HU) rat cerebral and mesenteric arteries. Four-week HU was used to simulate microgravity in rats. Vascular superoxide generation, protein and mRNA levels of Nox2/Nox4, and the activity of NADPH oxidase were examined in the present study. Compared with control rats, the levels of superoxide increased in cerebral (P<0.001) but not in mesenteric vascular smooth muscle cells. The protein and mRNA levels of Nox2 and Nox4 were upregulated significantly (P<0.001 and P<0.001 for Nox2, respectively; P<0.001 and P<0.001 for Nox4, respectively) in HU rat cerebral arteries but not in mesenteric arteries. NADPH oxidases were activated significantly by HU (P<0.001) in cerebral arteries but not in mesenteric arteries. Chronic treatment with mitochondria-targeted antioxidant mitoTEMPO attenuated superoxide levels (P<0.001), decreased the protein and mRNA expression levels of Nox2/Nox4 (P<0.01 and P<0.05 for Nox2, respectively; P<0.001 and P<0.001 for Nox4, respectively) and the activity of NADPH oxidase (P<0.001) in HU rat cerebral arteries, but exerted no effects on HU rat mesenteric arteries. Therefore, mitochondria regulated the expression and activity of NADPH oxidases during simulated microgravity. Both mitochondria and NADPH oxidase participated in vascular redox status regulation.
Collapse
|
405
|
Nox4 NADPH oxidase contributes to smooth muscle cell phenotypes associated with unstable atherosclerotic plaques. Redox Biol 2014; 2:642-50. [PMID: 24936437 PMCID: PMC4052526 DOI: 10.1016/j.redox.2014.04.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/12/2014] [Accepted: 04/13/2014] [Indexed: 11/23/2022] Open
Abstract
Plaque instability associated with acute coronary syndromes results in part from apoptosis and senescence of cells within the atherosclerotic (AS) lesion. Increased cellular oxidative stress has been proposed to contribute to plaque progression and changes in composition, leading to plaque instability. Our objective was to examine the role of NADPH oxidase in smooth muscle cell (SMC) phenotypes associated with an unstable plaque. Aortae were isolated from pre-lesion (8 weeks of age) and post-lesion (35 weeks of age) hypercholesterolemic mice (ApoE(-/-)/LDLR(-/-), AS), and age-matched normal C57BL/6J mice. We observed an age-dependent increase in reactive oxygen species (ROS) in aorta from AS mice, with evidence for elevated ROS prior to lesion development. Whereas macrophage infiltration was restricted to the lesion, oxidized lipids extended beyond the plaque and into the vessel wall. Consistent with these findings, we observed dynamic changes in the expression of NADPH oxidases in AS vessels. Specifically, Nox1 expression was increased early and decreased with lesion progression, while induction of Nox4 was a late event. Nox2 and p22(phox) were elevated throughout lesion development. Similar to observations in aortae, SMCs isolated from the lesion of AS aortae had decreased Nox1 and increased Nox4 levels as compared to SMCs from normal mice. AS SMCs demonstrated increased generation of ROS, cell cycle arrest, evidence of senescence, and increased susceptibility to apoptosis. Overexpression of Nox4 in normal SMCs recapitulated the phenotypes of the AS SMCs. We conclude that increased expression of Nox4 in AS may drive SMC phenotypes that lead to the plaque instability and rupture responsible for myocardial infarction and stroke.
Collapse
|
406
|
Dunn LL, Midwinter RG, Ni J, Hamid HA, Parish CR, Stocker R. New insights into intracellular locations and functions of heme oxygenase-1. Antioxid Redox Signal 2014; 20:1723-42. [PMID: 24180287 PMCID: PMC3961787 DOI: 10.1089/ars.2013.5675] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/01/2013] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Heme oxygenase-1 (HMOX1) plays a critical role in the protection of cells, and the inducible enzyme is implicated in a spectrum of human diseases. The increasing prevalence of cardiovascular and metabolic morbidities, for which current treatment approaches are not optimal, emphasizes the necessity to better understand key players such as HMOX1 that may be therapeutic targets. RECENT ADVANCES HMOX1 is a dynamic protein that can undergo post-translational and structural modifications which modulate HMOX1 function. Moreover, trafficking from the endoplasmic reticulum to other cellular compartments, including the nucleus, highlights that HMOX1 may play roles other than the catabolism of heme. CRITICAL ISSUES The ability of HMOX1 to be induced by a variety of stressors, in an equally wide variety of tissues and cell types, represents an obstacle for the therapeutic exploitation of the enzyme. Any capacity to modulate HMOX1 in cardiovascular and metabolic diseases should be tempered with an appreciation that HMOX1 may have an impact on cancer. Moreover, the potential for heme catabolism end products, such as carbon monoxide, to amplify the HMOX1 stress response should be considered. FUTURE DIRECTIONS A more complete understanding of HMOX1 modifications and the properties that they impart is necessary. Delineating these parameters will provide a clearer picture of the opportunities to modulate HMOX1 in human disease.
Collapse
Affiliation(s)
- Louise L. Dunn
- Vascular Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, Australia
- Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | | | - Jun Ni
- Vascular Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, Australia
- Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - Hafizah A. Hamid
- Vascular Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, Australia
- Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - Christopher R. Parish
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Roland Stocker
- Vascular Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, Australia
- Faculty of Medicine, The University of New South Wales, Sydney, Australia
| |
Collapse
|
407
|
Hecker L, Logsdon NJ, Kurundkar D, Kurundkar A, Bernard K, Hock T, Meldrum E, Sanders YY, Thannickal VJ. Reversal of persistent fibrosis in aging by targeting Nox4-Nrf2 redox imbalance. Sci Transl Med 2014; 6:231ra47. [PMID: 24718857 PMCID: PMC4545252 DOI: 10.1126/scitranslmed.3008182] [Citation(s) in RCA: 518] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The incidence and prevalence of pathological fibrosis increase with advancing age, although mechanisms for this association are unclear. We assessed the capacity for repair of lung injury in young (2 months) and aged (18 months) mice. Whereas the severity of fibrosis was not different between these groups, aged mice demonstrated an impaired capacity for fibrosis resolution. Persistent fibrosis in lungs of aged mice was characterized by the accumulation of senescent and apoptosis-resistant myofibroblasts. These cellular phenotypes were sustained by alterations in cellular redox homeostasis resulting from elevated expression of the reactive oxygen species-generating enzyme Nox4 [NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase-4] and an impaired capacity to induce the Nrf2 (NFE2-related factor 2) antioxidant response. Lung tissues from human subjects with idiopathic pulmonary fibrosis (IPF), a progressive and fatal lung disease, also demonstrated this Nox4-Nrf2 imbalance. Nox4 mediated senescence and apoptosis resistance in IPF fibroblasts. Genetic and pharmacological targeting of Nox4 in aged mice with established fibrosis attenuated the senescent, antiapoptotic myofibroblast phenotype and led to a reversal of persistent fibrosis. These studies suggest that loss of cellular redox homeostasis promotes profibrotic myofibroblast phenotypes that result in persistent fibrosis associated with aging. Our studies suggest that restoration of Nox4-Nrf2 redox balance in myofibroblasts may be a therapeutic strategy in age-associated fibrotic disorders, potentially able to resolve persistent fibrosis or even reverse its progression.
Collapse
Affiliation(s)
- Louise Hecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Birmingham VA Medical Center, Birmingham, AL 35233, USA
| | - Naomi J. Logsdon
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Deepali Kurundkar
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ashish Kurundkar
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen Bernard
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas Hock
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Yan Y. Sanders
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor J. Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
408
|
Valsartan slows the progression of diabetic nephropathy in db/db mice via a reduction in podocyte injury, and renal oxidative stress and inflammation. Clin Sci (Lond) 2014; 126:707-20. [PMID: 24195695 DOI: 10.1042/cs20130223] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Higher doses of AngII (angiotensin II) blockers are intended to optimize albuminuria reduction rather than for blood pressure control in chronic kidney disease. However, the long-term renoprotection of high-dose AngII blockers has yet to be defined. The present study sought to determine whether doses of ARB (AngII receptor blocker) that maximally reduce proteinuria could slow the progression of glomerulosclerosis in the uninephrectomized db/db mouse, a model of Type 2 diabetes. Untreated uninephrectomized db/db mice had normal blood pressure, but developed progressive albuminuria and mesangial matrix expansion between 18 and 22 weeks of age, which was associated with increased renal expression of TGFβ1 (transforming growth factor β1), PAI-1 (plasminogen-activator inhibitor-1), type IV collagen and FN (fibronectin). Treatment with valsartan in the drinking water of db/db mice from 18 to 22 weeks of age, at a dose that was determined previously to maximally reduce proteinuria, prevented the increases in albuminuria and the markers of renal fibrosis seen in untreated db/db mice. In addition, WT-1 (Wilms tumour protein-1)-immunopositive podocyte numbers were found to be lower in the untreated glomeruli of mice with diabetes. The expression of podocin and nephrin were continually decreased in mice with diabetes between 18 and 22 weeks of age. These changes are indicative of podocyte injury and the administration of valsartan ameliorated them substantially. Renal expression of TNFα (tumour necrosis factor α), MCP-1 (monocyte chemoattractant protein-1), Nox2 (NADPH oxidase 2), p22phox and p47phox and urine TBARS (thiobarbituric acid-reacting substance) levels, the markers of renal inflammation and oxidative stress, were increased during disease progression in mice with diabetes. Valsartan treatment was shown to reduce these markers. Thus high doses of valsartan not only reduce albuminuria maximally, but also halt the progression of the glomerulosclerosis resulting from Type 2 diabetes via a reduction in podocyte injury and renal oxidative stress and inflammation.
Collapse
|
409
|
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: 4.9] [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
|
410
|
Wang J, Hong Z, Zeng C, Yu Q, Wang H. NADPH oxidase 4 promotes cardiac microvascular angiogenesis after hypoxia/reoxygenation in vitro. Free Radic Biol Med 2014; 69:278-88. [PMID: 24480752 DOI: 10.1016/j.freeradbiomed.2014.01.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/29/2013] [Accepted: 01/21/2014] [Indexed: 02/06/2023]
Abstract
Microvascular endothelial cell dysfunction plays a key role in myocardial ischemia/reperfusion (I/R) injury, wherein reactive oxygen species (ROS)-dependent signaling is intensively involved. However, the roles of the various ROS sources remain unclear. This study sought to investigate the role of NADPH oxidase 4 (Nox4) in the cardiac microvascular endothelium in response to I/R injury. Adult rat cardiac microvascular endothelial cells (CMECs) were isolated and subjected to hypoxia/reoxygenation (H/R). Our results showed that Nox4 was highly expressed in CMECs, was significantly increased at both mRNA and protein levels after H/R injury, and contributed to H/R-stimulated increase in Nox activity and ROS generation. Downregulation of Nox4 by small interfering RNA transfection did not affect cell viability or ROS production under normoxia, but exacerbated H/R injury as evidenced by increased apoptosis and inhibited cell survival, migration, and angiogenesis after H/R. Nox4 inhibition also increased prolyl hydroxylase 2 (PHD2) expression and blocked H/R-induced increases in HIF-1α and VEGF expression. Pretreatment with DMOG, a specific competitive PHD inhibitor, upregulated HIF-1α and VEGF expression and significantly reversed Nox4 knockdown-induced injury. However, Nox2 was scarcely expressed and played a minimal role in CMEC survival and angiogenesis after H/R, though a modest upregulation of Nox2 was observed. In conclusion, this study demonstrated a previously unrecognized protective role of Nox4, a ROS-generating enzyme and the major Nox isoform in CMECs, against H/R injury by inhibiting apoptosis and promoting migration and angiogenesis via a PHD2-dependent upregulation of HIF-1/VEGF proangiogenic signaling.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhibo Hong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Chao Zeng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qiujun Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Haichang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
411
|
Physiological and pathological functions of NADPH oxidases during myocardial ischemia-reperfusion. Trends Cardiovasc Med 2014; 24:202-5. [PMID: 24880746 DOI: 10.1016/j.tcm.2014.03.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress, the presence of reactive oxygen species (ROS) in excess of the antioxidant capacity in the heart induces myocardial damage, accumulation of which leads to ischemic heart disease and heart failure. NADPH oxidase (Nox) 2 and 4 are the major sources of O2- and H2O2 in the heart and play a crucial role in the regulation of growth and death in cardiomyocytes. Both Nox2 and Nox4 are upregulated in response to ischemia-reperfusion (I/R), thereby contributing to ROS production and consequent myocardial injury. Suppression of either one of them can reduce ROS and I/R injury in the heart. Importantly, however, a minimum level of ROS production by either Nox2 or Nox4 is essential for the activation of HIF-1α and inhibition of PPARα during I/R, such that combined suppression of both Nox2 and Nox4 exacerbates myocardial I/R injury. Thus, either excessive activation or suppression of Noxs below physiological levels can induce cardiac injury. Here we discuss both detrimental and salutary functions of Nox isoforms during myocardial I/R.
Collapse
|
412
|
DiStasi MR, Unthank JL, Miller SJ. Nox2 and p47(phox) modulate compensatory growth of primary collateral arteries. Am J Physiol Heart Circ Physiol 2014; 306:H1435-43. [PMID: 24633549 DOI: 10.1152/ajpheart.00828.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of NADPH oxidase (Nox) in both the promotion and impairment of compensatory collateral growth remains controversial because the specific Nox and reactive oxygen species involved are unclear. The aim of this study was to identify the primary Nox and reactive oxygen species associated with early stage compensatory collateral growth in young, healthy animals. Ligation of the feed arteries that form primary collateral pathways in rat mesentery and mouse hindlimb was used to assess the role of Nox during collateral growth. Changes in mesenteric collateral artery Nox mRNA expression determined by real-time PCR at 1, 3, and 7 days relative to same-animal control arteries suggested a role for Nox subunits Nox2 and p47(phox). Administration of apocynin or Nox2ds-tat suppressed collateral growth in both rat and mouse models, suggesting the Nox2/p47(phox) interaction was involved. Functional significance of p47(phox) expression was assessed by evaluation of collateral growth in rats administered p47(phox) small interfering RNA and in p47(phox-/-) mice. Diameter measurements of collateral mesenteric and gracilis arteries at 7 and 14 days, respectively, indicated no significant collateral growth compared with control rats or C57BL/6 mice. Chronic polyethylene glycol-conjugated catalase administration significantly suppressed collateral development in rats and mice, implying a requirement for H2O2. Taken together, these results suggest that Nox2, modulated at least in part by p47(phox), mediates early stage compensatory collateral development via a process dependent upon peroxide generation. These results have important implications for the use of antioxidants and the development of therapies for peripheral arterial disease.
Collapse
Affiliation(s)
- Matthew R DiStasi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joseph L Unthank
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana; and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Steven J Miller
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana; and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
413
|
Peshavariya HM, Chan EC, Liu GS, Jiang F, Dusting GJ. Transforming growth factor-β1 requires NADPH oxidase 4 for angiogenesis in vitro and in vivo. J Cell Mol Med 2014; 18:1172-83. [PMID: 24629065 PMCID: PMC4508156 DOI: 10.1111/jcmm.12263] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/28/2014] [Indexed: 01/25/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a key physiological event in organ development and tissue responses to hypoxia but is also involved in pathophysiologies such as tumour growth and retinopathies. Understanding the molecular mechanisms involved is important to design strategies for therapeutic intervention. One important regulator of angiogenesis is transforming growth factor-β1 (TGF-β1). In addition, reactive oxygen species (ROS) and the ROS-forming NADPH oxidase type 4 (Nox4) have been implicated as additional regulators such as during hypoxia. Here, we show that both processes are indeed mechanistically linked. TGF-β1-stimulated Nox4 expression and ROS formation in endothelial cells. In cells from Nox4-deficient mice, TGF-β1-induced cell proliferation, migration and tube formation were abolished. In vivo, TGF-β1 stimulated growth of blood vessels into sponges implanted subcutaneously, and this angiogenesis was markedly reduced in Nox4 knockout mice. Thus, endothelial cells are regulated by a TGF-β1 signalling pathway involving Nox4-derived ROS to promote angiogenesis. In order to abrogate pathological angiogenesis triggered by a multitude of factors, such as TGF-β1 and hypoxia, Nox4 may thus be an ideal therapeutic target.
Collapse
Affiliation(s)
- Hitesh M Peshavariya
- Centre for Eye Research Australia, Department of Ophthalmology, University of Melbourne, East Melbourne, Victoria, Australia; O'Brien Institute, Fitzroy, Victoria, Australia
| | | | | | | | | |
Collapse
|
414
|
|
415
|
Bretón-Romero R, Lamas S. Hydrogen peroxide signaling in vascular endothelial cells. Redox Biol 2014; 2:529-34. [PMID: 24634835 PMCID: PMC3953958 DOI: 10.1016/j.redox.2014.02.005] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 02/20/2014] [Indexed: 12/27/2022] Open
Abstract
Redox signaling is implicated in different physiological and pathological events in the vasculature. Among the different reactive oxygen species, hydrogen peroxide (H2O2) is a very good candidate to perform functions as an intracellular messenger in the regulation of several biological events. In this review, we summarize the main physiological sources of H2O2 in the endothelium and the molecular mechanisms by which it is able to act as a signaling mediator in the vasculature.
Collapse
Affiliation(s)
- Rosa Bretón-Romero
- Centro de Biología Molecular 'Severo Ochoa' CSIC-UAM, Campus Universidad Autónoma, Nicolás Cabrera 1, Madrid E-28049, Spain
| | - Santiago Lamas
- Centro de Biología Molecular 'Severo Ochoa' CSIC-UAM, Campus Universidad Autónoma, Nicolás Cabrera 1, Madrid E-28049, Spain
| |
Collapse
|
416
|
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
|
417
|
Brandes RP, Weissmann N, Schröder K. Nox family NADPH oxidases in mechano-transduction: mechanisms and consequences. Antioxid Redox Signal 2014; 20:887-98. [PMID: 23682993 PMCID: PMC3924808 DOI: 10.1089/ars.2013.5414] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE The majority of cells in a multi-cellular organism are continuously exposed to ever-changing physical forces. Mechano-transduction links these events to appropriate reactions of the cells involving stimulation of signaling cascades, reorganization of the cytoskeleton and alteration of gene expression. RECENT ADVANCES Mechano-transduction alters the cellular redox balance and the formation of reactive oxygen species (ROS). Nicotine amide adenine dinucleotide reduced form (NADPH) oxidases of the Nox family are prominent ROS generators and thus, contribute to this stress-induced ROS formation. CRITICAL ISSUES Different types and patterns of mechano-stress lead to Nox-dependent ROS formation and Nox-mediated ROS formation contributes to cellular responses and adaptation to physical forces. Thereby, Nox enzymes can mediate vascular protection during physiological mechano-stress. Despite this, over-activation and induction of Nox enzymes and a subsequent substantial increase in ROS formation also promotes oxidative stress in pathological situations like disturbed blood flow or extensive stretch. FUTURE DIRECTIONS Individual protein targets of Nox-mediated redox-signaling will be identified to better understand the specificity of Nox-dependent ROS signaling in mechano-transduction. Nox-inhibitors will be tested to reduce cellular activation in response to mechano-stimuli.
Collapse
Affiliation(s)
- Ralf P Brandes
- 1 Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt , Frankfurt am Main, Germany
| | | | | |
Collapse
|
418
|
Brandes RP, Weissmann N, Schröder K. Redox-mediated signal transduction by cardiovascular Nox NADPH oxidases. J Mol Cell Cardiol 2014; 73:70-9. [PMID: 24560815 DOI: 10.1016/j.yjmcc.2014.02.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 11/30/2022]
Abstract
The only known function of the Nox family of NADPH oxidases is the production of reactive oxygen species (ROS). Some Nox enzymes show high tissue-specific expression and the ROS locally produced are required for synthesis of hormones or tissue components. In the cardiovascular system, Nox enzymes are low abundant and function as redox-modulators. By reacting with thiols, nitric oxide (NO) or trace metals, Nox-derived ROS elicit a plethora of cellular responses required for physiological growth factor signaling and the induction and adaptation to pathological processes. The interactions of Nox-derived ROS with signaling elements in the cardiovascular system are highly diverse and will be detailed in this article, which is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
Affiliation(s)
- Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Germany.
| | - Norbert Weissmann
- Giessen University Lung Center, Justus-Liebig-Universität, Gießen, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Germany
| |
Collapse
|
419
|
The Proatherogenic Effect of Chronic Nitric Oxide Synthesis Inhibition in ApoE-Null Mice Is Dependent on the Presence of PPAR α. PPAR Res 2014; 2014:124583. [PMID: 24587793 PMCID: PMC3920724 DOI: 10.1155/2014/124583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 10/14/2013] [Accepted: 10/18/2013] [Indexed: 12/17/2022] Open
Abstract
Inhibition of endothelial nitric oxide synthase (eNOS) accelerates atherosclerosis in ApoE-null mice by impairing the balance between angiotensin II (AII) and NO. Our previous data suggested a role for PPARα in the deleterious effect of the renin-angiotensin system (RAS). We tested the hypothesis that ApoE-null mice lacking PPARα (DKO mice) would be resistant to the proatherogenic effect of NOS inhibition. DKO mice fed a Western diet were immune to the 23% worsening in aortic sinus plaque area seen in the ApoE-null animals under 12 weeks of NOS inhibition with a subpressor dose of L-NAME, P = 0.002. This was accompanied by a doubling of reactive oxygen species (ROS-) generating aortic NADPH oxidase activity (a target of AII, which paralleled Nox1 expression) and by a 10-fold excess of the proatherogenic iNOS, P < 0.01. L-NAME also caused a doubling of aortic renin and angiotensinogen mRNA level in the ApoE-null mice but not in the DKO, and it upregulated eNOS in the DKO mice only. These data suggest that, in the ApoE-null mouse, PPARα contributes to the proatherogenic effect of unopposed RAS/AII action induced by L-NAME, an effect which is associated with Nox1 and iNOS induction, and is independent of blood pressure and serum lipids.
Collapse
|
420
|
Schulz E, Wenzel P, Münzel T, Daiber A. Mitochondrial redox signaling: Interaction of mitochondrial reactive oxygen species with other sources of oxidative stress. Antioxid Redox Signal 2014; 20:308-24. [PMID: 22657349 PMCID: PMC3887453 DOI: 10.1089/ars.2012.4609] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Oxidative stress is a well established hallmark of cardiovascular disease and there is strong evidence for a causal role of reactive oxygen and nitrogen species (RONS) therein. RECENT ADVANCES Improvement of cardiovascular complications by genetic deletion of RONS producing enzymes and overexpression of RONS degrading enzymes proved the involvement of these species in cardiovascular disease at a molecular level. Vice versa, overexpression of RONS producing enzymes as well as deletion of antioxidant enzymes was demonstrated to aggravate cardiovascular complications. CRITICAL ISSUES With the present overview we present and discuss different pathways how mitochondrial RONS interact (crosstalk) with other sources of oxidative stress, namely NADPH oxidases, xanthine oxidase and an uncoupled nitric oxide synthase. The potential mechanisms of how this crosstalk proceeds are discussed in detail. Several examples from the literature are summarized (including hypoxia, angiotensin II mediated vascular dysfunction, cellular starvation, nitrate tolerance, aging, hyperglycemia, β-amyloid stress and others) and the underlying mechanisms are put together to a more general concept of redox-based activation of different sources of RONS via enzyme-specific "redox switches". Mitochondria play a key role in this concept providing redox triggers for oxidative damage in the cardiovascular system but also act as amplifiers to increase the burden of oxidative stress. FUTURE DIRECTIONS Based on these considerations, the characterization of the role of mitochondrial RONS formation in cardiac disease as well as inflammatory processes but also the role of mitochondria as potential therapeutic targets in these pathophysiological states should be addressed in more detail in the future.
Collapse
Affiliation(s)
- Eberhard Schulz
- 1 2nd Medical Clinic, Molecular Cardiology, Medical Center of the Johannes Gutenberg University , Mainz, Germany
| | | | | | | |
Collapse
|
421
|
Kaludercic N, Mialet-Perez J, Paolocci N, Parini A, Di Lisa F. Monoamine oxidases as sources of oxidants in the heart. J Mol Cell Cardiol 2014; 73:34-42. [PMID: 24412580 DOI: 10.1016/j.yjmcc.2013.12.032] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 12/28/2013] [Accepted: 12/31/2013] [Indexed: 01/22/2023]
Abstract
Oxidative stress can be generated at several sites within the mitochondria. Among these, monoamine oxidase (MAO) has been described as a prominent source. MAOs are mitochondrial flavoenzymes responsible for the oxidative deamination of catecholamines, serotonin and biogenic amines, and during this process they generate H2O2 and aldehyde intermediates. The role of MAO in cardiovascular pathophysiology has only recently gathered some attention since it has been demonstrated that both H2O2 and aldehydes may target mitochondrial function and consequently affect function and viability of the myocardium. In the present review, we will discuss the role of MAO in catecholamine and serotonin clearance and cycling in relation to cardiac structure and function. The relevant contribution of each MAO isoform (MAO-A or -B) will be discussed in relation to mitochondrial dysfunction and myocardial injury. Finally, we will examine both beneficial effects of their pharmacological or genetic inhibition along with potential adverse effects observed at baseline in MAO knockout mice, as well as the deleterious effects following their over-expression specifically at cardiomyocyte level. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
Affiliation(s)
- Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padua, Italy
| | - Jeanne Mialet-Perez
- INSERM UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; Paul Sabatier University, Toulouse, France
| | | | - Angelo Parini
- INSERM UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; Paul Sabatier University, Toulouse, France
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR), Padua, Italy; Department of Biomedical Sciences, University of Padua, Italy.
| |
Collapse
|
422
|
Crowley SD. The cooperative roles of inflammation and oxidative stress in the pathogenesis of hypertension. Antioxid Redox Signal 2014; 20:102-20. [PMID: 23472597 PMCID: PMC3880899 DOI: 10.1089/ars.2013.5258] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
SIGNIFICANCE Innate and adaptive immunity play fundamental roles in the development of hypertension and its complications. As effectors of the cell-mediated immune response, myeloid cells and T lymphocytes protect the host organism from infection by attacking foreign intruders with bursts of reactive oxygen species (ROS). RECENT ADVANCES While these ROS may help to preserve the vascular tone and thereby protect against circulatory collapse in the face of overwhelming infection, aberrant elaboration of ROS triggered by immune cells in the absence of a hemodynamic insult can lead to pathologic increases in blood pressure. Conversely, misdirected oxidative stress in cardiovascular control organs, including the vasculature, the kidney, and the nervous system potentiates inflammatory responses, augmenting blood pressure elevation and inciting target organ damage. CRITICAL ISSUES Inflammation and oxidative stress thereby act as cooperative and synergistic partners in the pathogenesis of hypertension. FUTURE DIRECTIONS Pharmacologic interventions for hypertensive patients will need to exploit this robust bidirectional relationship between ROS generation and immune activation in cardiovascular control organs to maximize therapeutic benefit, while limiting off-target side effects.
Collapse
Affiliation(s)
- Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers , Durham, North Carolina
| |
Collapse
|
423
|
Abstract
General use and popularity of over-the-counter supplemental antioxidants have rapidly spread all over the world and are believed to promote cardiovascular health and wellbeing. However, there is a paucity of information and lack of proof that physiological and above-physiological levels of oxidants do harm at the cellular and organismal levels. Instead, several reports demonstrated that reduction in Reactive Oxygen Species (ROS) did not improve vascular function. Interestingly, recent studies show that increased ROS levels play protective role in vascular endothelium and may improve coronary endothelial function. In the current review, we introduce the concept that increased ROS levels, often seen in association with cardiovascular disease, probably is an endothelial-way or ‘oxidative response’ to cope with vascular pathology.
Collapse
Affiliation(s)
- M Ruhul Abid
- Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Frank W Sellke
- Cardiovascular Research Center, Department of Surgery, Rhode Island Hospital, Brown University Warren Alpert Medical School, Providence, RI, USA
| |
Collapse
|
424
|
Williams CR, Gooch JL. Calcineurin Aβ regulates NADPH oxidase (Nox) expression and activity via nuclear factor of activated T cells (NFAT) in response to high glucose. J Biol Chem 2013; 289:4896-905. [PMID: 24371139 DOI: 10.1074/jbc.m113.514869] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hypertrophy is an adaptive response that enables organs to appropriately meet increased functional demands. Previously, we reported that calcineurin (Cn) is required for glomerular and whole kidney hypertrophy in diabetic rodents (Gooch, J. L., Barnes, J. L., Garcia, S., and Abboud, H. E. (2003). Calcineurin is activated in diabetes and is required for glomerular hypertrophy and ECM accumulation. Am. J. Physiol. Renal Physiol. 284, F144-F154; Reddy, R. N., Knotts, T. L., Roberts, B. R., Molkentin, J. D., Price, S. R., and Gooch, J. L. (2011). Calcineurin Aβ is required for hypertrophy but not matrix expansion in the diabetic kidney. J. Cell Mol. Med. 15, 414-422). Because studies have also implicated the reactive oxygen species-generating enzymes NADPH oxidases (Nox) in diabetic kidney responses, we tested the hypothesis that Nox and Cn cooperate in a common signaling pathway. First, we examined the role of the two main isoforms of Cn in hypertrophic signaling. Using primary kidney cells lacking a catalytic subunit of Cn (CnAα(-/-) or CnAβ(-/-)), we found that high glucose selectively activates CnAβ, whereas CnAα is constitutively active. Furthermore, CnAβ but not CnAα mediates hypertrophy. Next, we found that chronic reactive oxygen species generation in response to high glucose is attenuated in CnAβ(-/-) cells, suggesting that Cn is upstream of Nox. Consistent with this, loss of CnAβ reduces basal expression and blocks high glucose induction of Nox2 and Nox4. Inhibition of nuclear factor of activated T cells (NFAT), a CnAβ-regulated transcription factor, decreases Nox2 and Nox4 expression, whereas NFAT overexpression increases Nox2 and Nox4, indicating that the CnAβ/NFAT pathway modulates Nox. These data reveal that the CnAβ/NFAT pathway regulates Nox and plays an important role in high glucose-mediated hypertrophic responses in the kidney.
Collapse
Affiliation(s)
- Clintoria R Williams
- From the Atlanta Veterans Administration Medical Center, Atlanta, Georgia 30033 and
| | | |
Collapse
|
425
|
Veit F, Pak O, Egemnazarov B, Roth M, Kosanovic D, Seimetz M, Sommer N, Ghofrani HA, Seeger W, Grimminger F, Brandes RP, Schermuly RT, Weissmann N. Function of NADPH oxidase 1 in pulmonary arterial smooth muscle cells after monocrotaline-induced pulmonary vascular remodeling. Antioxid Redox Signal 2013; 19:2213-31. [PMID: 23706097 DOI: 10.1089/ars.2012.4904] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AIMS Chronic hypoxia induces pulmonary hypertension (PH) that is concomitant with pulmonary vascular remodeling. Reactive oxygen species (ROS) are thought to play a major role in this. Recent findings suggest that ROS production by NADPH oxidase 4 (Nox4) is important in this remodeling. We investigated whether ROS production by Nox is also important in an inflammatory model of monocrotaline (MCT)-induced PH. We examined ROS production, their possible sources, and their impact on the function of pulmonary arterial smooth muscle cells (PASMC) isolated from MCT-treated and healthy rats. RESULTS MCT-PASMC showed increased intracellular superoxide production, migration, and proliferation compared with healthy controls due to increased Nox1 expression. A comparison of PASMC from MCT- and nontreated rats revealed an up-regulation of Sod2, Nrf2, cyclin D1, and matrix metalloproteinase-9 (MMP-9) as well as an increased phosphorylation of cofilin and extracellular signal-regulated kinases (Erk). Expression of Sod2, Nrf2, and cyclin D1 and phosphorylation of cofilin and Erk were Nox1 dependent. INNOVATION The role of ROS in PH is not fully understood. Mitochondria and Nox have been suggested as sources of altered ROS generation in PH, yet it remains unclear whether increased or decreased ROS contributes to the development of PH. Our studies provide evidence that for different triggers of PH, different Nox isoforms regulate proliferation and migration of PASMC. CONCLUSION In contrast to hypoxia-induced PH, Nox1 but not Nox4 is responsible for pathophysiological proliferation and migration of PASMC in an inflammatory model of MCT-induced PH via increased superoxide production. Thus, different Nox isoforms may be targeted in different forms of PH.
Collapse
Affiliation(s)
- Florian Veit
- 1 Excellencecluster Cardio-Pulmonary System (ECCPS), German Lung Center (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen , Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
426
|
|
427
|
Abstract
Despite the damaging effect on tissues at a high concentration, it has been gradually established that oxidative stress plays a positive role during angiogenesis. In adults, physiological or pathological angiogenesis is initiated by tissue demands for oxygen and nutrients, resulting in a hypoxia/reoxygenation cycle, which, in turn promotes the formation of reactive oxygen species (ROS). The ROS can be generated either endogenously, through mitochondrial electron transport chain reactions and nicotinamide adenine dinucleotide phosphate oxidase, or exogenously, resulting from exposure to environmental agents, such as ultraviolet or ionizing radiation. In many conditions, ROS promotes angiogenesis, either directly or via the generation of active oxidation products, including peroxidized lipids. The latter lipid metabolites are generated in excess during atherosclerosis, thereby linking atherogenic processes and pathological angiogenesis. Although the main mechanism of oxidative stress-induced angiogenesis involves hypoxia-inducible factor/vascular endothelial growth factor (VEGF) signaling, recent studies have identified several pathways that are VEGF-independent. This review aims to provide a summary of the past and present views on the role of oxidative stress as a mediator and modulator of angiogenesis, and to highlight newly identified mechanisms.
Collapse
|
428
|
Baltanás A, Miguel-Carrasco JL, San José G, Cebrián C, Moreno MU, Dotor J, Borrás-Cuesta F, López B, González A, Díez J, Fortuño A, Zalba G. A synthetic peptide from transforming growth factor-β₁ type III receptor inhibits NADPH oxidase and prevents oxidative stress in the kidney of spontaneously hypertensive rats. Antioxid Redox Signal 2013; 19:1607-18. [PMID: 23350688 DOI: 10.1089/ars.2012.4653] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS The NADPH oxidases constitute a major source of superoxide anion (·O2(-)) in hypertension. Several studies suggest an important role of NADPH oxidases in different effects mediated by transforming growth factor-β₁ (TGF-β₁). We investigated whether a chronic treatment with P144, a peptide synthesized from type III TGF-β₁ receptor, inhibited NADPH oxidases in the renal cortex of spontaneously hypertensive rats (SHR). RESULTS Here, we show that chronic administration of P144 significantly reduced the NADPH oxidase expression and activity as well as the oxidative stress observed in control vehicle-treated SHR (V-SHR). In addition, P144 was also able to reduce the significant increase in the renal fibrosis and in mRNA expression of different components of collagen metabolism, as well as in the levels of connective tissue growth factor observed in V-SHR. Finally, TGF-β₁-stimulated NRK52E exhibited a significant increase in NADPH oxidase expression and activity as well as a TGF-β₁-dependent intracellular pathway that were inhibited in the presence of P144. INNOVATION Our experimental evidence suggests that reversing oxidative stress may be therapeutically useful in preventing fibrosis-associated renal damage. We show here that (i) the TGF-β₁-NADPH oxidases axis is crucial in the development of fibrosis in an experimental hypertensive renal disease animal model, and (ii) the use of P144 reverses TGF-β₁-dependent NADPH oxidase activity; thus, P144 may be considered a novel therapeutic tool in kidney disease associated with hypertension. CONCLUSION We demonstrate that P144 inhibits NADPH oxidases and prevents oxidative stress in kidneys from hypertensive rats. Our data also suggest that these effects are associated with the renal antifibrotic effect of P144.
Collapse
Affiliation(s)
- Ana Baltanás
- 1 Division of Cardiovascular Sciences, Centre for Applied Medical Research, University of Navarra , Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
429
|
Anea CB, Zhang M, Chen F, Ali MI, Hart CMM, Stepp DW, Kovalenkov YO, Merloiu AM, Pati P, Fulton D, Rudic RD. Circadian clock control of Nox4 and reactive oxygen species in the vasculature. PLoS One 2013; 8:e78626. [PMID: 24205282 PMCID: PMC3808297 DOI: 10.1371/journal.pone.0078626] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 09/14/2013] [Indexed: 12/30/2022] Open
Abstract
Recent studies have shown that circadian clock disruption is associated with pathological remodeling in the arterial structure and vascular stiffness. Moreover, chronic circadian disruption is associated with dysfunction in endothelial responses and signaling. Reactive oxygen species have emerged as key regulators in vascular pathology. Previously, we have demonstrated that circadian clock dysfunction exacerbates superoxide production through eNOS uncoupling. To date, the impact of circadian clock mutation on vascular NADPH oxidase expression and function is not known. The goal in the current study was to determine if the circadian clock controls vascular Nox4 expression and hydrogen peroxide formation in arteries, particularly in endothelial and vascular smooth muscle cells. In aorta, there was an increase in hydrogen peroxide and Nox4 expression in mice with a dysfunctional circadian rhythm (Bmal1-KO mice). In addition, the Nox4 gene promoter is activated by the core circadian transcription factors. Lastly, in synchronized cultured human endothelial cells, Nox4 gene expression exhibited rhythmic oscillations. These data reveal that the circadian clock plays an important role in the control of Nox4 and disruption of the clock leads to subsequent production of reaction oxygen species.
Collapse
Affiliation(s)
- Ciprian B. Anea
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Maoxiang Zhang
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Feng Chen
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - M. Irfan Ali
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - C. Michael M. Hart
- Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Center, Atlanta, Georgia, United States of America
| | - David W. Stepp
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Physiology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Yevgeniy O. Kovalenkov
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - Ana-Maria Merloiu
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Paramita Pati
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - David Fulton
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - R. Daniel Rudic
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
430
|
Shafique E, Choy WC, Liu Y, Feng J, Cordeiro B, Lyra A, Arafah M, Yassin-Kassab A, Zanetti AVD, Clements RT, Bianchi C, Benjamin LE, Sellke FW, Abid MR. Oxidative stress improves coronary endothelial function through activation of the pro-survival kinase AMPK. Aging (Albany NY) 2013; 5:515-30. [PMID: 24018842 PMCID: PMC3765580 DOI: 10.18632/aging.100569] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Age-associated decline in cardiovascular function is believed to occur from the deleterious effects of reactive oxygen species (ROS). However, failure of recent clinical trials using antioxidants in patients with cardiovascular disease, and the recent findings showing paradoxical role for NADPH oxidase-derived ROS in endothelial function challenge this long-held notion against ROS. Here, we examine the effects of endothelium-specific conditional increase in ROS on coronary endothelial function. We have generated a novel binary (Tet-ON/OFF) conditional transgenic mouse (Tet-Nox2:VE-Cad-tTA) that induces endothelial cell (EC)-specific overexpression of Nox2/gp91 (NADPH oxidase) and 1.8±0.42-fold increase in EC-ROS upon tetracycline withdrawal (Tet-OFF). We examined ROS effects on EC signaling and function. First, we demonstrate that endothelium-dependent coronary vasodilation was significantly improved in Tet-OFF Nox2 compared to Tet-ON (control) littermates. Using EC isolated from mouse heart, we show that endogenous ROS increased eNOS activation and nitric oxide (NO) synthesis through activation of the survival kinase AMPK. Coronary vasodilation in Tet-OFF Nox2 animals was CaMKKβ-AMPK-dependent. Finally, we demonstrate that AMPK activation induced autophagy and thus, protected ECs from oxidant-induced cell death. Together, these findings suggest that increased ROS levels, often associated with cardiovascular conditions in advanced age, play a protective role in endothelial homeostasis by inducing AMPK-eNOS axis.
Collapse
Affiliation(s)
- Ehtesham Shafique
- Cardiovascular Research Center, Division of Cardiothoracic Surgery, Department of Surgery, Rhode Island Hospital, Providence, RI 02903, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
431
|
Sag CM, Wagner S, Maier LS. Role of oxidants on calcium and sodium movement in healthy and diseased cardiac myocytes. Free Radic Biol Med 2013; 63:338-49. [PMID: 23732518 DOI: 10.1016/j.freeradbiomed.2013.05.035] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 12/19/2022]
Abstract
In this review article we give an overview of current knowledge with respect to redox-sensitive alterations in Na(+) and Ca(2+) handling in the heart. In particular, we focus on redox-activated protein kinases including cAMP-dependent protein kinase A (PKA), protein kinase C (PKC), and Ca/calmodulin-dependent protein kinase II (CaMKII), as well as on redox-regulated downstream targets such as Na(+) and Ca(2+) transporters and channels. We highlight the pathological and physiological relevance of reactive oxygen species and some of its sources (such as NADPH oxidases, NOXes) for excitation-contraction coupling (ECC). A short outlook with respect to the clinical relevance of redox-dependent Na(+) and Ca(2+) imbalance will be given.
Collapse
Affiliation(s)
- Can M Sag
- Cardiovascular Division, The James Black Centre, King's College London, UK
| | | | | |
Collapse
|
432
|
Postprandial lipoproteins and the molecular regulation of vascular homeostasis. Prog Lipid Res 2013; 52:446-64. [DOI: 10.1016/j.plipres.2013.06.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 12/17/2022]
|
433
|
Sciarretta S, Zhai P, Shao D, Zablocki D, Nagarajan N, Terada LS, Volpe M, Sadoshima J. Activation of NADPH oxidase 4 in the endoplasmic reticulum promotes cardiomyocyte autophagy and survival during energy stress through the protein kinase RNA-activated-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/activating transcription factor 4 pathway. Circ Res 2013; 113:1253-64. [PMID: 24081881 DOI: 10.1161/circresaha.113.301787] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE Autophagy is an essential survival mechanism during energy stress in the heart. Oxidative stress is activated by energy stress, but its role in mediating autophagy is poorly understood. NADPH oxidase (Nox) 4 is an enzyme that generates reactive oxygen species (ROS) at intracellular membranes. Whether Nox4 acts as a sensor of energy stress to mediate activation of autophagy is unknown. OBJECTIVE We investigated whether Nox4 is involved in the regulation of autophagy and cell survival during energy stress in cardiomyocytes. METHODS AND RESULTS Production of ROS in cardiomyocytes was increased during glucose deprivation (GD) in a Nox4-dependent manner. Protein levels and the ROS-producing activity of Nox4 were increased in the endoplasmic reticulum (ER), but not in mitochondria, in response to GD. Selective knockdown of Nox4, but not Nox2, or selective reduction of ROS in the ER with ER-targeted catalase, but not mitochondria-targeted perioxiredoxin 3, abrogated GD-induced autophagy. Nox4 promoted autophagy during GD through activation of the protein kinase RNA-activated-like ER kinase pathway by suppression of prolyl hydroxylase 4. The decrease in cell survival during GD in the presence of Nox4 knockdown was rescued by reactivation of autophagy by Atg7 overexpression, indicating that the effect of Nox4 on cell survival is critically mediated through regulation of autophagy. Nox4 was activated during fasting and prolonged ischemia in the mouse heart, where Nox4 is also required for autophagy activation and cardioprotection. CONCLUSIONS Nox4 critically mediates autophagy in response to energy stress in cardiomyocytes by eliciting ROS in the ER and stimulating the protein kinase RNA-activated-like ER kinase signaling pathway.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.S., P.Z., D.S., D.Z., N.N., J.S.); IRCCS Neuromed, Pozzilli (IS), Italy (S.S., M.V.); Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas (L.S.T.); and Department of Clinical/Molecular Medicine, School of Medicine and Psychology, Sapienza University, Rome, Italy (M.V.)
| | | | | | | | | | | | | | | |
Collapse
|
434
|
Majzunova M, Dovinova I, Barancik M, Chan JYH. Redox signaling in pathophysiology of hypertension. J Biomed Sci 2013; 20:69. [PMID: 24047403 PMCID: PMC3815233 DOI: 10.1186/1423-0127-20-69] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/14/2013] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are products of normal cellular metabolism and derive from various sources in different cellular compartments. Oxidative stress resultant from imbalance between ROS generation and antioxidant defense mechanisms is important in pathogenesis of cardiovascular diseases, such as hypertension, heart failure, atherosclerosis, diabetes, and cardiac hypertrophy. In this review we focus on hypertension and address sources of cellular ROS generation, mechanisms involved in regulation of radical homeostasis, superoxide dismutase isoforms in pathophysiology of hypertension; as well as radical intracellular signaling and phosphorylation processes in proteins of the affected cardiovascular tissues. Finally, we discuss the transcriptional factors involved in redox-sensitive gene transcription and antioxidant response, as well as their roles in hypertension.
Collapse
Affiliation(s)
- Miroslava Majzunova
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Sienkiewiczova 1, 813 71 Bratislava, Slovakia.
| | | | | | | |
Collapse
|
435
|
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: 113] [Impact Index Per Article: 10.3] [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
|
436
|
Roos CM, Hagler M, Zhang B, Oehler EA, Arghami A, Miller JD. Transcriptional and phenotypic changes in aorta and aortic valve with aging and MnSOD deficiency in mice. Am J Physiol Heart Circ Physiol 2013; 305:H1428-39. [PMID: 23997094 DOI: 10.1152/ajpheart.00735.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The purpose of this study was to characterize changes in antioxidant and age-related gene expression in aorta and aortic valve with aging, and test the hypothesis that increased mitochondrial oxidative stress accelerates age-related endothelial and aortic valve dysfunction. Wild-type (MnSOD(+/+)) and manganese SOD heterozygous haploinsufficient (MnSOD(+/-)) mice were studied at 3 and 18 mo of age. In aorta from wild-type mice, antioxidant expression was preserved, although there were age-associated increases in Nox2 expression. Haploinsufficiency of MnSOD did not alter antioxidant expression in aorta, but increased expression of Nox2. When compared with that of aorta, age-associated reductions in antioxidant expression were larger in aortic valves from wild-type and MnSOD haploinsufficient mice, although Nox2 expression was unchanged. Similarly, sirtuin expression was relatively well-preserved in aorta from both genotypes, whereas expression of SIRT1, SIRT2, SIRT3, SIRT4, and SIRT6 were significantly reduced in the aortic valve. Expression of p16(ink4a), a marker of cellular senescence, was profoundly increased in both aorta and aortic valve from MnSOD(+/+) and MnSOD(+/-) mice. Functionally, we observed comparable age-associated reductions in endothelial function in aorta from both MnSOD(+/+) and MnSOD(+/-) mice. Interestingly, inhibition of NAD(P)H oxidase with apocynin or gp91ds-tat improved endothelial function in MnSOD(+/+) mice but significantly impaired endothelial function in MnSOD(+/-) mice at both ages. Aortic valve function was not impaired by aging or MnSOD haploinsufficiency. Changes in antioxidant and sirtuin gene expression with aging differ dramatically between aorta and aortic valve. Furthermore, although MnSOD does not result in overt cardiovascular dysfunction with aging, compensatory transcriptional responses to MnSOD deficiency appear to be tissue specific.
Collapse
Affiliation(s)
- Carolyn M Roos
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | |
Collapse
|
437
|
Sena CM, Pereira AM, Seiça R. Endothelial dysfunction - a major mediator of diabetic vascular disease. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2216-31. [PMID: 23994612 DOI: 10.1016/j.bbadis.2013.08.006] [Citation(s) in RCA: 534] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 07/31/2013] [Accepted: 08/20/2013] [Indexed: 12/18/2022]
Abstract
The vascular endothelium is a multifunctional organ and is critically involved in modulating vascular tone and structure. Endothelial cells produce a wide range of factors that also regulate cellular adhesion, thromboresistance, smooth muscle cell proliferation, and vessel wall inflammation. Thus, endothelial function is important for the homeostasis of the body and its dysfunction is associated with several pathophysiological conditions, including atherosclerosis, hypertension and diabetes. Patients with diabetes invariably show an impairment of endothelium-dependent vasodilation. Therefore, understanding and treating endothelial dysfunction is a major focus in the prevention of vascular complications associated with all forms of diabetes mellitus. The mechanisms of endothelial dysfunction in diabetes may point to new management strategies for the prevention of cardiovascular disease in diabetes. This review will focus on the mechanisms and therapeutics that specifically target endothelial dysfunction in the context of a diabetic setting. Mechanisms including altered glucose metabolism, impaired insulin signaling, low-grade inflammatory state, and increased reactive oxygen species generation will be discussed. The importance of developing new pharmacological approaches that upregulate endothelium-derived nitric oxide synthesis and target key vascular ROS-producing enzymes will be highlighted and new strategies that might prove clinically relevant in preventing the development and/or retarding the progression of diabetes associated vascular complications.
Collapse
Affiliation(s)
- Cristina M Sena
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Portugal; IBILI, Faculty of Medicine, University of Coimbra, Portugal.
| | | | | |
Collapse
|
438
|
Yong R, Chen XM, Shen S, Vijayaraj S, Ma Q, Pollock CA, Saad S. Plumbagin ameliorates diabetic nephropathy via interruption of pathways that include NOX4 signalling. PLoS One 2013; 8:e73428. [PMID: 23991195 PMCID: PMC3753271 DOI: 10.1371/journal.pone.0073428] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 07/30/2013] [Indexed: 02/07/2023] Open
Abstract
NADPH oxidase 4 (Nox4) is reported to be the major source of reactive oxygen species (ROS) in the kidneys during the early stages of diabetic nephropathy. It has been shown to mediate TGFβ1-induced differentiation of cardiac fibroblasts into myofibroblasts. Despite TGFβ1 being recognised as a mediator of renal fibrosis and functional decline role in diabetic nephropathy, the renal interaction between Nox 4 and TGFβ1 is not well characterised. The aim of this study was to investigate the role of Nox4 inhibition on TGFβ1-induced fibrotic responses in proximal tubular cells and in a mouse model of diabetic nephropathy. Immortalised human proximal tubular cells (HK2) were incubated with TGFβ1 ± plumbagin (an inhibitor of Nox4) or specific Nox4 siRNA. Collagen IV and fibronectin mRNA and protein expression were measured. Streptozotocin (STZ) induced diabetic C57BL/6J mice were administered plumbagin (2 mg/kg/day) or vehicle (DMSO; 50 µl/mouse) for 24 weeks. Metabolic, physiological and histological markers of nephropathy were determined. TGFβ1 increased Nox4 mRNA expression and plumbagin and Nox4 siRNA significantly inhibited TGF-β1 induced fibronectin and collagen IV expression in human HK2 cells. STZ-induced diabetic C57BL/6J mice developed physiological features of diabetic nephropathy at 24 weeks, which were reversed with concomitant plumbagin treatment. Histologically, plumbagin ameliorated diabetes induced upregulation of extracellular matrix protein expression compared to control. This study demonstrates that plumbagin ameliorates the development of diabetic nephropathy through pathways that include Nox4 signalling.
Collapse
Affiliation(s)
- Rachel Yong
- Department of Medicine, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Xin-Ming Chen
- Department of Medicine, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Sylvie Shen
- Department of Medicine, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Swarna Vijayaraj
- Department of Medicine, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Qing Ma
- Department of Medicine, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Carol A. Pollock
- Department of Medicine, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Sonia Saad
- Department of Medicine, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
439
|
Brüne B, Dehne N, Grossmann N, Jung M, Namgaladze D, Schmid T, von Knethen A, Weigert A. Redox control of inflammation in macrophages. Antioxid Redox Signal 2013; 19:595-637. [PMID: 23311665 PMCID: PMC3718318 DOI: 10.1089/ars.2012.4785] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 12/14/2012] [Accepted: 01/11/2013] [Indexed: 12/13/2022]
Abstract
Macrophages are present throughout the human body, constitute important immune effector cells, and have variable roles in a great number of pathological, but also physiological, settings. It is apparent that macrophages need to adjust their activation profile toward a steadily changing environment that requires altering their phenotype, a process known as macrophage polarization. Formation of reactive oxygen species (ROS), derived from NADPH-oxidases, mitochondria, or NO-producing enzymes, are not necessarily toxic, but rather compose a network signaling system, known as redox regulation. Formation of redox signals in classically versus alternatively activated macrophages, their action and interaction at the level of key targets, and the resulting physiology still are insufficiently understood. We review the identity, source, and biological activities of ROS produced during macrophage activation, and discuss how they shape the key transcriptional responses evoked by hypoxia-inducible transcription factors, nuclear-erythroid 2-p45-related factor 2 (Nrf2), and peroxisome proliferator-activated receptor-γ. We summarize the mechanisms how redox signals add to the process of macrophage polarization and reprogramming, how this is controlled by the interaction of macrophages with their environment, and addresses the outcome of the polarization process in health and disease. Future studies need to tackle the option whether we can use the knowledge of redox biology in macrophages to shape their mediator profile in pathophysiology, to accelerate healing in injured tissue, to fight the invading pathogens, or to eliminate settings of altered self in tumors.
Collapse
Affiliation(s)
- Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I-Pathobiochemistry, Goethe-University Frankfurt, Frankfurt, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
440
|
Lee DY, Wauquier F, Eid AA, Roman LJ, Ghosh-Choudhury G, Khazim K, Block K, Gorin Y. Nox4 NADPH oxidase mediates peroxynitrite-dependent uncoupling of endothelial nitric-oxide synthase and fibronectin expression in response to angiotensin II: role of mitochondrial reactive oxygen species. J Biol Chem 2013; 288:28668-86. [PMID: 23940049 DOI: 10.1074/jbc.m113.470971] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Activation of glomerular mesangial cells (MCs) by angiotensin II (Ang II) leads to extracellular matrix accumulation. Here, we demonstrate that, in MCs, Ang II induces endothelial nitric-oxide synthase (eNOS) uncoupling with enhanced generation of reactive oxygen species (ROS) and decreased production of NO. Ang II promotes a rapid increase in 3-nitrotyrosine formation, and uric acid attenuates Ang II-induced decrease in NO bioavailability, demonstrating that peroxynitrite mediates the effects of Ang II on eNOS dysfunction. Ang II rapidly up-regulates Nox4 protein. Inhibition of Nox4 abolishes the increase in ROS and peroxynitrite generation as well as eNOS uncoupling triggered by Ang II, indicating that Nox4 is upstream of eNOS. This pathway contributes to Ang II-mediated fibronectin accumulation in MCs. Ang II also elicits an increase in mitochondrial abundance of Nox4 protein, and the oxidase contributes to ROS production in mitochondria. Overexpression of mitochondrial manganese superoxide dismutase prevents the stimulatory effects of Ang II on mitochondrial ROS production, loss of NO availability, and MC fibronectin accumulation, whereas manganese superoxide dismutase depletion increases mitochondrial ROS, NO deficiency, and fibronectin synthesis basally and in cells exposed to Ang II. This work provides the first evidence that uncoupled eNOS is responsible for Ang II-induced MC fibronectin accumulation and identifies Nox4 and mitochondrial ROS as mediators of eNOS dysfunction. These data shed light on molecular processes underlying the oxidative signaling cascade engaged by Ang II and identify potential targets for intervention to prevent renal fibrosis.
Collapse
|
441
|
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: 13.5] [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
|
442
|
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: 1.7] [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
|
443
|
Trac D, Liu B, Pao AC, Thomas SV, Park M, Downs CA, Ma HP, Helms MN. Fulvene-5 inhibition of Nadph oxidases attenuates activation of epithelial sodium channels in A6 distal nephron cells. Am J Physiol Renal Physiol 2013; 305:F995-F1005. [PMID: 23863470 DOI: 10.1152/ajprenal.00098.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nadph oxidase 4 is an important cellular source of reactive oxygen species (ROS) generation in the kidney. Novel antioxidant drugs, such as Nox4 inhibitor compounds, are being developed. There is, however, very little experimental evidence for the biological role and regulation of Nadph oxidase isoforms in the kidney. Herein, we show that Fulvene-5 is an effective inhibitor of Nox-generated ROS and report the role of Nox isoforms in activating epithelial sodium channels (ENaC) in A6 distal nephron cells via oxidant signaling and cell stretch activation. Using single-channel patch-clamp analysis, we report that Fulvene-5 blocked the increase in ENaC activity that is typically observed with H2O2 treatment of A6 cells: average ENaC NPo values decreased from a baseline level of 1.04 ± 0.18 (means ± SE) to 0.25 ± 0.08 following Fulvene-5 treatment. H2O2 treatment failed to increase ENaC activity in the presence of Fulvene-5. Moreover, Fulvene-5 treatment of A6 cells blocked the osmotic cell stretch response of A6 cells, indicating that stretch activation of Nox-derived ROS plays an important role in ENaC regulation. Together, these findings indicate that Fulvene-5, and perhaps other classes of antioxidant inhibitors, may represent a novel class of compounds useful for the treatment of pathological disorders stemming from inappropriate ion channel activity, such as hypertension.
Collapse
Affiliation(s)
- David Trac
- Dept. of Pediatrics, 2015 Uppergate Dr., Atlanta, GA 30322.
| | | | | | | | | | | | | | | |
Collapse
|
444
|
Cubbon RM, Mercer BN, Sengupta A, Kearney MT. Importance of insulin resistance to vascular repair and regeneration. Free Radic Biol Med 2013; 60:246-63. [PMID: 23466555 DOI: 10.1016/j.freeradbiomed.2013.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/22/2013] [Accepted: 02/23/2013] [Indexed: 01/14/2023]
Abstract
Metabolic insulin resistance is apparent across a spectrum of clinical disorders, including obesity and diabetes, and is characterized by an adverse clustering of cardiovascular risk factors related to abnormal cellular responses to insulin. These disorders are becoming increasingly prevalent and represent a major global public health concern because of their association with significant increases in atherosclerosis-related mortality. Endogenous repair mechanisms are thought to retard the development of vascular disease, and a growing evidence base supports the adverse impact of the insulin-resistant phenotype upon indices of vascular repair. Beyond the impact of systemic metabolic changes, emerging data from murine studies also provide support for abnormal insulin signaling at the level of vascular cells in retarding vascular repair. Interrelated pathophysiological factors, including reduced nitric oxide bioavailability, oxidative stress, altered growth factor activity, and abnormal intracellular signaling, are likely to act in conjunction to impede vascular repair while also driving vascular damage. Understanding of these processes is shaping novel therapeutic paradigms that aim to promote vascular repair and regeneration, either by recruiting endogenous mechanisms or by the administration of cell-based therapies.
Collapse
Affiliation(s)
- Richard M Cubbon
- Multidisciplinary Cardiovascular Research Centre, LIGHT Laboratories, The University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | |
Collapse
|
445
|
Sturza A, Leisegang MS, Babelova A, Schröder K, Benkhoff S, Loot AE, Fleming I, Schulz R, Muntean DM, Brandes RP. Monoamine Oxidases Are Mediators of Endothelial Dysfunction in the Mouse Aorta. Hypertension 2013; 62:140-6. [DOI: 10.1161/hypertensionaha.113.01314] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Monoamine oxidases (MAOs) generate H
2
O
2
as a by-product of their catalytic cycle. Whether MAOs are mediators of endothelial dysfunction is unknown and was determined here in the angiotensin II and lipopolysaccharide-models of vascular dysfunction in mice. Quantitative real-time polymerase chain reaction revealed that mouse aortas contain enzymes involved in catecholamine generation and MAO-A and MAO-B mRNA. MAO-A and -B proteins could be detected by Western blot not only in mouse aortas but also in human umbilical vein endothelial cells. Ex vivo incubation of mouse aorta with recombinant MAO-A increased H
2
O
2
formation and induced endothelial dysfunction that was attenuated by polyethylene glycol-catalase and MAO inhibitors. In vivo lipopolysaccharide (8 mg/kg IP overnight) or angiotensin II (1 mg/kg per day, 2 weeks, minipump) treatment induced vascular MAO-A and -B expressions and resulted in attenuated endothelium-dependent relaxation of the aorta in response to acetylcholine. MAO inhibitors reduced the lipopolysaccharide- and angiotensin II–induced aortic reactive oxygen species formation by 50% (ferrous oxidation xylenol orange assay) and partially normalized endothelium-dependent relaxation. MAO-A and MAO-B inhibitors had an additive effect; combined application completely restored endothelium-dependent relaxation. To determine how MAO-dependent H
2
O
2
formation induces endothelial dysfunction, cyclic GMP was measured. Histamine stimulation of human umbilical vein endothelial cells to activate endothelial NO synthase resulted in an increase in cyclic GMP, which was almost abrogated by MAO-A exposure. MAO inhibition prevented this effect, suggesting that MAO-induced H
2
O
2
formation is sufficient to attenuate endothelial NO release. Thus, MAO-A and MAO-B are both expressed in the mouse aorta, induced by in vivo lipopolysaccharide and angiotensin II treatment and contribute via the generation of H
2
O
2
to endothelial dysfunction in vascular disease models.
Collapse
Affiliation(s)
- Adrian Sturza
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Matthias S. Leisegang
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Andrea Babelova
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Katrin Schröder
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Sebastian Benkhoff
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Annemarieke E. Loot
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Ingrid Fleming
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Rainer Schulz
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Danina M. Muntean
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| | - Ralf P. Brandes
- From the Institut für Kardiovaskuläre Physiologie (A.S., M.S.L., A.B., K.S., S.B., R.P.B.) and Institute for Vascular Signaling (A.E.L., I.F.), Goethe-Universität, Frankfurt, Germany; Department of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania (A.S., D.M.M.); Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.); and DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Germany (A.S., M.S.L., A.B., K.S., S.B., A.E.L
| |
Collapse
|
446
|
Sestrin 2 and AMPK connect hyperglycemia to Nox4-dependent endothelial nitric oxide synthase uncoupling and matrix protein expression. Mol Cell Biol 2013; 33:3439-60. [PMID: 23816887 DOI: 10.1128/mcb.00217-13] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mesangial matrix accumulation is an early feature of glomerular pathology in diabetes. Oxidative stress plays a critical role in hyperglycemia-induced glomerular injury. Here, we demonstrate that, in glomerular mesangial cells (MCs), endothelial nitric oxide synthase (eNOS) is uncoupled upon exposure to high glucose (HG), with enhanced generation of reactive oxygen species (ROS) and decreased production of nitric oxide. Peroxynitrite mediates the effects of HG on eNOS dysfunction. HG upregulates Nox4 protein, and inhibition of Nox4 abrogates the increase in ROS and peroxynitrite generation, as well as the eNOS uncoupling triggered by HG, demonstrating that Nox4 functions upstream from eNOS. Importantly, this pathway contributes to HG-induced MC fibronectin accumulation. Nox4-mediated eNOS dysfunction was confirmed in glomeruli of a rat model of type 1 diabetes. Sestrin 2-dependent AMP-activated protein kinase (AMPK) activation attenuates HG-induced MC fibronectin synthesis through blockade of Nox4-dependent ROS and peroxynitrite generation, with subsequent eNOS uncoupling. We also find that HG negatively regulates sestrin 2 and AMPK, thereby promoting Nox4-mediated eNOS dysfunction and increased fibronectin. These data identify a protective function for sestrin 2/AMPK and potential targets for intervention to prevent fibrotic injury in diabetes.
Collapse
|
447
|
Liu XM, Peyton KJ, Durante W. Physiological cyclic strain promotes endothelial cell survival via the induction of heme oxygenase-1. Am J Physiol Heart Circ Physiol 2013; 304:H1634-43. [PMID: 23604711 PMCID: PMC3680772 DOI: 10.1152/ajpheart.00872.2012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 04/12/2013] [Indexed: 12/14/2022]
Abstract
Endothelial cells (ECs) are constantly subjected to cyclic strain that arises from periodic change in vessel wall diameter as a result of pulsatile blood flow. Application of physiological levels of cyclic strain inhibits EC apoptosis; however, the underlying mechanism is not known. Since heme oxygenase-1 (HO-1) is a potent inhibitor of apoptosis, the present study investigated whether HO-1 contributes to the antiapoptotic action of cyclic strain. Administration of physiological cyclic strain (6% at 1 Hz) to human aortic ECs stimulated an increase in HO-1 activity, protein, and mRNA expression. The induction of HO-1 was preceded by a rise in reactive oxygen species (ROS) and Nrf2 protein expression. Cyclic strain also stimulated an increase in HO-1 promoter activity that was prevented by mutating the antioxidant responsive element in the promoter or by overexpressing dominant-negative Nrf2. In addition, the strain-mediated induction of HO-1 and activation of Nrf2 was abolished by the antioxidant N-acetyl-l-cysteine. Finally, application of cyclic strain blocked inflammatory cytokine-mediated EC death and apoptosis. However, the protective action of cyclic strain was reversed by the HO inhibitor tin protoporphyrin-IX and was absent in ECs isolated from HO-1-deficient mice. In conclusion, the present study demonstrates that a hemodynamically relevant level of cyclic strain stimulates HO-1 gene expression in ECs via the ROS-Nrf2 signaling pathway to inhibit EC death. The ability of cyclic strain to induce HO-1 expression may provide an important mechanism by which hemodynamic forces promote EC survival and vascular homeostasis.
Collapse
Affiliation(s)
- Xiao-ming Liu
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | | | | |
Collapse
|
448
|
Abstract
Oxidative stress has been linked to the pathogenesis of the major complications of diabetes in the kidney, the heart, the eye or the vasculature. NADPH oxidases of the Nox family are a major source of ROS (reactive oxygen species) and are critical mediators of redox signalling in cells from different organs afflicted by the diabetic milieu. In the present review, we provide an overview of the current knowledge related to the understanding of the role of Nox in the processes that control cell injury induced by hyperglycaemia and other predominant factors enhanced in diabetes, including the renin–angiotensin system, TGF-β (transforming growth factor-β) and AGEs (advanced glycation end-products). These observations support a critical role for Nox homologues in diabetic complications and indicate that NADPH oxidases are an important therapeutic target. Therefore the design and development of small-molecule inhibitors that selectively block Nox oxidases appears to be a reasonable approach to prevent or retard the complications of diabetes in target organs. The bioefficacy of these agents in experimental animal models is also discussed in the present review.
Collapse
|
449
|
Vázquez-Medina JP, Popovich I, Thorwald MA, Viscarra JA, Rodriguez R, Sonanez-Organis JG, Lam L, Peti-Peterdi J, Nakano D, Nishiyama A, Ortiz RM. Angiotensin receptor-mediated oxidative stress is associated with impaired cardiac redox signaling and mitochondrial function in insulin-resistant rats. Am J Physiol Heart Circ Physiol 2013; 305:H599-607. [PMID: 23771688 DOI: 10.1152/ajpheart.00101.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Activation of angiotensin receptor type 1 (AT1) contributes to NADPH oxidase (Nox)-derived oxidative stress during metabolic syndrome. However, the specific role of AT1 in modulating redox signaling, mitochondrial function, and oxidative stress in the heart remains more elusive. To test the hypothesis that AT1 activation increases oxidative stress while impairing redox signaling and mitochondrial function in the heart during diet-induced insulin resistance in obese animals, Otsuka Long Evans Tokushima Fatty (OLETF) rats (n = 8/group) were treated with the AT1 blocker (ARB) olmesartan for 6 wk. Cardiac Nox2 protein expression increased 40% in OLETF compared with age-matched, lean, strain-control Long Evans Tokushima Otsuka (LETO) rats, while mRNA and protein expression of the H₂O₂-producing Nox4 increased 40-100%. ARB treatment prevented the increase in Nox2 without altering Nox4. ARB treatment also normalized the increased levels of protein and lipid oxidation (nitrotyrosine, 4-hydroxynonenal) and increased the redox-sensitive transcription factor Nrf2 by 30% and the activity of antioxidant enzymes (SOD, catalase, GPx) by 50-70%. Citrate synthase (CS) and succinate dehydrogenase (SDH) activities decreased 60-70%, whereas cardiac succinate levels decreased 35% in OLETF compared with LETO, suggesting that mitochondrial function in the heart is impaired during obesity-induced insulin resistance. ARB treatment normalized CS and SDH activities, as well as succinate levels, while increasing AMPK and normalizing Akt, suggesting that AT1 activation also impairs cellular metabolism in the diabetic heart. These data suggest that the cardiovascular complications associated with metabolic syndrome may result from AT1 receptor-mediated Nox2 activation leading to impaired redox signaling, mitochondrial activity, and dysregulation of cellular metabolism in the heart.
Collapse
Affiliation(s)
- José Pablo Vázquez-Medina
- Department of Molecular and Cellular Biology, School of Natural Sciences, University of California Merced, Merced, California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
450
|
He C, Zhu H, Zhang W, Okon I, Wang Q, Li H, Le YZ, Xie Z. 7-Ketocholesterol induces autophagy in vascular smooth muscle cells through Nox4 and Atg4B. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:626-37. [PMID: 23770348 DOI: 10.1016/j.ajpath.2013.04.028] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 03/25/2013] [Accepted: 04/16/2013] [Indexed: 10/26/2022]
Abstract
Oxidized lipoproteins stimulate autophagy in advanced atherosclerotic plaques. However, the mechanisms underlying autophagy induction and the role of autophagy in atherogenesis remain to be determined. This study was designed to investigate the mechanisms by which 7-ketocholesterol (7-KC), a major component of oxidized lipoproteins, induces autophagy. This study was also designed to determine the effect of autophagy induction on apoptosis, a central event in the development of atherosclerosis. Exposure of human aortic smooth muscle cells to 7-KC increased autophagic flux. Autophagy induction was suppressed by treating the cells with either a reactive oxygen species scavenger or an antioxidant. Administration of 7-KC concomitantly up-regulated Nox4 expression, increased intracellular hydrogen peroxide levels, and inhibited autophagy-related gene 4B activity. Catalase overexpression to remove hydrogen peroxide or Nox4 knockdown with siRNA reduced intracellular hydrogen peroxide levels, restored autophagy-related gene 4B activity, and consequently attenuated 7-KC-induced autophagy. Moreover, inhibition of autophagy aggravated both endoplasmic reticulum (ER) stress and cell death in response to 7-KC. In contrast, up-regulation of autophagic activity by rapamycin had opposite effects. Finally, activation of autophagy by chronic rapamycin treatment attenuated ER stress, apoptosis, and atherosclerosis in apolipoprotein E knockout (ApoE(-/-)) mouse aortas. In conclusion, we demonstrate that up-regulation of autophagy is a cellular protective response that attenuates 7-KC-induced cell death in human aortic smooth muscle cells.
Collapse
Affiliation(s)
- Chaoyong He
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | | | | | |
Collapse
|