101
|
Floriano-Sánchez E, Castro-Marín M, Cárdenas-Rodríguez N, Lara-Padilla E. Evaluación de la expresión de la subunidad p22 phox de la NADPH oxidasa (NOX) en cáncer de próstata e hiperplasia prostática benigna: estudio comparativo. Actas Urol Esp 2010. [DOI: 10.1016/j.acuro.2010.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
102
|
Nitti M, Furfaro AL, Cevasco C, Traverso N, Marinari UM, Pronzato MA, Domenicotti C. PKC delta and NADPH oxidase in retinoic acid-induced neuroblastoma cell differentiation. Cell Signal 2010; 22:828-35. [PMID: 20074641 DOI: 10.1016/j.cellsig.2010.01.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 12/10/2009] [Accepted: 01/05/2010] [Indexed: 01/24/2023]
Abstract
The role of reactive oxygen species (ROS) in the regulation of signal transduction processes has been well established in many cell types and recently the fine tuning of redox signalling in neurons received increasing attention. With regard to this, the involvement of NADPH oxidase (NOX) in neuronal pathophysiology has been proposed but deserves more investigation. In the present study, we used SH-SY5Y neuroblastoma cells to analyse the role of NADPH oxidase in retinoic acid (RA)-induced differentiation, pointing out the involvement of protein kinase C (PKC) delta in the activation of NOX. Retinoic acid induces neuronal differentiation as revealed by the increased expression of MAP2, the decreased cell doubling rate, and the gain in neuronal morphological features and these events are accompanied by the increased expression level of PKC delta and p67(phox), one of the components of NADPH oxidase. Using DPI to inhibit NOX activity we show that retinoic acid acts through this enzyme to induce morphological changes linked to the differentiation. Moreover, using rottlerin to inhibit PKC delta or transfection experiments to overexpress it, we show that retinoic acid acts through this enzyme to induce MAP2 expression and to increase p67(phox) membrane translocation leading to NADPH oxidase activation. These findings identify the activation of PKC delta and NADPH oxidase as crucial steps in RA-induced neuroblastoma cell differentiation.
Collapse
Affiliation(s)
- Mariapaola Nitti
- Department of Experimental Medicine, University of Genoa, Italy.
| | | | | | | | | | | | | |
Collapse
|
103
|
Jacob A, Hack B, Chiang E, Garcia JGN, Quigg RJ, Alexander JJ. C5a alters blood-brain barrier integrity in experimental lupus. FASEB J 2010; 24:1682-8. [PMID: 20065106 DOI: 10.1096/fj.09-138834] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The blood-brain barrier (BBB) is a crucial anatomic location in the brain. Its dysfunction complicates many neurodegenerative diseases, from acute conditions, such as sepsis, to chronic diseases, such as systemic lupus erythematosus (SLE). Several studies suggest an altered BBB in lupus, but the underlying mechanism remains unknown. In the current study, we observed a definite loss of BBB integrity in MRL/MpJ-Tnfrsf6(lpr) (MRL/lpr) lupus mice by IgG infiltration into brain parenchyma. In line with this result, we examined the role of complement activation, a key event in this setting, in maintenance of BBB integrity. Complement activation generates C5a, a molecule with multiple functions. Because the expression of the C5a receptor (C5aR) is significantly increased in brain endothelial cells treated with lupus serum, the study focused on the role of C5a signaling through its G-protein-coupled receptor C5aR in brain endothelial cells, in a lupus setting. Reactive oxygen species production increased significantly in endothelial cells, in both primary cells and the bEnd3 cell line treated with lupus serum from MRL/lpr mice, compared with those treated with control serum from MRL(+/+) mice. In addition, increased permeability monitored by changes in transendothelial electrical resistance, cytoskeletal remodeling caused by actin fiber rearrangement, and increased iNOS mRNA expression were observed in bEnd3 cells. These disruptive effects were alleviated by pretreating cells with a C5a receptor antagonist (C5aRant) or a C5a antibody. Furthermore, the structural integrity of the vasculature in MRL/lpr brain was maintained by C5aR inhibition. These results demonstrate the regulation of BBB integrity by the complement system in a neuroinflammatory setting. For the first time, a novel role of C5a in the maintenance of BBB integrity is identified and the potential of C5a/C5aR blockade highlighted as a promising therapeutic strategy in SLE and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexander Jacob
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
104
|
Liu RM, Gaston Pravia KA. Oxidative stress and glutathione in TGF-beta-mediated fibrogenesis. Free Radic Biol Med 2010; 48:1-15. [PMID: 19800967 PMCID: PMC2818240 DOI: 10.1016/j.freeradbiomed.2009.09.026] [Citation(s) in RCA: 324] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 09/24/2009] [Accepted: 09/26/2009] [Indexed: 12/16/2022]
Abstract
Transforming growth factor beta (TGF-beta) is the most potent and ubiquitous profibrogenic cytokine, and its expression is increased in almost all the fibrotic diseases and in experimental fibrosis models. TGF-beta increases reactive oxygen species production and decreases the concentration of glutathione (GSH), the most abundant intracellular free thiol and an important antioxidant, which mediates many of the fibrogenic effects of TGF-beta in various types of cells. A decreased GSH concentration is also observed in human fibrotic diseases and in experimental fibrosis models. Although the biological significance of GSH depletion in the development of fibrosis remains obscure, GSH and N-acetylcysteine, a precursor of GSH, have been used in clinics for the treatment of fibrotic diseases. This review summarizes recent findings in the field to address the potential mechanism whereby oxidative stress mediates fibrogenesis induced by TGF-beta and the potential therapeutic value of antioxidant treatment in fibrotic diseases.
Collapse
Affiliation(s)
- R-M Liu
- Department of Environmental Health Sciences, School of Public Health, Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | |
Collapse
|
105
|
Burhans WC, Heintz NH. The cell cycle is a redox cycle: linking phase-specific targets to cell fate. Free Radic Biol Med 2009; 47:1282-93. [PMID: 19486941 DOI: 10.1016/j.freeradbiomed.2009.05.026] [Citation(s) in RCA: 261] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 05/19/2009] [Accepted: 05/22/2009] [Indexed: 11/22/2022]
Abstract
Reactive oxygen species (ROS) regulate the strength and duration of signaling through redox-dependent signal transduction pathways via the cyclic oxidation/reduction of cysteine residues in kinases, phosphatases, and other regulatory factors. Signaling circuits may be segregated in organelles or other subcellular domains with distinct redox states, permitting them to respond independently to changes in the oxidation state of two major thiol reductants, glutathione and thioredoxin. Studies in yeast, and in complex eukaryotes, show that oscillations in oxygen consumption, energy metabolism, and redox state are intimately integrated with cell cycle progression. Because signaling pathways play specific roles in different phases of the cell cycle and the hierarchy of redox-dependent regulatory checkpoints changes during cell cycle progression, the effects of ROS on cell fate vary during the cell cycle. In G1, ROS stimulate mitogenic pathways that control the activity of cyclin-dependent kinases (CDKs) and phosphorylation of the retinoblastoma protein (pRB), thereby regulating S-phase entry. In response to oxidative stress, Nrf2 and Foxo3a promote cell survival by inducing the expression of antioxidant enzymes and factors involved in cell cycle withdrawal, such as the cyclin-dependent kinase inhibitor (CKI) p27. In S phase, ROS induce S-phase arrest via PP2A-dependent dephosphorylation of pRB. In precancerous cells, unconstrained mitogenic signaling by activated oncogenes induces replication stress in S phase, which activates the DNA-damage response and induces cell senescence. A number of studies suggest that interactions of ROS with the G1 CDK/CKI network play a fundamental role in senescence, which is considered a barrier to tumorigenesis. Adaptive responses and loss of checkpoint proteins such as p53 and p16(INK4a) allow tumor cells to tolerate constitutive mitogenic signaling and enhanced production of ROS, leading to altered redox status in many fully transformed cells. Alterations in oxidant and energy metabolism of cancer cells have emerged as fertile ground for new therapeutic targets. The present challenge is to identify redox-dependent targets relevant to each cell cycle phase, to understand how these targets control fate decisions, and to describe the mechanisms that link metabolism to cell cycle progression.
Collapse
Affiliation(s)
- William C Burhans
- Department of Molecular & Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | |
Collapse
|
106
|
Nair DG, Funk CD. A cell-based assay for screening lipoxygenase inhibitors. Prostaglandins Other Lipid Mediat 2009; 90:98-104. [PMID: 19804839 DOI: 10.1016/j.prostaglandins.2009.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 09/21/2009] [Accepted: 09/24/2009] [Indexed: 12/11/2022]
Abstract
Lipoxygenases (LOX) form a family of lipid peroxidizing enzymes within the plant and animal kingdoms. In humans, six functional lipoxygenase isoforms have been identified. 5-LOX, "platelet-type" 12-LOX (p12-LOX) and 15-LOX type 1 (15-LOX1), originally identified in leukocytes, platelets, and reticulocytes, respectively, generate lipid mediators involved in host cellular functions and in the pathophysiology of asthma, cardiovascular diseases, and cancer. The pharmaceutical industry has reinvigorated their programs to develop novel LOX inhibitors in view of recent findings. However, high throughput LOX screening assays to test novel agents against these intracellular enzymes are limited. We describe a cell-based 96-well microplate fluorescence assay tested against several existing LOX inhibitors, and validate the assay by comparing known IC(50) values and HPLC analysis, which may provide a useful screen for novel LOX inhibitors.
Collapse
Affiliation(s)
- Dileep G Nair
- Department of Physiology and Biochemistry, Queen's University, Kingston, ON, Canada
| | | |
Collapse
|
107
|
Zhou G, Dada LA, Wu M, Kelly A, Trejo H, Zhou Q, Varga J, Sznajder JI. Hypoxia-induced alveolar epithelial-mesenchymal transition requires mitochondrial ROS and hypoxia-inducible factor 1. Am J Physiol Lung Cell Mol Physiol 2009; 297:L1120-30. [PMID: 19801454 DOI: 10.1152/ajplung.00007.2009] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Patients with acute lung injury develop hypoxia, which may lead to lung dysfunction and aberrant tissue repair. Recent studies have suggested that epithelial-mesenchymal transition (EMT) contributes to pulmonary fibrosis. We sought to determine whether hypoxia induces EMT in alveolar epithelial cells (AEC). We found that hypoxia induced the expression of alpha-smooth muscle actin (alpha-SMA) and vimentin and decreased the expression of E-cadherin in transformed and primary human, rat, and mouse AEC, suggesting that hypoxia induces EMT in AEC. Both severe hypoxia and moderate hypoxia induced EMT. The reactive oxygen species (ROS) scavenger Euk-134 prevented hypoxia-induced EMT. Moreover, hypoxia-induced expression of alpha-SMA and vimentin was prevented in mitochondria-deficient rho(0) cells, which are incapable of ROS production during hypoxia. CoCl(2) and dimethyloxaloylglycine, two compounds that stabilize hypoxia-inducible factor (HIF)-alpha under normoxia, failed to induce alpha-SMA expression in AEC. Furthermore, overexpression of constitutively active HIF-1alpha did not induce alpha-SMA. However, loss of HIF-1alpha or HIF-2alpha abolished induction of alpha-SMA mRNA during hypoxia. Hypoxia increased the levels of transforming growth factor (TGF)-beta1, and preincubation of AEC with SB431542, an inhibitor of the TGF-beta1 type I receptor kinase, prevented the hypoxia-induced EMT, suggesting that the process was TGF-beta1 dependent. Furthermore, both ROS and HIF-alpha were necessary for hypoxia-induced TGF-beta1 upregulation. Accordingly, we have provided evidence that hypoxia induces EMT of AEC through mitochondrial ROS, HIF, and endogenous TGF-beta1 signaling.
Collapse
|
108
|
Subcellular localization of Nox4 and regulation in diabetes. Proc Natl Acad Sci U S A 2009; 106:14385-90. [PMID: 19706525 DOI: 10.1073/pnas.0906805106] [Citation(s) in RCA: 390] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress is implicated in human diseases. Some of the oxidative pathways are harbored in the mitochondria. NAD(P)H oxidases have been identified not only in phagocytic but also in somatic cells. Nox4 is the most ubiquitous of these oxidases and is a major source of reactive oxygen species (ROS) in many cell types and in kidney tissue of diabetic animals. We generated specific Nox4 antibodies, and found that Nox4 localizes to mitochondria. (i) Immunoblot analysis in cultured mesangial cells and kidney cortex revealed that Nox4 is present in crude mitochondria, in mitochondria-enriched heavy fractions, and in purified mitochondria; (ii) immunofluorescence confocal microscopy also revealed that Nox4 localizes with the mitochondrial marker Mitotracker; and (iii) the mitochondrial localization prediction program MitoProt indicated that the probability score for Nox4 is identical to mitochondrial protein cytochrome c oxidase subunit IV. We also show that in purified mitochondria, siRNA-mediated knockdown of Nox4 significantly reduces NADPH oxidase activity in pure mitochondria and blocks glucose-induced mitochondrial superoxide generation. In a rat model of diabetes, mitochondrial Nox4 expression is increased in kidney cortex. Our data provide evidence that a functional Nox4 is present and regulated in mitochondria, indicating the existence of a previously undescribed source of ROS in this organelle.
Collapse
|
109
|
Churchman AT, Anwar AA, Li FYL, Sato H, Ishii T, Mann GE, Siow RCM. Transforming growth factor-beta1 elicits Nrf2-mediated antioxidant responses in aortic smooth muscle cells. J Cell Mol Med 2009; 13:2282-2292. [PMID: 19674192 DOI: 10.1111/j.1582-4934.2009.00874.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The anti-inflammatory properties of transforming growth factor-beta(1) (TGF-beta(1)) account for its protection against atherosclerotic plaque rupture. This study investigates whether activation of the Nrf2 (nuclear factor erythroid 2 [NF-E2]-related factor 2) transcription pathway is involved in TGF-beta(1) mediated induction of the antioxidant enzyme heme oxygenase-1 (HO-1) in smooth muscle cells (SMC). Human aortic smooth muscle cells (HAoSMC) or wild-type and Nrf2-deficient mouse (MAoSMC) aortic SMC were treated with TGF-beta(1) (2.5-10 ng/ml, 0-24 hrs). We report the first evidence that TGF-beta(1) induces Nrf2 mediated HO-1 expression and antioxidant response element activity, which was paralleled by enhanced superoxide production and expression of the NAD(P)H oxidase subunit p22(phox). TGF-beta(1) failed to induce HO-1 expression in MAoSMC derived from Nrf2-deficient mice, and HO-1 induction by TGF-beta(1) in HAoSMC was attenuated by inhibition of extracellular signal regulated kinase or c-jun-N-terminal kinase but not p38 mitogen activated protein kinase. Inhibition of NAD(P)H oxidase or scavenging of superoxide diminished HO-1 induction in response to TGF-beta(1). The oxidative stress agents glucose oxidase (GOx) and diethylmaleate enhanced TGF-beta(1) generation and HO-1 expression in HAoSMC, while antagonism of TGF-beta(1) signalling by adenoviral Smad7 overexpression attenuated their induction of HO-1. Pre-treatment of HAoSMC with TGF-beta(1) reduced nuclear translocation of the pro-apoptotic mediator p53 elicited by GOx. Our findings demonstrate that Nrf2 is a new target of TGF-beta(1) signalling in the vasculature which may contribute to the atheroprotective properties attributed to this growth factor.
Collapse
Affiliation(s)
| | - Anila A Anwar
- Cardiovascular Division, School of Medicine, King's College London, UK
| | - Francois Y L Li
- Cardiovascular Division, School of Medicine, King's College London, UK
| | - Hideyo Sato
- Department of Bioresources, Faculty of Agriculture, Yamagata University, Tsuruoka, Japan
| | - Tetsuro Ishii
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tuskuba, Japan
| | - Giovanni E Mann
- Cardiovascular Division, School of Medicine, King's College London, UK
| | - Richard C M Siow
- Cardiovascular Division, School of Medicine, King's College London, UK
| |
Collapse
|
110
|
Felton VM, Borok Z, Willis BC. N-acetylcysteine inhibits alveolar epithelial-mesenchymal transition. Am J Physiol Lung Cell Mol Physiol 2009; 297:L805-12. [PMID: 19648289 DOI: 10.1152/ajplung.00009.2009] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The ability of transforming growth factor-beta1 (TGF-beta1) to induce epithelial-mesenchymal transition (EMT) in alveolar epithelial cells (AEC) in vitro and in vivo, together with the demonstration of EMT in biopsies of idiopathic pulmonary fibrosis (IPF) patients, suggests a role for TGF-beta1-induced EMT in disease pathogenesis. We investigated the effects of N-acetylcysteine (NAC) on TGF-beta1-induced EMT in a rat epithelial cell line (RLE-6TN) and in primary rat alveolar epithelial cells (AEC). RLE-6TN cells exposed to TGF-beta1 for 5 days underwent EMT as evidenced by acquisition of a fibroblast-like morphology, downregulation of the epithelial-specific protein zonula occludens-1, and induction of the mesenchymal-specific proteins alpha-smooth muscle actin (alpha-SMA) and vimentin. These changes were inhibited by NAC, which also prevented Smad3 phosphorylation. Similarly, primary alveolar epithelial type II cells exposed to TGF-beta1 also underwent EMT that was prevented by NAC. TGF-beta1 decreased cellular GSH levels by 50-80%, whereas NAC restored them to approximately 150% of those found in TGF-beta1-treated cells. Treatment with glutathione monoethyl ester similarly prevented an increase in mesenchymal marker expression. Consistent with its role as an antioxidant and cellular redox stabilizer, NAC dramatically reduced intracellular reactive oxygen species production in the presence of TGF-beta1. Finally, inhibition of intracellular ROS generation during TGF-beta1 treatment prevented alveolar EMT, but treatment with H2O2 alone did not induce EMT. We conclude that NAC prevents EMT in AEC in vitro, at least in part through replenishment of intracellular GSH stores and limitation of TGF-beta1-induced intracellular ROS generation. We speculate that beneficial effects of NAC on pulmonary function in IPF may be mediated by inhibitory effects on alveolar EMT.
Collapse
Affiliation(s)
- V M Felton
- Heart and Lung Institute, St. Joseph's Hospital and Medical Center, Department of Pediatrics, University of Arizona College of Medicine, 500 W. Thomas Rd., Suite 500, Phoenix, AZ 85013, USA
| | | | | |
Collapse
|
111
|
RamachandraRao SP, Zhu Y, Ravasi T, McGowan TA, Toh I, Dunn SR, Okada S, Shaw MA, Sharma K. Pirfenidone is renoprotective in diabetic kidney disease. J Am Soc Nephrol 2009; 20:1765-75. [PMID: 19578007 DOI: 10.1681/asn.2008090931] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although several interventions slow the progression of diabetic nephropathy, current therapies do not halt progression completely. Recent preclinical studies suggested that pirfenidone (PFD) prevents fibrosis in various diseases, but the mechanisms underlying its antifibrotic action are incompletely understood. Here, we evaluated the role of PFD in regulation of the extracellular matrix. In mouse mesangial cells, PFD decreased TGF-beta promoter activity, reduced TGF-beta protein secretion, and inhibited TGF-beta-induced Smad2-phosphorylation, 3TP-lux promoter activity, and generation of reactive oxygen species. To explore the therapeutic potential of PFD, we administered PFD to 17-wk-old db/db mice for 4 wk. PFD treatment significantly reduced mesangial matrix expansion and expression of renal matrix genes but did not affect albuminuria. Using liquid chromatography with subsequent electrospray ionization tandem mass spectrometry, we identified 21 proteins unique to PFD-treated diabetic kidneys. Analysis of gene ontology and protein-protein interactions of these proteins suggested that PFD may regulate RNA processing. Immunoblotting demonstrated that PFD promotes dosage-dependent dephosphorylation of eukaryotic initiation factor, potentially inhibiting translation of mRNA. In conclusion, PFD is renoprotective in diabetic kidney disease and may exert its antifibrotic effects, in part, via inhibiting RNA processing.
Collapse
Affiliation(s)
- Satish P RamachandraRao
- Center for Renal Translational Medicine, Division of Nephrology and Hypertension, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Irwin DC, McCord JM, Nozik-Grayck E, Beckly G, Foreman B, Sullivan T, White M, T Crossno J, Bailey D, Flores SC, Majka S, Klemm D, van Patot MCT. A potential role for reactive oxygen species and the HIF-1alpha-VEGF pathway in hypoxia-induced pulmonary vascular leak. Free Radic Biol Med 2009; 47:55-61. [PMID: 19358884 PMCID: PMC2689923 DOI: 10.1016/j.freeradbiomed.2009.03.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 03/06/2009] [Accepted: 03/28/2009] [Indexed: 01/11/2023]
Abstract
Acute hypoxia causes pulmonary vascular leak and is involved in the pathogenesis of pulmonary edema associated with inflammation, acute altitude exposure, and other critical illnesses. Reactive oxygen species, HIF-1, and VEGF have all been implicated in various hypoxic pathologies, yet the ROS-HIF-1-VEGF pathway in pulmonary vascular leak has not been defined. We hypothesized that the ROS-HIF-1-VEGF pathway has an important role in producing hypoxia-induced pulmonary vascular leak. Human pulmonary artery endothelial cell (HPAEC) monolayers were exposed to either normoxia (21% O(2)) or acute hypoxia (3% O(2)) for 24 h and monolayer permeability and H(2)O(2), nuclear HIF-1alpha, and cytosolic VEGF levels were determined. HPAEC were treated with antioxidant cocktail (AO; ascorbate, glutathione, and alpha-tocopherol), HIF-1 siRNA, or the VEGF soluble binding protein fms-like tyrosine kinase-1 (sFlt-1) to delineate the role of the ROS-HIF-1-VEGF pathway in hypoxia-induced HPAEC leak. Additionally, mice exposed to hypobaric hypoxia (18,000 ft, 10% O(2)) were treated with the same antioxidant to determine if in vitro responses corresponded to in vivo hypoxia stress. Hypoxia increased albumin permeativity, H(2)O(2) production, and nuclear HIF-1alpha and cytosolic VEGF concentration. Treatment with an AO lowered the hypoxia-induced HPAEC monolayer permeability as well as the elevation of HIF-1alpha and VEGF. Treatment of hypoxia-induced HPAEC with either an siRNA designed against HIF-1alpha or the VEGF antagonist sFlt-1 decreased monolayer permeability. Mice treated with AO and exposed to hypobaric hypoxia (18,000 ft, 10% O(2)) had less pulmonary vascular leak than those that were untreated. Our data suggest that hypoxia-induced permeability is due, in part, to the ROS-HIF-1alpha-VEGF pathway.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Active Transport, Cell Nucleus/genetics
- Animals
- Antioxidants/pharmacology
- Capillary Permeability/drug effects
- Capillary Permeability/genetics
- Cell Nucleus/metabolism
- Cells, Cultured
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Hypoxia
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/immunology
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Pulmonary Artery/pathology
- RNA, Small Interfering/genetics
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Transcriptional Activation/drug effects
- Transcriptional Activation/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- David C Irwin
- University of Colorado Health Science Center, School of Medicine, Denver, CO 80262, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
The angiotensin II-AT1 receptor stimulates reactive oxygen species within the cell nucleus. Biochem Biophys Res Commun 2009; 384:149-54. [PMID: 19409874 DOI: 10.1016/j.bbrc.2009.04.126] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 04/15/2009] [Indexed: 12/26/2022]
Abstract
We and others have reported significant expression of the Ang II Type 1 receptor (AT1R) on renal nuclei; thus, the present study assessed the functional pathways and distribution of the intracellular AT1R on isolated nuclei. Ang II (1nM) stimulated DCF fluorescence, an intranuclear indicator of reactive oxygen species (ROS), while the AT1R antagonist losartan or the NADPH oxidase (NOX) inhibitor DPI abolished the increase in ROS. Dual labeling of nuclei with antibodies against nucleoporin 62 (Nup62) and AT1R or the NADPH oxidase isoform NOX4 revealed complete overlap of the Nup62 and AT1R (99%) by flow cytometry, while NOX4 was present on 65% of nuclei. Treatment of nuclei with a PKC agonist increased ROS while the PKC inhibitor GF109203X or PI3 kinase inhibitor LY294002 abolished Ang II stimulation of ROS. We conclude that the Ang II-AT1R-PKC axis may directly influence nuclear function within the kidney through a redox sensitive pathway.
Collapse
|
114
|
Chamberlain CG, Mansfield KJ, Cerra A. Glutathione and catalase suppress TGFbeta-induced cataract-related changes in cultured rat lenses and lens epithelial explants. Mol Vis 2009; 15:895-905. [PMID: 19421408 PMCID: PMC2676196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 04/27/2009] [Indexed: 10/24/2022] Open
Abstract
PURPOSE The damaging effects of oxidative stress and transforming growth factor-beta (TGFbeta)-induced transdifferentiation of lens epithelial cells have both been implicated independently in the etiology of cataract. The aim of this study was to investigate whether the presence of antioxidant systems in the lens influences the ability of lens epithelial cells to respond to TGFbeta. METHODS Whole lenses from young rats were cultured with or without TGFbeta in the presence or absence of reduced glutathione (GSH). Lens epithelial explants from weanling rats were used to investigate the effects of GSH and catalase on TGFbeta-induced cataract-related changes. Lenses were monitored for opacification for three to four days, photographed, and then processed for routine histology. Explants were assessed by phase contrast microscopy, enzyme-linked immunosorbent assay (ELISA) of alpha-smooth muscle actin (alphaSMA), and/or immunolocalization of alphaSMA and Pax6, markers for transdifferentiation and normal lens epithelial phenotype, respectively. RESULTS In cultured lenses, GSH strongly suppressed TGFbeta-induced opacification and subcapsular plaque formation. In explants, both GSH and catalase suppressed changes typically associated with TGFbeta-induced transdifferentiation including wrinkling of the lens capsule, cell-surface blebbing, apoptotic cell loss, induction of alphaSMA, and loss of Pax6 expression. CONCLUSIONS This study suggests that antioxidant systems present in the normal lens, which protect the epithelium against the damaging effects of reactive oxygen species, may also serve to protect it against the potentially cataractogenic effects of TGFbeta. Taken together with other recent studies, it also raises the possibility that TGFbeta may induce cataract-related changes in lens epithelial cells via release of hydrogen peroxide.
Collapse
|
115
|
Bacillus anthracis edema toxin suppresses human macrophage phagocytosis and cytoskeletal remodeling via the protein kinase A and exchange protein activated by cyclic AMP pathways. Infect Immun 2009; 77:2530-43. [PMID: 19307216 DOI: 10.1128/iai.00905-08] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bacillus anthracis, the etiological agent of anthrax, is a gram-positive spore-forming bacterium. It produces edema toxin (EdTx), a powerful adenylate cyclase that increases cyclic AMP (cAMP) levels in host cells. Because other cAMP-increasing agents inhibit key macrophage (MPhi) functions, such as phagocytosis, it was hypothesized that EdTx would exhibit similar suppressive activities. Our previous GeneChip data showed that EdTx downregulated MPhi genes involved in actin cytoskeleton remodeling, including protein kinase A (PKA). To further examine the role of EdTx during anthrax pathogenesis, we explored the hypothesis that EdTx treatment leads to deregulation of the cAMP-dependent PKA system, resulting in impaired cytoskeletal functions essential for MPhi activity. Our data revealed that EdTx significantly suppressed human MPhi phagocytosis of Ames spores. Cytoskeletal changes, such as decreased cell spreading and lowered F-actin content, were also observed for toxin-treated MPhis. Further, EdTx altered the protein levels and activity of PKA and exchange protein activated by cAMP (Epac), a recently identified cAMP-binding molecule. By using PKA- and Epac-selective cAMP analogs, we confirmed the involvement of both pathways in the inhibition of MPhi functions elicited by EdTx-generated cAMP. These results suggested that EdTx weakened the host immune response by increasing cAMP levels, which then signaled via PKA and Epac to cripple MPhi phagocytosis and interfered with cytoskeletal remodeling.
Collapse
|
116
|
Muller M. Cellular senescence: molecular mechanisms, in vivo significance, and redox considerations. Antioxid Redox Signal 2009; 11:59-98. [PMID: 18976161 DOI: 10.1089/ars.2008.2104] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cellular senescence is recognized as a critical cellular response to prolonged rounds of replication and environmental stresses. Its defining characteristics are arrested cell-cycle progression and the development of aberrant gene expression with proinflammatory behavior. Whereas the mechanistic events associated with senescence are generally well understood at the molecular level, the impact of senescence in vivo remains to be fully determined. In addition to the role of senescence as an antitumor mechanism, this review examines cellular senescence as a factor in organismal aging and age-related diseases, with particular emphasis on aberrant gene expression and abnormal paracrine signaling. Senescence as an emerging factor in tissue remodeling, wound repair, and infection is considered. In addition, the role of oxidative stress as a major mediator of senescence and the role of NAD(P)H oxidases and changes to intracellular GSH/GSSG status are reviewed. Recent findings indicate that senescence and the behavior of senescent cells are amenable to therapeutic intervention. As the in vivo significance of senescence becomes clearer, the challenge will be to modulate the adverse effects of senescence without increasing the risks of other diseases, such as cancer. The uncoupled relation between cell-cycle arrest and the senescent phenotype suggests that this is an achievable outcome.
Collapse
Affiliation(s)
- Michael Muller
- Centre for Education and Research on Ageing, ANZAC Research Institute, University of Sydney, Concord RG Hospital, Concord, Sydney, Australia.
| |
Collapse
|
117
|
Violi F, Basili S, Nigro C, Pignatelli P. Role of NADPH oxidase in atherosclerosis. Future Cardiol 2009; 5:83-92. [DOI: 10.2217/14796678.5.1.83] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Reactive oxidant species (ROS) seem to play a key role in the atherosclerotic process via a series of molecular changes that lead to macrophage infiltration in the endothelium and eventually to plaque formation. ROS are also implicated in arterial dysfunction via inactivation of nitric oxide, a potent vasodilator and antiaggregating molecule produced by the endothelium. Owing to the relevance of endothelial dysfunction and vascular inflammation in the process of human atherosclerosis, a lot of effort has been directed towards discovering the ROS-generating pathways implicated in the ROS upregulation. Amongst the enzymatic pathways, NADPH oxidase is the most important enzyme responsible for ROS formation in human vessels. Experimental and clinical studies suggested a role for this enzyme in initiation and progression of atherosclerotic disease. The purpose of this review is to analyze whether the basic and clinical studies are consistent with this hypothesis and to point out if determination of NADPH oxidase is useful in the setting of the atherosclerosis to predict its progression and clinical complications.
Collapse
Affiliation(s)
- Francesco Violi
- Division of Internal Medicine, Department of Experimental Medicine, University of Rome ‘La Sapienza’, Viale del Policlinico 155, 00161, Rome, Italy
| | - Stefania Basili
- Division of Internal Medicine, Department of Experimental Medicine, University of Rome ‘La Sapienza’, Viale del Policlinico 155, 00161, Rome, Italy
| | - Carmen Nigro
- Division of Internal Medicine, Department of Experimental Medicine, University of Rome ‘La Sapienza’, Viale del Policlinico 155, 00161, Rome, Italy
| | - Pasquale Pignatelli
- Division of Internal Medicine, Department of Experimental Medicine, University of Rome ‘La Sapienza’, Viale del Policlinico 155, 00161, Rome, Italy
| |
Collapse
|
118
|
Grenklo S, Hillberg L, Zhao Rathje LS, Pinaev G, Schutt CE, Lindberg U. Tropomyosin assembly intermediates in the control of microfilament system turnover. Eur J Cell Biol 2008; 87:905-20. [DOI: 10.1016/j.ejcb.2008.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 06/18/2008] [Accepted: 06/23/2008] [Indexed: 01/07/2023] Open
|
119
|
Thomas SR, Witting PK, Drummond GR. Redox control of endothelial function and dysfunction: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal 2008; 10:1713-65. [PMID: 18707220 DOI: 10.1089/ars.2008.2027] [Citation(s) in RCA: 282] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The endothelium is essential for the maintenance of vascular homeostasis. Central to this role is the production of endothelium-derived nitric oxide (EDNO), synthesized by the endothelial isoform of nitric oxide synthase (eNOS). Endothelial dysfunction, manifested as impaired EDNO bioactivity, is an important early event in the development of various vascular diseases, including hypertension, diabetes, and atherosclerosis. The degree of impairment of EDNO bioactivity is a determinant of future vascular complications. Accordingly, growing interest exists in defining the pathologic mechanisms involved. Considerable evidence supports a causal role for the enhanced production of reactive oxygen species (ROS) by vascular cells. ROS directly inactivate EDNO, act as cell-signaling molecules, and promote protein dysfunction, events that contribute to the initiation and progression of endothelial dysfunction. Increasing data indicate that strategies designed to limit vascular ROS production can restore endothelial function in humans with vascular complications. The purpose of this review is to outline the various ways in which ROS can influence endothelial function and dysfunction, describe the redox mechanisms involved, and discuss approaches for preventing endothelial dysfunction that may highlight future therapeutic opportunities in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Shane R Thomas
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | |
Collapse
|
120
|
Expression of NADPH oxidases and enhanced H(2)O(2)-generating activity in human coronary artery endothelial cells upon induction with tumor necrosis factor-alpha. Int Immunopharmacol 2008; 8:1377-85. [PMID: 18687299 DOI: 10.1016/j.intimp.2008.05.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2008] [Revised: 05/05/2008] [Accepted: 05/06/2008] [Indexed: 11/21/2022]
Abstract
Tumor necrosis factor (TNF)-alpha, which potentiates reactive oxygen species (ROS) generation, is crucial for the development of coronary arteritis and aneurysm in Kawasaki disease. We hypothesized that vascular NADPH oxidase (Nox) enzymes participate in the TNF-alpha-triggered endothelial damage through elevating ROS generation. Thus, we herein examine the expression of Nox enzymes in human coronary artery endothelial cells (HCAEC) and the effects of TNF-alpha on Nox-mediated ROS generation. We show that HCAEC in culture spontaneously generate H(2)O(2) at basal level (0.53 nmol/min/mg protein). In searching for Nox components responsible for the H(2)O(2) generation, two distinct isoforms of Nox4 are found expressed in HCAEC: the prototype Nox4A and the shorter Nox4B, respectively in the postnuclear supernatant and the nuclear fractions. Other expressed Nox family components are: as mRNAs, Nox4C, Nox4D, Nox1, p51(nox), and Racs; as mRNAs and proteins, Nox2, p22(phox), p47(phox), and p67(phox). The H(2)O(2)-generating activity increases up to three-fold upon inclusion of TNF-alpha in culture, concomitantly with augmented expressions of Nox4A, p22(phox), p47(phox) and p67(phox) proteins. Together, these results suggest that Nox2 and Nox4A enzymes are induced by TNF-alpha endowing HCAEC with enhanced ROS-generating activity, which may play a role in the initial endothelial dysfunction through oxidative stress.
Collapse
|
121
|
Sharma K, RamachandraRao S, Qiu G, Usui HK, Zhu Y, Dunn SR, Ouedraogo R, Hough K, McCue P, Chan L, Falkner B, Goldstein BJ. Adiponectin regulates albuminuria and podocyte function in mice. J Clin Invest 2008; 118:1645-56. [PMID: 18431508 PMCID: PMC2323186 DOI: 10.1172/jci32691] [Citation(s) in RCA: 290] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Accepted: 02/20/2008] [Indexed: 12/13/2022] Open
Abstract
Increased albuminuria is associated with obesity and diabetes and is a risk factor for cardiovascular and renal disease. However, the link between early albuminuria and adiposity remains unclear. To determine whether adiponectin, an adipocyte-derived hormone, is a communication signal between adipocytes and the kidney, we performed studies in a cohort of patients at high risk for diabetes and kidney disease as well as in adiponectin-knockout (Ad(-/-)) mice. Albuminuria had a negative correlation with plasma adiponectin in obese patients, and Ad(-/-) mice exhibited increased albuminuria and fusion of podocyte foot processes. In cultured podocytes, adiponectin administration was associated with increased activity of AMPK, and both adiponectin and AMPK activation reduced podocyte permeability to albumin and podocyte dysfunction, as evidenced by zona occludens-1 translocation to the membrane. These effects seemed to be caused by reduction of oxidative stress, as adiponectin and AMPK activation both reduced protein levels of the NADPH oxidase Nox4 in podocytes. Ad(-/-) mice treated with adiponectin exhibited normalization of albuminuria, improvement of podocyte foot process effacement, increased glomerular AMPK activation, and reduced urinary and glomerular markers of oxidant stress. These results suggest that adiponectin is a key regulator of albuminuria, likely acting through the AMPK pathway to modulate oxidant stress in podocytes.
Collapse
Affiliation(s)
- Kumar Sharma
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Satish RamachandraRao
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Gang Qiu
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Hitomi Kataoka Usui
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Yanqing Zhu
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Stephen R. Dunn
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Raogo Ouedraogo
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Kelly Hough
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Peter McCue
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lawrence Chan
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Bonita Falkner
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Barry J. Goldstein
- Translational Research in Kidney Disease, Division of Nephrology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.
Veterans Administration San Diego Healthcare System, La Jolla, California, USA.
Center for Novel Therapies in Kidney Disease, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Department of Medicine, and
Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
Division of Endocrinology, Diabetes, and Metabolic Diseases, Baylor College of Medicine, Houston, Texas, USA.
Center for Hypertension, Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
122
|
Xu H, Goettsch C, Xia N, Horke S, Morawietz H, Förstermann U, Li H. Differential roles of PKCalpha and PKCepsilon in controlling the gene expression of Nox4 in human endothelial cells. Free Radic Biol Med 2008; 44:1656-67. [PMID: 18291120 DOI: 10.1016/j.freeradbiomed.2008.01.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 11/30/2007] [Accepted: 01/19/2008] [Indexed: 11/22/2022]
Abstract
NADPH oxidases are major sources of superoxide in the vascular wall. This study investigates the role of protein kinase C (PKC) in regulating gene expression of NADPH oxidases. Treatment of human umbilical vein endothelial cells (HUVEC) and HUVEC-derived EA.hy 926 endothelial cells with phorbol 12-myristate 13-acetate (PMA) or phorbol 12,13-dibutyrate led to a PKC-dependent biphasic expression of the gp91phox homolog Nox4. A downregulation of Nox4 was observed at 6 h and an upregulation at 48 h after phorbol ester treatment. The early Nox4 downregulation was associated with a reduced superoxide production, whereas the late Nox4 upregulation was accompanied by a clear enhancement of superoxide. PMA activated the PKC isoforms alpha and epsilon in HUVEC and EA.hy 926 cells. Knockdown of PKCepsilon by siRNA prevented the early downregulation of Nox4, whereas knockdown of PKCalpha selectively abolished the late Nox4 upregulation. Vascular endothelial growth factor (VEGF), which activates PKCalpha but not PKCepsilon in HUVEC, increased Nox4 expression without the initial downregulation. VEGF-induced Nox4 upregulation was associated with an enhanced proliferation and angiogenesis of HUVEC. Both effects could be reduced by inhibition of NADPH oxidase. Thus, a selective inhibition/knockdown of PKCalpha may represent a novel therapeutic strategy for vascular disease.
Collapse
Affiliation(s)
- Hui Xu
- Department of Pharmacology, Johannes Gutenberg University, D-55131 Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
123
|
Koli K, Myllärniemi M, Keski-Oja J, Kinnula VL. Transforming growth factor-beta activation in the lung: focus on fibrosis and reactive oxygen species. Antioxid Redox Signal 2008; 10:333-42. [PMID: 17961070 DOI: 10.1089/ars.2007.1914] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Transforming growth factor-betas (TGF-beta) regulate a wide variety of cellular functions in normal development and are involved in both tissue homeostasis and disease pathogenesis. The regulation of the TGF-beta family of growth factors is unique because they are targeted to the extracellular matrix in a biologically inactive form. The release from pericellular matrices and the activation of TGF-beta are important mechanisms in several pathophysiologic conditions. Reactive oxygen species (ROS) can activate TGF-beta either directly or indirectly via the activation of proteases. In addition, TGF-beta itself induces ROS production as part of its signal-transduction pathway. The lung is a unique organ, because its structures act as boundaries between gaseous and aqueous phases, allowing the utilization of inhaled oxygen. However, this renders pulmonary tissues vulnerable to the toxic effects of inhaled air. The oxidant pathways are especially relevant in the lung, where TGF-beta is known to have a role in tissue repair and connective tissue turnover. In pulmonary fibrosis, TGF-beta activation is considered as a hallmark of disease progression. More recently, the oxidative effects of cigarette smoking have been found to activate TGF-beta in chronic obstructive pulmonary disease (COPD), a disease consisting of emphysema, airway fibrosis, and focal lung fibrosis.
Collapse
Affiliation(s)
- Katri Koli
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | |
Collapse
|
124
|
Wu MJ, Gu ZY, Sun W. Effects of hydrostatic pressure on cytoskeleton and BMP-2, TGF-beta, SOX-9 production in rat temporomandibular synovial fibroblasts. Osteoarthritis Cartilage 2008; 16:41-7. [PMID: 17631391 DOI: 10.1016/j.joca.2007.05.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2007] [Accepted: 05/29/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Recent experimental evidence has suggested that pressure may play an important role in the pathogenesis of arthritic diseases such as temporomandibular disorders (TMDs), rheumatic diseases and osteoarthritis. This study examines the effects of hydrostatic pressure (HP) on cytoskeleton and protein production of bone morphogenetic protein-2 (BMP-2), transforming growth factor-beta (TGF-beta) and the SRY HMG box related gene 9 (SOX-9) in synovial fibroblasts (SFs) of rat temporomandibular joint (TMJ). METHODS SFs derived from rat TMJ were grown to confluence in Dulbecco's modified Eagle medium supplemented with 15% fetal calf serum. The monolayer of SFs was subjected to different HPs (0, 30, 60, and 90kPa) by an in-house designed pressure chamber for 12h. Changes of cell morphology were observed by fluorescent microscope. Production of TGF-beta, BMP-2 and SOX-9 was examined by immunocytochemical assay and western blot. RESULTS Compared with the untreated control, the cellular actin configuration of SFs became elongated and more intense F-actin stress fiber staining was observed after HP loading. Exposure of SFs to HP for 12h resulted in significant up-regulation of BMP-2 by 46, 54, and 66% at 30, 60, and 90kPa, respectively, whilst TGF-beta increased by 11, 19, and 28% at 30, 60, and 90kPa, respectively. HP also induced the increase of SOX-9 by 72% at 30kPa and 83% at 60kPa, but only 54% at 90kPa. CONCLUSIONS The obtained data suggest that HP induced the alteration of cytoskeleton and bone-morphogenetic-related proteins' production of SFs, which may influence the pathological condition of TMDs.
Collapse
Affiliation(s)
- M-J Wu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Zhejiang University, Hangzhou, China
| | | | | |
Collapse
|
125
|
Xia C, Meng Q, Liu LZ, Rojanasakul Y, Wang XR, Jiang BH. Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor. Cancer Res 2007; 67:10823-30. [PMID: 18006827 DOI: 10.1158/0008-5472.can-07-0783] [Citation(s) in RCA: 357] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Reactive oxygen species (ROS) are associated with multiple cellular functions such as cell proliferation, differentiation, and apoptosis. However, the direct roles of endogenous ROS production still remain to be elucidated. In this study, we found that high levels of ROS were spontaneously produced by ovarian and prostate cancer cells. This elevated ROS production was inhibited by NADPH oxidase inhibitor diphenylene iodonium (DPI) and mitochondria electron chain inhibitor rotenone in the cells. To further analyze the source of ROS production, we found that ovarian cancer cells have much higher expression of NOX4 NADPH oxidase, and that specific inhibition of NADPH oxidase subunit p47(phox) diminished ROS production. To analyze the functional relevance of ROS production, we showed that ROS regulated hypoxia-inducible factor 1 (HIF-1) and vascular endothelial growth factor (VEGF) expression in ovarian cancer cells. Elevated levels of endogenous ROS were required for inducing angiogenesis and tumor growth. NOX4 knockdown in ovarian cancer cells decreased the levels of VEGF and HIF-1 alpha and tumor angiogenesis. This study suggests a new mechanism of higher ROS production in ovarian cancer cells and provides strong evidence that endogenous ROS play an important role for cancer cells to induce angiogenesis and tumor growth. This information may be useful to understand the new mechanism of cancer cells in inducing tumorigenesis and to develop new therapeutic strategy by targeting ROS signaling in human cancer in the future.
Collapse
Affiliation(s)
- Chang Xia
- Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia 26506, USA
| | | | | | | | | | | |
Collapse
|
126
|
Lindberg U, Karlsson R, Lassing I, Schutt CE, Höglund AS. The microfilament system and malignancy. Semin Cancer Biol 2007; 18:2-11. [PMID: 18024149 DOI: 10.1016/j.semcancer.2007.10.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Increased motile activity, increased rate of cell proliferation and removal of growth inhibiting cell-cell contacts are hallmarks of tumorigenesis. Activation of cell motility and migration is caused by activation of receptors, turning on the growth cycle. Increased expression of metalloproteinases, breaking cell:cell contacts and organ confines, allows the spread of malignant cancer cells to other sites in the organism. It has become increasingly clear that most transmembrane proteins (growth factor receptors, adhesion proteins and ion channels) are either permanently or transiently associated with the sub-membraneous system of actin microfilaments (MF), whose force generating capacity they control. Although there has been great progress in our understanding of the physiological importance of the MF-system, as will be exemplified in this issue of SCB, many aspects of actin microfilament formation and its regulation are still unclear. Redox control of the actin (MF)-system in cell motility and migration and its perturbations in pathophysiology, including cancer, is an emerging field of research.
Collapse
Affiliation(s)
- Uno Lindberg
- Department of Microbiology, Tumor Biology, and Cell Biology, The Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
127
|
Williams KJ, Qiu G, Usui HK, Dunn SR, McCue P, Bottinger E, Iozzo RV, Sharma K. Decorin deficiency enhances progressive nephropathy in diabetic mice. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1441-50. [PMID: 17884968 PMCID: PMC2043506 DOI: 10.2353/ajpath.2007.070079] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Decorin, a proteoglycan that inhibits active transforming growth factor-beta, is increased in diabetic nephropathy; however, its functional significance is unclear. In this study, we used low-dose streptozotocin to induce type 1 diabetes in wild-type (C57BL/6J Dcn(+/+)), Dcn(-/-), and Dcn(+/-) mice and studied the mice for up to 1 year of diabetes. Decorin gene dose had no effect on severity of diabetes; however, the Dcn(-/-) diabetic mice died significantly earlier than nondiabetic controls (57 versus 7.3% mortality). In contrast to wild-type diabetic mice, which failed to develop significant nephropathy, the Dcn(-/-) diabetic mice developed a significant increase in albuminuria and plasma creatinine and a concurrent decrease in circulating adiponectin levels. Interestingly, adiponectin levels at 6 months of diabetes were predictive of mortality in diabetic mice. Dcn(-/-) diabetic mice exhibited advanced glomerular lesions, including diffuse mesangial matrix accumulation and fibrin cap formation. By immunohistochemistry, Dcn(-/-) diabetic mice exhibited significant increases in glomerular transforming growth factor-beta, type I collagen, macrophage infiltration, and Nox4. We conclude that decorin is a natural protective factor against diabetic nephropathy and that the Dcn(-/-) diabetic mouse is a useful new model of progressive diabetic nephropathy.
Collapse
Affiliation(s)
- Kevin Jon Williams
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Serrander L, Cartier L, Bedard K, Banfi B, Lardy B, Plastre O, Sienkiewicz A, Fórró L, Schlegel W, Krause KH. NOX4 activity is determined by mRNA levels and reveals a unique pattern of ROS generation. Biochem J 2007; 406:105-14. [PMID: 17501721 PMCID: PMC1948990 DOI: 10.1042/bj20061903] [Citation(s) in RCA: 511] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
NOX4 is an enigmatic member of the NOX (NADPH oxidase) family of ROS (reactive oxygen species)-generating NADPH oxidases. NOX4 has a wide tissue distribution, but the physiological function and activation mechanisms are largely unknown, and its pharmacology is poorly understood. We have generated cell lines expressing NOX4 upon tetracycline induction. Tetracycline induced a rapid increase in NOX4 mRNA (1 h) followed closely (2 h) by a release of ROS. Upon tetracycline withdrawal, NOX4 mRNA levels and ROS release decreased rapidly (<24 h). In membrane preparations, NOX4 activity was selective for NADPH over NADH and did not require the addition of cytosol. The pharmacological profile of NOX4 was distinct from other NOX isoforms: DPI (diphenyleneiodonium chloride) and thioridazine inhibited the enzyme efficiently, whereas apocynin and gliotoxin did not (IC(50)>100 muM). The pattern of NOX4-dependent ROS generation was unique: (i) ROS release upon NOX4 induction was spontaneous without need for a stimulus, and (ii) the type of ROS released from NOX4-expressing cells was H(2)O(2), whereas superoxide (O(2)(-)) was almost undetectable. Probes that allow detection of intracellular O(2)(-) generation yielded differential results: DHE (dihydroethidium) fluorescence and ACP (1-acetoxy-3-carboxy-2,2,5,5-tetramethylpyrrolidine) ESR measurements did not detect any NOX4 signal, whereas a robust signal was observed with NBT. Thus NOX4 probably generates O(2)(-) within an intracellular compartment that is accessible to NBT (Nitro Blue Tetrazolium), but not to DHE or ACP. In conclusion, NOX4 has a distinct pharmacology and pattern of ROS generation. The close correlation between NOX4 mRNA and ROS generation might hint towards a function as an inducible NOX isoform.
Collapse
Affiliation(s)
- Lena Serrander
- Foundation for Medical Research, University of Geneva, 64 av de la Roseraie, 1205, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Zhu Y, Kataoka Usui H, Sharma K. Regulation of transforming growth factor beta in diabetic nephropathy: implications for treatment. Semin Nephrol 2007; 27:153-60. [PMID: 17418684 PMCID: PMC1948024 DOI: 10.1016/j.semnephrol.2007.01.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The recognition that the drivers of matrix accumulation is an appropriate therapeutic target for diabetic nephropathy is now accepted by the nephrology and pharmaceutical communities. Interventions focused around transforming growth factor-beta (TGF-beta) likely will be an important area of clinical investigation in the near future. Understanding the various pathways involved in stimulating TGF-beta in the diabetic kidney is of paramount importance in devising strategies to combat the development and progression of diabetic nephropathy. In this review we highlight the major pathways involved in stimulating TGF-beta production by increased glucose levels and discuss the therapeutic implications thereof.
Collapse
Affiliation(s)
| | | | - Kumar Sharma
- From The Center for Novel Therapies in Kidney Disease, Dorrance Hamilton Research Laboratories , Division of Nephrology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
130
|
Steinkamp-Fenske K, Bollinger L, Xu H, Yao Y, Horke S, Förstermann U, Li H. Reciprocal regulation of endothelial nitric-oxide synthase and NADPH oxidase by betulinic acid in human endothelial cells. J Pharmacol Exp Ther 2007; 322:836-42. [PMID: 17496167 DOI: 10.1124/jpet.107.123356] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide (NO) produced by endothelial NO synthase (eNOS) is a protective principle in the vasculature. Many cardiovascular diseases are associated with reduced NO bioactivity and eNOS uncoupling due to oxidative stress. Compounds that reverse eNOS uncoupling and increase eNOS expression are of therapeutic interest. Zizyphi Spinosi semen (ZSS) is one of the most widely used traditional Chinese herbs with protective effects on the cardiovascular system. In human umbilical vein endothelial cells (HUVEC) and HUVEC-derived EA.hy 926 cells, an extract of ZSS increased eNOS promoter activity, eNOS mRNA and protein expression, and NO production in a concentration- and time-dependent manner. Major ZSS constituents include saponins, such as jujuboside A and B, and pentacyclic triterpenes, such as betulin and betulinic acid. Jujuboside A, jujuboside B, or betulin had no significant effect on eNOS expression, whereas betulinic acid increased eNOS mRNA and protein expression in HUVEC and EA.hy 926 cells. Interestingly, betulinic acid also attenuated the expression of NADPH oxidase subunits Nox4 and p22phox, thereby reducing oxidative stress and improving eNOS function. Consequently, betulinic acid-treated endothelial cells showed an increased production of bioactive NO (as indicated by a higher efficacy in stimulating cGMP generation in RFL-6 reporter cells). Thus, betulinic acid possesses combined properties of eNOS up-regulation and NADPH oxidase down-regulation. Compounds such as betulinic acid may have a therapeutic potential in cardiovascular disease.
Collapse
|
131
|
Lassing I, Schmitzberger F, Björnstedt M, Holmgren A, Nordlund P, Schutt CE, Lindberg U. Molecular and structural basis for redox regulation of beta-actin. J Mol Biol 2007; 370:331-48. [PMID: 17521670 DOI: 10.1016/j.jmb.2007.04.056] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 04/18/2007] [Accepted: 04/18/2007] [Indexed: 12/20/2022]
Abstract
An essential consequence of growth factor-mediated signal transduction is the generation of intracellular H(2)O(2). It operates as a second messenger in the control of actin microfilament dynamics, causing rapid and dramatic changes in the morphology and motile activity of stimulated cells. Little is understood about the molecular mechanisms causing these changes in the actin system. Here, it is shown that H(2)O(2) acts directly upon several levels of this system, and some of the mechanistic effects are detailed. We describe the impact of oxidation on the polymerizability of non-muscle beta/gamma-actin and compare with that of muscle alpha-actin. Oxidation of beta/gamma-actin can cause a complete loss of polymerizability, crucially, reversible by the thioredoxin system. Further, oxidation of the actin impedes its interaction with profilin and causes depolymerization of filamentous actin. The effects of oxidation are critically dependent on the nucleotide state and the concentration of Ca(2+). We have determined the crystal structure of oxidized beta-actin to a resolution of 2.6 A. The arrangement in the crystal implies an antiparallel homodimer connected by an intermolecular disulfide bond involving cysteine 374. Our data indicate that this dimer forms under non-polymerizing and oxidizing conditions. We identify oxidation of cysteine 272 in the crystallized actin dimer, likely to a cysteine sulfinic acid. In beta/gamma-actin, this is the cysteine residue most reactive towards H(2)O(2) in solution, and we suggest plausible structural determinants for its reactivity. No other oxidative modification was obvious in the structure, highlighting the specificity of the oxidation by H(2)O(2). Possible consequences of the observed effects in a cellular context and their potential relevance are discussed.
Collapse
Affiliation(s)
- Ingrid Lassing
- Department of Microbiology, Tumor Biology, and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
132
|
Malik AN, Rossios C, Al-Kafaji G, Shah A, Page RA. Glucose regulation of CDK7, a putative thiol related gene, in experimental diabetic nephropathy. Biochem Biophys Res Commun 2007; 357:237-44. [PMID: 17416350 DOI: 10.1016/j.bbrc.2007.03.132] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2007] [Accepted: 03/22/2007] [Indexed: 10/23/2022]
Abstract
We previously described the identification of the 3'end of an unknown gene CDK7 using differential display which appeared to be up-regulated in diabetic kidneys [R.A. Page, C.A. Morris, J.D. Williams, C.J. von Ruhland, A.N. Malik, Isolation of diabetes-associated kidney genes using differential display, Biochem. Biophys. Res. Commun. 232 (1997) 49-53]. Here we show that CDK7 is a putative thiol related gene which is regulated by glucose in human and rat renal cells. CDK7 mRNA increased by >threefold in cultured human mesangial cells grown in high glucose for 4 days. In the kidneys of the GK rat, a model of type II diabetes, CDK7 showed a steady age-related increase in mRNA, increasing to >sixfold in 40 week GK rats compared to normoglycemic age-matched Wistar rat kidneys, this increase correlates with progressive hyperglycemia. CDK7 mRNA is widely expressed, showing particularly high levels of expression in rat and human liver, and encodes a putative 338 amino acids highly conserved peptide with several conserved domains, including a cys-pro-arg-cys domain conserved in 15 diverse species which is similar to the catalytic centre of thioredoxin, suggesting a role in oxidative stress.
Collapse
Affiliation(s)
- Afshan N Malik
- Division of Reproduction and Endocrinology, School of Biomedical and Health Sciences, King's College London, UK.
| | | | | | | | | |
Collapse
|
133
|
Abstract
Reactive oxygen species (ROS) are recently proposed to be involved in tumor metastasis which is a complicated processes including epithelial-mesenchymal transition (EMT), migration, invasion of the tumor cells and angiogenesis around the tumor lesion. ROS generation may be induced intracellularly, in either NADPH oxidase- or mitochondria-dependent manner, by growth factors and cytokines (such as TGFbeta and HGF) and tumor promoters (such as TPA) capable of triggering cell adhesion, EMT and migration. As a signaling messenger, ROS are able to oxidize the critical target molecules such as PKC and protein tyrosine phosphates (PTPs), which are relevant to tumor cell invasion. PKC contain multiple cysteine residues that can be oxidized and activated by ROS. Inactivation of multiple PTPs by ROS may relieve the tyrosine phosphorylation-dependent signaling. Two of the down-stream molecules regulated by ROS are MAPK and PAK. MAPKs cascades were established to be a major signal pathway for driving tumor cell metastasis, which are mediated by PKC, TGF-beta/Smad and integrin-mediated signaling. PAK is an effector of Rac-mediated cytoskeletal remodeling that is responsible for cell migration and angiogenesis. There are several transcriptional factors such as AP1, Ets, Smad and Snail regulating a lot of genes relevant to metastasis. AP-1 and Smad can be activated by PKC activator and TGF-beta1, respectively, in a ROS dependent manner. On the other hand, Est-1 can be upregulated by H2O2 via an antioxidant response element in the promoter. The ROS-regulated genes relevant to EMT and metastasis include E-cahedrin, integrin and MMP. Comprehensive understanding of the ROS-triggered signaling transduction, transcriptional activation and regulation of gene expressions will help strengthen the critical role of ROS in tumor progression and devising strategy for chemo-therapeutic interventions.
Collapse
Affiliation(s)
- Wen-Sheng Wu
- Department of Medical Technology, Tzu Chi University, No. 701, Chung Yang Rd, Sec 3, Hualien 970, Taiwan.
| |
Collapse
|
134
|
Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 2007; 87:245-313. [PMID: 17237347 DOI: 10.1152/physrev.00044.2005] [Citation(s) in RCA: 4976] [Impact Index Per Article: 276.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
For a long time, superoxide generation by an NADPH oxidase was considered as an oddity only found in professional phagocytes. Over the last years, six homologs of the cytochrome subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the phagocyte NADPH oxidase itself (NOX2/gp91(phox)), the homologs are now referred to as the NOX family of NADPH oxidases. These enzymes share the capacity to transport electrons across the plasma membrane and to generate superoxide and other downstream reactive oxygen species (ROS). Activation mechanisms and tissue distribution of the different members of the family are markedly different. The physiological functions of NOX family enzymes include host defense, posttranlational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. NOX enzymes also contribute to a wide range of pathological processes. NOX deficiency may lead to immunosuppresion, lack of otoconogenesis, or hypothyroidism. Increased NOX activity also contributes to a large number or pathologies, in particular cardiovascular diseases and neurodegeneration. This review summarizes the current state of knowledge of the functions of NOX enzymes in physiology and pathology.
Collapse
Affiliation(s)
- Karen Bedard
- Biology of Ageing Laboratories, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
135
|
Vitreous induces heme oxygenase-1 expression mediated by transforming growth factor-beta and reactive oxygen species generation in human retinal pigment epithelial cells. Mol Vis 2007; 13:66-78. [PMID: 17277740 PMCID: PMC2503184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
PURPOSE When human retinal pigment epithelial (RPE) cells come in contact with vitreous, they undergo changes in gene expression that include inflammatory and anti-oxidant responses. The effects of vitreous on expression of heme oxygenase-1 (HO-1), metallothionein (MT) -1a and -2a, and c-fos were investigated. Activator protein-1 (AP-1) binding sites are located in the promoter region of HO-1 and MT genes and the effects of vitreous on c-fos activity were investigated. METHODS Low passage cultures of human RPE cells were grown in the presence or absence of vitreous or transforming growth factor-beta (TGF-beta). The expression of HO-1 and MTs was measured by real time PCR and, in the case of HO-1, by immunoblotting and immunofluorescence microscopy. Specific inhibitors were used to investigate possible signaling pathways. The effect of vitreous on activation of AP-1 transcription factor was determined by immunoblotting, electrophoretic mobility shift assays, or immunofluorescence microscopy. RESULTS Incubation of RPE cells with vitreous resulted in increased expression of HO-1, MT-1a and MT-2a. TGF-beta caused an increase in HO-1 expression, although not to the extent mediated by vitreous, but had little effect on MT expression. Addition of inhibitors of TGF-beta signaling (SB431542 or TGF-beta-neutralizing antibodies) decreased the vitreous induction of HO-1. Several reactive oxygen species (ROS) quenchers inhibited the TGF-beta-induced or vitreous-induced elevation of HO-1 mRNA but had no effect on vitreous-mediated induction of MT expression. Inhibitors of the mitogen-activated protein kinase (p38MAPK; SB203580) and Jun N-terminal kinase (JNK; SP600125) pathways inhibited vitreous-induction of HO-1. C-fos, a component of AP-1 transcription factor complexes, exhibited increased expression and activation in the presence of vitreous. CONCLUSIONS TGF-beta, a known component of vitreous, can account for some but not all of the regulation of the anti-oxidant, anti-inflammatory HO-1 gene in human RPE cells, but it does not participate in the vitreous-mediated upregulation of MTs. Both vitreous and TGF-beta signals increased HO-1 expression via ROS but the latter were not involved in vitreous-mediated MT expression. Increased p38, JNK, and c-fos activation may be implicated in vitreous modulation of HO-1.
Collapse
|
136
|
Chiarugi P, Buricchi F. Protein tyrosine phosphorylation and reversible oxidation: two cross-talking posttranslation modifications. Antioxid Redox Signal 2007; 9:1-24. [PMID: 17115885 DOI: 10.1089/ars.2007.9.1] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In addition to protein phosphorylation, redox-dependent posttranslational modification of proteins is emerging as a key signaling system, conserved throughout evolution, and influencing many aspects of cellular homeostasis. Recent data have provided new insight about the interplay between phosphorylation- and redox-dependent signaling, and reactive oxygen species have been included among intracellular signal transducers of growth factor and extracellular matrix receptors. Both tyrosine phosphorylation and thiol oxidation are reversible and dynamic, and this review will particularly focus on the cross-talk between these posttranslational protein regulatory means. Although these modifications share their reversibility, their effects on enzymatic activity of protein tyrosine phosphatases (PTPs) and protein tyrosine kinases (PTKs) may be even opposite. Indeed, while tyrosine phosphorylation is frequently correlated to enzyme activation, thiol oxidation leads to inactivation of PTPs and to superactivation of PTKs. Several papers describe that both these modifications occur during the same input, (i.e., cell proliferation and motility induced by numerous growth factors and cytokines). The review will discuss several aspects of phosphorylation\oxidation interplay, describing both convergent and divergent features of the integrated and coordinated function of PTPs and PTKs during signaling.
Collapse
Affiliation(s)
- Paola Chiarugi
- Department of Biochemical Sciences, University of Florence Center for Research, Transfer and High Education, Florence, Italy.
| | | |
Collapse
|
137
|
|
138
|
Abstract
The phagocytic NADPH oxidase is recognized as a critical component of innate immunity, responsible for generation of microbicidal reactive oxygen species (ROS). This enzyme is one representative of the Nox family of oxidases (Nox1-Nox5, Duox1, and Duox2) that exhibit diverse expression patterns and appear to serve a variety of functions related to ROS generation. Mounting evidence now suggests that several of these novel oxidases also serve in host defense, particularly those showing high expression along epithelial surfaces exposed to the external environment. Within these sites, Nox enzymes tend to be located on apical cell surfaces and release ROS into extracellular environments, where they can be used by known antimicrobial peroxidases. Moreover, microbial factors were shown in several cases to cause higher ROS production, either by direct oxidase activation or by inducing higher oxidase expression. Several oxidases are also induced by immune cytokines, including interferon-gamma, interleukin (IL)-4, and IL-13. Although most of the evidence supporting host defense roles for mammalian nonphagocytic oxidases remains circumstantial, recent evidence indicates that Drosophila Duox plays a role in host resistance to infection. Finally, oxidative defense against invading pathogens appears to be an ancient protective mechanism, because related oxidases are known to participate in disease resistance in plants.
Collapse
Affiliation(s)
- Thomas L Leto
- Molecular Defenses Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA.
| | | |
Collapse
|
139
|
Grange L, Nguyen MVC, Lardy B, Derouazi M, Campion Y, Trocme C, Paclet MH, Gaudin P, Morel F. NAD(P)H oxidase activity of Nox4 in chondrocytes is both inducible and involved in collagenase expression. Antioxid Redox Signal 2006; 8:1485-96. [PMID: 16987005 DOI: 10.1089/ars.2006.8.1485] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Reactive oxygen species (ROS) are regulators of redox-sensitive cell signaling pathways. In osteoarthritis, human interleukin-1beta is implicated in cartilage destruction through an ROS-dependent matrix metalloproteinase production. To determine the molecular source of ROS production in the human IL-1beta (hIL-1beta)-sensitive chondrocyte immortalized cell line C-20/A4, transfected cells were constructed that overexpress NAD(P)H oxidases. First, RT-PCR analysis showed that the C-20/A4 cell line expressed Nox2, Nox4, p22( phox ), and p67( phox ), but not p47( phox ). It was found that ROS production by C-20/A4 chondrocytes does not depend on PMA and ionomycin activation. This indicates that Nox2 was not involved in the production of ROS. In C- 20/A4 cells that overexpress Nox4, hIL-1beta stimulated ROS production three times more than the normal production of C-20/A4 cells. Moreover, there was a fourfold increase in the production of collagenase (MMP-1) by chondrocytes that overexpress Nox4. Interestingly, MMP-1 production in cells that overexpress Nox2 was not sensitive to hIL-1beta. These data suggest that under hIL-1beta stimulation, C-20/A4 chondrocytes produce MMP-1 through a Nox4-mediated, ROS-dependent pathway.
Collapse
Affiliation(s)
- Laurent Grange
- GREPI EA 2938 UJF, Lab Enzymology/DBPC, Universitary Hospital A. Michallon, Grenoble, France [corrected]
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Alexandrova AY, Kopnin PB, Vasiliev JM, Kopnin BP. ROS up-regulation mediates Ras-induced changes of cell morphology and motility. Exp Cell Res 2006; 312:2066-73. [PMID: 16624288 DOI: 10.1016/j.yexcr.2006.03.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Revised: 03/06/2006] [Accepted: 03/08/2006] [Indexed: 12/31/2022]
Abstract
Expression of activated Ras causes an increase in intracellular content of reactive oxygen species (ROS). To determine the role of ROS up-regulation in mediation of Ras-induced morphological transformation and increased cell motility, we studied the effects of hydrogen peroxide and antioxidant NAC on morphology of REF52 rat fibroblasts and their ability to migrate into the wound in vitro. Treatment with low dosages of hydrogen peroxide leading to 1.5- to 2-fold increase in intracellular ROS levels induced changes of cell shape, actin cytoskeleton organization, cell adhesions and migration resembling those in Ras-transformed cells. On the other hand, treatment with NAC attenuating ROS up-regulation in cells with conditional or constitutive expression of activated Ras led to partial reversion of morphological transformation and decreased cell motility. The effect of ROS on cell morphology and motility probably results from modulation of activity of Rac1, Rho, and cofilin proteins playing a key role in regulation of actin dynamics. The obtained data are consistent with the idea that ROS up-regulation mediates two key events in Ras-induced morphological transformation and cell motility: it is responsible for Rac1 activation and is necessary (though insufficient) for realization of Ras-induced cofilin dephosphorylation.
Collapse
Affiliation(s)
- Antonina Y Alexandrova
- Institute of Carcinogenesis, Blokhin Memorial Russian Cancer Research Center, Kashirskoye shosse 24, 115478 Moscow, Russia
| | | | | | | |
Collapse
|