51
|
Liu RM, Desai LP. Reciprocal regulation of TGF-β and reactive oxygen species: A perverse cycle for fibrosis. Redox Biol 2015; 6:565-577. [PMID: 26496488 PMCID: PMC4625010 DOI: 10.1016/j.redox.2015.09.009] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/17/2015] [Accepted: 09/20/2015] [Indexed: 12/21/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is the most potent pro-fibrogenic cytokine and its expression is increased in almost all of fibrotic diseases. Although signaling through Smad pathway is believed to play a central role in TGF-β's fibrogenesis, emerging evidence indicates that reactive oxygen species (ROS) modulate TGF-β's signaling through different pathways including Smad pathway. TGF-β1 increases ROS production and suppresses antioxidant enzymes, leading to a redox imbalance. ROS, in turn, induce/activate TGF-β1 and mediate many of TGF-β's fibrogenic effects, forming a vicious cycle (see graphic flow chart on the right). Here, we review the current knowledge on the feed-forward mechanisms between TGF-β1 and ROS in the development of fibrosis. Therapeutics targeting TGF-β-induced and ROS-dependent cellular signaling represents a novel approach in the treatment of fibrotic disorders. TGF-β1 is the most potent ubiquitous profibrogenic cytokine. TGF- β 1 induces redox imbalance by ↑ ROS production and ↓ anti-oxidant defense system Redox imbalance, in turn, activates latent TGF-β1 and induces TGF-β1 expression. Redox imbalance also mediates many of TGF-β1’s profibrogenic effects
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmi ngham, Birmingham, AL, USA.
| | - Leena P Desai
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmi ngham, Birmingham, AL, USA
| |
Collapse
|
52
|
Ivanov AV, Smirnova OA, Petrushanko IY, Ivanova ON, Karpenko IL, Alekseeva E, Sominskaya I, Makarov AA, Bartosch B, Kochetkov SN, Isaguliants MG. HCV core protein uses multiple mechanisms to induce oxidative stress in human hepatoma Huh7 cells. Viruses 2015; 7:2745-70. [PMID: 26035647 PMCID: PMC4488712 DOI: 10.3390/v7062745] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/12/2015] [Accepted: 05/26/2015] [Indexed: 12/13/2022] Open
Abstract
Hepatitis C virus (HCV) infection is accompanied by the induction of oxidative stress, mediated by several virus proteins, the most prominent being the nucleocapsid protein (HCV core). Here, using the truncated forms of HCV core, we have delineated several mechanisms by which it induces the oxidative stress. The N-terminal 36 amino acids of HCV core induced TGF\(\upbeta\)1-dependent expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases 1 and 4, both of which independently contributed to the production of reactive oxygen species (ROS). The same fragment also induced the expression of cyclo-oxygenase 2, which, however, made no input into ROS production. Amino acids 37-191 of HCV core up-regulated the transcription of a ROS generating enzyme cytochrome P450 2E1. Furthermore, the same fragment induced the expression of endoplasmic reticulum oxidoreductin 1\(\upalpha\). The latter triggered efflux of Ca2+ from ER to mitochondria via mitochondrial Ca2+ uniporter, leading to generation of superoxide anions, and possibly also H2O2. Suppression of any of these pathways in cells expressing the full-length core protein led to a partial inhibition of ROS production. Thus, HCV core causes oxidative stress via several independent pathways, each mediated by a distinct region of the protein.
Collapse
Affiliation(s)
- Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, Moscow 119991, Russia.
| | - Olga A Smirnova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, Moscow 119991, Russia.
| | - Irina Y Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, Moscow 119991, Russia.
| | - Olga N Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, Moscow 119991, Russia.
| | - Inna L Karpenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, Moscow 119991, Russia.
| | - Ekaterina Alekseeva
- Latvian Biomedical Research and Study Center, Ratsupites 1, Riga LV1067, Latvia.
| | - Irina Sominskaya
- Latvian Biomedical Research and Study Center, Ratsupites 1, Riga LV1067, Latvia.
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, Moscow 119991, Russia.
| | - Birke Bartosch
- Inserm U1052, Cancer Research Center of Lyon, University of Lyon, 151, Cours A Thomas, 69424 Lyon Cedex, Lyon, France.
- DevWeCan Laboratories of Excellence Network (Labex), Lyon F-69000, France.
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, Moscow 119991, Russia.
| | - Maria G Isaguliants
- Ivanovsky Institute of Virology, Gamaleya str. 16, Moscow 123098, Russia.
- Kirchenstein Institute of Microbiology and Virology, Riga Stradins University, Ratsupites 5, Riga LV-1069, Latvia.
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Nobelsvägen 16, Stockholm 17177, Sweden.
| |
Collapse
|
53
|
Burke RM, Berk BC. The Role of PB1 Domain Proteins in Endothelial Cell Dysfunction and Disease. Antioxid Redox Signal 2015; 22:1243-56. [PMID: 25686626 DOI: 10.1089/ars.2014.6182] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE There are a limited number of proteins containing the Phox-Bem1 (PB1) protein interaction domain, and almost all of them play some role in endothelial cell (EC) function, health, and homeostasis. RECENT ADVANCES Most of these proteins have been shown to physically interact through PB1-PB1 binding and, as such, are linked together to form complexes that are responsive to hemodynamic force. These complexes range from redox regulation to inflammation to autophagy and back, and they employ multiple feedback mechanisms that are reliant on PB1 domain proteins. CRITICAL ISSUES Pathologic roles for PB1 domain-containing proteins have been demonstrated in multiple diseases, including vascular disease, cancer, liver disease, and myriad other concerns. Findings cited in this review show that dimerization of PB1 proteins exerts novel effects on EC function that may be important in multiple cardiovascular diseases, including atherosclerosis, thrombosis, inflammation, and hypertension. FUTURE DIRECTIONS As mechanistic understanding of the component pathways (redox regulation, cell polarity, inflammation, atheroprotection, and autophagy) is continually increasing, the larger picture of how these pathways interact with one another is evolving rapidly. We can now evaluate the PB1 domain proteins as a family in the context of multiple phenotypic readouts in EC function as well as evaluate them as drug targets against disease.
Collapse
Affiliation(s)
- Ryan M Burke
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester Medical Center , Rochester, New York
| | | |
Collapse
|
54
|
Stanicka J, Russell EG, Woolley JF, Cotter TG. NADPH oxidase-generated hydrogen peroxide induces DNA damage in mutant FLT3-expressing leukemia cells. J Biol Chem 2015; 290:9348-61. [PMID: 25697362 DOI: 10.1074/jbc.m113.510495] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Indexed: 11/06/2022] Open
Abstract
Internal tandem duplication of the FMS-like tyrosine kinase (FLT3-ITD) receptor is present in 20% of acute myeloid leukemia (AML) patients and it has been associated with an aggressive AML phenotype. FLT3-ITD expressing cell lines have been shown to generate increased levels of reactive oxygen species (ROS) and DNA double strand breaks (DSBs). However, the molecular basis of how FLT3-ITD-driven ROS leads to the aggressive form of AML is not clearly understood. Our group has previously reported that inhibition of FLT3-ITD signaling results in post-translational down-regulation of p22(phox), a small membrane-bound subunit of the NADPH oxidase (NOX) complex. Here we demonstrated that 32D cells, a myeloblast-like cell line transfected with FLT3-ITD, have a higher protein level of p22(phox) and p22(phox)-interacting NOX isoforms than 32D cells transfected with the wild type FLT3 receptor (FLT3-WT). The inhibition of NOX proteins, p22(phox), and NOX protein knockdowns caused a reduction in ROS, as measured with a hydrogen peroxide (H2O2)-specific dye, peroxy orange 1 (PO1), and nuclear H2O2, as measured with nuclear peroxy emerald 1 (NucPE1). These reductions in the level of H2O2 following the NOX knockdowns were accompanied by a decrease in the number of DNA DSBs. We showed that 32D cells that express FLT3-ITD have a higher level of both oxidized DNA and DNA DSBs than their wild type counterparts. We also observed that NOX4 and p22(phox) localize to the nuclear membrane in MV4-11 cells expressing FLT3-ITD. Taken together these data indicate that NOX and p22(phox) mediate the ROS production from FLT3-ITD that signal to the nucleus causing genomic instability.
Collapse
Affiliation(s)
- Joanna Stanicka
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Eileen G Russell
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - John F Woolley
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Thomas G Cotter
- From the Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| |
Collapse
|
55
|
Brennan-Minnella AM, Won SJ, Swanson RA. NADPH oxidase-2: linking glucose, acidosis, and excitotoxicity in stroke. Antioxid Redox Signal 2015; 22:161-74. [PMID: 24628477 PMCID: PMC4281853 DOI: 10.1089/ars.2013.5767] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Neuronal superoxide production contributes to cell death in both glutamate excitotoxicity and brain ischemia (stroke). NADPH oxidase-2 (NOX2) is the major source of neuronal superoxide production in these settings, and regulation of NOX2 activity can thereby influence outcome in stroke. RECENT ADVANCES Reduced NOX2 activity can rescue cells from oxidative stress and cell death that otherwise occur in excitotoxicity and ischemia. NOX2 activity is regulated by several factors previously shown to affect outcome in stroke, including glucose availability, intracellular pH, protein kinase ζ/δ, casein kinase 2, phosphoinositide-3-kinase, Rac1/2, and phospholipase A2. The newly identified functions of these factors as regulators of NOX2 activity suggest alternative mechanisms for their effects on ischemic brain injury. CRITICAL ISSUES Key aspects of these regulatory influences remain unresolved, including the mechanisms by which rac1 and phospholipase activities are coupled to N-methyl-D-aspartate (NMDA) receptors, and whether superoxide production by NOX2 triggers subsequent superoxide production by mitochondria. FUTURE DIRECTIONS It will be important to establish whether interventions targeting the signaling pathways linking NMDA receptors to NOX2 in brain ischemia can provide a greater neuroprotective efficacy or a longer time window to treatment than provided by NMDA receptor blockade alone. It will likewise be important to determine whether dissociating superoxide production from the other signaling events initiated by NMDA receptors can mitigate the deleterious effects of NMDA receptor blockade.
Collapse
|
56
|
Schröder K. NADPH oxidases in bone homeostasis and osteoporosis. Cell Mol Life Sci 2015; 72:25-38. [PMID: 25167924 PMCID: PMC11114015 DOI: 10.1007/s00018-014-1712-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 02/06/2023]
Abstract
Bone formation and degradation are perfectly coordinated. In case of an imbalance of these processes diseases occur associated with exaggerated formation of new bone or bone loss as in osteoporosis. Most studies investigating osteoporosis either focus on osteoblast or osteoclast function and differentiation. Both processes have been suggested to be affected by reactive oxygen species (ROS). Besides a potentially harmful role of ROS, these small molecules are important second messengers. The family of NADPH oxidases produces ROS in a controlled and targeted manner, to specifically regulate signal transduction. This review will highlight the role of reactive oxygen species in bone cell differentiation and bone-loss associated disease with a special focus on osteoporosis and NADPH oxidases as specialized sources of ROS.
Collapse
Affiliation(s)
- Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität, Universität Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany,
| |
Collapse
|
57
|
Verstraeten SV, Fraga CG, Oteiza PI. Interactions of flavan-3-ols and procyanidins with membranes: mechanisms and the physiological relevance. Food Funct 2014; 6:32-41. [PMID: 25418533 DOI: 10.1039/c4fo00647j] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Flavonoids are a type of phenolic compound widely present in edible plants. A great number of health benefits have been ascribed to flavonoid consumption in the human population. However, the molecular mechanisms involved in such effects remain to be identified. The flavan-3-ols (-)-epicatechin and (+)-catechin, and their related oligomers (procyanidins) have been thoroughly studied because of their capacity to interact with cell membranes. Starting with these interactions, procyanidins could modulate multiple biochemical processes, such as enzyme activities, receptor-ligand binding, membrane-initiated cell signaling, and molecule transport across membranes. This review focuses on molecular aspects of procyanidin interactions with membrane lipid components, and the resulting protection of the membranes against mechanical and/or oxidative damage, resulting in the maintenance of cell functions.
Collapse
Affiliation(s)
- Sandra V Verstraeten
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | | | | |
Collapse
|
58
|
Meitzler JL, Antony S, Wu Y, Juhasz A, Liu H, Jiang G, Lu J, Roy K, Doroshow JH. NADPH oxidases: a perspective on reactive oxygen species production in tumor biology. Antioxid Redox Signal 2014; 20:2873-89. [PMID: 24156355 PMCID: PMC4026372 DOI: 10.1089/ars.2013.5603] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Reactive oxygen species (ROS) promote genomic instability, altered signal transduction, and an environment that can sustain tumor formation and growth. The NOX family of NADPH oxidases, membrane-bound epithelial superoxide and hydrogen peroxide producers, plays a critical role in the maintenance of immune function, cell growth, and apoptosis. The impact of NOX enzymes in carcinogenesis is currently being defined and may directly link chronic inflammation and NOX ROS-mediated tumor formation. RECENT ADVANCES Increased interest in the function of NOX enzymes in tumor biology has spurred a surge of investigative effort to understand the variability of NOX expression levels in tumors and the effect of NOX activity on tumor cell proliferation. These initial efforts have demonstrated a wide variance in NOX distribution and expression levels across numerous cancers as well as in common tumor cell lines, suggesting that much remains to be discovered about the unique role of NOX-related ROS production within each system. Progression from in vitro cell line studies toward in vivo tumor tissue screening and xenograft models has begun to provide evidence supporting the importance of NOX expression in carcinogenesis. CRITICAL ISSUES A lack of universally available, isoform-specific antibodies and animal tumor models of inducible knockout or over-expression of NOX isoforms has hindered progress toward the completion of in vivo studies. FUTURE DIRECTIONS In vivo validation experiments and the use of large, existing gene expression data sets should help define the best model systems for studying the NOX homologues in the context of cancer.
Collapse
Affiliation(s)
- Jennifer L Meitzler
- 1 Laboratory of Molecular Pharmacology of the Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Paik YH, Kim J, Aoyama T, De Minicis S, Bataller R, Brenner DA. Role of NADPH oxidases in liver fibrosis. Antioxid Redox Signal 2014; 20:2854-72. [PMID: 24040957 PMCID: PMC4026397 DOI: 10.1089/ars.2013.5619] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Hepatic fibrosis is the common pathophysiologic process resulting from chronic liver injury, characterized by the accumulation of an excessive extracellular matrix. Multiple lines of evidence indicate that oxidative stress plays a pivotal role in the pathogenesis of liver fibrosis. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) is a multicomponent enzyme complex that generates reactive oxygen species (ROS) in response to a wide range of stimuli. In addition to phagocytic NOX2, there are six nonphagocytic NOX proteins. RECENT ADVANCES In the liver, NOX is functionally expressed both in the phagocytic form and in the nonphagocytic form. NOX-derived ROS contributes to various kinds of liver disease caused by alcohol, hepatitis C virus, and toxic bile acids. Recent evidence indicates that both phagocytic NOX2 and nonphagocytic NOX isoforms, including NOX1 and NOX4, mediate distinct profibrogenic actions in hepatic stellate cells, the main fibrogenic cell type in the liver. The critical role of NOX in hepatic fibrogenesis provides a rationale to assess pharmacological NOX inhibitors that treat hepatic fibrosis in patients with chronic liver disease. CRITICAL ISSUES Although there is compelling evidence indicating a crucial role for NOX-mediated ROS generation in hepatic fibrogenesis, little is known about the expression, subcellular localization, regulation, and redox signaling of NOX isoforms in specific cell types in the liver. Moreover, the exact mechanism of NOX-mediated fibrogenic signaling is still largely unknown. FUTURE DIRECTIONS A better understanding through further research about NOX-mediated fibrogenic signaling may enable the development of novel anti-fibrotic therapy using NOX inhibition strategy. Antio
Collapse
Affiliation(s)
- Yong-Han Paik
- 1 Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine , Seoul, Korea
| | | | | | | | | | | |
Collapse
|
60
|
Abstract
SIGNIFICANCE Understanding isoform- and context-specific subcellular Nox reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase compartmentalization allows relevant functional inferences. This review addresses the interplay between Nox NADPH oxidases and the endoplasmic reticulum (ER), an increasingly evident player in redox pathophysiology given its role in redox protein folding and stress responses. RECENT ADVANCES Catalytic/regulatory transmembrane subunits are synthesized in the ER and their processing includes folding, N-glycosylation, heme insertion, p22phox heterodimerization, as shown for phagocyte Nox2. Dual oxidase (Duox) maturation also involves the regulation by ER-resident Duoxa2. The ER is the activation site for some isoforms, typically Nox4, but potentially other isoforms. Such location influences redox/Nox-mediated calcium signaling regulation via ER targets, such as sarcoendoplasmic reticulum calcium ATPase (SERCA). Growing evidence suggests that Noxes are integral signaling elements of the unfolded protein response during ER stress, with Nox4 playing a dual prosurvival/proapoptotic role in this setting, whereas Nox2 enhances proapoptotic signaling. ER chaperones such as protein disulfide isomerase (PDI) closely interact with Noxes. PDI supports growth factor-dependent Nox1 activation and mRNA expression, as well as migration in smooth muscle cells, and PDI overexpression induces acute spontaneous Nox activation. CRITICAL ISSUES Mechanisms of PDI effects include possible support of complex formation and RhoGTPase activation. In phagocytes, PDI supports phagocytosis, Nox activation, and redox-dependent interactions with p47phox. Together, the results implicate PDI as possible Nox organizer. FUTURE DIRECTIONS We propose that convergence between Noxes and ER may have evolutive roots given ER-related functional contexts, which paved Nox evolution, namely calcium signaling and pathogen killing. Overall, the interplay between Noxes and the ER may provide relevant insights in Nox-related (patho)physiology.
Collapse
Affiliation(s)
- Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine , São Paulo, Brazil
| | | | | |
Collapse
|
61
|
De Deken X, Corvilain B, Dumont JE, Miot F. Roles of DUOX-mediated hydrogen peroxide in metabolism, host defense, and signaling. Antioxid Redox Signal 2014; 20:2776-93. [PMID: 24161126 DOI: 10.1089/ars.2013.5602] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Among the NADPH oxidases, the dual oxidases, DUOX1 and DUOX2, constitute a distinct subfamily initially called thyroid oxidases, based on their high level of expression in thyroid tissue. Genetic alterations causing inherited hypothyroidism clearly demonstrate their physiological implication in thyroid hormonogenesis. However, a growing list of biological functions triggered by DUOX-dependent reactive oxygen species (ROS) in highly differentiated mucosae have recently emerged. RECENT ADVANCES A role of DUOX enzymes as ROS providers for lactoperoxidase-mediated killing of invading pathogens has been well established and a role in bacteria chemorepulsion has been proposed. Control of DUOX expression and activity by inflammatory molecules and immune receptor activation consolidates their contributions to innate immune defense of mucosal surfaces. Recent studies conducted in ancestral organisms have identified effectors of DUOX redox signaling involved in wound healing including epithelium regeneration and leukocyte recruitment. Moreover, local generation of hydrogen peroxide (H2O2) by DUOX has also been suggested to constitute a positive feedback loop to promote receptor signaling activation. CRITICAL ISSUES A correct balance between H2O2 generation and detoxification mechanisms must be properly maintained to avoid oxidative damages. Overexpression of DUOX genes has been associated with an increasing number of chronic inflammatory diseases. Furthermore, H2O2-mediated DNA damage supports a mutagenic function promoting tumor development. FUTURE DIRECTIONS Despite the high sequence similarity shared between DUOX1 and DUOX2, the two isoforms present distinct regulations, tissue expression and catalytic functions. The phenotypic characterization of novel DUOX/DUOXA invalidated animal models will be very useful for defining their medical importance in pathological conditions.
Collapse
Affiliation(s)
- Xavier De Deken
- Faculté de Médecine, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB) , Brussels, Belgium
| | | | | | | |
Collapse
|
62
|
Abstract
SIGNIFICANCE Reactive oxygen species (ROS) play a critical role in vascular disease. While there are many possible sources of ROS, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases play a central role. They are a source of "kindling radicals," which affect other enzymes, such as nitric oxide synthase endothelial nitric oxide synthase or xanthine oxidase. This is important, as risk factors for atherosclerosis (hypertension, diabetes, hypercholesterolemia, and smoking) regulate the expression and activity of NADPH oxidases in the vessel wall. RECENT ADVANCES There are seven isoforms in mammals: Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2. Nox1, Nox2, Nox4, and Nox5 are expressed in endothelium, vascular smooth muscle cells, fibroblasts, or perivascular adipocytes. Other homologues have not been found or are expressed at very low levels; their roles have not been established. Nox1/Nox2 promote the development of endothelial dysfunction, hypertension, and inflammation. Nox4 may have a role in protecting the vasculature during stress; however, when its activity is increased, it may be detrimental. Calcium-dependent Nox5 has been implicated in oxidative damage in human atherosclerosis. CRITICAL ISSUES NADPH oxidase-derived ROS play a role in vascular pathology as well as in the maintenance of normal physiological vascular function. We also discuss recently elucidated mechanisms such as the role of NADPH oxidases in vascular protection, vascular inflammation, pulmonary hypertension, tumor angiogenesis, and central nervous system regulation of vascular function and hypertension. FUTURE DIRECTIONS Understanding the role of individual oxidases and interactions between homologues in vascular disease is critical for efficient pharmacological regulation of vascular NADPH oxidases in both the laboratory and clinical practice.
Collapse
Affiliation(s)
- Anna Konior
- 1 Department of Internal Medicine, Jagiellonian University School of Medicine , Cracow, Poland
| | | | | | | |
Collapse
|
63
|
Bernard K, Hecker L, Luckhardt TR, Cheng G, Thannickal VJ. NADPH oxidases in lung health and disease. Antioxid Redox Signal 2014; 20:2838-53. [PMID: 24093231 PMCID: PMC4026303 DOI: 10.1089/ars.2013.5608] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The evolution of the lungs and circulatory systems in vertebrates ensured the availability of molecular oxygen (O2; dioxygen) for aerobic cellular metabolism of internal organs in large animals. O2 serves as the physiologic terminal acceptor of mitochondrial electron transfer and of the NADPH oxidase (Nox) family of oxidoreductases to generate primarily water and reactive oxygen species (ROS), respectively. RECENT ADVANCES The purposeful generation of ROS by Nox family enzymes suggests important roles in normal physiology and adaptation, most notably in host defense against invading pathogens and in cellular signaling. CRITICAL ISSUES However, there is emerging evidence that, in the context of chronic stress and/or aging, Nox enzymes contribute to the pathogenesis of a number of lung diseases. FUTURE DIRECTIONS Here, we review evolving functions of Nox enzymes in normal lung physiology and emerging pathophysiologic roles in lung disease.
Collapse
Affiliation(s)
- Karen Bernard
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | | | | | | | | |
Collapse
|
64
|
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
|
65
|
Tan C, Day R, Bao S, Turk J, Zhao QD. Group VIA phospholipase A2 mediates enhanced macrophage migration in diabetes mellitus by increasing expression of nicotinamide adenine dinucleotide phosphate oxidase 4. Arterioscler Thromb Vasc Biol 2014; 34:768-78. [PMID: 24482376 DOI: 10.1161/atvbaha.113.302847] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We previously demonstrated that nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) mediates increased monocyte priming and chemotaxis under conditions of diabetic metabolic stress, and emerging data indicate that group VIA phospholipase A2 (iPLA2β) also participates in regulating monocyte chemotaxis. Here, we examined relationships between iPLA2β expression and Nox4 action in mouse peritoneal macrophages subjected to diabetic metabolic stress. APPROACH AND RESULTS Increased iPLA2β expression and activity were observed in macrophages from low-density lipoprotein receptor knockout mice that were fed a high-fat diet, and this was associated with time-dependent increases in atherosclerotic lesion size and macrophage content. Incubating macrophages with 30 mmol/L D-glucose, 100 μg/mL low-density lipoprotein, or both (D-glucose+low-density lipoprotein) induced a robust increase in iPLA2β expression and activity and in cell migration in response to monocyte chemoattractant protein-1. The increases in iPLA2β activity and cell migration were prevented by a bromoenol lactone iPLA2β suicide inhibitor or an iPLA2β antisense oligonucleotide. Incubating macrophages under conditions that mimic diabetic metabolic stress ex vivo resulted in increased Nox4 expression and activity and hydrogen peroxide generation compared with controls. Bromoenol lactone prevented those effects without affecting Nox2 expression. Nox4 inhibition eliminated diabetic metabolic stress-induced acceleration of macrophage migration. Lysophosphatidic acid restored Nox4 expression, hydrogen peroxide generation, and migration to bromoenol lactone-treated cells, and a lysophosphatidic acid receptor antagonist abrogated iPLA2β-mediated increases in Nox4 expression. CONCLUSIONS Taken together, these observations identify iPLA2β and lysophosphatidic acid derived from its action as critical in regulating macrophage Nox4 activity and migration in the diabetic state in vivo and under similar conditions ex vivo.
Collapse
Affiliation(s)
- Chunyan Tan
- From the Department of Medicine, University of Texas Health Science Center, San Antonio (C.T., R.D., Q.D.Z.); and Department of Medicine, Washington University School of Medicine, St. Louis, MO (S.B., J.T.)
| | | | | | | | | |
Collapse
|
66
|
Ullevig SL, Kim HS, Nguyen HN, Hambright WS, Robles AJ, Tavakoli S, Asmis R. Ursolic acid protects monocytes against metabolic stress-induced priming and dysfunction by preventing the induction of Nox4. Redox Biol 2014; 2:259-66. [PMID: 24494201 PMCID: PMC3909821 DOI: 10.1016/j.redox.2014.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 01/03/2014] [Indexed: 01/11/2023] Open
Abstract
AIMS Dietary supplementation with ursolic acid (UA) prevents monocyte dysfunction in diabetic mice and protects mice against atherosclerosis and loss of renal function. The goal of this study was to determine the molecular mechanism by which UA prevents monocyte dysfunction induced by metabolic stress. METHODS AND RESULTS Metabolic stress sensitizes or "primes" human THP-1 monocytes and murine peritoneal macrophages to the chemoattractant MCP-1, converting these cells into a hyper-chemotactic phenotype. UA protected THP-1 monocytes and peritoneal macrophages against metabolic priming and prevented their hyper-reactivity to MCP-1. UA blocked the metabolic stress-induced increase in global protein-S-glutathionylation, a measure of cellular thiol oxidative stress, and normalized actin-S-glutathionylation. UA also restored MAPK phosphatase-1 (MKP1) protein expression and phosphatase activity, decreased by metabolic priming, and normalized p38 MAPK activation. Neither metabolic stress nor UA supplementation altered mRNA or protein levels of glutaredoxin-1, the principal enzyme responsible for the reduction of mixed disulfides between glutathione and protein thiols in these cells. However, the induction of Nox4 by metabolic stress, required for metabolic priming, was inhibited by UA in both THP-1 monocytes and peritoneal macrophages. CONCLUSION UA protects THP-1 monocytes against dysfunction by suppressing metabolic stress-induced Nox4 expression, thereby preventing the Nox4-dependent dysregulation of redox-sensitive processes, including actin turnover and MAPK-signaling, two key processes that control monocyte migration and adhesion. This study provides a novel mechanism for the anti-inflammatory and athero- and renoprotective properties of UA and suggests that dysfunctional blood monocytes may be primary targets of UA and related compounds.
Collapse
Key Words
- Atherosclerosis
- GSH, reduced glutathione
- Grx, glutaredoxin
- HFD, high-fat diet
- HG, high d-glucose
- LDL, low-density lipoprotein
- MAPK, mitogen-activated protein kinase
- MCP-1, monocyte chemoattractant protein-1
- MKP-1, MAPK phosphatase-1
- Monocyte
- Nox4
- Nox4, NADPH oxidase 4
- OA, oleanolic acid
- PSSG, protein–glutathione mixed disulfide
- ROS, reactive oxygen species
- S-glutathionylation
- UA, ursolic acid
- Ursolic acid
Collapse
Affiliation(s)
- Sarah L. Ullevig
- Department of Kinesiology, Health, and Nutrition, University of Texas at San Antonio, United States
| | - Hong Seok Kim
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center, San Antonio, United States
| | - Huynh Nga Nguyen
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, United States
| | - William S. Hambright
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, United States
| | - Andrew J. Robles
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, United States
| | - Sina Tavakoli
- Department of Radiology, University of Texas Health Science Center, San Antonio, United States
| | - Reto Asmis
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center, San Antonio, United States
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, United States
- Department of Radiology, University of Texas Health Science Center, San Antonio, United States
| |
Collapse
|
67
|
Verstraeten SV, Jaggers GK, Fraga CG, Oteiza PI. Procyanidins can interact with Caco-2 cell membrane lipid rafts: Involvement of cholesterol. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2646-53. [DOI: 10.1016/j.bbamem.2013.07.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/12/2013] [Accepted: 07/16/2013] [Indexed: 11/28/2022]
|
68
|
Ullevig S, Kim HS, Asmis R. S-glutathionylation in monocyte and macrophage (dys)function. Int J Mol Sci 2013; 14:15212-32. [PMID: 23887649 PMCID: PMC3759857 DOI: 10.3390/ijms140815212] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 06/15/2013] [Accepted: 06/18/2013] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease involving the accumulation of monocytes and macrophages in the vascular wall. Monocytes and macrophages play a central role in the initiation and progression of atherosclerotic lesion development. Oxidative stress, which occurs when reactive oxygen species (ROS) overwhelm cellular antioxidant systems, contributes to the pathophysiology of many chronic inflammatory diseases, including atherosclerosis. Major targets of ROS are reactive thiols on cysteine residues in proteins, which when oxidized can alter cellular processes, including signaling pathways, metabolic pathways, transcription, and translation. Protein-S-glutathionylation is the process of mixed disulfide formation between glutathione (GSH) and protein thiols. Until recently, protein-S-glutathionylation was associated with increased cellular oxidative stress, but S-glutathionylation of key protein targets has now emerged as a physiologically important redox signaling mechanism, which when dysregulated contributes to a variety of disease processes. In this review, we will explore the role of thiol oxidative stress and protein-S-glutathionylation in monocyte and macrophage dysfunction as a mechanistic link between oxidative stress associated with metabolic disorders and chronic inflammatory diseases, including atherosclerosis.
Collapse
Affiliation(s)
- Sarah Ullevig
- Department of Biochemistry, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
| | - Hong Seok Kim
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
| | - Reto Asmis
- Department of Biochemistry, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-210-567-3411; Fax: +1-210-567-3719
| |
Collapse
|
69
|
Lee CF, Ullevig S, Kim HS, Asmis R. Regulation of Monocyte Adhesion and Migration by Nox4. PLoS One 2013; 8:e66964. [PMID: 23825596 PMCID: PMC3688996 DOI: 10.1371/journal.pone.0066964] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023] Open
Abstract
We showed that metabolic disorders promote thiol oxidative stress in monocytes, priming monocytes for accelerated chemokine-induced recruitment, and accumulation at sites of vascular injury and the progression of atherosclerosis. The aim of this study was to identify both the source of reactive oxygen species (ROS) responsible for thiol oxidation in primed and dysfunctional monocytes and the molecular mechanisms through which ROS accelerate the migration and recruitment of monocyte-derived macrophages. We found that Nox4, a recently identified NADPH oxidase in monocytes and macrophages, localized to focal adhesions and the actin cytoskeleton, and associated with phospho-FAK, paxillin, and actin, implicating Nox4 in the regulation of monocyte adhesion and migration. We also identified Nox4 as a new, metabolic stress-inducible source of ROS that controls actin S-glutathionylation and turnover in monocytes and macrophages, providing a novel mechanistic link between Nox4-derived H2O2 and monocyte adhesion and migration. Actin associated with Nox4 was S-glutathionylated, and Nox4 association with actin was enhanced in metabolically-stressed monocytes. Metabolic stress induced Nox4 and accelerated monocyte adhesion and chemotaxis in a Nox4-dependent mechanism. In conclusion, our data suggest that monocytic Nox4 is a central regulator of actin dynamics, and induction of Nox4 is the rate-limiting step in metabolic stress-induced monocyte priming and dysfunction associated with accelerated atherosclerosis and the progression of atherosclerotic plaques.
Collapse
Affiliation(s)
- Chi Fung Lee
- Department of Biochemistry, School of Health Professions, University of Texas Health Science Center at San Antonio, Texas, United States of America
| | - Sarah Ullevig
- Department of Biochemistry, School of Health Professions, University of Texas Health Science Center at San Antonio, Texas, United States of America
| | - Hong Seok Kim
- Department of Clinical Laboratory Sciences, School of Health Professions, University of Texas Health Science Center at San Antonio, Texas, United States of America
| | - Reto Asmis
- Department of Biochemistry, School of Health Professions, University of Texas Health Science Center at San Antonio, Texas, United States of America
- Department of Clinical Laboratory Sciences, School of Health Professions, University of Texas Health Science Center at San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
70
|
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: 3.9] [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
|
71
|
Vázquez-Medina JP, Soñanez-Organis JG, Rodriguez R, Viscarra JA, Nishiyama A, Crocker DE, Ortiz RM. Prolonged fasting activates Nrf2 in post-weaned elephant seals. ACTA ACUST UNITED AC 2013; 216:2870-8. [PMID: 23619404 DOI: 10.1242/jeb.081927] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Elephant seals naturally experience prolonged periods of absolute food and water deprivation (fasting). In humans, rats and mice, prolonged food deprivation activates the renin-angiotensin system (RAS) and increases oxidative damage. In elephant seals, prolonged fasting activates RAS without increasing oxidative damage likely due to an increase in antioxidant defenses. The mechanism leading to the upregulation of antioxidant defenses during prolonged fasting remains elusive. Therefore, we investigated whether prolonged fasting activates the redox-sensitive transcription factor Nrf2, which controls the expression of antioxidant genes, and if such activation is potentially mediated by systemic increases in RAS. Blood and skeletal muscle samples were collected from seals fasting for 1, 3, 5 and 7 weeks. Nrf2 activity and nuclear content increased by 76% and 167% at week 7. Plasma angiotensin II (Ang II) and transforming growth factor β (TGF-β) were 5000% and 250% higher at week 7 than at week 1. Phosphorylation of Smad2, an effector of Ang II and TGF signaling, increased by 120% at week 7 and by 84% in response to intravenously infused Ang II. NADPH oxidase 4 (Nox4) mRNA expression, which is controlled by smad proteins, increased 430% at week 7, while Nox4 protein expression, which can activate Nrf2, was 170% higher at week 7 than at week 1. These results demonstrate that prolonged fasting activates Nrf2 in elephant seals and that RAS stimulation can potentially result in increased Nox4 through Smad phosphorylation. The results also suggest that Nox4 is essential to sustain the hormetic adaptive response to oxidative stress in fasting seals.
Collapse
|
72
|
Xi G, Shen XC, Wai C, Clemmons DR. Recruitment of Nox4 to a plasma membrane scaffold is required for localized reactive oxygen species generation and sustained Src activation in response to insulin-like growth factor-I. J Biol Chem 2013; 288:15641-53. [PMID: 23612968 DOI: 10.1074/jbc.m113.456046] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nox4-derived ROS is increased in response to hyperglycemia and is required for IGF-I-stimulated Src activation. This study was undertaken to determine the mechanism by which Nox4 mediates sustained Src activation. IGF-I stimulated sustained Src activation, which occurred primarily on the SHPS-1 scaffold protein. In vitro oxidation experiments indicated that Nox4-derived ROS was able to oxidize Src when they are in close proximity, and Src oxidation leads to its activation. Therefore we hypothesized that Nox4 recruitment to the plasma membrane scaffold SHPS-1 allowed localized ROS generation to mediate sustained Src oxidation and activation. To determine the mechanism of Nox4 recruitment, we analyzed the role of Grb2, a component of the SHPS-1 signaling complex. We determined that Nox4 Tyr-491 was phosphorylated after IGF-I stimulation and was responsible for Nox4 binding to the SH2 domain of Grb2. Overexpression of a Nox4 mutant, Y491F, prevented Nox4/Grb2 association. Importantly, it also prevented Nox4 recruitment to SHPS-1. The role of Grb2 was confirmed using a Pyk2 Y881F mutant, which blocked Grb2 recruitment to SHPS-1. Cells expressing this mutant had impaired Nox4 recruitment to SHPS-1. IGF-I-stimulated downstream signaling and biological actions were also significantly impaired in Nox4 Y491F-overexpressing cells. Disruption of Nox4 recruitment to SHPS-1 in aorta from diabetic mice inhibited IGF-I-stimulated Src oxidation and activation as well as cell proliferation. These findings provide insight into the mechanism by which localized Nox4-derived ROS regulates the sustained activity of a tyrosine kinase that is critical for mediating signal transduction and biological actions.
Collapse
Affiliation(s)
- Gang Xi
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
73
|
Yin W, Voit EO. Function and design of the Nox1 system in vascular smooth muscle cells. BMC SYSTEMS BIOLOGY 2013; 7:20. [PMID: 23497394 PMCID: PMC3606394 DOI: 10.1186/1752-0509-7-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 02/18/2013] [Indexed: 11/24/2022]
Abstract
Background Recent studies have demonstrated that the activation of NADPH oxidase 1 (Nox1) plays an important role in the control of reactive oxygen species and their involvement in vascular physiology and pathophysiology. In order to function properly, Nox1 needs to be available in an optimal state, where it is ready to respond appropriately and efficiently to upstream signals. It must also be able to return quickly to this state as soon as the input signal disappears. While Nox1 activation has been discussed extensively in recent years, mechanisms for enzyme disassembly and proper subunit recovery have not received the same attention and therefore require investigation. Results We study the Nox1 system in vascular smooth smucle cells and propose four potential disassembly mechanisms. The analysis consists primarily of large-scale Monte-Carlo simulations whose results are essentially independent of specific parameter values. The computational analysis shows that a specific profile of subunit concentrations is crucial for optimal functioning and responsiveness of the system to input signals. Specifically, free p47phox and inactive Rac1 should be dominant under unstimulated resting conditions, and the proteolytic disassembly pathway should have a low flux, as it is relatively inefficient. The computational results also reveal that the optimal design of the three subunit recovery pathways depends on the intracellular settings of the pathway and that the response speeds of key reversible reactions within the pathway are of great importance. Conclusions Our results provide a systematic basis for understanding the dynamics of Nox1 and yield novel insights into its crucially important disassembly mechanisms. The rigorous comparisons of the relative importance of four potential disassembly pathways demonstrate that disassembly via proteolysis is the least effective mechanism. The relative significance of the other three recovery pathways varies among different scenarios. It is greatly affected by the required response speed of the system and depends critically on appropriate flux balances between forward and reverse reactions. Our findings are predictive and pose novel hypotheses that should be validated with future experiments.
Collapse
Affiliation(s)
- Weiwei Yin
- The Wallace H, Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | |
Collapse
|
74
|
Strategies for anti-fibrotic therapies. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1088-103. [PMID: 23266403 DOI: 10.1016/j.bbadis.2012.12.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/07/2012] [Accepted: 12/08/2012] [Indexed: 02/07/2023]
Abstract
The fibrotic diseases encompass a wide spectrum of entities including such multisystemic diseases as systemic sclerosis, nephrogenic systemic fibrosis and sclerodermatous graft versus host disease, as well as organ-specific disorders such as pulmonary, liver, and kidney fibrosis. Collectively, given the wide variety of affected organs, the chronic nature of the fibrotic processes, and the large number of individuals suffering their devastating effects, these diseases pose one of the most serious health problems in current medicine and a serious economic burden to society. Despite these considerations there is currently no accepted effective treatment. However, remarkable progress has been achieved in the elucidation of their pathogenesis including the identification of the critical role of myofibroblasts and the determination of molecular mechanisms that result in the transcriptional activation of the genes responsible for the fibrotic process. Here we review the origin of the myofibroblast and discuss the crucial regulatory pathways involving multiple growth factors and cytokines that participate in the pathogenesis of the fibrotic process. Potentially effective therapeutic strategies based upon this new information are considered in detail and the major challenges that remain and their possible solutions are presented. It is expected that translational efforts devoted to convert this new knowledge into novel and effective anti-fibrotic drugs will be forthcoming in the near future. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
|
75
|
Chen F, Haigh S, Barman S, Fulton DJR. From form to function: the role of Nox4 in the cardiovascular system. Front Physiol 2012; 3:412. [PMID: 23125837 PMCID: PMC3485577 DOI: 10.3389/fphys.2012.00412] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 10/04/2012] [Indexed: 01/15/2023] Open
Abstract
The NADPH oxidase (Nox) family of proteins is comprised of seven members, including Noxes1–5 and the Duoxes 1 and 2. Nox4 is readily distinguished from the other Nox isoforms by its high level of expression in cardiovascular tissues and unique enzymatic properties. Nox4 is constitutively active and the amount of reactive oxygen species (ROS) contributed by Nox4 is primarily regulated at the transcriptional level although there is recent evidence for post-translational control. Nox4 emits a different pattern of ROS and its subcellular localizations, tissue distribution and influence over signaling pathways is different from the other Nox enzymes. Previous investigations have revealed that Nox4 is involved in oxygen sensing, vasomotor control, cellular proliferation, differentiation, migration, apoptosis, senescence, fibrosis, and angiogenesis. Elevated expression of Nox4 has been reported in a number of cardiovascular diseases, including atherosclerosis, pulmonary fibrosis, and hypertension, cardiac failure and ischemic stroke. However, many important questions remain regarding the functional significance of Nox4 in health and disease, including the role of Nox4 subcellular localization and its downstream targets. The goal of this review is to summarize the recent literature on the genetic and enzymatic regulation, subcellular localization, signaling pathways, and the role of Nox4 in cardiovascular disease states.
Collapse
Affiliation(s)
- Feng Chen
- Vascular Biology Center, Georgia Health Sciences University Augusta, GA, USA
| | | | | | | |
Collapse
|
76
|
Apocynin-treatment reverses hyperoxaluria induced changes in NADPH oxidase system expression in rat kidneys: a transcriptional study. PLoS One 2012; 7:e47738. [PMID: 23091645 PMCID: PMC3473023 DOI: 10.1371/journal.pone.0047738] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/14/2012] [Indexed: 11/25/2022] Open
Abstract
Purpose We have previously shown that production of reactive oxygen species (ROS) is an important contributor to renal injury and inflammation following exposure to oxalate (Ox) or calcium-oxalate (CaOx) crystals. The present study was conducted, utilizing global transcriptome analyses, to determine the effect of Apocynin on changes in the NADPH oxidase system activated in kidneys of rats fed a diet leading to hyperoxaluria and CaOx crystal deposition. Approach Age-, sex- and weight-matched rats were either fed regular rat chow or regular rat chow supplemented with 5% w/w hydroxy-L-proline (HLP). Half of the rats on the HLP diet were also placed on Apocynin-supplemented H2O. After 28 days, each rat was euthanized, their kidneys freshly explanted and dissected to obtain both cortex and medulla tissues. Total RNA was extracted from each tissue and subjected to genomic microarrays to obtain global transcriptome data. KEGG was used to identify gene clusters with differentially expressed genes. Immunohistochemistry was used to confirm protein expressions of selected genes. Results Genes encoding both membrane- and cytosolic-NADPH oxidase complex-associated proteins, together with p21rac and Rap1a, were coordinately up-regulated significantly in both renal medulla and cortex tissues in the HLP-fed rats compared to normal healthy untreated controls. Activation of NADPH oxidase appears to occur via the angiotensin-II/angiotensin-II receptor-2 pathway, although the DAG-PKC pathway of neutrophils may also contribute. Immuno histochemical staining confirmed up-regulated gene expressions. Simultaneously, genes encoding ROS scavenger proteins were down-regulated. HLP-fed rats receiving Apocynin had a complete reversal in the differential-expression of the NADPH oxidase system genes, despite showing similar levels of hyperoxaluria. Conclusions A strong up-regulation of an oxidative/respiratory burst involving the NADPH oxidase system, activated via the angiotensin-II and most likely the DAG-PKC pathways, occurs in kidneys of hyperoxaluric rats. Apocynin treatment reversed this activation without affecting the levels of hyperoxaluria.
Collapse
|
77
|
Kahles T, Brandes RP. NADPH oxidases as therapeutic targets in ischemic stroke. Cell Mol Life Sci 2012; 69:2345-63. [PMID: 22618244 PMCID: PMC11114534 DOI: 10.1007/s00018-012-1011-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 11/06/2011] [Accepted: 04/20/2012] [Indexed: 01/07/2023]
Abstract
Reactive oxygen species (ROS) act physiologically as signaling molecules. In pathological conditions, such as ischemic stroke, ROS are released in excessive amounts and upon reperfusion exceed the body's antioxidant detoxifying capacity. This process leads to brain tissue damage during reoxygenation. Consequently, antioxidant strategies have long been suggested as a therapy for experimental stroke, but clinical trials have not yet been able to promote the translation of this concept into patient treatment regimens. As an evolution of this concept, recent studies have targeted the sources of ROS generation-rather than ROS themselves. In this context, NADPH oxidases have been identified as important generators of ROS in the cerebral vasculature under both physiological conditions in general and during ischemia/reoxygenation in particular. Inhibition of NADPH oxidases or genetic deletion of certain NADPH oxidase isoforms has been found to considerably reduce ischemic injury in experimental stroke. This review focuses on recent advances in the understanding of NADPH oxidase-mediated tissue injury in the cerebral vasculature, particularly at the level of the blood-brain barrier, and highlights promising inhibitory strategies that target the NADPH oxidases.
Collapse
Affiliation(s)
- Timo Kahles
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität, Frankfurt, Germany.
| | | |
Collapse
|
78
|
Altenhöfer S, Kleikers PWM, Radermacher KA, Scheurer P, Rob Hermans JJ, Schiffers P, Ho H, Wingler K, Schmidt HHHW. The NOX toolbox: validating the role of NADPH oxidases in physiology and disease. Cell Mol Life Sci 2012; 69:2327-43. [PMID: 22648375 PMCID: PMC3383958 DOI: 10.1007/s00018-012-1010-9] [Citation(s) in RCA: 288] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are cellular signals but also disease triggers; their relative excess (oxidative stress) or shortage (reductive stress) compared to reducing equivalents are potentially deleterious. This may explain why antioxidants fail to combat diseases that correlate with oxidative stress. Instead, targeting of disease-relevant enzymatic ROS sources that leaves physiological ROS signaling unaffected may be more beneficial. NADPH oxidases are the only known enzyme family with the sole function to produce ROS. Of the catalytic NADPH oxidase subunits (NOX), NOX4 is the most widely distributed isoform. We provide here a critical review of the currently available experimental tools to assess the role of NOX and especially NOX4, i.e. knock-out mice, siRNAs, antibodies, and pharmacological inhibitors. We then focus on the characterization of the small molecule NADPH oxidase inhibitor, VAS2870, in vitro and in vivo, its specificity, selectivity, and possible mechanism of action. Finally, we discuss the validation of NOX4 as a potential therapeutic target for indications including stroke, heart failure, and fibrosis.
Collapse
Affiliation(s)
- Sebastian Altenhöfer
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Pamela W. M. Kleikers
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Kim A. Radermacher
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | | | - J. J. Rob Hermans
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Paul Schiffers
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Heidi Ho
- National Stroke Research Institute, Melbourne, VIC Australia
| | - Kirstin Wingler
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Harald H. H. W. Schmidt
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
79
|
The 1027th target candidate in stroke: Will NADPH oxidase hold up? EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2012; 4:11. [PMID: 22625431 PMCID: PMC3403875 DOI: 10.1186/2040-7378-4-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 05/24/2012] [Indexed: 02/07/2023]
Abstract
As recently reviewed, 1026 neuroprotective drug candidates in stroke research have all failed on their road towards validation and clinical translation, reasons being quality issues in preclinical research and publication bias. Quality control guidelines for preclinical stroke studies have now been established. However, sufficient understanding of the underlying mechanisms of neuronal death after stroke that could be possibly translated into new therapies is lacking. One exception is the hypothesis that cellular death is mediated by oxidative stress. Oxidative stress is defined as an excess of reactive oxygen species (ROS) derived from different possible enzymatic sources. Among these, NADPH oxidases (NOX1-5) stand out as they represent the only known enzyme family that has no other function than to produce ROS. Based on data from different NOX knockout mouse models in ischemic stroke, the most relevant isoform appears to be NOX4. Here we discuss the state-of-the-art of this target with respect to stroke and open questions that need to be addressed on the path towards clinical translation.
Collapse
|
80
|
Lassègue B, San Martín A, Griendling KK. Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system. Circ Res 2012; 110:1364-90. [PMID: 22581922 PMCID: PMC3365576 DOI: 10.1161/circresaha.111.243972] [Citation(s) in RCA: 611] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 03/09/2012] [Indexed: 02/07/2023]
Abstract
The NADPH oxidase (Nox) enzymes are critical mediators of cardiovascular physiology and pathophysiology. These proteins are expressed in virtually all cardiovascular cells, and regulate such diverse functions as differentiation, proliferation, apoptosis, senescence, inflammatory responses and oxygen sensing. They target a number of important signaling molecules, including kinases, phosphatases, transcription factors, ion channels, and proteins that regulate the cytoskeleton. Nox enzymes have been implicated in many different cardiovascular pathologies: atherosclerosis, hypertension, cardiac hypertrophy and remodeling, angiogenesis and collateral formation, stroke, and heart failure. In this review, we discuss in detail the biochemistry of Nox enzymes expressed in the cardiovascular system (Nox1, 2, 4, and 5), their roles in cardiovascular cell biology, and their contributions to disease development.
Collapse
Affiliation(s)
- Bernard Lassègue
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
81
|
Laurindo FRM, Pescatore LA, Fernandes DDC. Protein disulfide isomerase in redox cell signaling and homeostasis. Free Radic Biol Med 2012; 52:1954-69. [PMID: 22401853 DOI: 10.1016/j.freeradbiomed.2012.02.037] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 02/23/2012] [Accepted: 02/24/2012] [Indexed: 12/16/2022]
Abstract
Thiol proteins may potentially act as redox signaling adaptor proteins, adjusting reactive oxygen species intermediates to specific signals and redox signals to cell homeostasis. In this review, we discuss redox effects of protein disulfide isomerase (PDI), a thioredoxin superfamily oxidoreductase from the endoplasmic reticulum (ER). Abundantly expressed PDI displays ubiquity, interactions with redox and nonredox proteins, versatile effects, and several posttranslational modifications. The PDI family contains >20 members with at least some apparent complementary actions. PDI has oxidoreductase, isomerase, and chaperone effects, the last not directly dependent on its thiols. PDI is a converging hub for pathways of disulfide bond introduction into ER-processed proteins, via hydrogen peroxide-generating mechanisms involving the oxidase Ero1α, as well as hydrogen peroxide-consuming reactions involving peroxiredoxin IV and the novel peroxidases Gpx7/8. PDI is a candidate pathway for coupling ER stress to oxidant generation. Emerging information suggests a convergence between PDI and Nox family NADPH oxidases. PDI silencing prevents Nox responses to angiotensin II and inhibits Akt phosphorylation in vascular cells and parasite phagocytosis in macrophages. PDI overexpression spontaneously enhances Nox activation and expression. In neutrophils, PDI redox-dependently associates with p47phox and supports the respiratory burst. At the cell surface, PDI exerts transnitrosation, thiol reductase, and apparent isomerase activities toward targets including adhesion and matrix proteins and proteases. Such effects mediate redox-dependent adhesion, coagulation/thrombosis, immune functions, and virus internalization. The route of PDI externalization remains elusive. Such multiple redox effects of PDI may contribute to its conspicuous expression and functional role in disease, rendering PDI family members putative redox cell signaling adaptors.
Collapse
Affiliation(s)
- Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, 05403-000 São Paulo, Brazil.
| | | | | |
Collapse
|
82
|
Takac I, Schröder K, Brandes RP. The Nox family of NADPH oxidases: friend or foe of the vascular system? Curr Hypertens Rep 2012; 14:70-8. [PMID: 22071588 DOI: 10.1007/s11906-011-0238-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NADPH (nicotinamide adenine dinucleotide phosphate) oxidases are important sources of reactive oxygen species (ROS). In the vascular system, ROS can have both beneficial and detrimental effects. Under physiologic conditions, ROS are involved in signaling pathways that regulate vascular tone as well as cellular processes like proliferation, migration and differentiation. However, high doses of ROS, which are produced after induction or activation of NADPH oxidases in response to cardiovascular risk factors and inflammation, contribute to the development of endothelial dysfunction and vascular disease. In vascular cells, the NADPH oxidase isoforms Nox1, Nox2, Nox4, and Nox5 are expressed, which differ in their activity, response to stimuli, and the type of ROS released. This review focuses on the specific role of different NADPH oxidase isoforms in vascular physiology and their potential contributions to vascular diseases.
Collapse
Affiliation(s)
- Ina Takac
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt am Main, Germany
| | | | | |
Collapse
|
83
|
Qian J, Chen F, Kovalenkov Y, Pandey D, Moseley MA, Foster MW, Black SM, Venema RC, Stepp DW, Fulton DJR. Nitric oxide reduces NADPH oxidase 5 (Nox5) activity by reversible S-nitrosylation. Free Radic Biol Med 2012; 52:1806-19. [PMID: 22387196 PMCID: PMC3464050 DOI: 10.1016/j.freeradbiomed.2012.02.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 02/21/2012] [Accepted: 02/22/2012] [Indexed: 12/20/2022]
Abstract
The NADPH oxidases (Noxs) are a family of transmembrane oxidoreductases that produce superoxide and other reactive oxygen species (ROS). Nox5 was the last of the conventional Nox isoforms to be identified and is a calcium-dependent enzyme that does not depend on accessory subunits for activation. Recently, Nox5 was shown to be expressed in human blood vessels and therefore the goal of this study was to determine whether nitric oxide (NO) can modulate Nox5 activity. Endogenously produced NO potently inhibited basal and stimulated Nox5 activity and this inhibition was reversible with chronic, but not acute, exposure to L-NAME. Nox5 activity was reduced by NO donors, iNOS, and eNOS and in endothelial cells and LPS-stimulated smooth muscle cells in a manner dependent on NO concentration. ROS production was diminished by NO in an isolated enzyme activity assay replete with surplus calcium and NADPH. There was no evidence for NO-dependent changes in tyrosine nitration, glutathiolation, or phosphorylation of Nox5. In contrast, there was evidence for the increased nitrosylation of Nox5 as determined by the biotin-switch assay and mass spectrometry. Four S-nitrosylation sites were identified and of these, mutation of C694 dramatically lowered Nox5 activity, NO sensitivity, and biotin labeling. Furthermore, coexpression of the denitrosylation enzymes thioredoxin 1 and GSNO reductase prevented NO-dependent inhibition of Nox5. The potency of NO against other Nox enzymes was in the order Nox1 ≥ Nox3 > Nox5 > Nox2, whereas Nox4 was refractory. Collectively, these results suggest that endogenously produced NO can directly S-nitrosylate and inhibit the activity of Nox5.
Collapse
Affiliation(s)
- Jin Qian
- Vascular Biology Center, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Bevilacqua E, Gomes SZ, Lorenzon AR, Hoshida MS, Amarante-Paffaro AM. NADPH oxidase as an important source of reactive oxygen species at the mouse maternal-fetal interface: putative biological roles. Reprod Biomed Online 2012; 25:31-43. [PMID: 22560120 DOI: 10.1016/j.rbmo.2012.03.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 03/21/2012] [Accepted: 03/22/2012] [Indexed: 11/30/2022]
Abstract
Oxygen derivatives that comprise the large family of reactive oxygen species (ROS) are actively involved in placental biology. They are generated at the maternal-fetal interface at the level of decidual, trophoblast and mesenchymal components. In normal conditions, ROS produced in low concentrations participate in different functions as signalling molecules, regulating activation of redox-sensitive transcription factors and protein kinases involved in cell survival, proliferation and apoptosis, hence much of cell functioning. Physiological ROS generation is also associated with such defence mechanisms as phagocytosis and microbiocidal activities. In mice, particularly but not exclusively, trophoblast cells phagocytose intensively during implantation and post-implantation periods and express enzymic machinery to address a ROS-producing response to changes in the environment. The cells directly associated with ROS production are trophoblast giant cells, which mediate each and every relationship with the maternal organism. In this review, the production of ROS by the implanting mouse trophoblast is discussed, focusing on NADPH oxidase expression, regulatory mechanisms and similarities with NOX2 from phagocytes. Some of the current controversies are assessed by attempting to integrate data from studies in human trophoblast and mouse models.
Collapse
Affiliation(s)
- Estela Bevilacqua
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
85
|
Choi J. Oxidative stress, endogenous antioxidants, alcohol, and hepatitis C: pathogenic interactions and therapeutic considerations. Free Radic Biol Med 2012; 52:1135-50. [PMID: 22306508 DOI: 10.1016/j.freeradbiomed.2012.01.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 01/04/2012] [Accepted: 01/12/2012] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) is a blood-borne pathogen that was identified as an etiologic agent of non-A, non-B hepatitis in 1989. HCV is estimated to have infected at least 170 million people worldwide. The majority of patients infected with HCV do not clear the virus and become chronically infected, and chronic HCV infection increases the risk for hepatic steatosis, cirrhosis, and hepatocellular carcinoma. HCV induces oxidative/nitrosative stress from multiple sources, including inducible nitric oxide synthase, the mitochondrial electron transport chain, hepatocyte NAD(P)H oxidases, and inflammation, while decreasing glutathione. The cumulative oxidative burden is likely to promote both hepatic and extrahepatic conditions precipitated by HCV through a combination of local and more distal effects of reactive species, and clinical, animal, and in vitro studies strongly point to a role of oxidative/nitrosative stress in HCV-induced pathogenesis. Oxidative stress and hepatopathogenesis induced by HCV are exacerbated by even low doses of alcohol. Alcohol and reactive species may have other effects on hepatitis C patients such as modulation of the host immune system, viral replication, and positive selection of HCV sequence variants that contribute to antiviral resistance. This review summarizes the current understanding of redox interactions of HCV, outlining key experimental findings, directions for future research, and potential applications to therapy.
Collapse
Affiliation(s)
- Jinah Choi
- Department of Molecular Cell Biology, School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA.
| |
Collapse
|
86
|
Schröder K, Zhang M, Benkhoff S, Mieth A, Pliquett R, Kosowski J, Kruse C, Luedike P, Michaelis UR, Weissmann N, Dimmeler S, Shah AM, Brandes RP. Nox4 is a protective reactive oxygen species generating vascular NADPH oxidase. Circ Res 2012; 110:1217-25. [PMID: 22456182 DOI: 10.1161/circresaha.112.267054] [Citation(s) in RCA: 496] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE The function of Nox4, a source of vascular H(2)O(2), is unknown. Other Nox proteins were identified as mediators of endothelial dysfunction. OBJECTIVE We determined the function of Nox4 in situations of increased stress induced by ischemia or angiotensin II with global and tamoxifen-inducible Nox4(-/-) mice. METHODS AND RESULTS Nox4 was highly expressed in the endothelium and contributed to H(2)O(2) formation. Nox4(-/-) mice exhibited attenuated angiogenesis (femoral artery ligation) and PEG-catalase treatment in control mice had a similar effect. Tube formation in cultured Nox4(-/-) lung endothelial cells (LECs) was attenuated and restored by low concentrations of H(2)O(2,) whereas PEG-catalase attenuated tube formation in control LECs. Angiotensin II infusion was used as a model of oxidative stress. Compared to wild-type, aortas from inducible Nox4-deficient animals had development of increased inflammation, media hypertrophy, and endothelial dysfunction. Mechanistically, loss of Nox4 resulted in reduction of endothelial nitric oxide synthase expression, nitric oxide production, and heme oxygenase-1 (HO-1) expression, which was associated with apoptosis and inflammatory activation. HO-1 expression is controlled by Nrf-2. Accordingly, Nox4-deficient LECs exhibited reduced Nrf-2 protein level and deletion of Nox4 reduced Nrf-2 reporter gene activity. In vivo treatment with hemin, an inducer of HO-1, blocked the vascular hypertrophy induced by Nox4 deletion in the angiotensin II infusion model and carbon monoxide, the product of HO-1, blocked the Nox4-deletion-induced apoptosis in LECs. CONCLUSION Endogenous Nox4 protects the vasculature during ischemic or inflammatory stress. Different from Nox1 and Nox2, this particular NADPH oxidase therefore may have a protective vascular function.
Collapse
Affiliation(s)
- Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Frankfurt, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Streeter J, Thiel W, Brieger K, Miller Jr. FJ. Opportunity Nox: The Future of NADPH Oxidases as Therapeutic Targets in Cardiovascular Disease. Cardiovasc Ther 2012; 31:125-37. [DOI: 10.1111/j.1755-5922.2011.00310.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
88
|
Amanso AM, Griendling KK. Differential roles of NADPH oxidases in vascular physiology and pathophysiology. Front Biosci (Schol Ed) 2012; 4:1044-64. [PMID: 22202108 DOI: 10.2741/s317] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) are produced by all vascular cells and regulate the major physiological functions of the vasculature. Production and removal of ROS are tightly controlled and occur in discrete subcellular locations, allowing for specific, compartmentalized signaling. Among the many sources of ROS in the vessel wall, NADPH oxidases are implicated in physiological functions such as control of vasomotor tone, regulation of extracellular matrix and phenotypic modulation of vascular smooth muscle cells. They are involved in the response to injury, whether as an oxygen sensor during hypoxia, as a regulator of protein processing, as an angiogenic stimulus, or as a mechanism of wound healing. These enzymes have also been linked to processes leading to disease development, including migration, proliferation, hypertrophy, apoptosis and autophagy. As a result, NADPH oxidases participate in atherogenesis, systemic and pulmonary hypertension and diabetic vascular disease. The role of ROS in each of these processes and diseases is complex, and a more full understanding of the sources, targets, cell-specific responses and counterbalancing mechanisms is critical for the rational development of future therapeutics.
Collapse
Affiliation(s)
- Angelica M Amanso
- Department of Medicine, Division of Cardiology, Emory University, Division of Cardiology, Atlanta, GA 30322, USA
| | | |
Collapse
|
89
|
Da Silva M, Jaggers GK, Verstraeten SV, Erlejman AG, Fraga CG, Oteiza PI. Large procyanidins prevent bile-acid-induced oxidant production and membrane-initiated ERK1/2, p38, and Akt activation in Caco-2 cells. Free Radic Biol Med 2012; 52:151-9. [PMID: 22074817 DOI: 10.1016/j.freeradbiomed.2011.10.436] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 09/30/2011] [Accepted: 10/07/2011] [Indexed: 11/24/2022]
Abstract
Procyanidins are oligomers of flavanol subunits present in large amounts in fruits and vegetables. Their consumption is associated with health benefits against colonic inflammation and colorectal cancer (CRC). Large procyanidins (with more than three subunits) are not absorbed by intestinal epithelial cells but could exert biological actions through their interactions with the cell membrane. This study investigated the capacity of hexameric procyanidins (Hex) to prevent oncogenic events initiated by deoxycholic acid (DCA), a secondary bile acid linked to the promotion of CRC. Hex interacted with Caco-2 cell membranes preferentially at the water-lipid interface. Hex (2.5-20 μM) inhibited DCA-triggered increase in cellular calcium, NADPH oxidase activation, and oxidant production. DCA promoted the activation of protein kinase B (Akt), of the mitogen-activated protein kinases ERK1/2 and p38, and of the downstream transcription factor AP-1. This activation was not triggered by calcium or oxidant increases. Hex caused a dose-dependent inhibition of DCA-mediated activation of all these signals. DCA also triggered alterations in the cell monolayer morphology and apoptotic cell death, events that were delayed by Hex. The capacity of large procyanidins to interact with the cell membrane and prevent those cell membrane-associated events can in part explain the beneficial effects of procyanidins on CRC.
Collapse
Affiliation(s)
- Mathieu Da Silva
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | | | | | | | | | | |
Collapse
|
90
|
Ullevig S, Zhao Q, Lee CF, Seok Kim H, Zamora D, Asmis R. NADPH oxidase 4 mediates monocyte priming and accelerated chemotaxis induced by metabolic stress. Arterioscler Thromb Vasc Biol 2011; 32:415-26. [PMID: 22095986 DOI: 10.1161/atvbaha.111.238899] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Metabolic disorders increase monocyte chemoattractant protein-1 (MCP-1)-induced monocyte chemotaxis in mice. The goal of this study was to determine the molecular mechanisms responsible for the enhanced responsiveness of monocytes to chemoattractants induced by metabolic stress. METHODS AND RESULTS Chronic exposure of monocytes to diabetic conditions induced by human LDL plus high D-glucose concentrations (LDL+HG) promoted NADPH Oxidase 4 (Nox4) expression, increased intracellular H(2)O(2) formation, stimulated protein S-glutathionylation, and increased chemotaxis in response to MCP-1, platelet-derived growth factor B, and RANTES. Both H(2)O(2) added exogenously and overexpression of Nox4 mimicked LDL+HG-induced monocyte priming, whereas Nox4 knockdown protected monocytes against metabolic stress-induced priming and accelerated chemotaxis. Exposure of monocytes to LDL+HG promoted the S-glutathionylation of actin, decreased the F-actin/G-actin ratio, and increased actin remodeling in response to MCP-1. Preventing LDL+HG-induced protein S-glutathionylation by overexpressing glutaredoxin 1 prevented monocyte priming and normalized monocyte chemotaxis in response to MCP-1. Induction of hypercholesterolemia and hyperglycemia in C57BL/6 mice promoted Nox4 expression and protein S-glutathionylation in macrophages, and increased macrophage recruitment into MCP-1-loaded Matrigel plugs implanted subcutaneous in these mice. CONCLUSIONS By increasing actin-S-glutathionylation and remodeling, metabolic stress primes monocytes for chemoattractant-induced transmigration and recruitment to sites of vascular injury. This Nox4-dependent process provides a novel mechanism through which metabolic disorders promote atherogenesis.
Collapse
Affiliation(s)
- Sarah Ullevig
- Clinical Laboratory Sciences, School of Health Professions, University of Texas Health Science Center at San Antonio, 8403 Floyd Curl Drive, MC 6246, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | |
Collapse
|
91
|
Ghouleh IA, Khoo NK, Knaus UG, Griendling KK, Touyz RM, Thannickal VJ, Barchowsky A, Nauseef WM, Kelley EE, Bauer PM, Darley-Usmar V, Shiva S, Cifuentes-Pagano E, Freeman BA, Gladwin MT, Pagano PJ. Oxidases and peroxidases in cardiovascular and lung disease: new concepts in reactive oxygen species signaling. Free Radic Biol Med 2011; 51:1271-88. [PMID: 21722728 PMCID: PMC3205968 DOI: 10.1016/j.freeradbiomed.2011.06.011] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 06/06/2011] [Accepted: 06/07/2011] [Indexed: 12/17/2022]
Abstract
Reactive oxygen species (ROS) are involved in numerous physiological and pathophysiological responses. Increasing evidence implicates ROS as signaling molecules involved in the propagation of cellular pathways. The NADPH oxidase (Nox) family of enzymes is a major source of ROS in the cell and has been related to the progression of many diseases and even environmental toxicity. The complexity of this family's effects on cellular processes stems from the fact that there are seven members, each with unique tissue distribution, cellular localization, and expression. Nox proteins also differ in activation mechanisms and the major ROS detected as their product. To add to this complexity, mounting evidence suggests that other cellular oxidases or their products may be involved in Nox regulation. The overall redox and metabolic status of the cell, specifically the mitochondria, also has implications on ROS signaling. Signaling of such molecules as electrophilic fatty acids has an impact on many redox-sensitive pathologies and thus, as anti-inflammatory molecules, contributes to the complexity of ROS regulation. This review is based on the proceedings of a recent international Oxidase Signaling Symposium at the University of Pittsburgh's Vascular Medicine Institute and Department of Pharmacology and Chemical Biology and encompasses further interaction and discussion among the presenters.
Collapse
Affiliation(s)
- Imad Al Ghouleh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Nicholas K.H. Khoo
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| | - Ulla G. Knaus
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Kathy K. Griendling
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA
| | - Rhian M. Touyz
- Ottawa Hospital Research Institute, Univ of Ottawa, Ottawa, Ontario, Canada
| | - Victor J. Thannickal
- Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Aaron Barchowsky
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - William M. Nauseef
- Inflammation Program, Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
- Veterans Administration Medical Center, Iowa City, IA
| | - Eric E. Kelley
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA
| | - Phillip M. Bauer
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Bruce A. Freeman
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| | - Mark T. Gladwin
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
- Department of Pulmonary, Allergy & Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Patrick J. Pagano
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
92
|
Craige SM, Chen K, Pei Y, Li C, Huang X, Chen C, Shibata R, Sato K, Walsh K, Keaney JF. NADPH oxidase 4 promotes endothelial angiogenesis through endothelial nitric oxide synthase activation. Circulation 2011; 124:731-40. [PMID: 21788590 DOI: 10.1161/circulationaha.111.030775] [Citation(s) in RCA: 221] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
UNLABELLED BACKGROUND- Reactive oxygen species serve signaling functions in the vasculature, and hypoxia has been associated with increased reactive oxygen species production. NADPH oxidase 4 (Nox4) is a reactive oxygen species-producing enzyme that is highly expressed in the endothelium, yet its specific role is unknown. We sought to determine the role of Nox4 in the endothelial response to hypoxia. METHODS AND RESULTS Hypoxia induced Nox4 expression both in vitro and in vivo and overexpression of Nox4 was sufficient to promote endothelial proliferation, migration, and tube formation. To determine the in vivo relevance of our observations, we generated transgenic mice with endothelial-specific Nox4 overexpression using the vascular endothelial cadherin promoter (VECad-Nox4 mice). In vivo, the VECad-Nox4 mice had accelerated recovery from hindlimb ischemia and enhanced aortic capillary sprouting. Because endothelial nitric oxide synthase (eNOS) is involved in endothelial angiogenic responses and eNOS is activated by reactive oxygen species, we probed the effect of Nox4 on eNOS. In cultured endothelial cells overexpressing Nox4, we observed a significant increase in eNOS protein expression and activity. To causally address the link between eNOS and Nox4, we crossed our transgenic Nox4 mice with eNOS(-/-) mice. Aortas from these mice did not demonstrate enhanced aortic sprouting, and VECad-Nox4 mice on the eNOS(-/-) background did not demonstrate enhanced recovery from hindlimb ischemia. CONCLUSIONS Collectively, we demonstrate that augmented endothelial Nox4 expression promotes angiogenesis and recovery from hypoxia in an eNOS-dependent manner.
Collapse
Affiliation(s)
- Siobhan M Craige
- University of Massachusetts Medical School, Department of Medicine/Division of Cardiovascular Medicine, Worcester, MA 01605, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Brewer AC, Murray TVA, Arno M, Zhang M, Anilkumar NP, Mann GE, Shah AM. Nox4 regulates Nrf2 and glutathione redox in cardiomyocytes in vivo. Free Radic Biol Med 2011; 51:205-15. [PMID: 21554947 PMCID: PMC3112490 DOI: 10.1016/j.freeradbiomed.2011.04.022] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 03/21/2011] [Accepted: 04/11/2011] [Indexed: 12/16/2022]
Abstract
NADPH oxidase-4 (Nox4) is an important modulator of redox signaling that is inducible at the level of transcriptional expression in multiple cell types. By contrast to other Nox enzymes, Nox4 is continuously active without requiring stimulation. We reported recently that expression of Nox4 is induced in the adult heart as an adaptive stress response to pathophysiological insult. To elucidate the potential downstream target(s) regulated by Nox4, we performed a microarray screen to assess the transcriptomes of transgenic (tg) mouse hearts in which Nox4 was overexpressed. The screen revealed a significant increase in the expression of many antioxidant and detoxifying genes regulated by Nrf2 in tg compared to wild-type (wt) mouse hearts, and this finding was subsequently confirmed by Q-PCR. Expression of glutathione biosynthetic and recycling enzymes was increased in tg hearts and associated with higher levels of both GSH and the ratio of reduced:oxidised GSH, compared to wt hearts. The increases in expression of the antioxidant genes and the changes in glutathione redox effected by Nox4 were ablated in an Nrf2-null genetic background. These data therefore demonstrate that Nox4 can activate the Nrf2-regulated pathway, and suggest a potential role for Nox4 in the regulation of GSH redox in cardiomyocytes.
Collapse
Key Words
- nadph, nicotinamide adenine dinucleotide phosphate
- nrf2, nf-e2-related factor 2
- q-pcr, quantitative polymerase chain reaction
- er, endoplasmic reticulum
- eb, embryoid body
- αmhc, α myosin heavy chain
- βmhc, β myosin heavy chain
- mlc2v, myosin regulatory light chain 2
- rt, reverse transcriptase
- dtt, dithiothreitol
- page, polyacrylamide gel electrophoresis
- ecl, enhanced chemiluminescence
- pbs, phosphate-buffered saline
- pvdf, polyvinylidene difluoride
- sem, standard error of the mean
- elisa, enzyme-linked immunosorbent serologic assay
- nox4
- nrf2
- cardiomyocytes
- glutathione
- reactive oxygen species
Collapse
Affiliation(s)
- Alison C Brewer
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London, UK.
| | | | | | | | | | | | | |
Collapse
|
94
|
Ray R, Murdoch CE, Wang M, Santos CX, Zhang M, Alom-Ruiz S, Anilkumar N, Ouattara A, Cave AC, Walker SJ, Grieve DJ, Charles RL, Eaton P, Brewer AC, Shah AM. Endothelial Nox4 NADPH Oxidase Enhances Vasodilatation and Reduces Blood Pressure In Vivo. Arterioscler Thromb Vasc Biol 2011; 31:1368-76. [DOI: 10.1161/atvbaha.110.219238] [Citation(s) in RCA: 252] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective—
Increased reactive oxygen species (ROS) production is involved in the pathophysiology of endothelial dysfunction. NADPH oxidase-4 (Nox4) is a ROS-generating enzyme expressed in the endothelium, levels of which increase in pathological settings. Recent studies indicate that it generates predominantly hydrogen peroxide (H
2
O
2
), but its role in vivo remains unclear.
Methods and Results—
We generated transgenic mice with endothelium-targeted Nox4 overexpression (Tg) to study the in vivo role of Nox4. Tg demonstrated significantly greater acetylcholine- or histamine-induced vasodilatation than wild-type littermates. This resulted from increased H
2
O
2
production and H
2
O
2
-induced hyperpolarization but not altered nitric oxide bioactivity. Tg had lower systemic blood pressure than wild-type littermates, which was normalized by antioxidants.
Conclusion—
Endothelial Nox4 exerts potentially beneficial effects on vasodilator function and blood pressure that are attributable to H
2
O
2
production. These effects contrast markedly with those reported for Nox1 and Nox2, which involve superoxide-mediated inactivation of nitric oxide. Our results suggest that therapeutic strategies to modulate ROS production in vascular disease may need to separately target individual Nox isoforms.
Collapse
Affiliation(s)
- Robin Ray
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Colin E. Murdoch
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Minshu Wang
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Celio X. Santos
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Min Zhang
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Sara Alom-Ruiz
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Narayana Anilkumar
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Alexandre Ouattara
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Alison C. Cave
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Simon J. Walker
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - David J. Grieve
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Rebecca L. Charles
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Philip Eaton
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Alison C. Brewer
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Ajay M. Shah
- From the Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| |
Collapse
|
95
|
Chen F, Pandey D, Chadli A, Catravas JD, Chen T, Fulton DJR. Hsp90 regulates NADPH oxidase activity and is necessary for superoxide but not hydrogen peroxide production. Antioxid Redox Signal 2011; 14:2107-19. [PMID: 21194376 PMCID: PMC3085945 DOI: 10.1089/ars.2010.3669] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The goal of this study was to identify whether heat-shock protein 90 (Hsp90) regulates the production of superoxide and other reactive oxygen species from the NADPH oxidases (Nox). We found that pharmacological and genetic inhibition of Hsp90 directly reduced Nox5-derived superoxide without secondarily modifying signaling events. Coimmunoprecipitation and bioluminescence resonance energy transfer studies suggest that the C-terminus of Nox5 binds to Hsp90. Long-term Hsp90 inhibition reduced Nox5 expression and provides further evidence that Nox5 is an Hsp90 client protein. Inhibitors of Hsp90 also reduced superoxide from Nox1, Nox2 (neutrophils), and Nox3. However, Nox4, which emits only hydrogen peroxide, was unaffected by Hsp90 inhibitors. Hydrogen peroxide production from the other Nox enzymes was not affected by short-term inhibition of Hsp90, but long-term inhibition reduced production of all reactive oxygen species coincident with loss of enzyme expression. Expression of chimeric Nox enzymes consisting of N-terminal Nox1 or Nox3 and C-terminal Nox4 resulted in only hydrogen peroxide formation that was insensitive to Hsp90 inhibitors. We conclude that Hsp90 binds to the C-terminus of Noxes1-3 and 5 and is necessary for enzyme stability and superoxide production. Hsp90 does not bind to the C-terminus of Nox4 and is not required for hydrogen peroxide formation.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Sciences, Xi'an Jiaotong University School of Medicine, Xi'an Shaanxi, P.R. China
| | | | | | | | | | | |
Collapse
|
96
|
Santos CX, Anilkumar N, Zhang M, Brewer AC, Shah AM. Redox signaling in cardiac myocytes. Free Radic Biol Med 2011; 50:777-93. [PMID: 21236334 PMCID: PMC3049876 DOI: 10.1016/j.freeradbiomed.2011.01.003] [Citation(s) in RCA: 237] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 01/05/2011] [Accepted: 01/05/2011] [Indexed: 02/07/2023]
Abstract
The heart has complex mechanisms that facilitate the maintenance of an oxygen supply-demand balance necessary for its contractile function in response to physiological fluctuations in workload as well as in response to chronic stresses such as hypoxia, ischemia, and overload. Redox-sensitive signaling pathways are centrally involved in many of these homeostatic and stress-response mechanisms. Here, we review the main redox-regulated pathways that are involved in cardiac myocyte excitation-contraction coupling, differentiation, hypertrophy, and stress responses. We discuss specific sources of endogenously generated reactive oxygen species (e.g., mitochondria and NADPH oxidases of the Nox family), the particular pathways and processes that they affect, the role of modulators such as thioredoxin, and the specific molecular mechanisms that are involved-where this knowledge is available. A better understanding of this complex regulatory system may allow the development of more specific therapeutic strategies for heart diseases.
Collapse
Key Words
- aif, apoptosis-inducing factor
- arc, apoptosis repressor with caspase recruitment domain
- camkii, calmodulin kinase ii
- ctgf, connective tissue growth factor
- eb, embryoid body
- ecc, excitation–contraction coupling
- er, endoplasmic reticulum
- es, embryonic stem
- etc, electron transport chain
- g6pdh, glucose-6-phosphate dehydrogenase
- gpcr, g-protein-coupled receptor
- hdac, histone deacetylase
- hif, hypoxia-inducible factor
- mao-a, monoamine oxidase-a
- mi, myocardial infarction
- mmp, matrix metalloproteinase
- mptp, mitochondrial permeability transition pore
- mtdna, mitochondrial dna
- ncx, na/ca exchanger
- nos, nitric oxide synthase
- phd, prolyl hydroxylase dioxygenase
- pka, protein kinase a
- pkc, protein kinase c
- pkg, protein kinase g
- ros, reactive oxygen species
- ryr, ryanodine receptor
- serca, sarcoplasmic reticulum calcium atpase
- sr, sarcoplasmic reticulum
- trx1, thioredoxin1
- tnfα, tumor necrosis factor-α
- vegf, vascular endothelial growth factor
- cardiac myocyte
- reactive oxygen species
- redox signaling
- hypertrophy
- heart failure
- nadph oxidase
- mitochondria
- free radicals
Collapse
|
97
|
New insight into the Nox4 subcellular localization in HEK293 cells: First monoclonal antibodies against Nox4. Biochimie 2011; 93:457-68. [DOI: 10.1016/j.biochi.2010.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 11/03/2010] [Indexed: 11/17/2022]
|
98
|
Takac I, Schröder K, Zhang L, Lardy B, Anilkumar N, Lambeth JD, Shah AM, Morel F, Brandes RP. The E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4. J Biol Chem 2011; 286:13304-13. [PMID: 21343298 DOI: 10.1074/jbc.m110.192138] [Citation(s) in RCA: 414] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In contrast to the NADPH oxidases Nox1 and Nox2, which generate superoxide (O(2)(·-)), Nox4 produces hydrogen peroxide (H(2)O(2)). We constructed chimeric proteins and mutants to address the protein region that specifies which reactive oxygen species is produced. Reactive oxygen species were measured with luminol/horseradish peroxidase and Amplex Red for H(2)O(2) versus L-012 and cytochrome c for O(2)(·-). The third extracytosolic loop (E-loop) of Nox4 is 28 amino acids longer than that of Nox1 or Nox2. Deletion of E-loop amino acids only present in Nox4 or exchange of the two cysteines in these stretches switched Nox4 from H(2)O(2) to O(2)(·-) generation while preserving expression and intracellular localization. In the presence of an NO donor, the O(2)()-producing Nox4 mutants, but not wild-type Nox4, generated peroxynitrite, excluding artifacts of the detection system as the apparent origin of O(2)(·-). In Cos7 cells, in which Nox4 partially localizes to the plasma membrane, an antibody directed against the E-loop decreased H(2)O(2) but increased O(2)(·-) formation by Nox4 without affecting Nox1-dependent O(2)(·-) formation. The E-loop of Nox4 but not Nox1 and Nox2 contains a highly conserved histidine that could serve as a source for protons to accelerate spontaneous dismutation of superoxide to form H(2)O(2). Mutation of this but not of four other conserved histidines also switched Nox4 from H(2)O(2) to O(2)(·-) formation. Thus, H(2)O(2) formation is an intrinsic property of Nox4 that involves its E-loop. The structure of the E-loop may hinder O(2)(·-) egress and/or provide a source for protons, allowing dismutation to form H(2)O(2).
Collapse
Affiliation(s)
- Ina Takac
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60596 Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Spencer NY, Yan Z, Boudreau RL, Zhang Y, Luo M, Li Q, Tian X, Shah AM, Davisson RL, Davidson B, Banfi B, Engelhardt JF. Control of hepatic nuclear superoxide production by glucose 6-phosphate dehydrogenase and NADPH oxidase-4. J Biol Chem 2011; 286:8977-87. [PMID: 21212270 DOI: 10.1074/jbc.m110.193821] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Redox-regulated signal transduction is coordinated by spatially controlled production of reactive oxygen species within subcellular compartments. The nucleus has long been known to produce superoxide (O(2)(·-)); however, the mechanisms that control this function remain largely unknown. We have characterized molecular features of a nuclear superoxide-producing system in the mouse liver. Using electron paramagnetic resonance, we investigated whether several NADPH oxidases (NOX1, 2, and 4) and known activators of NOX (Rac1, Rac2, p22(phox), and p47(phox)) contribute to nuclear O(2)(·-) production in isolated hepatic nuclei. Our findings demonstrate that NOX4 most significantly contributes to hepatic nuclear O(2)(·-) production that utilizes NADPH as an electron donor. Although NOX4 protein immunolocalized to both nuclear membranes and intranuclear inclusions, fluorescent detection of NADPH-dependent nuclear O(2)(·-) predominantly localized to the perinuclear space. Interestingly, NADP(+) and G6P also induced nuclear O(2)(·-) production, suggesting that intranuclear glucose-6-phosphate dehydrogenase (G6PD) can control NOX4 activity through nuclear NADPH production. Using G6PD mutant mice and G6PD shRNA, we confirmed that reductions in nuclear G6PD enzyme decrease the ability of hepatic nuclei to generate O(2)(·-) in response to NADP(+) and G6P. NOX4 and G6PD protein were also observed in overlapping microdomains within the nucleus. These findings provide new insights on the metabolic pathways for substrate regulation of nuclear O(2)(·-) production by NOX4.
Collapse
Affiliation(s)
- Netanya Y Spencer
- Department of Anatomy and Cell Biology, College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Zhang N, Andresen BT, Zhang C. Inflammation and reactive oxygen species in cardiovascular disease. World J Cardiol 2010; 2:408-10. [PMID: 21191541 PMCID: PMC3011135 DOI: 10.4330/wjc.v2.i12.408] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 09/04/2010] [Accepted: 09/11/2010] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) have long been proposed to be mediators of experimental cardiovascular pathology. There is also a wealth of data indicating that ROS are involved in clinical cardiovascular pathology. However, multiple clinical studies have shown little benefit from anti-oxidant treatments, whereas nearly all experimental studies have shown a marked effect of anti-oxidant therapy. One reason for this discrepancy is that ROS are produced through multiple different mechanisms of which some are clinically beneficial; thus, in a defined experimental system where predominately pathological ROS are generated does not mimic a clinical setting where there are likely to be multiple ROS generating systems producing beneficial and pathological ROS. Simple inhibition of ROS would not be expected to have the same result in these two situations; ergo, it is important to understand the molecular mechanism underlying the production of ROS so that clinical treatments can be tailored to target the pathological production of ROS. One such example of this in cardiovascular biology is tissue specific inflammation-mediated ROS generation. This and the following series of articles discuss the current understanding of the role of ROS in cardiovascular disease, specifically focusing on the molecular mechanisms of ROS generation and the actions of ROS within the cardiovascular system. Although there are still many areas with regard to the effects of ROS in the cardiovascular system that are not completely understood, there is a wealth of data suggesting that blocking pathological ROS production is likely to have beneficial clinical effects compared to traditional anti-oxidants.
Collapse
Affiliation(s)
- Nannan Zhang
- Nannan Zhang, Cuihua Zhang, Department of Internal Medicine1, Medical Pharmacology and Physiology, Nutrition and Exercise Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 652114, United States
| | | | | |
Collapse
|