1
|
Carvalho LRRA, Shimari M, Boeder AM, Zhuge Z, Cai M, Leijding C, Gastaldello S, Kleschyov AL, Schiffer TA, Guimarães DD, Picozzi G, Lund LH, Fellström B, Weitzberg E, Lundberg JO, Hagberg CE, Pironti G, Andersson DC, Carlström M. A novel model of cardiovascular-kidney-metabolic syndrome combining unilateral nephrectomy and high-salt-sugar-fat diet in mice. Lab Anim (NY) 2024; 53:336-346. [PMID: 39438661 PMCID: PMC11519006 DOI: 10.1038/s41684-024-01457-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The aim of this study was to explore biological interaction and pathophysiology mechanisms in a new mouse model of cardiovascular-kidney-metabolic (CKM) syndrome, induced by chronic moderate renal failure in combination with consumption of a customized Western diet rich in carbohydrates, fat and salt. Male C57BL/6J mice were subjected to unilateral nephrectomy, fed a customized Western diet rich not only in sugar and fat but also in salt, and followed for 12 weeks or 20 weeks. Sham-operated mice on a standard chow served as healthy controls. Body composition, weight gain, glucose metabolism, fat distribution, blood pressure, cardiac function, vascular reactivity, renal function, inflammation and mitochondrial function were measured and combined with biochemical and histopathological analyses. The novel triple-hit model of CKM syndrome showed signs and symptoms of metabolic syndrome, disturbed glucose metabolism, impaired adipocyte physiology and fat redistribution, cardiovascular dysfunction, renal damage and dysfunction, systemic inflammation, elevated blood pressure and cardiac remodeling. The pathological changes were more pronounced in mice after prolonged exposure for 20 weeks, but no deaths occurred. In the present mouse model of CKM syndrome, profound and significant metabolic, cardiac, vascular and renal dysfunctions and injuries emerged by using a Western diet rich not only in fat and carbohydrates but also in salt. This multisystem disease model could be used for mechanistic studies and the evaluation of new therapeutic strategies.
Collapse
Affiliation(s)
| | - Miho Shimari
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ariela Maína Boeder
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Zhengbing Zhuge
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Min Cai
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Leijding
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Stefano Gastaldello
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Andrei L Kleschyov
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Gaia Picozzi
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lars H Lund
- Department of Medicine, Cardiology Unit, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Bengt Fellström
- Department of Medical Science, Renal Unit, University Hospital, Uppsala, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gianluigi Pironti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Cardiology Unit, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Daniel C Andersson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Heart, Vascular and Neurology Theme, Cardiology Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Totoń-Żurańska J, Mikolajczyk TP, Saju B, Guzik TJ. Vascular remodelling in cardiovascular diseases: hypertension, oxidation, and inflammation. Clin Sci (Lond) 2024; 138:817-850. [PMID: 38920058 DOI: 10.1042/cs20220797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024]
Abstract
Optimal vascular structure and function are essential for maintaining the physiological functions of the cardiovascular system. Vascular remodelling involves changes in vessel structure, including its size, shape, cellular and molecular composition. These changes result from multiple risk factors and may be compensatory adaptations to sustain blood vessel function. They occur in diverse cardiovascular pathologies, from hypertension to heart failure and atherosclerosis. Dynamic changes in the endothelium, fibroblasts, smooth muscle cells, pericytes or other vascular wall cells underlie remodelling. In addition, immune cells, including macrophages and lymphocytes, may infiltrate vessels and initiate inflammatory signalling. They contribute to a dynamic interplay between cell proliferation, apoptosis, migration, inflammation, and extracellular matrix reorganisation, all critical mechanisms of vascular remodelling. Molecular pathways underlying these processes include growth factors (e.g., vascular endothelial growth factor and platelet-derived growth factor), inflammatory cytokines (e.g., interleukin-1β and tumour necrosis factor-α), reactive oxygen species, and signalling pathways, such as Rho/ROCK, MAPK, and TGF-β/Smad, related to nitric oxide and superoxide biology. MicroRNAs and long noncoding RNAs are crucial epigenetic regulators of gene expression in vascular remodelling. We evaluate these pathways for potential therapeutic targeting from a clinical translational perspective. In summary, vascular remodelling, a coordinated modification of vascular structure and function, is crucial in cardiovascular disease pathology.
Collapse
Affiliation(s)
- Justyna Totoń-Żurańska
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz P Mikolajczyk
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Blessy Saju
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| | - Tomasz J Guzik
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
3
|
Mendes Furtado M, Lima Rocha JÉ, da Silva Mendes AV, Mello Neto RS, Brito AKDS, Sena de Almeida JOC, Rodrigues Queiroz EI, de Sousa França JV, Cunha Sales ALDC, Gomes Vasconcelos A, Felix Cabral W, de Oliveira Lopes L, Souza do Carmo I, Souza Kückelhaus SA, de Souza de Almeida Leite JR, Nunes AMV, Rizzo MDS, Citó AMDGL, Fortes Lustosa AKM, Lucarini M, Durazzo A, Martins MDCDCE, Arcanjo DDR. Effects of ω-3 PUFA-Rich Oil Supplementation on Cardiovascular Morphology and Aortic Vascular Reactivity of Adult Male Rats Submitted to an Hypercholesterolemic Diet. BIOLOGY 2022; 11:biology11020202. [PMID: 35205069 PMCID: PMC8869584 DOI: 10.3390/biology11020202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 01/27/2023]
Abstract
Simple Summary Currently, processed and ultraprocessed foods represent a significant component of the diet of modern societies, increasing the risk of developing obesity, diabetes and atherosclerosis. Therefore, replacing saturated fats with mono- and polyunsaturated fats, such as omega-3 polyunsaturated fatty acids (ω-3 PUFAs), has been considered as a dietary strategy to reduce clinical events related to atherosclerosis. In the present study, the effects of 56-day ω-3 PUFA-rich oil supplementation on liver function, lipid profile, and oxidative stress in hypercholesterolemic rats were investigated, as well as its impact on cardiovascular health. Interestingly, we observed a positive effect in reducing hepatic markers, preserving cardiovascular morphology, and increasing vasodilator responsiveness. These findings contribute to the generation of consistent recommendations for the therapeutic use of ω-3 PUFAs in the treatment of atherosclerosis, leading to a consequent reduction in related morbidity and mortality. Abstract Atherosclerosis is a cardiovascular disease associated with abnormalities of vascular functions. The consumption of mono- and polyunsaturated fatty acids can be considered a strategy to reduce clinical events related to atherosclerosis. In the present study, we investigated the effects of supplementation with 310 mg of ω-3 PUFAs (2:1 eicosapentaenoic/docosahexaenoic acids) for 56 days on rats with hypercholesterolemia induced by a diet containing cholesterol (0.1%), cholic acid (0.5%), and egg yolk. Serum biochemical parameters were determined by the enzymatic colorimetric method. Assessment of vascular effects was performed by analysis of histological sections of the heart and aortic arch stained with hematoxylin and eosin and vascular reactivity of the aorta artery. We observed that treatment with ω-3 PUFAs did not promote alterations in lipid profile. On the other hand, we documented a favorable reduction in liver biomarkers, as well as contributions to the preservation of heart and aortic arch morphologies. Interestingly, the vascular reactivity of rat thoracic aortic preparations was improved after treatment with ω-3 PUFAs, with a decrease in hyperreactivity to phenylephrine and increased vasorelaxation promoted by acetylcholine. Our findings suggest that the supplementation of hypercholesterolemic rats with ω-3 PUFAs promoted improvement in liver and vascular endothelial function as well as preserving heart and aortic tissue, reinforcing the early health benefits of ω-3 PUFAs in the development of atherosclerotic plaque and further related events.
Collapse
Affiliation(s)
- Mariely Mendes Furtado
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, PI, Brazil
| | - Joana Érica Lima Rocha
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, PI, Brazil
| | | | - Renato Sampaio Mello Neto
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, PI, Brazil
| | | | | | | | | | - Ana Lina de Carvalho Cunha Sales
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, PI, Brazil
- University Hospital, Federal University of Piauí, Teresina 64049-550, PI, Brazil
| | - Andreanne Gomes Vasconcelos
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Wanessa Felix Cabral
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Luana de Oliveira Lopes
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, DF, Brazil
| | | | - Selma Aparecida Souza Kückelhaus
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, DF, Brazil
| | | | - Adriana Maria Viana Nunes
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, PI, Brazil
| | | | | | | | - Massimo Lucarini
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy
| | - Alessandra Durazzo
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy
| | | | | |
Collapse
|
4
|
Abstract
S-nitrosothiols (RSNO) are involved in post-translational modifications of many proteins analogous to protein phosphorylation. In addition, RSNO have many physiological roles similar to nitric oxide ((•)NO), which are presumably involving the release of (•)NO from the RSNO. However, the much longer life span in biological systems for RSNO than (•)NO suggests a dominant role for RSNO in mediating (•)NO bioactivity. RSNO are detected in plasma in low nanomolar levels in healthy human subjects. These RSNO are believed to be redirecting the (•)NO to the vasculature. However, the mechanism for the formation of RSNO in vivo has not been established. We have reviewed the reactions of (•)NO with oxygen, metalloproteins, and free radicals that can lead to the formation of RSNO and have evaluated the potential for each mechanism to provide a source for RSNO in vivo.
Collapse
Affiliation(s)
- Enika Nagababu
- Molecular Dynamics Section, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Rm No. 5B131, Baltimore, MD, 21224, USA,
| | | |
Collapse
|
5
|
Coen M, Gabbiani G, Bochaton-Piallat ML. Myofibroblast-mediated adventitial remodeling: an underestimated player in arterial pathology. Arterioscler Thromb Vasc Biol 2012; 31:2391-6. [PMID: 21868702 DOI: 10.1161/atvbaha.111.231548] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The arterial adventitia has been long considered an essentially supportive tissue; however, more and more data suggest that it plays a major role in the modulation of the vascular tone by complex interactions with structures located within intima and media. The purpose of this review is to summarize these data and to describe the mechanisms involved in adventitia/media and adventitia/intima cross-talk. In response to a plethora of stimuli, the adventitia undergoes remodeling processes, resulting in positive (adaptive) remodeling, negative (constrictive) remodeling, or both. The differentiation of the adventitial fibroblast into myofibroblast (MF), a key player of wound healing and fibrosis development, is a hallmark of negative remodeling; this can lead to vessel stenosis and thus contribute to major cardiovascular diseases. The mechanisms of fibroblast-to-MF differentiation and the role of the MF in adventitial remodeling are highlighted herein.
Collapse
Affiliation(s)
- Matteo Coen
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
6
|
Kajita M, Murata T, Horiguchi K, Iizuka M, Hori M, Ozaki H. iNOS expression in vascular resident macrophages contributes to circulatory dysfunction of splanchnic vascular smooth muscle contractions in portal hypertensive rats. Am J Physiol Heart Circ Physiol 2010; 300:H1021-31. [PMID: 21193589 DOI: 10.1152/ajpheart.00563.2009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Portal hypertension, a major complication of cirrhosis, is caused by both increased portal blood flow due to arterial vasodilation and augmented intrahepatic vascular resistance due to sinusoidal constriction. In this study, we examined the possible involvement of resident macrophages in the tone regulation of splanchnic blood vessels using bile duct ligated (BDL) portal hypertensive rats and an in vitro organ culture method. In BDL cirrhosis, the number of ED2-positive resident macrophages increased by two- to fourfold in the vascular walls of the mesenteric artery and extrahepatic portal vein compared with those in sham-operated rats. Many ED1-positive monocytes were also recruited into this area. The expression of inducible nitric oxide (NO) synthase (iNOS) mRNA was increased in the vascular tissues isolated from BDL rats, and accordingly, nitrate/nitrite production was increased. Immunohistochemistry revealed that iNOS was largely expressed in ED1-positive and ED2-positive cells. We further analyzed the effect of iNOS expression on vascular smooth muscle contraction using an in vitro organ culture system. iNOS mRNA expression and nitrate production significantly increased in vascular tissues (without endothelium) incubated with 1 μg/ml lipopolysaccharide (LPS) for 6 h. Immunohistochemistry indicated that iNOS was largely expressed in ED2-positive resident macrophages. α-Adrenergic-stimulated contractility of the mesenteric artery was greatly suppressed by LPS treatment and was restored by N(G)-nitro-L-arginine methyl ester (NO synthase inhibitor); in contrast, portal vein contractility was largely unaffected by LPS. Sodium nitroprusside (NO donor) and 8-bromo-cGMP showed greater contractile inhibition in the mesenteric artery than in the portal vein with decreasing myosin light chain phosphorylation. In the presence of an α-adrenergic agonist, the mesenteric artery cytosolic Ca(2+) level was greatly reduced by sodium nitroprusside; however, the portal vein Ca(2+) level was largely unaffected. These results suggest that the induction of iNOS in monocytes/macrophages contributes to a hypercirculatory state in the cirrhosis model rat in which the imbalance of the responsiveness of visceral vascular walls to NO (mesenteric artery >> portal vein) may account for the increased portal venous flow in portal hypertension.
Collapse
Affiliation(s)
- Masahiro Kajita
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
7
|
Hong H, Sun J, Cai W. Multimodality imaging of nitric oxide and nitric oxide synthases. Free Radic Biol Med 2009; 47:684-98. [PMID: 19524664 DOI: 10.1016/j.freeradbiomed.2009.06.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 05/28/2009] [Accepted: 06/10/2009] [Indexed: 01/27/2023]
Abstract
Nitric oxide (NO) and NO synthases (NOSs) are crucial factors in many pathophysiological processes such as inflammation, vascular/neurological function, and many types of cancer. Noninvasive imaging of NO or NOS can provide new insights in understanding these diseases and facilitate the development of novel therapeutic strategies. In this review, we will summarize the current state-of-the-art multimodality imaging in detecting NO and NOSs, including optical (fluorescence, chemiluminescence, and bioluminescence), electron paramagnetic resonance (EPR), magnetic resonance (MR), and positron emission tomography (PET). With continued effort over the last several years, these noninvasive imaging techniques can now reveal the biodistribution of NO or NOS in living subjects with high fidelity which will greatly facilitate scientists/clinicians in the development of new drugs and/or patient management. Lastly, we will also discuss future directions/applications of NO/NOS imaging. Successful development of novel NO/NOS imaging agents with optimal in vivo stability and desirable pharmacokinetics for clinical translation will enable the maximum benefit in patient management.
Collapse
Affiliation(s)
- Hao Hong
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705-2275, USA
| | | | | |
Collapse
|
8
|
Pricci M, Bourget JM, Robitaille H, Porro C, Soleti R, Mostefai HA, Auger FA, Martinez MC, Andriantsitohaina R, Germain L. Applications of human tissue-engineered blood vessel models to study the effects of shed membrane microparticles from T-lymphocytes on vascular function. Tissue Eng Part A 2009; 15:137-45. [PMID: 18925833 DOI: 10.1089/ten.tea.2007.0360] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Microparticles (MPs) are membrane vesicles harboring cell surface proteins and containing cytoplasmic components of the original cell. High levels of circulating MPs have been detected in pathological states associated with vascular dysfunction. We took advantage of the self-assembly method of tissue engineering to produce in vitro three vascular constructs from human vascular smooth muscle cells and fibroblasts to investigate the role of the adventitia in the modulation of vascular tone by MPs, comparing the contractile response of each of these constructs to histamine. The first two were composed of an adventitia (tissue-engineered vascular adventitia (TEVA)) or a media (tissue-engineered vascular media (TEVM)) solely, and the third one contained a media and an adventitia (tissue-engineered vascular media and adventitia (TEVMA)). In the three constructs, the results show that histamine induces contraction insensitive to blockade of inducible nitric oxide (NO) synthase (iNOS) and cyclooxygenase-2 (COX-2) and not affected by MP treatment. MPs decreased NO production and nuclear factor (NF)-kappaB expression but did not affect superoxide anion (O(2)(-)) release in TEVA. MPs enhanced NF-kappaB expression but did not affect iNOS and COX-2 expression or NO or O(2)(-) release in TEVM. In TEVMA, MPs did not enhance NF-kappaB expression, but COX-2 expression was higher, and O(2)(-) release was lower. Thus, MPs affected NO, O(2)(-), NF-kappaB, and COX-2 in a subtle fashion to maintain the contractile response to histamine. The use of tissue-engineered vascular constructs results in a better understanding of the effect of MPs on human adventitia and media.
Collapse
Affiliation(s)
- Maria Pricci
- Institut National de la Santé et de la Recherche Médicale 771, CNRS UMR 6214, Faculté de Médecine, Angers, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Hetrick EM, Schoenfisch MH. Analytical chemistry of nitric oxide. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2009; 2:409-33. [PMID: 20636069 PMCID: PMC3563389 DOI: 10.1146/annurev-anchem-060908-155146] [Citation(s) in RCA: 225] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Nitric oxide (NO) is the focus of intense research primarily because of its wide-ranging biological and physiological actions. To understand its origin, activity, and regulation, accurate and precise measurement techniques are needed. Unfortunately, analytical assays for monitoring NO are challenged by NO's unique chemical and physical properties, including its reactivity, rapid diffusion, and short half-life. Moreover, NO concentrations may span the picomolar-to-micromolar range in physiological milieus, requiring techniques with wide dynamic response ranges. Despite such challenges, many analytical techniques have emerged for the detection of NO. Herein, we review the most common spectroscopic and electrochemical methods, with a focus on the underlying mechanism of each technique and on approaches that have been coupled with modern analytical measurement tools to create novel NO sensors.
Collapse
Affiliation(s)
- Evan M. Hetrick
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
10
|
Sjakste N, Baumane L, Boucher JL, Dzintare M, Meirena D, Sjakste J, Lauberte L, Kalvinsh I. Effects of γ-Butyrobetaine and Mildronate on Nitric Oxide Production in Lipopolysaccharide-Treated Rats. Basic Clin Pharmacol Toxicol 2008. [DOI: 10.1111/j.1742-7843.2004.pto_940108.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
11
|
Malyshev E, Tazi KA, Moreau R, Lebrec D. Discrepant effects of inducible nitric oxide synthase modulation on systemic and splanchnic endothelial nitric oxide synthase activity and expression in cirrhotic rats. J Gastroenterol Hepatol 2007; 22:2195-201. [PMID: 18031380 DOI: 10.1111/j.1440-1746.2006.04608.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Arterial vasodilatation, which is a major factor in the pathogenesis of the hyperkinetic circulatory state and portal hypertension in cirrhosis, is due to arterial nitric oxide (NO) overproduction secondary to endothelial NO synthase (eNOS) and inducible NOS (iNOS) upregulation. However, in cirrhosis, the respective roles of eNOS and iNOS isoforms in NO overproduction are still unknown and the effect of iNOS modulation on eNOS activity and expression has not been evaluated in the systemic or splanchnic vessels. The aim of this study was to evaluate the effects of modulating aortic and superior mesenteric arteries (SMA) iNOS on arterial eNOS activity and expression in rats with cirrhosis. METHODS eNOS and iNOS protein expression and eNOS activity (assessed by its phosphorylation at serine 1177) were measured in the aortas and SMA in untreated and treated cirrhotic rats with lipopolysaccharide (LPS), N-iminoethyl-L-lysine (L-NIL), a selective iNOS inhibitor, and LPS plus L-NIL. RESULTS LPS administration significantly increased eNOS and iNOS protein expression and eNOS activity in the aortas of both sham-operated and cirrhotic rats. However, in SMA, LPS administration induced a decrease in eNOS protein expression and activity and an increase in iNOS protein expression. CONCLUSION The results of this study may explain the worsening of the hyperdynamic state in cirrhosis during septic shock by direct LPS-induced eNOS activation in large systemic vessels, and its inhibition in concomitant small splanchnic vasculature by iNOS synthesized NO.
Collapse
Affiliation(s)
- Eugene Malyshev
- INSERM U773, Research Center Bichat Beaujon CRB3, Beaujon Hospital, Clichy, France
| | | | | | | |
Collapse
|
12
|
Atar S, Siegel RJ, Akel R, Ye Y, Lin Y, Modi SA, Sewani A, Tuero E, Birnbaum Y. Ultrasound at 27 kHz increases tissue expression and activity of nitric oxide synthases in acute limb ischemia in rabbits. ULTRASOUND IN MEDICINE & BIOLOGY 2007; 33:1483-8. [PMID: 17507145 DOI: 10.1016/j.ultrasmedbio.2007.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2006] [Revised: 03/05/2007] [Accepted: 03/19/2007] [Indexed: 05/15/2023]
Abstract
Transcutaneous low-frequency ultrasound (US) preserves myocardial and skeletal muscle viability by increasing tissue perfusion through an undefined nitric oxide (NO)-dependent mechanism. We have examined whether US increases tissue expression and activity of the three nitric oxide synthase (NOS) isoforms: endothelial (eNOS), neuronal (nNOS) and inducible (iNOS). The two femoral arteries of four New Zealand rabbits were ligated for a total of 120 min. After 60 min of ligation, transcutaneous low-frequency US (27 kHz, 0.13 W/cm2) was applied for 60 min to one thigh, while the contra-lateral artery served as a control (total ischemia time=120 min). Calcium-dependent (cNOS) and -independent (ciNOS) NOS activity, and concentration of total eNOS, ser-1177 phosphorylated eNOS (P-eNOS), nNOS and iNOS were then determined in the gracilis muscle. Compared with the control, US application significantly increased cNOS activity [3.34+/-0.28 versus 3.87+/-0.10x1000 counts per minute (cpm), respectively, p=0.031] and ciNOS activity (1.99+/-0.09 versus 3.26+/-0.68 cpm, respectively, p<0.001). Western immunoblotting revealed a significant increase in protein content of both iNOS (184.5+/-1.08%; p<0.0001) and P-eNOS (381.5+/-2.47%; p<0.001), with only a small increase in total eNOS and nNOS expression. In conclusion, application of transcutaneous low-frequency US to ischemic muscular tissue significantly increases both cNOS and ciNOS activity by increasing eNOS phosphorylation and iNOS expression, respectively.
Collapse
Affiliation(s)
- Shaul Atar
- Division of Cardiology, University of Texas Medical Branch, Galveston, TX 77555-0553, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Lartaud I, Faure S, Tabellion A, Resende AC, Nadaud S, Bagrel D, Capdeville-Atkinson C, Atkinson J. Melatonin counteracts the loss of agonist-evoked contraction of aortic rings induced by incubation. Fundam Clin Pharmacol 2007; 21:273-9. [PMID: 17521296 DOI: 10.1111/j.1472-8206.2007.00484.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Incubation of aortic rings in a culture medium produces phenomena similar to those observed with aging, i.e. oxidative stress and inflammation leading to increased nitric oxide (NO)-mediated dilation and decreased arterial sensitivity to vasoconstrictor agents. We evaluated whether melatonin protects aortic rings from such a decrease in vasoreactivity. Two concentrations of melatonin were used: 10(-8) M, EC50 for vascular MT1-MT2 receptors, and 10(-5) M, reported as anti-oxidant. Anti-oxidant capacity, inducible nitric oxide synthase (iNOS) expression and isometric contraction of thoracic aorta rings (Wistar rats) evoked by norepinephrine (NE) were assessed. Three days of incubation of aortic rings induced iNOS expression and a fall in NE-evoked contraction. When melatonin was added to the organ bath, it (10(-5) M) increased (+96%, P < 0.05), but did not restore (compared with freshly isolated rings) NE-evoked contraction. Three days of treatment with melatonin increased (10(-8) M, +99%) or restored (10(-5) M, +216%) NE-evoked contraction (compared with freshly isolated rings). The beneficial effects of 10(-8) and 10(-5) M melatonin on NE-evoked contraction were abolished in the presence of luzindole (2 x 10(-6) M, a melatonin receptor antagonist). The incubation-induced increase in iNOS expression was reduced following 3 days of melatonin administration (10(-8) and 10(-5) M). Melatonin (10(-5) M) increased catalase activity (6550 +/- 256, P < 0.05 vs. nontreated fresh aortic rings 5554 +/- 444 nmol min(-1) mg protein(-1)). In conclusion, melatonin counteracts the incubation-induced loss of agonist-evoked contraction of aortic rings by a specific receptor-mediated phenomenon involving iNOS expression; at higher melatonin concentrations, an anti-oxidant effect is probably also involved.
Collapse
Affiliation(s)
- Isabelle Lartaud
- Pharmacology Laboratory, Faculté de Pharmacie, Université Henri Poincaré Nancy-1, 5 rue Albert Lebrun, 54001 Nancy Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Kleschyov AL, Wenzel P, Munzel T. Electron paramagnetic resonance (EPR) spin trapping of biological nitric oxide. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 851:12-20. [PMID: 17070113 DOI: 10.1016/j.jchromb.2006.10.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Revised: 10/05/2006] [Accepted: 10/06/2006] [Indexed: 10/24/2022]
Abstract
Nitric oxide (NO) is a free radical species with multiple physiological functions. Because of low concentrations and short half-life of NO, its direct measurement in living tissues remains a difficult task. Electron paramagnetic resonance (EPR) spin trapping is probably one of the best suitable platforms for development of new methods for quantification of biological NO. The most reliable EPR-based approaches developed so far are based on the reaction of NO with various iron complexes, both intrinsic and exogenously applied. This review is focused on the current state and perspectives of EPR spin trapping for experimental and clinical NO biology.
Collapse
Affiliation(s)
- Andrei L Kleschyov
- Second Department of Medicine, Johannes Gutenberg University of Mainz, Mainz 55131, Germany.
| | | | | |
Collapse
|
15
|
Jia YX, Pan CS, Yang JH, Liu XH, Yuan WJ, Zhao J, Tang CS, Qi YF. ALTERED l-ARGININE/NITRIC OXIDE SYNTHASE/NITRIC OXIDE PATHWAY IN THE VASCULAR ADVENTITIA OF RATS WITH SEPSIS. Clin Exp Pharmacol Physiol 2006; 33:1202-8. [PMID: 17184502 DOI: 10.1111/j.1440-1681.2006.04498.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
1. In recent studies, the vascular adventitia has been established as an important source of inducible nitric oxide synthase (iNOS) and subsequent nitric oxide (NO) production, even more powerful than the media in response to certain inflammatory factors, such as lipopolysaccharide (LPS). The adventitia has an independent L-arginine (L-Arg)/NOS/NO pathway and is involved in the regulation of vascular function. In the present study, we explored the changes in and the pathophysiological significance of the L-Arg/NOS/NO pathway in the adventitia of rats with sepsis. 2. Sepsis was induced by caecal ligation and puncture in order to observe changes in L-Arg transport, NOS gene expression and activity and NO generation in the vascular adventitia to determine the mechanism of activation of the L-Arg/NOS/NO pathway. 3. Severe sepsis resulted in severe disturbance of haemodynamic features, with decreased mean arterial blood pressure, brachycardia and inhibited cardiac function (decreased left ventricular +/-dP/dt(max)). Left ventricular end-diastolic pressure was elevated threefold (P < 0.01) under anaesthesia. Rats with sepsis showed severe glucopenia and lacticaemia. Plasma levels of the inflammatory factors macrophage chemoattractant protein-1 and interleukin-8 were increased five- and 29-fold, respectively (P < 0.01). 4. In the adventitia of the thoracic and abdominal aortas, the L-Arg/NO pathway was similarly characterized: the uptake of [(3)H]-L-Arg was Na(+) independent, with the peak occurring at approximately 40 min incubation. Total NOS activity was largely calcium independent (> 90%). The V(max) of L-Arg transport in the sepsis group was increased by 83.5% (P < 0.01), but the K(m) value was not significantly different compared with controls. 5. The mRNA levels of cationic amino acid transporter (CAT)-1 and CAT-2B in the sepsis group were increased by 86 and 62%, respectively (both P < 0.01). Inducible NOS activity was increased 2.8-fold compared with controls (P < 0.01) and iNOS mRNA levels were elevated approximately sixfold (P < 0.01). The NO levels in the plasma and incubation media (incubation for 40 min) in the sepsis group were increased by 144 and 273%, respectively (both P < 0.01). 6. The Arg/NOS/NO pathway was activated in the vascular adventitia of rats with sepsis shock. The L-Arg/NOS/NO pathway in the aortic adventitia may play an important role in the pathogenesis of sepsis and septic shock.
Collapse
Affiliation(s)
- Yue Xia Jia
- Institute of Cardiovascular Research, Peking University First Hospital, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Lakshminrusimha S, Russell JA, Gugino SF, Ryan RM, Mathew B, Nielsen LC, Morin FC. Adjacent bronchus attenuates pulmonary arterial contractility. Am J Physiol Lung Cell Mol Physiol 2006; 291:L473-8. [PMID: 16581826 DOI: 10.1152/ajplung.00313.2005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bronchus-derived relaxing factor (BrDRF) decreases contractility of newborn rat pulmonary arteries (PA) and is dependent on nitric oxide (NO) synthesis. In vivo, this factor appears to gain access via the adventitial side of the PA. However, the adventitia has been reported to be a barrier to NO. We studied the effect of an adjacent bronchus on PA contractility to norepinephrine in nine juvenile lambs in the presence and absence of inhibitors of the NO pathway (LNA, ODQ, and Rp-8-Br-PET-cGMPS), cytochrome P-450 inhibitor (17-ODYA), perivascular nerve activity blocker (TTX), and superoxide scavenger (tiron), and following disruption of bronchial epithelium. We also evaluated whether BrDRF was effective on both the endothelial and/or adventitial side of PA. Fifth-generation PA rings with and without an attached bronchus were contracted in standard baths with norepinephrine. PA were dissected, cut open, and placed in a sided chamber in which adventitial and endothelial sides of the PA were exposed to unattached bronchus separately. Norepinephrine (10−8 to 10−5 M) contractions were expressed as a fraction of maximal KCl (118 mM) contractions. Norepinephrine contractions were significantly reduced by the presence of an attached bronchus, an effect reversed by pretreatment with LNA, ODQ, and Rp-8-Br-PET-cGMPS, and removal of bronchial epithelium. Unattached bronchus in the bath perfusing the adventitial side was effective in inhibiting the contractile response in PA. NO gas relaxed PA when administered on the endothelial side only. We speculate that BrDRF is a diffusible factor that crosses the adventitia and stimulates production of NO within the PA.
Collapse
Affiliation(s)
- Satyan Lakshminrusimha
- Department of Pediatrics, State University of New York at Buffalo, Center for Developmental Biology of the Lung, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Kleschyov AL, Strand S, Schmitt S, Gottfried D, Skatchkov M, Sjakste N, Daiber A, Umansky V, Munzel T. Dinitrosyl-iron triggers apoptosis in Jurkat cells despite overexpression of Bcl-2. Free Radic Biol Med 2006; 40:1340-8. [PMID: 16631524 DOI: 10.1016/j.freeradbiomed.2005.12.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Revised: 11/17/2005] [Accepted: 12/01/2005] [Indexed: 11/26/2022]
Abstract
Cells expressing the cytokine-inducible NO synthase are known to trigger apoptosis in neighboring cells. Paramagnetic dinitrosyl nonheme iron complexes (DNIC) were found in tumor tissue about 40 years ago; however, the role of these NO(+)-bearing species is not completely understood. In the human Jurkat leukemia cell line, the application of the model complex DNIC-thiosulfate (50-200 microM) induced apoptosis (defined by phosphatidylserine externalization) in a concentration- and time-dependent manner. In Jurkat cells, the pan-caspase inhibitor, zVADfmk (50 microM), and/or stable transfection of antiapoptotic protein, Bcl-2, was unable to afford protection against DNIC-induced apoptosis. The membrane-impermeable metal chelator, N-methyl-D-glucamine dithiocarbamate (MGD; 200 microM), in the presence of DNIC significantly increased apoptosis, but had no effect on its own. Electron paramagnetic resonance studies showed that MGD led to rapid transformation of the extracellular DNIC into the stable impermeable NO-Fe-MGD complex and to a burst-type release of nitrosonium (NO(+)) equivalents in the extracellular space. These results suggest that in Jurkat cells, DNIC-thiosulfate induces Bcl-2- and caspase-independent apoptosis, which is probably secondary to local nitrosative stress at the cell surface. We hypothesize that the local release of nonheme Fe-NO species by activated macrophages may play a role in the killing of malignant cells that have high Bcl-2 levels.
Collapse
Affiliation(s)
- Andrei L Kleschyov
- II-Department of Medicine, Johannes Gutenberg University, Mainz, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
August M, Wingerter O, Oelze M, Wenzel P, Kleschyov AL, Daiber A, Mülsch A, Münzel T, Tsilimingas N. Mechanisms underlying dysfunction of carotid arteries in genetically hyperlipidemic rabbits. Nitric Oxide 2006; 15:241-51. [PMID: 16597505 DOI: 10.1016/j.niox.2006.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 02/20/2006] [Accepted: 02/21/2006] [Indexed: 11/20/2022]
Abstract
In the present study we compared the vascular reactivity and integrity of the nitric oxide (NO)-cyclic 3',5'-guanosine monophopsphate (cGMP) pathway in carotid arteries of hyper- and normolipidemic rabbits. Vasodilation to acetylcholine, nitroglycerin, and sodium nitroprusside was desensitized in hyperlipidemia, but the nitroprusside-induced relaxation was normalized by an NO synthase inhibitor in endothelium-intact and -denuded vessels. Hyperlipidemic carotid arteries exhibited increased basal NO (detected by EPR spin-trapping) and reactive oxygen species formation (detected by chemiluminescence), whereas acetylcholine-induced NO formation was nearly abolished. Hyperlipidemia increased NADPH-dependent superoxide formation in carotid membranes, and carotid cryosections stained with the fluorescent dye dihydroethidium revealed increased endothelial and medial reactive oxygen species formation. Hyperlipidemia elicited macrophage invasion into the carotid wall, as detected by a dot-immunoblot. The basal activity of cGMP-dependent proteinkinase, the nitroprusside-stimulated activity of soluble guanylyl cyclase, and its protein expression were decreased by hyperlipidemia. The cGMP phosphodiesterase activity was marginally increased by hyperlipidemia, such that the ratio of cGMP-forming vs. -degrading capacity was decreased by 2-fold. Hyperlipidemia triggers infiltration of macrophages into the carotid wall and endothelial as well as smooth muscle superoxide formation. Consequently, relaxation of the carotid arteries are impaired due to smooth muscle and endothelial dysfunction.
Collapse
Affiliation(s)
- Michael August
- University Hospital Eppendorf, Division of Cardiology, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yang JH, Pan CS, Jia YX, Zhang J, Zhao J, Pang YZ, Yang J, Tang CS, Qi YF. Intermedin1-53 activates l-arginine/nitric oxide synthase/nitric oxide pathway in rat aortas. Biochem Biophys Res Commun 2006; 341:567-72. [PMID: 16434024 DOI: 10.1016/j.bbrc.2006.01.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2006] [Accepted: 01/05/2006] [Indexed: 10/25/2022]
Abstract
Intermedin (IMD), a novel member of the calcitonin/calcitonin gene-related peptide (CGRP) family, has similar or more potent vasodilatory and hypotensive actions than adrenomedullin (ADM) and CGRP. The present study was designed to observe the effects of synthetic rat IMD1-53 on L-arginine (L-Arg) cellular transport, nitric oxide synthase (NOS) activity, and nitric oxide (NO) production in the isolated rat aortic ring to illustrate its direct effect on the L-Arg/NOS/NO pathway in vasculature. IMD1-53 significantly increased NO production and cNOS activity in rat aortas and was more potent than equivalent ADM. But the peptides of both IMD and ADM had no effect on inducible NOS expression and activity. Otherwise, IMD1-53 induced a concentration-dependent increase in [3H]L-Arg transport and its effect was more potent than that of an equivalent concentration of ADM. Semiquantitative RT-PCR revealed that IMD1-53 significantly increased cationic amino acid transport (CAT)-1 and CAT-2B mRNA expression, and its effect was similar to that of ADM. All these results suggest that IMD1-53 might regulate vessel function homeostasis via upregulating the L-Arg/NOS/NO pathway.
Collapse
Affiliation(s)
- Jing-Hui Yang
- Institute of Cardiovascular Research, Peking University First Hospital, Beijing 100034, China
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gunnett CA, Lund DD, McDowell AK, Faraci FM, Heistad DD. Mechanisms of Inducible Nitric Oxide Synthase–Mediated Vascular Dysfunction. Arterioscler Thromb Vasc Biol 2005; 25:1617-22. [PMID: 15933248 DOI: 10.1161/01.atv.0000172626.00296.ba] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Inducible nitric oxide synthase (iNOS) is expressed in arteries during inflammation and may contribute to vascular dysfunction. Effects of gene transfer of iNOS to carotid arteries were examined in vitro in the absence of systemic inflammation to allow examination of mechanisms by which iNOS impairs contraction and relaxation.
Methods and Results—
After gene transfer of iNOS with an adenovirus (AdiNOS), constrictor responses to phenylephrine (PE) and U46619 were impaired. After AdiNOS, inhibition of soluble guanylate cyclase (sGC) with 1H-[1,2,4]oxadiazolo-[4,3,2]quinoxalin-1-one (ODQ) reduced the EC
50
for PE from 4.33±0.78 μmol/L to 1.15±0.43 μmol/L (mean±SEM). These results imply that iNOS impairs contraction by activation of the NO/cGMP pathway. Relaxation to acetylcholine (ACh) also was impaired after AdiNOS. Sepiapterin (300 μmol/L), the precursor for tetrahydrobiopterin (BH
4
), improved relaxation to Ach. Because BH
4
is an essential cofactor for production of NO by both iNOS and endothelial nitric oxide synthase (eNOS), these results suggest that iNOS may reduce production of NO by eNOS by limiting availability of BH
4
. Next, we examined effects of expression of iNOS in endothelium and adventitia. Selective expression of iNOS in endothelium, but not adventitia, impaired contraction to phenylephrine and relaxation to acetylcholine.
Conclusions—
We conclude that: (1) iNOS may impair contraction in part by activation of sGC; (2) iNOS impairs relaxation, at least in part, by limiting availability of BH
4
; and (3) expression of iNOS in endothelium may be a more important mediator of vascular dysfunction than expression of iNOS in adventitia.
Collapse
Affiliation(s)
- C A Gunnett
- Department of Internal Medicine, University of Iowa Carver College of Medicine, VA Medical Center, Iowa City, IA, USA
| | | | | | | | | |
Collapse
|
21
|
Beranova P, Schott C, Chalupsky K, Kleschyov AL, Stoclet JC, Muller B. Role of the adventitia in the cyclic GMP-mediated relaxant effect of N-hydroxy-L-arginine in rat aorta. J Vasc Res 2005; 42:331-6. [PMID: 15976507 DOI: 10.1159/000086489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Accepted: 04/23/2005] [Indexed: 11/19/2022] Open
Abstract
N(omega)-hydroxy-L-arginine (L-NOHA), the stable intermediate of the nitric oxide synthase (NOS)-catalyzed reaction, can induce NO/cyclic GMP-dependent relaxation in the rat aorta, in an endothelium- and NOS-independent manner. In this study, the role of the adventitia in the endothelium-independent effect of L-NOHA was investigated. Despite a decrease in norepinephrine (NE)-induced precontraction, adventitia removal in the rat aorta did not markedly alter the relaxant effect of forskolin, S-nitroso-N-acetylpenicillamine or glyceryl trinitrate. In contrast, both inhibition of NE-induced contraction and relaxation of NE-precontracted rings produced by L-NOHA were diminished in the absence of adventitia. Moreover, exposure to L-NOHA significantly enhanced the cyclic GMP level in the media of the aorta with, but not without adventitia. These findings demonstrate the role of the adventitia in the L-NOHA-induced decrease in tone and increase in cyclic GMP in the endothelium-denuded rat aorta. They suggest that NO or an NO-related compound formed from L-NOHA in the adventitia may produce paracrine effects.
Collapse
Affiliation(s)
- Petra Beranova
- Pharmacologie et Physico-Chimie, UMR CNRS 7034, Université Louis Pasteur de Strasbourg, France
| | | | | | | | | | | |
Collapse
|
22
|
Lin L, Ding WH, Jiang W, Zhang YG, Qi YF, Yuan WJ, Tang CS. Urotensin-II activates L-arginine/nitric oxide pathway in isolated rat aortic adventitia. Peptides 2004; 25:1977-84. [PMID: 15501530 DOI: 10.1016/j.peptides.2004.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2004] [Revised: 07/21/2004] [Accepted: 07/21/2004] [Indexed: 10/26/2022]
Abstract
Urotensin-II (U-II), a cyclic peptide widely expressed in blood vessels, has diverse vascular actions that range from potent vasoconstriction to vasodilation. Although, U-II-induced vasodilation has been shown to be partially dependent on nitric oxide (NO), the involvement of vascular adventitia-derived NO, remains unknown. The present study aimed to elucidate the activation of U-II on L-arginine/NO pathway in isolated rat aortic adventitia. In adventitia of thoracic and abdominal aortas, the l-arginine/NO pathway was similarly characterized: the uptake of l-[(3)H]arginine was Na(+)-independent, with the peak occurring over around 40 min incubation; the total NO synthase (NOS) activity was mostly calcium-independent (>90%), and significantly inhibited by a specific iNOS inhibitor AMT; the production of NO metabolites nitrate and nitrite (NO(x)) was stimulated by L-arginine but not by D-arginine. In aortic adventitia exposed to rat U-II (10(-9) and 10(-8)M) for 6 h, the V(max) of l-[(3)H]arginine uptake over 40 min incubation was significantly increased, while the K(m) of l-[(3)H]arginine uptake showed no significant change. Besides, the iNOS mRNA level was up-regulated, the total NOS activity, largely calcium-independent, was significantly induced, and the NO(x) production was significantly stimulated by U-II. According to the same protocol as U-II, the positive control lipopolysaccharide (LPS, 10 microg/ml), which had been established to activate adventitial L-arginine/NO pathway, increased l-[(3)H]arginine uptake, iNOS activity and NO(x) production to a greater extent than U-II. In addition, the total NOS activities induced by 3 and 6h incubation of U-II and LPS were significantly inhibited by a specific inhibitor of protein synthesis, actinomycin D. In conclusion, the results showed that rat U-II activated L-arginine/NOS/NO pathway in rat aortic adventitia, suggesting a potential contributive role of adventitia-derived NO in the vasodilator response of U-II.
Collapse
Affiliation(s)
- Li Lin
- Department of Physiology, Second Military Medical University, Shanghai 200433, PR China.
| | | | | | | | | | | | | |
Collapse
|
23
|
Sjakste N, Baumane L, Boucher JL, Dzintare M, Meirena D, Sjakste J, Lauberte L, Kalvinsh I. Effects of gamma-Butyrobetaine and Mildronate on Nitric Oxide Production in Lipopolysaccharide-Treated Rats. ACTA ACUST UNITED AC 2004. [DOI: 10.1111/j.1742-7843.2004.pto940108.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
24
|
Barclay JK, Murrant CL, Woodley NE, Reading SA. Potential Interactions Among Vascular and Muscular Functional Compartments During Active Hyperemia. ACTA ACUST UNITED AC 2003; 28:737-53. [PMID: 14710524 DOI: 10.1139/h03-056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The increase in blood flow that accompanies the start of contractions (active hyperemia) is a complex phenomenon involving a fast phase in which blood flow increases quickly and then slows or decreases (seek phase) before stabilizing at a flow corresponding to the metabolic rate (matched phase). This pattern of blood flow change involves contributions from a flow-induced increase in flow, a response to short periods of occlusion or partial occlusion due to force generated by the muscle contraction, and metabolism. Even denervated, the vascular bed, which consists of endothelial cells, vascular smooth muscle cells, and an adventitial layer that has significant secretory potential, is able to coordinate the response pattern. Within the vascular wall, communication is possible bidirectionally across the wall and also along the wall in a retrograde or upstream direction. The signals involved, which range from endothelial cell products such as nitric oxide and endothelin to adenosine, a skeletal muscle metabolite, appear to be situation- and time-dependent. In addition to the communication potential within and along the vascular wall, signals from the vascular system are able to exert inotropic effects on mammalian skeletal muscle. Key words: bidirectional signaling, postcontraction hyperemia, flow-induced flow changes, signal plasticity
Collapse
Affiliation(s)
- Jack K Barclay
- Dept. of Human Biology and Nutritional Sciences, Univ. of Guelph, Guelph, ON, N1G 2W1
| | | | | | | |
Collapse
|
25
|
Alencar JL, Lobysheva I, Geffard M, Sarr M, Schott C, Schini-Kerth VB, Nepveu F, Stoclet JC, Muller B. Role of S-nitrosation of cysteine residues in long-lasting inhibitory effect of nitric oxide on arterial tone. Mol Pharmacol 2003; 63:1148-58. [PMID: 12695543 DOI: 10.1124/mol.63.5.1148] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
S-Nitrosation of cysteine residues plays an important role in nitric oxide (NO) signaling and transport. The aim of the present study was to investigate the role of S-nitrosothiols as a storage form of NO, which may account for the long-lasting effects in the vasculature. Rat aorta exposed to S-nitrosoglutathione (GSNO) displayed, even after washout of the drug, a persistent increase in cysteine-NO residues (detected by immunostaining using an antiserum that selectively recognized S-nitrosoproteins) and in NO content (detected by NO spin-trapping), a persistent attenuation of the effect of vasoconstrictors, and a relaxant response upon addition of low molecular weight (LMW) thiols. Rat mesenteric and porcine coronary artery exposed in vitro to GSNO, as well as aorta and mesenteric arteries removed from rats treated in vivo with GSNO, displayed similar modifications of contraction. In isolated aorta exposed to GSNO, the decrease of the contractile response and the relaxant effect of LMW thiols were both blunted by NO scavengers (oxyhemoglobin or 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide) or by a cyclic GMP-dependent protein kinase inhibitor (Rp-8-bromoguanosine-3',5'-cyclic monophosphorothioate). In these arteries, mercuric chloride (which cleaves the cysteine-NO bond) exerted a transient relaxation, completely abolished the one of LMW thiols, and blunted the increase in cysteine-NO residues and NO content. Together, these data support the idea that S-nitrosation of cysteine residues is involved in long-lasting effects of NO on arterial tone. They suggest that S-nitrosation of tissue thiols is a mechanism of formation of local NO stores from which biologically active NO can subsequently be released.
Collapse
Affiliation(s)
- Jacicarlos L Alencar
- Faculté de Pharmacie, Pharmacologie & Physico-Chimie, Université Louis Pasteur, Centre National de la Recherche Scientifique Unité Mixte Recherche 7034, 67401 Illkirch, France
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tabernero A, Reimund JM, Chasserot S, Muller CD, Andriantsitohaina R. Cyclooxygenase-2 expression and role of vasoconstrictor prostanoids in small mesenteric arteries from patients with Crohn's disease. Circulation 2003; 107:1407-10. [PMID: 12642362 DOI: 10.1161/01.cir.0000055321.13957.17] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The present study investigates the vascular reactivity and the involvement of nitric oxide and prostanoids in regulating vasoconstriction of small mesenteric arteries from patients with Crohn's disease (CD) to understand the vascular component of this pathology. METHODS AND RESULTS An increased production of proinflammatory cytokines (tumor necrosis factor-alpha and interleukins 1beta, 6, and 8) has been observed in biopsy specimens of inflammatory intestinal mucosa. However, contractile responses of small mesenteric arteries from CD patients in response to norepinephrine were not changed ex vivo when compared with controls. Exposure to either the nitric oxide synthase inhibitor N(G)-nitro-L-arginine or the cyclooxygenase (COX) inhibitor indomethacin did not modify contractions induced by norepinephrine in either control or CD patients. However, in the latter, the specific COX-2 inhibitor N-(2-cyclohexyloxy-4-nitrophenyl) methanesulfonamide significantly attenuated norepinephrine-induced vasoconstriction. Furthermore, immunohistochemical analysis showed marked COX-2 expression in the whole arterial wall of vessels from CD patients. Vessels from control patients exhibited weak COX-2 staining in the adventitial and endothelial layers only. CONCLUSIONS The above results provide direct evidence for COX-2 expression in small mesenteric arteries from CD patients. They also shed new light on the involvement of vasoconstrictor metabolites of COX in regulating contraction of these arteries. Of particular interest is the balance between vasoconstrictor products from COX-2 and unidentified vasodilatory products that maintained vascular reactivity in a physiological range despite an increase of circulatory cytokines in patients with CD.
Collapse
Affiliation(s)
- Antonia Tabernero
- Pharmacologie et Physico-Chimie des Interactions Cellulaires et Moléculaires, Illkirch, France
| | | | | | | | | |
Collapse
|
27
|
Eguchi D, D'Uscio LV, Wambi C, Weiler D, Kovesdi I, O'Brien T, Katusic ZS. Inhibitory effect of recombinant iNOS gene expression on vasomotor function of canine basilar artery. Am J Physiol Heart Circ Physiol 2002; 283:H2560-6. [PMID: 12388312 DOI: 10.1152/ajpheart.00415.2002] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study was designed to determine the effect of recombinant inducible nitric oxide (NO) synthase (iNOS) gene expression on vasomotor function in cerebral arteries. Isolated canine basilar arteries were exposed ex vivo (30 min at 37 degrees C) to an adenoviral vector [10(7), 10(8), or 10(9) plaque-forming units (pfu)/ml] encoding either the iNOS gene or the beta-galactosidase reporter gene. Twenty-four hours after transduction, Western blot analysis demonstrated expression of iNOS protein only in iNOS (10(9) pfu/ml)-transduced arteries. Immunohistochemical analysis localized iNOS expression predominantly in adventitia. Vascular reactivity of isolated basilar arteries was studied by isometric force recording. Concentration-response curves to UTP (10(-9)-10(-3) M) and diethylaminodiazen-1-ium-1,2-dioate (10(-10)-10(-5) M) were significantly shifted to the right in iNOS gene (10(9) pfu/ml)-transduced rings compared with control and beta-galactosidase-transduced rings (P < 0.05, n = 5-6). Endothelium-dependent relaxation to bradykinin was significantly attenuated in iNOS-transduced rings (P < 0.001, n = 8). The basal level of cGMP and superoxide anion (O(2)(-).) production were elevated in iNOS-transduced rings (P < 0.05, n = 7 for cGMP; P < 0.01, n = 6-9 for O(2)(-). production). Our results suggest that expression of recombinant iNOS in cerebral arteries reduces vasomotor reactivity to both vasoconstrictor and vasodilator agonists. Attenuation of contractions is most likely due to functional antagonism between UTP and cGMP. Reduction of endothelium-dependent relaxation to bradykinin appears to be mediated in part by reduced reactivity of smooth muscle cells to NO.
Collapse
Affiliation(s)
- Daihiko Eguchi
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Maffei A, Poulet R, Vecchione C, Colella S, Fratta L, Frati G, Trimarco V, Trimarco B, Lembo G. Increased basal nitric oxide release despite enhanced free radical production in hypertension. J Hypertens 2002; 20:1135-42. [PMID: 12023683 DOI: 10.1097/00004872-200206000-00025] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Although in hypertension a defect in stimulated nitric oxide (NO) is well established, little is known about basal NO levels. Thus, we measured directly in vessels from normotensive [Wistar-Kyoto (WKY)] rats and spontaneously hypertensive rats (SHR) both basal and stimulated NO production using a novel technique [4,5-diaminofluorescein (DAF-2) fluorescence]. METHODS Isolated vessels were exposed to the fluorescent probe DAF-2. After the technique was validated with increasing doses of acetylcholine in the presence and absence of NG-nitro-L-arginine methyl ester (l-NAME), we measured NO production in vessels from WKY rats and SHR in the same experimental setting. Finally, to explore the impact of reactive oxygen species (ROS) on NO release, we analysed the effect of an antioxidant, such as ascorbic acid, on basal and stimulated NO in aortic rings of WKY rats and SHR. RESULTS Aortic rings from SHR exhibited a higher basal NO production and a lower responsiveness to agonist-induced NO release as compared with those observed in WKY rats. Also in resistance vessels such as mesenteric arteries, basal NO production was higher in hypertension. In hypertensive rats, ascorbic acid was able to further increase basal NO release and recovered the impaired stimulated NO production, whereas no effect was detected in normotensive rats. CONCLUSIONS Our data reveal an increased basal NO availability in hypertension despite the increased production of ROS, suggesting a greater complexity in hypertensive endothelial dysfunction when the analysis is focused on direct NO measurement.
Collapse
|
29
|
Schulze-Bauer CAJ, Regitnig P, Holzapfel GA. Mechanics of the human femoral adventitia including the high-pressure response. Am J Physiol Heart Circ Physiol 2002; 282:H2427-40. [PMID: 12003855 DOI: 10.1152/ajpheart.00397.2001] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adventitial mechanics were studied on the basis of adventitial tube tests and associated stress analyses utilizing a thin-walled model. Inflation tests of 11 nonstenotic human femoral arteries (79.3 +/- 8.2 yr, means +/- SD) were performed during autopsy. Adventitial tubes were separated anatomically and underwent cyclic, quasistatic extension-inflation tests using physiological pressures and high pressures up to 100 kPa. Associated circumferential and axial stretches were typically <20%, indicating "adventitiosclerosis." Adventitias behaved nearly elastically for both loading domains, demonstrating high tensile strengths (>1 MPa). The anisotropic and strongly nonlinear mechanical responses were represented appropriately by two-dimensional Fung-type stored-energy functions. At physiological pressure (13.3 kPa), adventitias carry ~25% of the pressure load in situ, whereas their circumferential and axial stresses were similar to the total wall stresses (~50 kPa in both directions), supporting a "uniform stress hypothesis." At higher pressures, they became the mechanically predominant layer, carrying >50% of the pressure load. These significant load-carrying capabilities depended strongly on circumferential and axial in-vessel prestretches (mean values: 0.95 and 1.08). On the basis of these results, the mechanical role of the adventitia at physiological and hypertensive states and during balloon angioplasty was characterized.
Collapse
Affiliation(s)
- Christian A J Schulze-Bauer
- Institute for Structural Analysis, Computational Biomechanics, Graz University of Technology, 8010 Graz, 8035 Graz, Austria.
| | | | | |
Collapse
|
30
|
Wagner AH, Schwabe O, Hecker M. Atorvastatin inhibition of cytokine-inducible nitric oxide synthase expression in native endothelial cells in situ. Br J Pharmacol 2002; 136:143-9. [PMID: 11976279 PMCID: PMC1762109 DOI: 10.1038/sj.bjp.0704678] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Animal experimental studies have demonstrated that inducible nitric oxide synthase (iNOS) expression correlates with neointima formation and is prevented by HMG-CoA reductase inhibitors (statins). In the present study we have investigated the underlying mechanism of action of these drugs in isolated segments of the rat aorta. Western blot analysis and immunohistochemistry revealed that tumour necrosis factor alpha (TNFalpha) plus interferon-gamma (IFNgamma) synergistically induce iNOS gene expression in the endothelium but not in the smooth muscle of these segments while constitutive endothelial NO synthase (eNOS) abundance was markedly reduced. Pre-treatment with 1 - 10 microM atorvastatin, cerivastatin or pravastatin decreased TNFalpha plus IFNgamma stimulated iNOS expression in the endothelium irrespective of the presence of the HMG-CoA reductase product mevalonate (400 microM). Electrophoretic mobility shift assay experiments confirmed that the combination of TNFalpha plus IFNgamma causes activation of the transcription factors STAT-1 and NF-kappaB in native endothelial cells. Neutralization of these transcription factors by employing the corresponding decoy oligonucleotides confirmed their involvement in TNFalpha plus IFNgamma stimulated iNOS expression. Translocation of both transcription factors was attenuated by atorvastatin, and this effect was insensitive to exogenous mevalonate. The present findings thus demonstrate a specific HMG-CoA reductase-independent inhibitory effect of statins, namely atorvastatin, on cytokine-stimulated transcription factor activation in native endothelial cells in situ and the subsequent expression of a gene product implicated in vascular inflammation. This effect may be therapeutically relevant and in addition provide an explanation for the reported rapid onset of action of these drugs in humans.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Atorvastatin
- Cells, Cultured
- Cytokines/metabolism
- Cytokines/pharmacology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Electrophoretic Mobility Shift Assay
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Heptanoic Acids/pharmacology
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Immunohistochemistry
- In Vitro Techniques
- Interferon-gamma/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type II
- Oligonucleotides/pharmacology
- Pyrroles/pharmacology
- Rats
- Rats, Wistar
- STAT1 Transcription Factor
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Andreas H Wagner
- Department of Cardiovascular Physiology, University of Goettingen School of Medicine, Humboldtallee 23, 37073 Goettingen, Germany
| | - Oliver Schwabe
- Department of Cardiovascular Physiology, University of Goettingen School of Medicine, Humboldtallee 23, 37073 Goettingen, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, University of Goettingen School of Medicine, Humboldtallee 23, 37073 Goettingen, Germany
- Author for correspondence:
| |
Collapse
|
31
|
Kleschyov AL, Hubert G, Munzel T, Stoclet JC, Bucher B. Low molecular mass dinitrosyl nonheme-iron complexes up-regulate noradrenaline release in the rat tail artery. BMC Pharmacol 2002; 2:3. [PMID: 11872148 PMCID: PMC65697 DOI: 10.1186/1471-2210-2-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2001] [Accepted: 02/08/2002] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Dinitrosyl nonheme-iron complexes can appear in cells and tissues overproducing nitric oxide. It is believed that due to their chemical nature these species may be implicated in certain pathophysiological events. We studied the possible role of low molecular mass dinitrosyl iron complexes in the control of noradrenaline release in electrically stimulated rat tail artery. RESULTS A model complex, dinitrosyl-iron-thiosulfate (at 1-10 microM) produced a concentration-dependent enhancement of electrical field stimulated [3H]noradrenaline release (up to 2 fold). At the same time, dinitrosyl-iron-thiosulfate inhibited neurogenic vasoconstriction, consistent with its nitric oxide donor properties. A specific inhibitor of cyclic GMP dependent protein kinase, Rp-8pCPT-cGMPS, partially inhibited the effect of dinitrosyl-iron-thiosulfate on neurogenic vasoconstriction, but not on [3H]noradrenaline release. Another model complex, dinitrosyl-iron-cysteine (at 3 microM) elicited similar responses as dinitrosyl-iron-thiosulfate. Conventional NO and NO+ donors such as sodium nitroprusside, S-nitroso-L-cysteine or S-nitroso-glutathione (at 10 microM) had no effect on [3H]noradrenaline release, though they potently decreased electrically-induced vasoconstriction. The "false complex", iron(II)-thiosulfate showed no activity. CONCLUSIONS Low molecular mass iron dinitrosyl complexes can up-regulate the stimulation-evoked release of vascular [3H]noradrenaline, apparently independently of their NO donor properties. This finding may have important implications in inflammatory tissues.
Collapse
Affiliation(s)
- Andrei L Kleschyov
- Pharmacologie et Physicochimie des Interactions Cellulaires et Moléculaires, CNRS, UMR 7034, Université Louis Pasteur de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France
- University Hospital Hamburg-Eppendorf, Division of Cardiology, Hamburg 20246, Germany
| | - Gilles Hubert
- Pharmacologie et Physicochimie des Interactions Cellulaires et Moléculaires, CNRS, UMR 7034, Université Louis Pasteur de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France
| | - Thomas Munzel
- University Hospital Hamburg-Eppendorf, Division of Cardiology, Hamburg 20246, Germany
| | - Jean-Claude Stoclet
- Pharmacologie et Physicochimie des Interactions Cellulaires et Moléculaires, CNRS, UMR 7034, Université Louis Pasteur de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France
| | - Bernard Bucher
- Pharmacologie et Physicochimie des Interactions Cellulaires et Moléculaires, CNRS, UMR 7034, Université Louis Pasteur de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France
| |
Collapse
|
32
|
Periadventitial Drug Delivery for the Prevention of Restenosis: Rationale and Results. J Vasc Interv Radiol 2002. [DOI: 10.1016/s1051-0443(02)70167-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|