51
|
Montezano AC, Nguyen Dinh Cat A, Rios FJ, Touyz RM. Angiotensin II and vascular injury. Curr Hypertens Rep 2014; 16:431. [PMID: 24760441 DOI: 10.1007/s11906-014-0431-2] [Citation(s) in RCA: 307] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vascular injury, characterized by endothelial dysfunction, structural remodelling, inflammation and fibrosis, plays an important role in cardiovascular diseases. Cellular processes underlying this include altered vascular smooth muscle cell (VSMC) growth/apoptosis, fibrosis, increased contractility and vascular calcification. Associated with these events is VSMC differentiation and phenotypic switching from a contractile to a proliferative/secretory phenotype. Inflammation, associated with macrophage infiltration and increased expression of redox-sensitive pro-inflammatory genes, also contributes to vascular remodelling. Among the many factors involved in vascular injury is Ang II. Ang II, previously thought to be the sole biologically active downstream peptide of the renin-angiotensin system (RAS), is converted to smaller peptides, [Ang III, Ang IV, Ang-(1-7)], that are functional and that modulate vascular tone and structure. The actions of Ang II are mediated via signalling pathways activated upon binding to AT1R and AT2R. AT1R activation induces effects through PLC-IP3-DAG, MAP kinases, tyrosine kinases, tyrosine phosphatases and RhoA/Rho kinase. Ang II elicits many of its (patho)physiological actions by stimulating reactive oxygen species (ROS) generation through activation of vascular NAD(P)H oxidase (Nox). ROS in turn influence redox-sensitive signalling molecules. Here we discuss the role of Ang II in vascular injury, focusing on molecular mechanisms and cellular processes. Implications in vascular remodelling, inflammation, calcification and atherosclerosis are highlighted.
Collapse
Affiliation(s)
- Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | | | | |
Collapse
|
52
|
Eirin A, Lerman A, Lerman LO. Mitochondria: a pathogenic paradigm in hypertensive renal disease. Hypertension 2014; 65:264-70. [PMID: 25403611 DOI: 10.1161/hypertensionaha.114.04598] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Alfonso Eirin
- From the Divisions of Nephrology and Hypertension (A.E., L.O.L.) and Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- From the Divisions of Nephrology and Hypertension (A.E., L.O.L.) and Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- From the Divisions of Nephrology and Hypertension (A.E., L.O.L.) and Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
53
|
Wojtera E, Konior A, Fedoryszak-Kuśka N, Beręsewicz A. Obligatory role of intraluminal O2- in acute endothelin-1 and angiotensin II signaling to mediate endothelial dysfunction and MAPK activation in guinea-pig hearts. Int J Mol Sci 2014; 15:19417-43. [PMID: 25350109 PMCID: PMC4264120 DOI: 10.3390/ijms151119417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/30/2014] [Accepted: 10/08/2014] [Indexed: 01/07/2023] Open
Abstract
We hypothesized that, due to a cross-talk between cytoplasmic O2--sources and intraluminally expressed xanthine oxidase (XO), intraluminal O2- is instrumental in mediating intraluminal (endothelial dysfunction) and cytosolic (p38 and ERK1/2 MAPKs phosphorylation) manifestations of vascular oxidative stress induced by endothelin-1 (ET-1) and angiotensin II (AT-II). Isolated guinea-pig hearts were subjected to 10-min agonist perfusion causing a burst of an intraluminal O2-. ET-1 antagonist, tezosentan, attenuated AT-II-mediated O2-, indicating its partial ET-1 mediation. ET-1 and Ang-T (AT-II+tezosentan) triggered intraluminal O2-, endothelial dysfunction, MAPKs and p47phox phosphorylation, and NADPH oxidase (Nox) and XO activation. These effects were: (i) prevented by blocking PKC (chelerythrine), Nox (apocynin), mitochondrial ATP-dependent K+ channel (5-HD), complex II (TTFA), and XO (allopurinol); (ii) mimicked by the activation of Nox (NADH); and mitochondria (diazoxide, 3-NPA) and (iii) the effects by NADH were prevented by 5-HD, TTFA and chelerythrine, and those by diazoxide and 3-NPA by apocynin and chelerythrine, suggesting that the agonists coactivate Nox and mitochondria, which further amplify their activity via PKC. The effects by ET-1, Ang-T, NADH, diazoxide, and 3-NPA were opposed by blocking intraluminal O2- (SOD) and XO, and were mimicked by XO activation (hypoxanthine). Apocynin, TTFA, chelerythrine, and SOD opposed the effects by hypoxanthine. In conclusion, oxidative stress by agonists involves cellular inside-out and outside-in signaling in which Nox-mitochondria-PKC system and XO mutually maintain their activities via the intraluminal O2-.
Collapse
Affiliation(s)
- Emilia Wojtera
- Department of Clinical Physiology, Postgraduate Medical School, Warsaw 01-813, Poland.
| | - Anna Konior
- Department of Clinical Physiology, Postgraduate Medical School, Warsaw 01-813, Poland.
| | | | - Andrzej Beręsewicz
- Department of Clinical Physiology, Postgraduate Medical School, Warsaw 01-813, Poland.
| |
Collapse
|
54
|
Pal R, Palmieri M, Loehr JA, Li S, Abo-Zahrah R, Monroe TO, Thakur PB, Sardiello M, Rodney GG. Src-dependent impairment of autophagy by oxidative stress in a mouse model of Duchenne muscular dystrophy. Nat Commun 2014; 5:4425. [PMID: 25028121 PMCID: PMC4101811 DOI: 10.1038/ncomms5425] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/17/2014] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal degenerative muscle disease resulting from mutations in the dystrophin gene. Increased oxidative stress and altered Ca(2+) homeostasis are hallmarks of dystrophic muscle. While impaired autophagy has recently been implicated in the disease process, the mechanisms underlying the impairment have not been elucidated. Here we show that nicotinamide adenine dinucleotide phosphatase (Nox2)-induced oxidative stress impairs both autophagy and lysosome formation in mdx mice. Persistent activation of Src kinase leads to activation of the autophagy repressor mammalian target of rapamycin (mTOR) via PI3K/Akt phosphorylation. Inhibition of Nox2 or Src kinase reduces oxidative stress and partially rescues the defective autophagy and lysosome biogenesis. Genetic downregulation of Nox2 activity in the mdx mouse decreases reactive oxygen species (ROS) production, abrogates defective autophagy and rescues histological abnormalities and contractile impairment. Our data highlight mechanisms underlying the pathogenesis of DMD and identify NADPH oxidase and Src kinase as potential therapeutic targets.
Collapse
Affiliation(s)
- Rituraj Pal
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Michela Palmieri
- Department of Molecular, Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - James A Loehr
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Shumin Li
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Reem Abo-Zahrah
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Tanner O Monroe
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Poulami B Thakur
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Marco Sardiello
- Department of Molecular, Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
55
|
Yao M, Rogers NM, Csányi G, Rodriguez AI, Ross MA, St Croix C, Knupp H, Novelli EM, Thomson AW, Pagano PJ, Isenberg JS. Thrombospondin-1 activation of signal-regulatory protein-α stimulates reactive oxygen species production and promotes renal ischemia reperfusion injury. J Am Soc Nephrol 2014; 25:1171-86. [PMID: 24511121 PMCID: PMC4033366 DOI: 10.1681/asn.2013040433] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 10/29/2013] [Indexed: 01/08/2023] Open
Abstract
Ischemia reperfusion injury (IRI) causes tissue and organ injury, in part, through alterations in tissue blood flow and the production of reactive oxygen species. The cell surface receptor signal-regulatory protein-α (SIRP-α) is expressed on inflammatory cells and suppresses phagocytosis, but the function of SIRP-α in IRI has not been determined. We reported previously that the matricellular protein thrombospondin-1 is upregulated in IRI. Here, we report a novel interaction between thrombospondin-1 and SIRP-α on nonphagocytic cells. In cell-free experiments, thrombospondin-1 bound SIRP-α. In vascular smooth muscle cells and renal tubular epithelial cells, treatment with thrombospondin-1 led to phosphorylation of SIRP-α and downstream activation of Src homology domain 2-containing phosphatase-1. Thrombospondin-1 also stimulated phosphorylation of p47(phox) (an organizer subunit for nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1/2) and increased production of superoxide, both of which were abrogated by knockdown or antibody blockade of SIRP-α. In rodent aortic rings, treatment with thrombospondin-1 increased the production of superoxide and inhibited nitric oxide-mediated vasodilation in a SIRP-α-dependent manner. Renal IRI upregulated the thrombospondin-1-SIRP-α signaling axis and was associated with increased superoxide production and cell death. A SIRP-α antibody that blocks thrombospondin-1 activation of SIRP-α mitigated the effects of renal IRI, increasing blood flow, suppressing production of reactive oxygen species, and preserving cellular architecture. A role for CD47 in SIRP-α activation in these pathways is also described. Overall, these results suggest that thrombospondin-1 binding to SIRP-α on nonphagocytic cells activates NADPH oxidase, limits vasodilation, and promotes renal IRI.
Collapse
Affiliation(s)
| | | | - Gábor Csányi
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology
| | - Andres I Rodriguez
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology
| | | | | | | | | | | | - Patrick J Pagano
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology
| | - Jeffrey S Isenberg
- Vascular Medicine Institute, Starzl Transplantation Institute, Department of Pharmacology and Chemical Biology, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
56
|
Brandes RP, Weissmann N, Schröder K. Redox-mediated signal transduction by cardiovascular Nox NADPH oxidases. J Mol Cell Cardiol 2014; 73:70-9. [PMID: 24560815 DOI: 10.1016/j.yjmcc.2014.02.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 11/30/2022]
Abstract
The only known function of the Nox family of NADPH oxidases is the production of reactive oxygen species (ROS). Some Nox enzymes show high tissue-specific expression and the ROS locally produced are required for synthesis of hormones or tissue components. In the cardiovascular system, Nox enzymes are low abundant and function as redox-modulators. By reacting with thiols, nitric oxide (NO) or trace metals, Nox-derived ROS elicit a plethora of cellular responses required for physiological growth factor signaling and the induction and adaptation to pathological processes. The interactions of Nox-derived ROS with signaling elements in the cardiovascular system are highly diverse and will be detailed in this article, which is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
Affiliation(s)
- Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Germany.
| | - Norbert Weissmann
- Giessen University Lung Center, Justus-Liebig-Universität, Gießen, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Germany
| |
Collapse
|
57
|
Kröller-Schön S, Steven S, Kossmann S, Scholz A, Daub S, Oelze M, Xia N, Hausding M, Mikhed Y, Zinßius E, Mader M, Stamm P, Treiber N, Scharffetter-Kochanek K, Li H, Schulz E, Wenzel P, Münzel T, Daiber A. Molecular mechanisms of the crosstalk between mitochondria and NADPH oxidase through reactive oxygen species-studies in white blood cells and in animal models. Antioxid Redox Signal 2014; 20:247-66. [PMID: 23845067 PMCID: PMC3887465 DOI: 10.1089/ars.2012.4953] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 06/19/2013] [Accepted: 07/08/2013] [Indexed: 12/21/2022]
Abstract
AIMS Oxidative stress is involved in the development of cardiovascular disease. There is a growing body of evidence for a crosstalk between different enzymatic sources of oxidative stress. With the present study, we sought to determine the underlying crosstalk mechanisms, the role of the mitochondrial permeability transition pore (mPTP), and its link to endothelial dysfunction. RESULTS NADPH oxidase (Nox) activation (oxidative burst and translocation of cytosolic Nox subunits) was observed in response to mitochondrial reactive oxygen species (mtROS) formation in human leukocytes. In vitro, mtROS-induced Nox activation was prevented by inhibitors of the mPTP, protein kinase C, tyrosine kinase cSrc, Nox itself, or an intracellular calcium chelator and was absent in leukocytes with p47phox deficiency (regulates Nox2) or with cyclophilin D deficiency (regulates mPTP). In contrast, the crosstalk in leukocytes was amplified by mitochondrial superoxide dismutase (type 2) (MnSOD(+/-)) deficiency. In vivo, increases in blood pressure, degree of endothelial dysfunction, endothelial nitric oxide synthase (eNOS) dysregulation/uncoupling (e.g., eNOS S-glutathionylation) or Nox activity, p47phox phosphorylation in response to angiotensin-II (AT-II) in vivo treatment, or the aging process were more pronounced in MnSOD(+/-) mice as compared with untreated controls and improved by mPTP inhibition by cyclophilin D deficiency or sanglifehrin A therapy. INNOVATION These results provide new mechanistic insights into what extent mtROS trigger Nox activation in phagocytes and cardiovascular tissue, leading to endothelial dysfunction. CONCLUSIONS Our data show that mtROS trigger the activation of phagocytic and cardiovascular NADPH oxidases, which may have fundamental implications for immune cell activation and development of AT-II-induced hypertension.
Collapse
Affiliation(s)
- Swenja Kröller-Schön
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sebastian Steven
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sabine Kossmann
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Center of Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Alexander Scholz
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Steffen Daub
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Oelze
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael Hausding
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Yuliya Mikhed
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Elena Zinßius
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael Mader
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Paul Stamm
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Nicolai Treiber
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | | | - Huige Li
- Department of Pharmacology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Eberhard Schulz
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Philip Wenzel
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Center of Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Münzel
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Daiber
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
58
|
Liu CC, Karimi Galougahi K, Weisbrod RM, Hansen T, Ravaie R, Nunez A, Liu YB, Fry N, Garcia A, Hamilton EJ, Sweadner KJ, Cohen RA, Figtree GA. Oxidative inhibition of the vascular Na+-K+ pump via NADPH oxidase-dependent β1-subunit glutathionylation: implications for angiotensin II-induced vascular dysfunction. Free Radic Biol Med 2013; 65:563-572. [PMID: 23816524 PMCID: PMC4474148 DOI: 10.1016/j.freeradbiomed.2013.06.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 02/07/2023]
Abstract
Glutathionylation of the Na(+)-K(+) pump's β1-subunit is a key molecular mechanism of physiological and pathophysiological pump inhibition in cardiac myocytes. Its contribution to Na(+)-K(+) pump regulation in other tissues is unknown, and cannot be assumed given the dependence on specific β-subunit isoform expression and receptor-coupled pathways. As Na(+)-K(+) pump activity is an important determinant of vascular tone through effects on [Ca(2+)]i, we have examined the role of oxidative regulation of the Na(+)-K(+) pump in mediating angiotensin II (Ang II)-induced increases in vascular reactivity. β1-subunit glutathione adducts were present at baseline and increased by exposure to Ang II in rabbit aortic rings, primary rabbit aortic vascular smooth muscle cells (VSMCs), and human arterial segments. In VSMCs, Ang II-induced glutathionylation was associated with marked reduction in Na(+)-K(+)ATPase activity, an effect that was abolished by the NADPH oxidase inhibitory peptide, tat-gp91ds. In aortic segments, Ang II-induced glutathionylation was associated with decreased K(+)-induced vasorelaxation, a validated index of pump activity. Ang II-induced oxidative inhibition of Na(+)-K(+) ATPase and decrease in K(+)-induced relaxation were reversed by preincubation of VSMCs and rings with recombinant FXYD3 protein that is known to facilitate deglutathionylation of β1-subunit. Knock-out of FXYD1 dramatically decreased K(+)-induced relaxation in a mouse model. Attenuation of Ang II signaling in vivo by captopril (8 mg/kg/day for 7 days) decreased superoxide-sensitive DHE levels in the media of rabbit aorta, decreased β1-subunit glutathionylation, and enhanced K(+)-induced vasorelaxation. Ang II inhibits the Na(+)-K(+) pump in VSMCs via NADPH oxidase-dependent glutathionylation of the pump's β1-subunit, and this newly identified signaling pathway may contribute to altered vascular tone. FXYD proteins reduce oxidative inhibition of the Na(+)-K(+) pump and may have an important protective role in the vasculature under conditions of oxidative stress.
Collapse
Affiliation(s)
- Chia-Chi Liu
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Keyvan Karimi Galougahi
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, Australia
| | - Robert M Weisbrod
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Thomas Hansen
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Ramtin Ravaie
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Andrea Nunez
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Yi B Liu
- Laboratory Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Natasha Fry
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Alvaro Garcia
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Elisha J Hamilton
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Kathleen J Sweadner
- Laboratory Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Gemma A Figtree
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, Australia.
| |
Collapse
|
59
|
Aggarwal S, Gross CM, Sharma S, Fineman JR, Black SM. Reactive oxygen species in pulmonary vascular remodeling. Compr Physiol 2013; 3:1011-34. [PMID: 23897679 DOI: 10.1002/cphy.c120024] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The pathogenesis of pulmonary hypertension is a complex multifactorial process that involves the remodeling of pulmonary arteries. This remodeling process encompasses concentric medial thickening of small arterioles, neomuscularization of previously nonmuscular capillary-like vessels, and structural wall changes in larger pulmonary arteries. The pulmonary arterial muscularization is characterized by vascular smooth muscle cell hyperplasia and hypertrophy. In addition, in uncontrolled pulmonary hypertension, the clonal expansion of apoptosis-resistant endothelial cells leads to the formation of plexiform lesions. Based upon a large number of studies in animal models, the three major stimuli that drive the vascular remodeling process are inflammation, shear stress, and hypoxia. Although, the precise mechanisms by which these stimuli impair pulmonary vascular function and structure are unknown, reactive oxygen species (ROS)-mediated oxidative damage appears to play an important role. ROS are highly reactive due to their unpaired valence shell electron. Oxidative damage occurs when the production of ROS exceeds the quenching capacity of the antioxidant mechanisms of the cell. ROS can be produced from complexes in the cell membrane (nicotinamide adenine dinucleotide phosphate-oxidase), cellular organelles (peroxisomes and mitochondria), and in the cytoplasm (xanthine oxidase). Furthermore, low levels of tetrahydrobiopterin (BH4) and L-arginine the rate limiting cofactor and substrate for endothelial nitric oxide synthase (eNOS), can cause the uncoupling of eNOS, resulting in decreased NO production and increased ROS production. This review will focus on the ROS generation systems, scavenger antioxidants, and oxidative stress associated alterations in vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | |
Collapse
|
60
|
Kim YM, Jin J, Choi JA, Cho SN, Lim YJ, Lee JH, Seo JY, Chen HY, Rha KS, Song CH. Staphylococcus aureus enterotoxin B-induced endoplasmic reticulum stress response is associated with chronic rhinosinusitis with nasal polyposis. Clin Biochem 2013; 47:96-103. [PMID: 24216216 DOI: 10.1016/j.clinbiochem.2013.10.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/30/2013] [Accepted: 10/31/2013] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Staphylococcus aureus enterotoxin B (SEB) might participate in the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP). However, the exact mechanism of polyp formation in CRSwNP remains unclear. Since the endoplasmic reticulum (ER) stress response is closely associated with chronic inflammation, we investigated the association between ER stress and SEB in the pathogenesis of CRSwNP. DESIGN AND METHODS Twenty-three CRSwNP patients with eosinophilic polyps (EP) or non-eosinophilic polyps (NEP) and 10 healthy subjects who were undergoing septoplasty were enrolled in this study. ER stress response was investigated using immunohistochemical staining and Western blotting. RESULTS We show in this study that there are significantly more SEB-positive cells and higher production of reactive oxygen species (ROS) in the epithelial layer of EP than NEP or control tissue. Both SEB and protein A were detected strongly in tissues from patients with CRSwNP. We observed SEB induced the ER stress response in RPMI 2650 cells. GRP78 elevation by SEB was reduced by ROS scavenger pretreatment. In addition, the induction of GRP78 and p47 phox was increased significantly in EP compared with NEP or control mucosa. CONCLUSIONS SEB may induce ER stress via ROS production in CRSwNP. Therefore, we suggest that SEB-induced ER stress may play important roles in the pathogenesis of nasal polyposis.
Collapse
Affiliation(s)
- Yong-Min Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-721, South Korea
| | - Jun Jin
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-721, South Korea; Department of Otorhinolaryngology, Yanbian University Hospital, Jilin Yanji 133000, China
| | - Ji-Ae Choi
- Department of Microbiology, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea; Infection Signaling Network Research, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Soo-Na Cho
- Department of Microbiology, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea; Infection Signaling Network Research, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Yun-Ji Lim
- Department of Microbiology, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea; Infection Signaling Network Research, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Jung-Hwan Lee
- Department of Microbiology, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea; Infection Signaling Network Research, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Jee Yeon Seo
- Department of Microbiology, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea; Infection Signaling Network Research, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea
| | - Hai Yue Chen
- Department of Pathology, Yanbian University Hospital, Jilin Yanji 133000, China
| | - Ki-Sang Rha
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-721, South Korea
| | - Chang-Hwa Song
- Department of Microbiology, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea; Infection Signaling Network Research, College of Medicine, Chungnam National University, Daejeon 301-747, South Korea.
| |
Collapse
|
61
|
Wagner B, Gorin Y. Src tyrosine kinase mediates platelet-derived growth factor BB-induced and redox-dependent migration in metanephric mesenchymal cells. Am J Physiol Renal Physiol 2013; 306:F85-97. [PMID: 24197068 DOI: 10.1152/ajprenal.00371.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The adult kidney is derived from the interaction between the metanephric blastema and the ureteric bud. Platelet-derived growth factor (PDGF) receptor β is essential for the development of the mature glomerular tuft, as mice deficient for this receptor lack mesangial cells. This study investigated the role of Src tyrosine kinase in PDGF-mediated reactive oxygen species (ROS) generation and migration of metanephric mesenchymal cells (MMCs). Cultured embryonic MMCs from wild-type and PDGF receptor-deficient embryos were established. Migration was determined via wound-healing assay. Unlike PDGF AA, PDGF BB-induced greater migration in MMCs with respect to control. This was abrogated by neutralizing an antibody to PDGF BB. Phosphatidylinositol 3-kinase (PI3K) inhibitors suppressed PDGF BB-induced migration. Conversely, mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitors had no effect. Src inhibitors inhibited PDGF-induced cell migration, PI3K activity, and Akt phosphorylation. Adenoviral dominant negative Src (AD DN Src) abrogated PDGF BB-induced Akt phosphorylation. Hydrogen peroxide stimulated cell migration. PDGF BB-induced wound closure was inhibited by the antioxidants N-acetyl-l-cysteine, tiron, and the flavoprotein inhibitor diphenyleneiodonium. These cells express the NADPH oxidase homolog Nox4. Inhibiting Nox4 with antisense oligonucleotides or small interfering RNA (siRNA) suppressed PDGF-induced wound closure. Inhibition of Src with siRNA reduced PDGF BB-induced ROS generation as assessed by 2',7'-dichlorodihydrofluorescein diacetate fluorescence. Furthermore, PDGF BB-stimulated ROS generation and migration were similarly suppressed by Ad DN Src. In MMCs, PDGF BB-induced migration is mediated by PI3K and Src in a redox-dependent manner involving Nox4. Src may be upstream to PI3K and Nox4.
Collapse
Affiliation(s)
- Brent Wagner
- South Texas Veterans Health Care System, Div. of Nephrology MC 7882, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900.
| | | |
Collapse
|
62
|
Nguyen Dinh Cat A, Montezano AC, Burger D, Touyz RM. Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid Redox Signal 2013; 19:1110-20. [PMID: 22530599 PMCID: PMC3771549 DOI: 10.1089/ars.2012.4641] [Citation(s) in RCA: 354] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Angiotensin II (Ang II) influences the function of many cell types and regulates many organ systems, in large part through redox-sensitive processes. In the vascular system, Ang II is a potent vasoconstrictor and also promotes inflammation, hypertrophy, and fibrosis, which are important in vascular damage and remodeling in cardiovascular diseases. The diverse actions of Ang II are mediated via Ang II type 1 and Ang II type 2 receptors, which couple to various signaling molecules, including NADPH oxidase (Nox), which generates reactive oxygen species (ROS). ROS are now recognized as signaling molecules, critically placed in pathways activated by Ang II. Mechanisms linking Nox and Ang II are complex and not fully understood. RECENT ADVANCES Ang II regulates vascular cell production of ROS through various recently characterized Noxs, including Nox1, Nox2, Nox4, and Nox5. Activation of these Noxs leads to ROS generation, which in turn influences many downstream signaling targets of Ang II, including MAP kinases, RhoA/Rho kinase, transcription factors, protein tyrosine phosphatases, and tyrosine kinases. Activation of these redox-sensitive pathways regulates vascular cell growth, inflammation, contraction, and senescence. CRITICAL ISSUES Although there is much evidence indicating a role for Nox/ROS in Ang II function, there is still a paucity of information on how Ang II exerts cell-specific effects through ROS and how Nox isoforms are differentially regulated by Ang II. Moreover, exact mechanisms whereby ROS induce oxidative modifications of signaling molecules mediating Ang II actions remain elusive. FUTURE DIRECTIONS Future research should elucidate these issues to better understand the significance of Ang II and ROS in vascular (patho) biology.
Collapse
Affiliation(s)
- Aurelie Nguyen Dinh Cat
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| | - Augusto C. Montezano
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| | - Dylan Burger
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| | - Rhian M. Touyz
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
63
|
Nazarewicz RR, Dikalova AE, Bikineyeva A, Dikalov SI. Nox2 as a potential target of mitochondrial superoxide and its role in endothelial oxidative stress. Am J Physiol Heart Circ Physiol 2013; 305:H1131-40. [PMID: 23955717 DOI: 10.1152/ajpheart.00063.2013] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Superoxide (O2(·-)) production by the NADPH oxidases is implicated in the pathogenesis of many cardiovascular diseases, including hypertension. We have previously shown that activation of NADPH oxidases increases mitochondrial O2(·-) which is inhibited by the ATP-sensitive K(+) channel (mitoKATP) inhibitor 5-hydroxydecanoic acid and that scavenging of mitochondrial or cytoplasmic O2(·-) inhibits hypertension. We hypothesized that mitoKATP-mediated mitochondrial O2(·-) potentiates cytoplasmic O2(·-) by stimulation of NADPH oxidases. In this work we studied Nox isoforms as a potential target of mitochondrial O2(·-). We tested contribution of reverse electron transfer (RET) from complex II to complex I in mitochondrial O2(·-) production and NADPH oxidase activation in human aortic endothelial cells. Activation of mitoKATP with low dose of diazoxide (100 nM) decreased mitochondrial membrane potential (tetramethylrhodamine methyl ester probe) and increased production of mitochondrial and cytoplasmic O2(·-) measured by site-specific probes and mitoSOX. Inhibition of RET with complex II inhibitor (malonate) or complex I inhibitor (rotenone) attenuated the production of mitochondrial and cytoplasmic O2(·-). Supplementation with a mitochondria-targeted SOD mimetic (mitoTEMPO) or a mitochondria-targeted glutathione peroxidase mimetic (mitoEbselen) inhibited production of mitochondrial and cytoplasmic O2(·-). Inhibition of Nox2 (gp91ds) or Nox2 depletion with small interfering RNA but not Nox1, Nox4, or Nox5 abolished diazoxide-induced O2(·-) production in the cytoplasm. Treatment of angiotensin II-infused mice with RET inhibitor dihydroethidium (malate) significantly reduced blood pressure. Our study suggests that mitoKATP-mediated mitochondrial O2(·-) stimulates cytoplasmic Nox2, contributing to the development of endothelial oxidative stress and hypertension.
Collapse
Affiliation(s)
- Rafal R Nazarewicz
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | |
Collapse
|
64
|
Choi SR, Roh DH, Yoon SY, Kang SY, Moon JY, Kwon SG, Choi HS, Han HJ, Beitz AJ, Oh SB, Lee JH. Spinal sigma-1 receptors activate NADPH oxidase 2 leading to the induction of pain hypersensitivity in mice and mechanical allodynia in neuropathic rats. Pharmacol Res 2013; 74:56-67. [PMID: 23732704 DOI: 10.1016/j.phrs.2013.05.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 05/18/2013] [Accepted: 05/26/2013] [Indexed: 01/27/2023]
Abstract
We have recently demonstrated that spinal sigma-1 receptors (Sig-1Rs) mediate pain hypersensitivity in mice and neuropathic pain in rats. In this study, we examine the role of NADPH oxidase 2 (Nox2)-induced reactive oxygen species (ROS) on Sig-1R-induced pain hypersensitivity and the induction of chronic neuropathic pain. Neuropathic pain was produced by chronic constriction injury (CCI) of the right sciatic nerve in rats. Mechanical allodynia and thermal hyperalgesia were evaluated in mice and CCI-rats. Western blotting and dihydroethidium (DHE) staining were performed to assess the changes in Nox2 activation and ROS production in spinal cord, respectively. Direct activation of spinal Sig-1Rs with the Sig-1R agonist, PRE084 induced mechanical allodynia and thermal hyperalgesia, which were dose-dependently attenuated by pretreatment with the ROS scavenger, NAC or the Nox inhibitor, apocynin. PRE084 also induced an increase in Nox2 activation and ROS production, which were attenuated by pretreatment with the Sig-1R antagonist, BD1047 or apocynin. CCI-induced nerve injury produced an increase in Nox2 activation and ROS production in the spinal cord, all of which were attenuated by intrathecal administration with BD1047 during the induction phase of neuropathic pain. Furthermore, administration with BD1047 or apocynin reversed CCI-induced mechanical allodynia during the induction phase, but not the maintenance phase. These findings demonstrate that spinal Sig-1Rs modulate Nox2 activation and ROS production in the spinal cord, and ultimately contribute to the Sig-1R-induced pain hypersensitivity and the peripheral nerve injury-induced induction of chronic neuropathic pain.
Collapse
Affiliation(s)
- Sheu-Ran Choi
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 151-742, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Soe NN, Sowden M, Baskaran P, Smolock EM, Kim Y, Nigro P, Berk BC. Cyclophilin A is required for angiotensin II-induced p47phox translocation to caveolae in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2013; 33:2147-53. [PMID: 23846495 DOI: 10.1161/atvbaha.113.301894] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Angiotensin II (AngII) signal transduction in vascular smooth muscle cells (VSMC) is mediated by reactive oxygen species (ROS). Cyclophilin A (CyPA) is a ubiquitously expressed cytosolic protein that possesses peptidyl-prolyl cis-trans isomerase activity, scaffold function, and significantly enhances AngII-induced ROS production in VSMC. We hypothesized that CyPA regulates AngII-induced ROS generation by promoting translocation of NADPH oxidase cytosolic subunit p47phox to caveolae of the plasma membrane. APPROACH AND RESULTS Overexpression of CyPA in CyPA-deficient VSMC (CyPA(-/-)VSMC) significantly increased AngII-stimulated ROS production. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors (VAS2870 or diphenylene iodonium) significantly attenuated AngII-induced ROS production in CyPA and p47phox-overexpressing CyPA(-/-)VSMC. Cell fractionation and sucrose gradient analyses showed that AngII-induced p47phox plasma membrane translocation, specifically to the caveolae, was reduced in CyPA(-/-)VSMC compared with wild-type-VSMC. Immunofluorescence studies demonstrated that AngII increased p47phox and CyPA colocalization and translocation to the plasma membrane. In addition, immunoprecipitation of CyPA followed by immunoblotting of p47phox and actin showed that AngII increased CyPA and p47phox interaction. AngII-induced p47phox and actin cell cytoskeleton association was attenuated in CyPA(-/-)VSMC. Mechanistically, inhibition of p47phox phosphorylation and phox homology domain deletion attenuated CyPA and p47phox interaction. Finally, cyclosporine A and CyPA-peptidyl-prolyl cis-trans isomerase mutant, R55A, inhibited AngII-stimulated CyPA and p47phox association in VSMC, suggesting that peptidyl-prolyl cis-trans isomerase activity was required for their interaction. CONCLUSIONS These findings provide the mechanism by which CyPA is an important regulator for AngII-induced ROS generation in VSMC through interaction with p47phox and cell cytoskeleton, which enhances the translocation of p47phox to caveolae.
Collapse
Affiliation(s)
- Nwe Nwe Soe
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | | | | | | | | | | |
Collapse
|
66
|
Spencer NY, Zhou W, Li Q, Zhang Y, Luo M, Yan Z, Lynch TJ, Abbott D, Banfi B, Engelhardt JF. Hepatocytes produce TNF-α following hypoxia-reoxygenation and liver ischemia-reperfusion in a NADPH oxidase- and c-Src-dependent manner. Am J Physiol Gastrointest Liver Physiol 2013; 305:G84-94. [PMID: 23639811 PMCID: PMC3725690 DOI: 10.1152/ajpgi.00430.2012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell line studies have previously demonstrated that hypoxia-reoxygenation (H/R) leads to the production of NADPH oxidase 1 and 2 (NOX1 and NOX2)-dependent reactive oxygen species (ROS) required for the activation of c-Src and NF-κB. We now extend these studies into mouse models to evaluate the contribution of hepatocytes to the NOX- and c-Src-dependent TNF-α production that follows H/R in primary hepatocytes and liver ischemia-reperfusion (I/R). In vitro, c-Src-deficient primary hepatocytes produced less ROS and TNF-α following H/R compared with controls. In vivo, c-Src-KO mice also had impaired TNF-α and NF-κB responses following partial lobar liver I/R. Studies in NOX1 and p47phox knockout primary hepatocytes demonstrated that both NOX1 and p47phox are partially required for H/R-mediated TNF-α production. To further investigate the involvement of NADPH oxidases in the production of TNF-α following liver I/R, we performed additional in vivo experiments in knockout mice deficient for NOX1, NOX2, p47phox, Rac1, and/or Rac2. Cumulatively, these results demonstrate that NOX2 and its activator subunits (p47phox and Rac) control the secretion of TNF-α by the liver following I/R. Interestingly, in the absence of Kupffer cells and NOX2, NOX1 played a dominant role in TNF-α production following hepatic I/R. However, NOX1 deletion alone had little effect on I/R-induced TNF-α. Thus Kupffer cell-derived factors and NOX2 act to suppress hepatic NOX1-dependent TNF-α production. We conclude that c-Src and NADPH oxidase components are necessary for redox-mediated production of TNF-α following liver I/R and that hepatocytes play an important role in this process.
Collapse
Affiliation(s)
- Netanya Y. Spencer
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Weihong Zhou
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Qiang Li
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Yulong Zhang
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Meihui Luo
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Ziying Yan
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Thomas J. Lynch
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Duane Abbott
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Botond Banfi
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| |
Collapse
|
67
|
Affiliation(s)
- Carmine Savoia
- Carmine Savoia is Associate Professor of Cardiology, Cardiology Unit and Chair, Clinical and Molecular Medicine Department, Sapienza University of Rome, Italy. After a fellowship in Ernesto Schiffrin’s laboratory, he has continued his research on pathophysiology of hypertension, vascular remodeling and cardiovascular damage, the renin–angiotensin–aldosterone system, and clinical studies in diabetic and/or hypertensive patients
| | - Ernesto L Schiffrin
- Ernesto L Schiffrin is Physician-in-Chief, Jewish General Hospital, Canada Research Chair in Hypertension and Vascular Research, Lady Davis Institute for Medical Research, and Professor and Vice-Chair (Research), Department of Medicine, McGill University (Montreal, Canada). His research deals with vascular remodeling in hypertension, renal and cardiometabolic diseases, from mice to humans, and the influence of the renin–angiotensin–aldosterone and endothelin systems, nuclear receptors and immunity on
| |
Collapse
|
68
|
Abstract
Oxidative stress has been linked to the pathogenesis of the major complications of diabetes in the kidney, the heart, the eye or the vasculature. NADPH oxidases of the Nox family are a major source of ROS (reactive oxygen species) and are critical mediators of redox signalling in cells from different organs afflicted by the diabetic milieu. In the present review, we provide an overview of the current knowledge related to the understanding of the role of Nox in the processes that control cell injury induced by hyperglycaemia and other predominant factors enhanced in diabetes, including the renin–angiotensin system, TGF-β (transforming growth factor-β) and AGEs (advanced glycation end-products). These observations support a critical role for Nox homologues in diabetic complications and indicate that NADPH oxidases are an important therapeutic target. Therefore the design and development of small-molecule inhibitors that selectively block Nox oxidases appears to be a reasonable approach to prevent or retard the complications of diabetes in target organs. The bioefficacy of these agents in experimental animal models is also discussed in the present review.
Collapse
|
69
|
Affiliation(s)
- Augusto C. Montezano
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Rhian M. Touyz
- From the British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
70
|
Schroeter MR, Leifheit-Nestler M, Hubert A, Schumann B, Glückermann R, Eschholz N, Krüger N, Lutz S, Hasenfuss G, Konstantinides S, Schäfer K. Leptin promotes neointima formation and smooth muscle cell proliferation via NADPH oxidase activation and signalling in caveolin-rich microdomains. Cardiovasc Res 2013; 99:555-65. [PMID: 23723060 DOI: 10.1093/cvr/cvt126] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIMS Apolipoprotein E (apoE) may act as a vasculoprotective factor by promoting plasma lipid clearance and cholesterol efflux. Moreover, apoE accumulates at sites of vascular injury and modulates the effect of growth factors on smooth muscle cells (SMCs). Experimental data suggested that hypothalamic apoE expression is reduced in obesity and associated with leptin resistance. In this study, we examined the role of apoE in mediating the effects of leptin on vascular lesion formation. METHODS AND RESULTS Leptin was administered to apoE knockout (apoE-/-) mice via osmotic pumps to increase its circulating levels. Morphometric analysis revealed that leptin did not alter neointima formation and failed to increase α-actin- or PCNA-immunopositive SMCs after vascular injury. Similar findings were obtained after analysis of atherosclerotic lesions. Comparison of apoE-/-, wild-type, or LDL receptor-/- mice and functional analyses in aortic SMCs from WT or apoE-/- mice or human arterial SMCs after treatment with small interfering (si)RNA or heparinase revealed that leptin requires the presence of apoE, expressed, secreted and bound to the cell surface, to fully activate leptin receptor signalling and to promote SMC proliferation and neointima formation. Mechanistically, leptin induced the phosphorylation and membrane translocation of caveolin (cav)-1, and apoE down-regulation or caveolae disruption inhibited the leptin-induced p47phox activation, ROS formation and SMC proliferation. Finally, leptin failed to increase neointima formation in mice lacking cav-1. CONCLUSION Our findings suggest that apoE mediates the effects of leptin on vascular lesion formation by stabilizing cav-1-enriched cell membrane microdomains in SMCs, thus allowing NADPH oxidase assembly and ROS-mediated mitogenic signalling.
Collapse
Affiliation(s)
- Marco R Schroeter
- Department of Cardiology and Pulmonary Medicine, University Medical Center Göttingen, Robert Koch Strasse 40, Göttingen D-37075, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
de Queiroz TM, Monteiro MMO, Braga VA. Angiotensin-II-derived reactive oxygen species on baroreflex sensitivity during hypertension: new perspectives. Front Physiol 2013; 4:105. [PMID: 23717285 PMCID: PMC3651964 DOI: 10.3389/fphys.2013.00105] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 04/24/2013] [Indexed: 12/12/2022] Open
Abstract
Hypertension is a multifactorial disorder, which has been associated with the reduction in baroreflex sensitivity (BRS) and autonomic dysfunction. Several studies have revealed that increased reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate [NAD(P)H] oxidase, following activation of type 1 receptor (AT1R) by Angiotensin-(Ang) II, the main peptide of the Renin–Angiotensin–Aldosterone System (RAAS), is the central mechanism involved in Ang-II-derived hypertension. In the present review, we will discuss the role of Ang II and oxidative stress in hypertension, the relationship between the BRS and the genesis of hypertension and how the oxidative stress triggers baroreflex dysfunction in several models of hypertension. Finally, we will describe some novel therapeutic drugs for improving the BRS during hypertension.
Collapse
Affiliation(s)
- Thyago M de Queiroz
- Department of Biotechnology, Biotechnology Center, Federal University of Paraiba João Pessoa, Brazil
| | | | | |
Collapse
|
72
|
Takada S, Kinugawa S, Hirabayashi K, Suga T, Yokota T, Takahashi M, Fukushima A, Homma T, Ono T, Sobirin MA, Masaki Y, Mizushima W, Kadoguchi T, Okita K, Tsutsui H. Angiotensin II receptor blocker improves the lowered exercise capacity and impaired mitochondrial function of the skeletal muscle in type 2 diabetic mice. J Appl Physiol (1985) 2013; 114:844-57. [DOI: 10.1152/japplphysiol.00053.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
NAD(P)H oxidase-induced oxidative stress is at least in part involved with lowered exercise capacity and impaired mitochondrial function in high-fat diet (HFD)-induced diabetic mice. NAD(P)H oxidase can be activated by activation of the renin-angiotensin system. We investigated whether ANG II receptor blocker can improve exercise capacity in diabetic mice. C57BL/6J mice were fed a normal diet (ND) or HFD, and each group of mice was divided into two groups: treatment with or without olmesartan (OLM; 3 mg·kg−1·day−1 in the drinking water). The following groups of mice were studied: ND, ND+OLM, HFD, and HFD+OLM ( n = 10 for each group). After 8 wk, HFD significantly increased body weight, plasma glucose, and insulin compared with ND, and OLM did not affect these parameters in either group. Exercise capacity, as determined by treadmill tests, was significantly reduced in HFD, and this reduction was ameliorated in HFD+OLM. ADP-dependent mitochondrial respiration was significantly decreased, and NAD(P)H oxidase activity and superoxide production by lucigenin chemiluminescence were significantly increased in skeletal muscle from HFD, which were attenuated by OLM. There were no such effects by OLM in ND. We concluded that OLM ameliorated the decrease in exercise capacity in diabetic mice via improvement in mitochondrial function and attenuation of oxidative stress in skeletal muscle. These data may have a clinical impact on exercise capacity in the medical treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Shingo Takada
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Research Fellow of the Japan Society for the Promotion of Science
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kagami Hirabayashi
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tadashi Suga
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Research Fellow of the Japan Society for the Promotion of Science
| | - Takashi Yokota
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masashige Takahashi
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Arata Fukushima
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tsuneaki Homma
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Taisuke Ono
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | - Yoshihiro Masaki
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Wataru Mizushima
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tomoyasu Kadoguchi
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Koichi Okita
- Graduate School of Program in Lifelong Learning Studies, Hokusho University, Ebetsu, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
73
|
Popolo A, Autore G, Pinto A, Marzocco S. Oxidative stress in patients with cardiovascular disease and chronic renal failure. Free Radic Res 2013; 47:346-56. [PMID: 23438723 DOI: 10.3109/10715762.2013.779373] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Oxidative response regulates many physiological response in human health, but if not properly regulated it could also lead to a number of deleterious effects. The importance of oxidative stress injury depends on the molecular target, the severity of the stress, and the mechanism by which the oxidative stress is imposed: it has been implicated in several diseases including cancer, neurodegenerative diseases, malaria, rheumatoid arthritis and cardiovascular and kidney disease. Most of the common diseases, such as hypertension, atherosclerosis, heart failure, and renal dysfunction, are associated with vascular functional and structural alterations including endothelial dysfunction, altered contractility, and vascular remodeling. Common to these processes is increased bioavailability of reactive oxygen species (ROS), decreased nitric oxide (NO) levels, and reduced antioxidant capacity. Oxidative processes are up-regulated also in patients with chronic renal failure (CRF) and seem to be a cause of elevated risk of morbidity and mortality in these patients. In this review, we highlight the role of oxidative stress in cardiovascular and renal disease.
Collapse
Affiliation(s)
- A Popolo
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | | | | | | |
Collapse
|
74
|
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: 10] [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
|
75
|
Reactive oxygen species in vascular formation and development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:374963. [PMID: 23401740 PMCID: PMC3564431 DOI: 10.1155/2013/374963] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/29/2012] [Accepted: 12/29/2012] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) are derived from the metabolism of oxygen and are traditionally viewed as toxic byproducts that cause damage to biomolecules. It is now becoming widely acknowledged that ROS are key modulators in a variety of biological processes and pathological states. ROS mediate key signaling transduction pathways by reversible oxidation of certain signaling components and are involved in the signaling of growth factors, G-protein-coupled receptors, Notch, and Wnt and its downstream cascades including MAPK, JAK-STAT, NF-κB, and PI3K/AKT. Vascular formation and development is one of the most important events during embryogenesis and is vital for postnasal tissue repair. In this paper, we will discuss how ROS regulate different steps in vascular development, including smooth muscle cell differentiation, angiogenesis, endothelial progenitor cells recruitment, and vascular cell migration.
Collapse
|
76
|
Csányi G, Yao M, Rodríguez AI, Al Ghouleh I, Sharifi-Sanjani M, Frazziano G, Huang X, Kelley EE, Isenberg JS, Pagano PJ. Thrombospondin-1 regulates blood flow via CD47 receptor-mediated activation of NADPH oxidase 1. Arterioscler Thromb Vasc Biol 2012; 32:2966-73. [PMID: 23087362 DOI: 10.1161/atvbaha.112.300031] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Although the matricellular protein thrombospondin-1 (TSP1) is highly expressed in the vessel wall in response to injury, its pathophysiological role in the development of vascular disease is poorly understood. This study was designed to test the hypothesis that TSP1 stimulates reactive oxygen species production in vascular smooth muscle cells and induces vascular dysfunction by promoting oxidative stress. METHODS AND RESULTS Nanomolar concentrations of TSP1 found in human vascular disease robustly stimulated superoxide (O(2)(•-)) levels in vascular smooth muscle cells at both cellular and tissue level as measured by cytochrome c and electron paramagnetic resonance. A peptide mimicking the C terminus of TSP1 known to specifically bind CD47 recapitulated this response. Transcriptional knockdown of CD47 and a monoclonal inhibitory CD47 antibody abrogated TSP1-triggered O(2)(•-) in vitro and ex vivo. TSP1 treatment of vascular smooth muscle cells activated phospholipase C and protein kinase C, resulting in phosphorylation of the NADPH oxidase organizer subunit p47(phox) and subsequent Nox1 activation, leading to impairment of arterial vasodilatation ex vivo. Further, we observed that blockade of CD47 and NADPH oxidase 1 gene silencing in vivo in rats improves TSP1-induced impairment of tissue blood flow after ischemia reperfusion. CONCLUSIONS Our data suggest a highly regulated process of reactive oxygen species stimulation and blood flow regulation promoted through a direct TSP1/CD47-mediated activation of Nox1. This is the first report, to our knowledge, of a matricellular protein acting as a ligand for NADPH oxidase activation and through specific engagement of integrin-associated protein CD47.
Collapse
Affiliation(s)
- Gábor Csányi
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
The vascular phenotypes in hypertension: Relation with the natural history of hypertension. ACTA ACUST UNITED AC 2012; 1:56-67. [PMID: 20409833 DOI: 10.1016/j.jash.2006.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Accepted: 11/10/2006] [Indexed: 11/23/2022]
Abstract
The different vascular phenotypes found in hypertension comprise different aspects. They may be clinical, diagnostic, structural, mechanical, functional, cellular and extracellular, signaling and molecular, proteomic, and gene expression phenotypes. In this manuscript the emphasis will be on the various structure, mechanics, dysfunction, and cell and signaling changes that can be demonstrated in hypertension, and particularly in human hypertension. The phenotype relates to the natural history of hypertension, increasingly elucidated on the basis of cohort studies. The evolution from pre-hypertension to diastolic, systolic, and systo-diastolic hypertension may have a vascular substratum that could explain, in part, the prevalence of each of these phenotypes. The potential for intervention to prevent the passage from pre-hypertension to hypertension thanks to therapies that modulate the development of vascular remodeling is highlighted.
Collapse
|
78
|
Chi PL, Chen YW, Hsiao LD, Chen YL, Yang CM. Heme oxygenase 1 attenuates interleukin-1β-induced cytosolic phospholipase A2 expression via a decrease in NADPH oxidase/reactive oxygen species/activator protein 1 activation in rheumatoid arthritis synovial fibroblasts. ACTA ACUST UNITED AC 2012; 64:2114-25. [PMID: 22231145 DOI: 10.1002/art.34371] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Reactive oxygen species (ROS) produced by cytokines induce the expression of inflammatory mediators in rheumatoid arthritis (RA). Heme oxygenase 1 (HO-1) exerts an antiinflammatory effect. The aim of this study was to examine the mechanisms underlying interleukin-1β (IL-1β)-induced cytosolic phospholipase A2 (cPLA2) expression through ROS generation as modulated by HO-1 in RA synovial fibroblasts (RASFs). METHODS IL-1β-induced ROS generation was determined by flow cytometry. The involvement of MAPKs and NADPH oxidase (NOX)/ROS in IL-1β-induced cPLA2 expression was investigated using pharmacologic inhibitors and transfection with small interfering RNAs (siRNAs) and was analyzed by Western blotting and promoter assay. Overexpression of HO-1 was performed by transfection of RASFs with a recombinant adenovirus containing human HO-1 plasmid. SCID mice with inflammation caused by IL-1β were infected with adenovirus containing HO-1. Histologic characterization of joint inflammation and local expression of cPLA2 were evaluated after treatment. RESULTS IL-1β-induced cPLA2 expression was mediated through NOX activation/ROS production, which was attenuated by N-acetylcysteine (NAC; a scavenger of ROS), the inhibitors of NOX (diphenyleneiodonium chloride and apocynin), MEK-1/2 (U0126), and JNK-1/2 (SP600125), transfection with the respective siRNAs, and the overexpression of HO-1 in RASFs. IL-1β-induced cPLA2 expression was mediated through recruitment of activator protein 1 (AP-1) to the cPLA2 promoter region, which was attenuated by NAC and overexpression of HO-1. Furthermore, HO-1 overexpression inhibited IL-1β-mediated cPLA2 expression in SCID mice. CONCLUSION In RASFs, IL-1β induced cPLA2 expression via activation of p42/p44 MAPK and JNK-1/2, leading to p47phox phosphorylation, ROS production, and AP-1 activation. The induction of HO-1 exerted protective effects on the pathogenesis of RA.
Collapse
Affiliation(s)
- Pei-Ling Chi
- Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | | | | | | | | |
Collapse
|
79
|
Lv P, Miao SB, Shu YN, Dong LH, Liu G, Xie XL, Gao M, Wang YC, Yin YJ, Wang XJ, Han M. Phosphorylation of Smooth Muscle 22α Facilitates Angiotensin II–Induced ROS Production Via Activation of the PKCδ-P47
phox
Axis Through Release of PKCδ and Actin Dynamics and Is Associated With Hypertrophy and Hyperplasia of Vascular Smooth Muscle Cells In Vitro and In Vivo. Circ Res 2012; 111:697-707. [DOI: 10.1161/circresaha.112.272013] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
We have demonstrated that smooth muscle (SM) 22α inhibits cell proliferation via blocking Ras-ERK1/2 signaling in vascular smooth muscle cells (VSMCs) and in injured arteries. The recent study indicates that SM22α disruption can independently promote arterial inflammation through activation of reactive oxygen species (ROS)-mediated NF-κB pathways. However, the mechanisms by which SM22α controls ROS production have not been characterized.
Objective:
To investigate how SM22α disruption promotes ROS production and to characterize the underlying mechanisms.
Methods and Results:
ROS level was measured by dihydroethidium staining for superoxide and TBA assay for malondialdehyde, respectively. We showed that downregulation and phosphorylation of SM22α were associated with angiotensin (Ang) II–induced increase in ROS production in VSMCs of rats and human. Ang II induced the phosphorylation of SM22α at Serine 181 in an Ang II type 1 receptor–PKCδ pathway–dependent manner. Phosphorylated SM22α activated the protein kinase C (PKC)δ-p47
phox
axis via 2 distinct pathways: (1) disassociation of PKCδ from SM22α, and in turn binding to p47
phox
, in the early stage of Ang II stimulation; and (2) acceleration of SM22α degradation through ubiquitin-proteasome, enhancing PKCδ membrane translocation via induction of actin cytoskeletal dynamics in later oxidative stress. Inhibition of SM22α phosphorylation abolished the Ang II–activated PKCδ-p47
phox
axis and inhibited the hypertrophy and hyperplasia of VSMCs in vitro and in vivo, accompanied with reduction of ROS generation.
Conclusions:
These findings indicate that the disruption of SM22α plays pivotal roles in vascular oxidative stress. PKCδ-mediated SM22α phosphorylation is a novel link between actin cytoskeletal remodeling and oxidative stress and may be a potential target for the development of new therapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Pin Lv
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Sui-Bing Miao
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Ya-Nan Shu
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Li-Hua Dong
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - George Liu
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Xiao-Li Xie
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Min Gao
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Yu-Can Wang
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Ya-Juan Yin
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Xiao-Juan Wang
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Mei Han
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| |
Collapse
|
80
|
Queisser N, Schupp N. Aldosterone, oxidative stress, and NF-κB activation in hypertension-related cardiovascular and renal diseases. Free Radic Biol Med 2012; 53:314-27. [PMID: 22609249 DOI: 10.1016/j.freeradbiomed.2012.05.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 05/07/2012] [Accepted: 05/07/2012] [Indexed: 02/07/2023]
Abstract
The mineralocorticoid aldosterone regulates electrolyte and fluid balance and is involved in blood pressure homoeostasis. Classically, it binds to its intracellular mineralocorticoid receptor to induce expression of proteins influencing the reabsorption of sodium and water in the distal nephron. Aldosterone gained special attention when large clinical studies showed that blocking its receptor in patients with cardiovascular diseases reduced their mortality. These patients present increased plasma aldosterone levels. The exact mechanisms of the potential toxic effects of aldosterone leading to cardiovascular damage are not known yet. The observation of reduced nitric oxide bioavailability in hyperaldosteronism implied the generation of oxidative stress by aldosterone. Subsequent studies confirmed the increase of oxidative stress markers in patients with chronic heart failure and in animal models of hyperaldosteronism. The effects of reactive oxygen species have been related to the activation of transcription factors, such as NF-κB. This review summarizes the present-day knowledge of aldosterone-induced oxidative stress and NF-κB activation in humans and different experimental models.
Collapse
Affiliation(s)
- Nina Queisser
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | | |
Collapse
|
81
|
Chignalia AZ, Schuldt EZ, Camargo LL, Montezano AC, Callera GE, Laurindo FR, Lopes LR, Avellar MCW, Carvalho MHC, Fortes ZB, Touyz RM, Tostes RC. Testosterone Induces Vascular Smooth Muscle Cell Migration by NADPH Oxidase and c-Src–Dependent Pathways. Hypertension 2012; 59:1263-71. [DOI: 10.1161/hypertensionaha.111.180620] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Andreia Z. Chignalia
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Elke Z. Schuldt
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Lívia L. Camargo
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Augusto C. Montezano
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Gláucia E. Callera
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Francisco R. Laurindo
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Lucia R. Lopes
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Maria Christina W. Avellar
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Maria Helena C. Carvalho
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Zuleica B. Fortes
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Rhian M. Touyz
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| | - Rita C. Tostes
- From the Institute of Biomedical Sciences (A.Z.C., E.Z.S., L.L.C., L.R.L., M.H.C.C., Z.B.F., R.C.T.), and Heart Institute, School of Medicine (A.Z.C., F.R.L.), University of São Paulo, São Paulo, Brazil; Kidney Research Centre (A.Z.C., E.Z.S., A.C.M., G.E.C., R.M.T.), Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Section of Experimental Endocrinology (M.C.W.A.), Department of Pharmacology, Federal University of São Paulo, Sao Paulo, Brazil; Department of
| |
Collapse
|
82
|
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: 617] [Impact Index Per Article: 47.5] [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
|
83
|
Reactive Oxygen Species Modulation of Na/K-ATPase Regulates Fibrosis and Renal Proximal Tubular Sodium Handling. Int J Nephrol 2012; 2012:381320. [PMID: 22518311 PMCID: PMC3299271 DOI: 10.1155/2012/381320] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 11/07/2011] [Indexed: 01/11/2023] Open
Abstract
The Na/K-ATPase is the primary force regulating renal sodium handling and plays a key role in both ion homeostasis and blood pressure regulation. Recently, cardiotonic steroids (CTS)-mediated Na/K-ATPase signaling has been shown to regulate fibrosis, renal proximal tubule (RPT) sodium reabsorption, and experimental Dahl salt-sensitive hypertension in response to a high-salt diet. Reactive oxygen species (ROS) are an important modulator of nephron ion transport. As there is limited knowledge regarding the role of ROS-mediated fibrosis and RPT sodium reabsorption through the Na/K-ATPase, the focus of this review is to examine the possible role of ROS in the regulation of Na/K-ATPase activity, its signaling, fibrosis, and RPT sodium reabsorption.
Collapse
|
84
|
Kassab A, Piwowar A. Cell oxidant stress delivery and cell dysfunction onset in type 2 diabetes. Biochimie 2012; 94:1837-48. [PMID: 22333037 DOI: 10.1016/j.biochi.2012.01.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 01/25/2012] [Indexed: 01/18/2023]
Abstract
Most known pathways of diabetic complications involve oxidative stress. The mitochondria electron transport chain is a significant source of reactive oxygen species (ROS) in insulin secretory cells, insulin peripheral sensitive cells and endothelial cells. Elevated intracellular glucose level induces tricarboxylic acid cycle electron donor overproduction and mitochondrial proton gradient increase leading to an increase in electron transporter lifetime. Subsequently, the electrons leaked combine with respiratory oxygen (O(2)) resulting in superoxide anion ((•)O(2)(-)) production. Advanced glycation end products derive ROS via interaction with their receptors. Elevated diacylglycerol and ROS activate the protein kinase C pathway which, in turn, activates NADPH oxidases. A vicious circle of pathway derived ROS installs. Pathologic pathways induced ROS are activated and persistent though glycemia returns to normal due to hyperglycemia memory. Endothelial nitric oxide synthase may produce both superoxide anion ((•)O(2)(-)) and nitric oxide (NO) leading to peroxynitrite ((•)ONOO(-)) generation. Homocysteine is also implicated in oxidative stress pathogenesis. In this paper we have highlighted the pathologic mechanisms of ROS on atherosclerosis, renal dysfunction, retina dysfunction and nerve dysfunction in type 2 diabetes. Cell oxidant stress delivery have pivotal role in cell dysfunction onset and progression of angiopathies but an early introduction of good glycemic control may protect cells more efficiently than antioxidants.
Collapse
Affiliation(s)
- Asma Kassab
- Biochemistry Laboratory, CHU Farhat Hached, Sousse, Tunisia.
| | | |
Collapse
|
85
|
Kim JM, Heo HS, Ha YM, Ye BH, Lee EK, Choi YJ, Yu BP, Chung HY. Mechanism of Ang II involvement in activation of NF-κB through phosphorylation of p65 during aging. AGE (DORDRECHT, NETHERLANDS) 2012; 34:11-25. [PMID: 21318332 PMCID: PMC3260361 DOI: 10.1007/s11357-011-9207-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 01/03/2011] [Indexed: 05/30/2023]
Abstract
Angiotensin II (Ang II), a major effector of the renin-angiotensin system, is now recognized as a pro-inflammatory mediator. This Ang II signaling, which causes transcription of pro-inflammatory genes, is regulated through nuclear factor-κB (NF-κB). At present, the molecular mechanisms underlying the effect of aging on Ang II signaling and NF-κB activation are not fully understood. The purpose of this study was to document altered molecular events involved in age-related changes in Ang II signaling and NF-κB activation. Experimentations were carried out using kidney tissues from Fischer 344 rats at 6, 12, 18, and 24 months of age, and the rat endothelial cell line, YPEN-1 for the detailed molecular work. Results show that increases in Ang II and Ang II type 1 receptor during aging were accompanied by the generation of reactive species. Increased Ang II activated NF-κB by phosphorylating IκBα and p65. Increased phosphorylation of p65 at Ser 536 was mediated by the enhanced phosphorylation of IκB kinase αβ, while phosphorylation site Ser 276 of p65 was mediated by upregulated mitogen-activated and stress-activated protein kinase-1. These altered molecular events in aged animals were partly verified by experiments using YPEN-1 cells. Collectively, our findings provide molecular insights into the pro-inflammatory actions of Ang II, actions that influence the phosphorylation of p65-mediated NF-κB activation during aging. Our study demonstrates the age-related pleiotropic nature of the physiologically important Ang II can change into a deleterious culprit that contributes to an increased incidence of many chronic diseases such as atherosclerosis, diabetes, and dementia.
Collapse
Affiliation(s)
- Ji Min Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Hyoung-Sam Heo
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Young Mi Ha
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Byeong Hyeok Ye
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Eun Kyeong Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Yeon Ja Choi
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Byung Pal Yu
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78229-3900 USA
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| |
Collapse
|
86
|
Usatyuk PV, Singleton PA, Pendyala S, Kalari SK, He D, Gorshkova IA, Camp SM, Moitra J, Dudek SM, Garcia JGN, Natarajan V. Novel role for non-muscle myosin light chain kinase (MLCK) in hyperoxia-induced recruitment of cytoskeletal proteins, NADPH oxidase activation, and reactive oxygen species generation in lung endothelium. J Biol Chem 2012; 287:9360-75. [PMID: 22219181 DOI: 10.1074/jbc.m111.294546] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We recently demonstrated that hyperoxia (HO) activates lung endothelial cell NADPH oxidase and generates reactive oxygen species (ROS)/superoxide via Src-dependent tyrosine phosphorylation of p47(phox) and cortactin. Here, we demonstrate that the non-muscle ~214-kDa myosin light chain (MLC) kinase (nmMLCK) modulates the interaction between cortactin and p47(phox) that plays a role in the assembly and activation of endothelial NADPH oxidase. Overexpression of FLAG-tagged wild type MLCK in human pulmonary artery endothelial cells enhanced interaction and co-localization between cortactin and p47(phox) at the cell periphery and ROS production, whereas abrogation of MLCK using specific siRNA significantly inhibited the above. Furthermore, HO stimulated phosphorylation of MLC and recruitment of phosphorylated and non-phosphorylated cortactin, MLC, Src, and p47(phox) to caveolin-enriched microdomains (CEM), whereas silencing nmMLCK with siRNA blocked recruitment of these components to CEM and ROS generation. Exposure of nmMLCK(-/-) null mice to HO (72 h) reduced ROS production, lung inflammation, and pulmonary leak compared with control mice. These results suggest a novel role for nmMLCK in hyperoxia-induced recruitment of cytoskeletal proteins and NADPH oxidase components to CEM, ROS production, and lung injury.
Collapse
Affiliation(s)
- Peter V Usatyuk
- Department of Pharmacology, University of Illinois, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Yang LL, Li DY, Zhang YB, Zhu MY, Chen D, Xu TD. Salvianolic acid A inhibits angiotensin II-induced proliferation of human umbilical vein endothelial cells by attenuating the production of ROS. Acta Pharmacol Sin 2012; 33:41-8. [PMID: 22101169 PMCID: PMC4010265 DOI: 10.1038/aps.2011.133] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Accepted: 09/15/2011] [Indexed: 12/12/2022]
Abstract
AIM To investigate the action of salvianolic acid A (SalA) on angiotensin II (Ang II)-induced proliferation of human umbilical vein endothelial cells (HUVECs) and the possible signaling pathways mediating this action. METHODS Cell proliferation was examined with MTT assay. The expression levels of Src phosphorylation (phospho-Src), Akt phosphorylation (phospho-Akt), and NADPH oxidase 4 (Nox4) in HUVECs were determined by Western blot. The production of reactive oxygen species (ROS) was estimated using fluorescence-activated cell sorting (FACS). RESULTS SalA (6.25-50 μmol/L) did not affect the viability of HUVECs. Treatment of HUVECs with Ang II (1 μmol/L) markedly increased the cell viability; pretreatment of HUVECs with SalA (12.5, 25 and 50 μmol/L) prevented Ang II-induced increase of the cell viability in a concentration-dependent manner. Treatment of HUVECs with Ang II (1 μmol/L) markedly up-regulated the protein expression levels of phospho-Src, phospho-Akt (473) and Nox4; pretreatment of HUVECs with SalA (12.5, 25 and 50 μmol/L) blocked all the effects in a concentration-dependent manner. Treatment of HUVECs with Ang II (1 μmol/L) dramatically increased ROS production in HUVECs; pretreatment of HUVECs with SalA (12.5, 25 and 50 μmol/L) blocked the ROS production in a concentration-dependent manner. CONCLUSION SalA inhibits Ang II-induced proliferation of HUVECs via reducing the expression levels of phospho-Src and phospho-Akt (473), thereby attenuating the production of ROS.
Collapse
Affiliation(s)
- Luan-luan Yang
- Institute of Cardiovascular Disease, Xuzhou Medical College, Xuzhou 221002, China
| | - Dong-ye Li
- Institute of Cardiovascular Disease, Xuzhou Medical College, Xuzhou 221002, China
| | - Yan-bin Zhang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| | - Man-yi Zhu
- Institute of Cardiovascular Disease, Xuzhou Medical College, Xuzhou 221002, China
| | - Dan Chen
- Institute of Cardiovascular Disease, Xuzhou Medical College, Xuzhou 221002, China
| | - Tong-da Xu
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, China
| |
Collapse
|
88
|
Lai EY, Solis G, Luo Z, Carlstrom M, Sandberg K, Holland S, Wellstein A, Welch WJ, Wilcox CS. p47(phox) is required for afferent arteriolar contractile responses to angiotensin II and perfusion pressure in mice. Hypertension 2011; 59:415-20. [PMID: 22184329 DOI: 10.1161/hypertensionaha.111.184291] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Myogenic and angiotensin contractions of afferent arterioles generate reactive oxygen species. Resistance vessels express neutrophil oxidase-2 and -4. Angiotensin II activates p47(phox)/neutrophil oxidase-2, whereas it downregulates NOX-4. Therefore, we tested the hypothesis that p47(phox) enhances afferent arteriolar angiotensin contractions. Angiotensin II infusion in p47(phox) +/+ but not -/- mice increased renal cortical NADPH oxidase activity (7±1-12±1 [P<0.01] versus 5±1-7±1 10(3) · RLU · min(-1) · μg protein(-1) [P value not significant]), mean arterial pressure (77±2-91±2 [P<0.005] versus 74±2-77±1 mm Hg [P value not significant]), and renal vascular resistance (7.5±0.4-10.1±0.7 [P<0.01] versus 7.9±0.4-8.3±0.4 mm Hg/mL · min(-1) · gram kidney weight(-1) [P value not significant]). Afferent arterioles from p47(phox) -/- mice had a lesser myogenic response (3.1±0.4 versus 1.4±0.2 dynes · cm(-1) · mm Hg(-1); P<0.02) and a lesser (P<0.05) contraction to 10(-6) M angiotensin II (diameter change +/+: 9.3±0.2-3.4±0.6 μm versus -/-: 9.9±0.6-7.5±0.4 μm). Angiotensin and increased perfusion pressure generated significantly (P<0.05) more reactive oxygen species in p47(phox) +/+ than -/- arterioles. Angiotensin II infusion increased the maximum responsiveness of afferent arterioles from p47(phox) +/+ mice to 10(-6) M angiotensin II yet decreased the response in p47(phox) -/- mice. The angiotensin infusion increased the sensitivity to angiotensin II only in p47(phox) +/+ mice. We conclude that p47(phox) is required to enhance renal NADPH oxidase activity and basal afferent arteriolar myogenic and angiotensin II contractions and to switch afferent arteriolar tachyphylaxis to sensitization to angiotensin during a prolonged angiotensin infusion. These effects likely contribute to hypertension and renal vasoconstriction during infusion of angiotensin II.
Collapse
Affiliation(s)
- En Yin Lai
- Hypertension, Kidney, and Vascular Research Center, Georgetown University Medical Center, NW, Washington, DC 20007, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Chan YC, Leung PS. The Renin-angiotensin system and reactive oxygen species: implications in pancreatitis. Antioxid Redox Signal 2011; 15:2743-55. [PMID: 21644836 DOI: 10.1089/ars.2011.4071] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE The renin-angiotensin system (RAS) is a circulating hormonal system involved in the regulation of blood pressure and circulating fluid electrolytes. Recent findings have revealed that locally generated angiotensin (Ang) II plays a pivotal role in normal physiology as well as pathophysiology in various tissues and organs, including the pancreas. This review article summarizes current progress that has been made in elucidating the putative roles of Ang II in both acute and chronic pancreatitis. RECENT ADVANCES A convergence of evidence suggests that the underlying mechanism may involve reactive oxygen species (ROS)-generating systems, such as nicotinamide adenine dinucleotide phosphate oxidase, and subsequent elevation of proinflammatory and profibrogenic gene expression as well as protein activity. More importantly, Ang II-induced ROS interacts with other ROS-generating systems to positively feed-forward the ROS-induced signaling. CRITICAL ISSUES AND FUTURE DIRECTIONS Advances in basic research indicate that RAS blockers may provide potential therapeutic role for the management of pancreatic inflammation and, more importantly, pancreatitis-associated complications. Genetic alterations resulting from a malfunction in the epigenetic control of pancreatic RAS could be a causative factor in the development of pancreatitis.
Collapse
Affiliation(s)
- Yuk Cheung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin New Teritories, Hong Kong, China
| | | |
Collapse
|
90
|
Knock GA, Ward JPT. Redox regulation of protein kinases as a modulator of vascular function. Antioxid Redox Signal 2011; 15:1531-47. [PMID: 20849377 DOI: 10.1089/ars.2010.3614] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Reactive oxygen species (ROS) are continuously generated in vascular tissues by various oxidoreductase enzymes. They contribute to normal cell signaling, and modulate vascular smooth muscle tone and endothelial permeability in response to physiological agonists and to various cellular stresses and environmental factors, such as hypoxia. While concentrations of ROS are normally tightly controlled by cellular redox buffer systems, if produced in excess they may contribute to vascular disease. Protein kinases are essential components of most cell signaling pathways, including those involving ROS. The functioning of several members of this highly diverse group of enzymes, which include receptor and nonreceptor tyrosine kinases, protein kinase C, mitogen-activated kinases, and Rho-kinase, are modified by ROS, either through direct oxidative modification or indirectly through modification of associated proteins such as tyrosine phosphatases and monomeric G proteins. In this review, we discuss the molecular mechanisms of redox modification of these proteins, the downstream pathways affected, the often complex interaction between major kinase pathways, and feedback to ROS production itself. We also discuss complicating factors such as differential actions of superoxide anion and hydrogen peroxide, questions concerning concentration dependence, and the significance of signaling microdomains.
Collapse
Affiliation(s)
- Greg A Knock
- Division of Asthma, Allergy, and Lung Biology, King's College London, Stamford Street, London, United Kingdom.
| | | |
Collapse
|
91
|
Savoia C, Sada L, Zezza L, Pucci L, Lauri FM, Befani A, Alonzo A, Volpe M. Vascular inflammation and endothelial dysfunction in experimental hypertension. Int J Hypertens 2011; 2011:281240. [PMID: 21915370 PMCID: PMC3170891 DOI: 10.4061/2011/281240] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 07/12/2011] [Indexed: 12/11/2022] Open
Abstract
Essential hypertension is characterized by increased peripheral vascular resistance to blood flow. The endothelium is a crucial regulator of vascular tone. Its function is impaired in patients with hypertension, with reduced vasodilation, increased vascular tone associated with a proinflammatory and prothrombotic state. Low-grade inflammation localized in vascular tissue is therefore recognized as an important contributor to the pathophysiology of hypertension, to the initiation and progression of atherosclerosis as well as to the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Carmine Savoia
- Cardiology Unit, Clinical and Molecular Medicine Department, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1037/1039, 00189 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Influence of obesity and metabolic dysfunction on the endothelial control in the coronary circulation. J Mol Cell Cardiol 2011; 52:840-7. [PMID: 21889942 DOI: 10.1016/j.yjmcc.2011.08.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/17/2011] [Accepted: 08/18/2011] [Indexed: 02/07/2023]
Abstract
Diseases of the coronary circulation remain the leading cause of death in Western society despite impressive advances in diagnosis, pharmacotherapy and post-event management. Part of this statistic likely stems from a parallel increase in the prevalence of obesity and metabolic dysfunction, both significant risk factors for coronary disease. Obesity and diabetes pose unique challenges for the heart and their impact on the coronary vasculature remains incompletely understood. The vascular endothelium is a major interface between arterial function and the physical and chemical components of blood flow. Proper function of the endothelium is necessary to preserve hemostasis, maintain vascular tone and limit the extent of vascular diseases such as atherosclerosis. Given its central role in vascular health, endothelial dysfunction has been the source of considerable research interest in diabetes and obesity. In the current review, we will examine the pathologic impact of obesity and diabetes on coronary function and the extent to which these two factors impact endothelial function. This article is part of a Special Issue entitled "Coronary Blood Flow".
Collapse
|
93
|
Zhang HB, Wen JK, Zhang J, Miao SB, Ma GY, Wang YY, Zheng B, Han M. Flavonoids from Inula britannica reduces oxidative stress through inhibiting expression and phosphorylation of p47(phox) in VSMCs. PHARMACEUTICAL BIOLOGY 2011; 49:815-820. [PMID: 21500971 DOI: 10.3109/13880209.2010.550055] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
CONTEXT Inula britanica Linn. (Compositae) is a traditional Chinese medicinal herb that has been used to treat bronchitis and inflammation. The total flavonoid extracts (TFEs) isolated from its flowers can inhibit neointimal formation induced by balloon injury in vivo. OBJECTIVE To investigate the mechanism by which TFE suppresses oxidative stress generation and the subsequent inflammation response in vitro. MATERIALS AND METHODS The cultured vascular smooth muscle cells (VSMCs) form rats were exposed to oxidative stress following pretreatment with or without TFE at different concentration. Then, fluorescence staining was used to detect superoxide anion (O₂(˙-)) production, and the lever of maleic dialdehyde (MDA) and superoxide dismutase (SOD) was measured at the same time. Furthermore, tumor necrosis factor-α (TNF-α) was measured by enzyme linked immunosorbent assay (ELISA), reverse transcription-PCR and western blot were performed to detect the expression activity of p47(phox) gene, and immunoprecipitation was used to test the level of p47(phox) phosphorylation. RESULTS TFE inhibited the production of O₂(˙-) induced by H₂O₂ in VSMCs, with decrease in secretion of TNF-α; elevated the activity of SOD in the medium, similar to the effect of quercetin; reduced the level of MDA in culture medium of VSMCs. The pretreatment with TFE resulted in decrease the level of p47(phox) mRNA and protein, and even p47(phox) phosphorylation in VSMCs, compared with H₂O₂ control. DISCUSSION AND CONCLUSION These findings demonstrate that TFE is capable of attenuating the oxidative stress generation and the subsequent inflammation response via preventing the overexpression and activation of p47(phox) and the increased TNF-α secretion in VSMCs in vitro.
Collapse
Affiliation(s)
- Hong-Bing Zhang
- Department of Biochemistry and Molecular Biology, Institute of Basic Medicine, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, PR China
| | | | | | | | | | | | | | | |
Collapse
|
94
|
Campos RR, Oliveira-Sales EB, Nishi EE, Boim MA, Dolnikoff MS, Bergamaschi CT. The role of oxidative stress in renovascular hypertension. Clin Exp Pharmacol Physiol 2011; 38:144-52. [PMID: 20678153 DOI: 10.1111/j.1440-1681.2010.05437.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
1. There is mounting evidence that increased oxidative stress and sympathetic nerve activity play important roles in renovascular hypertension. In the present review, we focus on the importance of oxidative stress in two distinct populations of neurons involved with cardiovascular regulation: those of the rostral ventrolateral medulla (RVLM) and those of the paraventricular nucleus of the hypothalamus (PVN) in the maintenance of sympathoexcitation and hypertension in two kidney-one clip (2K1C) hypertensive rats. Furthermore, the role of oxidative stress in the clipped kidney is also discussed. 2. In the studies reviewed in this article, it was found that hypertension and renal sympathoexcitation in 2K1C rats were associated with an increase in Angiotensin II type one receptor (AT(1) ) expression and in oxidative markers within the RVLM, PVN and in the clipped kidneys of 2K1C rats. Furthermore, acute or chronic anti-oxidant treatment decreased blood pressure and sympathetic activity, and improved the baroreflex control of heart rate and renal sympathetic nerve activity in 2K1C rats. Tempol or vitamin C administration in the RVLM, PVN or systemically all reduced blood pressure and renal sympathetic activity. Cardiovascular improvement in response to chronic anti-oxidant treatment was associated with a downregulation of AT(1) receptors, as well as oxidative markers in the central nuclei and clipped kidney. 3. The data discussed in the present review support the idea that an increase in oxidative stress within the RVLM, PVN and in the ischaemic kidney plays a major role in the maintenance of sympathoexcitation and hypertension in 2K1C rats.
Collapse
Affiliation(s)
- Ruy R Campos
- Department of Physiology, Cardiovascular Division Department of Medicine, Nephrology Division, Federal University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
95
|
Kim JM, Uehara Y, Choi YJ, Ha YM, Ye BH, Yu BP, Chung HY. Mechanism of attenuation of pro-inflammatory Ang II-induced NF-κB activation by genistein in the kidneys of male rats during aging. Biogerontology 2011; 12:537-50. [DOI: 10.1007/s10522-011-9345-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 06/15/2011] [Indexed: 01/13/2023]
|
96
|
Abstract
Hypertension is associated with vascular changes characterised by remodelling, endothelial dysfunction and hyperreactivity. Cellular processes underlying these perturbations include altered vascular smooth muscle cell growth and apoptosis, fibrosis, hypercontractility and calcification. Inflammation, associated with macrophage infiltration and increased expression of redox-sensitive pro-inflammatory genes, also contributes to vascular remodelling. Many of these features occur with ageing, and the vascular phenotype in hypertension is considered a phenomenon of ‘premature vascular ageing’. Among the many factors involved in the hypertensive vascular phenotype, angiotensin II (Ang II) is especially important. Ang II, previously thought to be the sole effector of the renin–angiotensin system (RAS), is converted to smaller peptides [Ang III, Ang IV, Ang-(1-7)] that are biologically active in the vascular system. Another new component of the RAS is the (pro)renin receptor, which signals through Ang-II-independent mechanisms and might influence vascular function. Ang II mediates effects through complex signalling pathways on binding to its G-protein-coupled receptors (GPCRs) AT1R and AT2R. These receptors are regulated by the GPCR-interacting proteins ATRAP, ARAP1 and ATIP. AT1R activation induces effects through the phospholipase C pathway, mitogen-activated protein kinases, tyrosine kinases/phosphatases, RhoA/Rhokinase and NAD(P)H-oxidase-derived reactive oxygen species. Here we focus on recent developments and new research trends related to Ang II and the RAS and involvement in the hypertensive vascular phenotype.
Collapse
|
97
|
Han Y, Yuan N, Zhang SJ, Gao J, Shi Z, Zhou YB, Gao XY, Zhu GQ. c-Src in paraventricular nucleus modulates sympathetic activity and cardiac sympathetic afferent reflex in renovascular hypertensive rats. Pflugers Arch 2011; 461:437-46. [PMID: 21340460 DOI: 10.1007/s00424-011-0932-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 12/23/2010] [Accepted: 02/01/2011] [Indexed: 10/18/2022]
Abstract
Enhanced cardiac sympathetic afferent reflex (CSAR) contributes to sympathetic activation in renovascular hypertension. The study was to determine whether c-Src in paraventricular nucleus (PVN) is involved in the enhanced CSAR and sympathetic activation in hypertensive rats induced by two-kidney one-clip (2K1C). At the end of the fourth week after 2K1C surgery, renal sympathetic nerve activity (RSNA) was recorded in anesthetized rats with baroreceptor denervation and vagotomy. The CSAR was evaluated by the RSNA response to epicardial application of capsaicin. In the PVN, c-Src activity was higher in 2K1C rats than sham-operated (Sham) rats while c-Src expression was not. Epicardial application of capsaicin or PVN microinjection of angiotensin II (Ang II) increased c-Src activity more in 2K1C than Sham rats. PVN microinjection of selective Src family kinase inhibitor 4-Amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazol [3,4-D] pyrimidine (PP2) or 2,3-Dihydro-N,N-dimethyl-2-oxo-3-[(4,5,6,7-tetrahydro-1 H-indol-2-yl)methylene]-1 H-indole-5-sulfonamide (SU6656) abolished the CSAR and decreased RSNA more in 2K1C than Sham rats. The Ang II-induced RSNA and CSAR enhancement was abolished by PP2 or SU6656 pretreatment in 2K1C and Sham rats. NAD(P)H oxidase activity and superoxide anion level in PVN were higher in 2K1C rats, which was attenuated by PP2 but increased by epicardial application of capsaicin or PVN microinjection of Ang II. The effects of capsaicin or Ang II were abolished by PP2. These results indicate that c-Src in the PVN is involved in the enhanced CSAR and sympathetic activation in renovascular hypertension, and mediates the excitatory effects of Ang II in the PVN on the CSAR and sympathetic activity via NAD(P)H oxidase-derived generation of superoxide anions.
Collapse
Affiliation(s)
- Ying Han
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Gu J, Wang JJ, Yan J, Cui CF, Wu WH, Li L, Wang ZS, Yu M, Gao N, Liu L, Ouyang DS. Effects of lignans extracted from Eucommia ulmoides and aldose reductase inhibitor epalrestat on hypertensive vascular remodeling. JOURNAL OF ETHNOPHARMACOLOGY 2011; 133:6-13. [PMID: 20817083 DOI: 10.1016/j.jep.2010.08.055] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 08/23/2010] [Accepted: 08/26/2010] [Indexed: 05/04/2023]
Abstract
AIM OF THE STUDY To investigate the effects of lignans extracted from Eucommia ulmoides and epalrestat on vascular remodeling in spontaneously hypertensive rats. MATERIALS AND METHODS Ten-week-old male spontaneously hypertensive rats were randomly divided into 3 groups (12 rats each group), and treated orally with 100 mg/kg/d of captopril (an angiotensin-converting enzyme inhibitor), 100 mg/kg/d of epalrestat (an aldose reductase inhibitor) and 300 mg/kg/d of lignans by gavage daily for 16 weeks, respectively. Sex-, age-, and number-matched spontaneously hypertensive rats and normotensive Wistar Kyoto rats, were treated with distilled water (vehicle) as controls. The rats were weighed weekly. Mean arterial blood pressure and heart rate were measured periodically by non-invasive blood pressure monitoring. They were sacrificed at the end of experiment (26-week-old). Superior mesenteric artery and aorta were isolated for determination of histomorphometry and the expression of aldose reductase by immunohistochemistry. RESULTS Captopril and lignans, but not epalrestat, decreased mean arterial blood pressure in spontaneously hypertensive rats. Vascular remodeling was improved in all three treated groups by histomorphometry. CONCLUSIONS Both lignans and epalrestat reversed hypertensive vascular remodeling. Aldose reductase played a vital role in the pathologic process of hypertensive vascular remodeling rather than elevation of blood pressure. These data suggested that aldose reductase could be a new therapeutic target for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Juan Gu
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Whitehead NP, Yeung EW, Froehner SC, Allen DG. Skeletal muscle NADPH oxidase is increased and triggers stretch-induced damage in the mdx mouse. PLoS One 2010; 5:e15354. [PMID: 21187957 PMCID: PMC3004864 DOI: 10.1371/journal.pone.0015354] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/11/2010] [Indexed: 11/19/2022] Open
Abstract
Recent studies have shown that oxidative stress contributes to the pathogenesis of muscle damage in dystrophic (mdx) mice. In this study we have investigated the role of NADPH oxidase as a source of the oxidative stress in these mice. The NADPH oxidase subunits gp91phox, p67phox and rac 1 were increased 2–3 fold in tibilais anterior muscles from mdx mice compared to wild type. Importantly, this increase occurred in 19 day old mice, before the onset of muscle necrosis and inflammation, suggesting that NADPH oxidase is an important source of oxidative stress in mdx muscle. In muscles from 9 week old mdx mice, gp91phox and p67phox were increased 3–4 fold and NADPH oxidase superoxide production was 2 times greater than wild type. In single fibers from mdx muscle NADPH oxidase subunits were all located on or near the sarcolemma, except for p67phox,which was expressed in the cytosol. Pharmacological inhibition of NADPH oxidase significantly reduced the intracellular Ca2+ rise following stretched contractions in mdx single fibers, and also attenuated the loss of muscle force. These results suggest that NADPH oxidase is a major source of reactive oxygen species in dystrophic muscle and its enhanced activity has a stimulatory effect on stretch-induced Ca2+ entry, a key mechanism for muscle damage and functional impairment.
Collapse
Affiliation(s)
- Nicholas P Whitehead
- Discipline of Physiology, Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia.
| | | | | | | |
Collapse
|
100
|
Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, Koschinsky ML, Harkewicz R, Witztum JL, Tsimikas S, Golenbock D, Moore KJ, Tabas I. Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab 2010; 12:467-82. [PMID: 21035758 PMCID: PMC2991104 DOI: 10.1016/j.cmet.2010.09.010] [Citation(s) in RCA: 367] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 07/09/2010] [Accepted: 08/02/2010] [Indexed: 02/06/2023]
Abstract
Macrophage apoptosis in advanced atheromata, a key process in plaque necrosis, involves the combination of ER stress with other proapoptotic stimuli. We show here that oxidized phospholipids, oxidized LDL, saturated fatty acids (SFAs), and lipoprotein(a) trigger apoptosis in ER-stressed macrophages through a mechanism requiring both CD36 and Toll-like receptor 2 (TLR2). In vivo, macrophage apoptosis was induced in SFA-fed, ER-stressed wild-type but not Cd36⁻(/)⁻ or Tlr2⁻(/)⁻ mice. For atherosclerosis, we combined TLR2 deficiency with that of TLR4, which can also promote apoptosis in ER-stressed macrophages. Advanced lesions of fat-fed Ldlr⁻(/)⁻ mice transplanted with Tlr4⁻(/)⁻Tlr2⁻(/)⁻ bone marrow were markedly protected from macrophage apoptosis and plaque necrosis compared with WT →Ldlr⁻(/)⁻ lesions. These findings provide insight into how atherogenic lipoproteins trigger macrophage apoptosis in the setting of ER stress and how TLR activation might promote macrophage apoptosis and plaque necrosis in advanced atherosclerosis.
Collapse
Affiliation(s)
- Tracie A Seimon
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|