1
|
Lin PK, Davis GE. Extracellular Matrix Remodeling in Vascular Disease: Defining Its Regulators and Pathological Influence. Arterioscler Thromb Vasc Biol 2023; 43:1599-1616. [PMID: 37409533 PMCID: PMC10527588 DOI: 10.1161/atvbaha.123.318237] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
Because of structural and cellular differences (ie, degrees of matrix abundance and cross-linking, mural cell density, and adventitia), large and medium-sized vessels, in comparison to capillaries, react in a unique manner to stimuli that induce vascular disease. A stereotypical vascular injury response is ECM (extracellular matrix) remodeling that occurs particularly in larger vessels in response to injurious stimuli, such as elevated angiotensin II, hyperlipidemia, hyperglycemia, genetic deficiencies, inflammatory cell infiltration, or exposure to proinflammatory mediators. Even with substantial and prolonged vascular damage, large- and medium-sized arteries, persist, but become modified by (1) changes in vascular wall cellularity; (2) modifications in the differentiation status of endothelial cells, vascular smooth muscle cells, or adventitial stem cells (each can become activated); (3) infiltration of the vascular wall by various leukocyte types; (4) increased exposure to critical growth factors and proinflammatory mediators; and (5) marked changes in the vascular ECM, that remodels from a homeostatic, prodifferentiation ECM environment to matrices that instead promote tissue reparative responses. This latter ECM presents previously hidden matricryptic sites that bind integrins to signal vascular cells and infiltrating leukocytes (in coordination with other mediators) to proliferate, invade, secrete ECM-degrading proteinases, and deposit injury-induced matrices (predisposing to vessel wall fibrosis). In contrast, in response to similar stimuli, capillaries can undergo regression responses (rarefaction). In summary, we have described the molecular events controlling ECM remodeling in major vascular diseases as well as the differential responses of arteries versus capillaries to key mediators inducing vascular injury.
Collapse
Affiliation(s)
- Prisca K. Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| |
Collapse
|
2
|
The adventitia in arterial development, remodeling, and hypertension. Biochem Pharmacol 2022; 205:115259. [PMID: 36150432 DOI: 10.1016/j.bcp.2022.115259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022]
Abstract
The adventitia receives input signals from the vessel wall, the immune system, perivascular nerves and from surrounding tissues to generate effector responses that regulate structural and mechanical properties of blood vessels. It is a complex and dynamic tissue that orchestrates multiple functions for vascular development, homeostasis, repair, and disease. The purpose of this review is to highlight recent advances in our understanding of the origins, phenotypes, and functions of adventitial and perivascular cells with particular emphasis on hypertensive vascular remodeling.
Collapse
|
3
|
Kuhn P, Bubel M, Jennewein M, Guthörl S, Pohlemann T, Oberringer M. Dose-dependent dominance: How cell densities design stromal cell functions during soft tissue healing. Cell Biochem Funct 2022; 40:439-450. [PMID: 35707856 DOI: 10.1002/cbf.3705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/15/2022] [Accepted: 04/10/2022] [Indexed: 11/06/2022]
Abstract
Regular soft tissue healing relies on the well-organized interaction of different stromal cell types with endothelial cells. However, spatiotemporal conditions might provoke high densities of one special stromal cell type, potentially leading to impaired healing. Detailed knowledge of the functions of rivaling stromal cell types aiming for tissue contraction and stabilization as well as vascular support is mandatory. By the application of an in vitro approach comprising the evaluation of cell proliferation, cell morphology, myofibroblastoid differentiation, and cytokine release, we verified a density-dependent modulation of these functions among juvenile and adult fibroblasts, pericytes, and adipose-derived stem cells during their interaction with microvascular endothelial cells in cocultures. Results indicate that juvenile fibroblasts rather support angiogenesis via paracrine regulation at the early stage of healing, a role potentially compromised in adult fibroblasts. In contrast, pericytes showed a more versatile character aiming at angiogenesis, vessel stabilization, and tissue contraction. Such a universal character was even more pronounced among adipose-derived stem cells. The explicit knowledge of the characteristic functions of stromal cell types is a prerequisite for the development of new analytical and therapeutic approaches for impaired soft tissue healing. The present study delivers new considerations concerning the roles of rivaling stromal cell types within a granulation tissue, pointing to extraordinary properties of pericytes and adipose-derived stem cells.
Collapse
Affiliation(s)
- Philipp Kuhn
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Monika Bubel
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Martina Jennewein
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Silke Guthörl
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Tim Pohlemann
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Martin Oberringer
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
4
|
Li R, Qi Y, Yuan Q, Xu L, Gao M, Xu Y, Han X, Yin L, Liu C. Protective effects of dioscin against isoproterenol-induced cardiac hypertrophy via adjusting PKCε/ERK-mediated oxidative stress. Eur J Pharmacol 2021; 907:174277. [PMID: 34171391 DOI: 10.1016/j.ejphar.2021.174277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/24/2022]
Abstract
Cardiac hypertrophy (CH) plays a central role in cardiac remodeling and is an independent risk factor for cardiac events. It is imperative to find drugs with protective effect on CH. Dioscin, one natural product, shows various pharmacological activities, and PKCepsilon (PKCε) plays an important role in the physiological hypertrophic responses. Thus, we aimed to investigate the possible protective effect of dioscin on CH through PKCε. In the present study, the isoproterenol (ISO)-induced H9C2 cells and primary cardiomyocytes models, and the ISO-induced rat model were established, and the pharmacodynamics and mechanism of dioscin were investigated. In vitro results prompted that, dioscin significantly improved ISO-induced cardiomyocyte hypertrophy, decreased the levels of cell size, protein content of single cell, reactive oxygen species, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), beta-myosin heavy chain (β-MHC). Moreover, in vivo, changes in histopathological of the animals caused by ISO are improved by dioscin. And dioscin decreased the index of CH and the levels of CK, MDA, LDH, and increased the levels of GSH, SOD and GSH-Px. Mechanism research showed that dioscin inhibited the expression levels of PKCε, and affected the expression levels of p-MEK, p-ERK, Nrf2, Keap1 and HO-1 to inhibit oxidative stress. In addition, the results of ISO-induced CH in PKCε siRNA transfected H9C2 cells and C57BL/6 mice further showed that the protective effect of dioscin on CH, which was mediated by inhibition of PKCε/ERK signal pathway. In summary, dioscin can effectively inhibit CH by regulating PKCε-mediated oxidative stress, which should be considered as one potent candidate for new drug research and development to treat CH in the future.
Collapse
Affiliation(s)
- Ruomiao Li
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Qianhui Yuan
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China.
| | - Chuntong Liu
- Pharmaceutical Department, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, China.
| |
Collapse
|
5
|
Ezrokhi M, Zhang Y, Luo S, Cincotta AH. Time-of-Day-Dependent Effects of Bromocriptine to Ameliorate Vascular Pathology and Metabolic Syndrome in SHR Rats Held on High Fat Diet. Int J Mol Sci 2021; 22:ijms22116142. [PMID: 34200262 PMCID: PMC8201259 DOI: 10.3390/ijms22116142] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 12/31/2022] Open
Abstract
The treatment of type 2 diabetes patients with bromocriptine-QR, a unique, quick release micronized formulation of bromocriptine, improves glycemic control and reduces adverse cardiovascular events. While the improvement of glycemic control is largely the result of improved postprandial hepatic glucose metabolism and insulin action, the mechanisms underlying the drug's cardioprotective effects are less well defined. Bromocriptine is a sympatholytic dopamine agonist and reduces the elevated sympathetic tone, characteristic of metabolic syndrome and type 2 diabetes, which potentiates elevations of vascular oxidative/nitrosative stress, known to precipitate cardiovascular disease. Therefore, this study investigated the impact of bromocriptine treatment upon biomarkers of vascular oxidative/nitrosative stress (including the pro-oxidative/nitrosative stress enzymes of NADPH oxidase 4, inducible nitric oxide (iNOS), uncoupled endothelial nitric oxide synthase (eNOS), the pro-inflammatory/pro-oxidative marker GTP cyclohydrolase 1 (GTPCH 1), and the pro-vascular health enzyme, soluble guanylate cyclase (sGC) as well as the plasma level of thiobarbituric acid reactive substances (TBARS), a circulating marker of systemic oxidative stress), in hypertensive SHR rats held on a high fat diet to induce metabolic syndrome. Inasmuch as the central nervous system (CNS) dopaminergic activities both regulate and are regulated by CNS circadian pacemaker circuitry, this study also investigated the time-of-day-dependent effects of bromocriptine treatment (10 mg/kg/day at either 13 or 19 h after the onset of light (at the natural waking time or late during the activity period, respectively) among animals held on 14 h daily photoperiods for 16 days upon such vascular biomarkers of vascular redox state, several metabolic syndrome parameters, and mediobasal hypothalamic (MBH) mRNA expression levels of neuropeptides neuropeptide Y (NPY) and agouti-related protein (AgRP) which regulate the peripheral fuel metabolism and of mRNA expression of other MBH glial and neuronal cell genes that support such metabolism regulating neurons in this model system. Such bromocriptine treatment at ZT 13 improved (reduced) biomarkers of vascular oxidative/nitrosative stress including plasma TBARS level, aortic NADPH oxidase 4, iNOS and GTPCH 1 levels, and improved other markers of coupled eNOS function, including increased sGC protein level, relative to controls. However, bromocriptine treatment at ZT 19 produced no improvement in either coupled eNOS function or sGC protein level. Moreover, such ZT 13 bromocriptine treatment reduced several metabolic syndrome parameters including fasting insulin and leptin levels, as well as elevated systolic and diastolic blood pressure, insulin resistance, body fat store levels and liver fat content, however, such effects of ZT 19 bromocriptine treatment were largely absent versus control. Finally, ZT 13 bromocriptine treatment reduced MBH NPY and AgRP mRNA levels and mRNA levels of several MBH glial cell/neuronal genes that code for neuronal support/plasticity proteins (suggesting a shift in neuronal structure/function to a new metabolic control state) while ZT 19 treatment reduced only AgRP, not NPY, and was with very little effect on such MBH glial cell genes expression. These findings indicate that circadian-timed bromocriptine administration at the natural circadian peak of CNS dopaminergic activity (that is diminished in insulin resistant states), but not outside this daily time window when such CNS dopaminergic activity is naturally low, produces widespread improvements in biomarkers of vascular oxidative stress that are associated with the amelioration of metabolic syndrome and reductions in MBH neuropeptides and gene expressions known to facilitate metabolic syndrome. These results of such circadian-timed bromocriptine treatment upon vascular pathology provide potential mechanisms for the observed marked reductions in adverse cardiovascular events with circadian-timed bromocriptine-QR therapy (similarly timed to the onset of daily waking as in this study) of type 2 diabetes subjects and warrant further investigations into related mechanisms and the potential application of such intervention to prediabetes and metabolic syndrome patients as well.
Collapse
|
6
|
Pagano PJ, Cifuentes-Pagano E. The Enigmatic Vascular NOX: From Artifact to Double Agent of Change: Arthur C. Corcoran Memorial Lecture - 2019. Hypertension 2021; 77:275-283. [PMID: 33390049 DOI: 10.1161/hypertensionaha.120.13897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NOXs (NADPH oxidases) comprise a family of proteins whose primary function is the production of reactive oxygen species, namely, superoxide anion and hydrogen peroxide. The prototype first being discovered and characterized in neutrophils, multiple NOXs are now known to be broadly expressed in cell and organ systems and whose phylogeny spans countless life forms beginning with prokaryotes. This long-enduring evolutionary conservation underscores the importance of fundamental NOX functions. This review chronicles a personal perspective of the field beginning with the discovery of NOXs in the vasculature and the advances achieved through the years as to our understanding of their mechanisms of action and role in oxidative stress and disease. Furthermore, applications of isoform-selective inhibitors to dissect the role of NOX isozymes in vascular biology, focusing on inflammation, pulmonary hypertension, and aging are described.
Collapse
Affiliation(s)
- Patrick J Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| |
Collapse
|
7
|
Rysz J, Franczyk B, Ławiński J, Gluba-Brzózka A. Oxidative Stress in ESRD Patients on Dialysis and the Risk of Cardiovascular Diseases. Antioxidants (Basel) 2020; 9:antiox9111079. [PMID: 33153174 PMCID: PMC7693989 DOI: 10.3390/antiox9111079] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease is highly prevalent worldwide. The decline of renal function is associated with inadequate removal of a variety of uremic toxins that exert detrimental effects on cells functioning, thus affecting the cardiovascular system. The occurrence of cardiovascular aberrations in CKD is related to the impact of traditional risk factors and non-traditional CKD-associated risk factors, including anemia; inflammation; oxidative stress; the presence of some uremic toxins; and factors related to the type, frequency of dialysis and the composition of dialysis fluid. Cardiovascular diseases are the most frequent cause for the deaths of patients with all stages of renal failure. The kidney is one of the vital sources of antioxidant enzymes, therefore, the impairment of this organ is associated with decreased levels of these enzymes as well as increased levels of pro-oxidants. Uremic toxins have been shown to play a vital role in the onset of oxidative stress. Hemodialysis itself also enhances oxidative stress. Elevated oxidative stress has been demonstrated to be strictly related to kidney and cardiac damage as it aggravates kidney dysfunction and induces cardiac hypertrophy. Antioxidant therapies may prove to be beneficial since they can decrease oxidative stress, reduce uremic cardiovascular toxicity and improve survival.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-419 Łódź, Poland; (J.R.); (B.F.)
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-419 Łódź, Poland; (J.R.); (B.F.)
| | - Janusz Ławiński
- Department of Urology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-419 Łódź, Poland; (J.R.); (B.F.)
- Correspondence: ; Tel.: +48-42-639-3750
| |
Collapse
|
8
|
Abstract
The microcirculation maintains tissue homeostasis through local regulation of blood flow and oxygen delivery. Perturbations in microvascular function are characteristic of several diseases and may be early indicators of pathological changes in the cardiovascular system and in parenchymal tissue function. These changes are often mediated by various reactive oxygen species and linked to disruptions in pathways such as vasodilation or angiogenesis. This overview compiles recent advances relating to redox regulation of the microcirculation by adopting both cellular and functional perspectives. Findings from a variety of vascular beds and models are integrated to describe common effects of different reactive species on microvascular function. Gaps in understanding and areas for further research are outlined. © 2020 American Physiological Society. Compr Physiol 10:229-260, 2020.
Collapse
Affiliation(s)
- Andrew O Kadlec
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - David D Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Medicine-Division of Cardiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
9
|
Strassheim D, Gerasimovskaya E, Irwin D, Dempsey EC, Stenmark K, Karoor V. RhoGTPase in Vascular Disease. Cells 2019; 8:E551. [PMID: 31174369 PMCID: PMC6627336 DOI: 10.3390/cells8060551] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022] Open
Abstract
Ras-homologous (Rho)A/Rho-kinase pathway plays an essential role in many cellular functions, including contraction, motility, proliferation, and apoptosis, inflammation, and its excessive activity induces oxidative stress and promotes the development of cardiovascular diseases. Given its role in many physiological and pathological functions, targeting can result in adverse effects and limit its use for therapy. In this review, we have summarized the role of RhoGTPases with an emphasis on RhoA in vascular disease and its impact on endothelial, smooth muscle, and heart and lung fibroblasts. It is clear from the various studies that understanding the regulation of RhoGTPases and their regulators in physiology and pathological conditions is required for effective targeting of Rho.
Collapse
Affiliation(s)
- Derek Strassheim
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - David Irwin
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Edward C Dempsey
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA.
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Vijaya Karoor
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
10
|
Kilic-Toprak E, Kilic-Erkek O, Abban-Mete G, Caner V, Baris IC, Turhan G, Kucukatay V, Senol H, Kuru O, Bor-Kucukatay M. Contribution of Heme Oxygenase 2 to Blood Pressure Regulation in Response to Swimming Exercise and Detraining in Spontaneously Hypertensive Rats. Med Sci Monit 2018; 24:5851-5859. [PMID: 30132448 PMCID: PMC6116639 DOI: 10.12659/msm.908992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background We aimed to determine the effects of exercise followed by detraining on systolic blood pressure (SBP), heme oxygenase 2 (HO-2) expression, and carboxyhemoglobin (COHb) concentration in spontaneously hypertensive rats (SHR) to explain the role of carbon monoxide (CO) in this process. Material/Methods Animals were randomized into exercised and detrained groups. Corresponding sedentary rats were grouped as Time 1–2. Swimming of 60 min/5 days/week for 10 weeks was applied. Detraining rats discontinued training for an additional 5 weeks. Gene and protein expressions were determined by real-time PCR and immunohistochemistry. Results Aorta HO-2 histological scores (HSCORE) of hypertensive rats were lower, while SBP was higher. Swimming caused enhancement of HO-2 immunostaining in aorta endothelium and adventitia of SHR. Exercise induced elevation of blood COHb index in SHR. Synchronous BP lowering effect of exercise was observed. HO-2 mRNA expression, HSCORE, and blood COHb index were unaltered during detraining, while SBP was still low in SHR. Conclusions CO synthesized by HO-2 at least partly plays a role in SBP regulation in the SHR- and BP-lowering effect of exercise. Regular exercise with short-term pauses may be advised to both hypertensives and individuals who are at risk.
Collapse
Affiliation(s)
- Emine Kilic-Toprak
- Department of Physiology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Ozgen Kilic-Erkek
- Department of Physiology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Gulcin Abban-Mete
- Department of Histology-Embryology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Vildan Caner
- Department of Medical Genetics, Faculty of Medicine Kinikli, Pamukkale University, Denizli, Turkey
| | - Ikbal Cansu Baris
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Gurkan Turhan
- Department of Histology-Embryology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Vural Kucukatay
- Department of Physiology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Hande Senol
- Department of Biostatistics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Oktay Kuru
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Muğla Sıtkı Koçman University, Muğla, Turkey
| | - Melek Bor-Kucukatay
- Department of Physiology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
11
|
Lefferts WK, Sperry SD, Jorgensen RS, Kasprowicz AG, Skilton MR, Figueroa A, Heffernan KS. Carotid stiffness, extra-media thickness and visceral adiposity in young adults. Atherosclerosis 2017; 265:140-146. [DOI: 10.1016/j.atherosclerosis.2017.08.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 07/14/2017] [Accepted: 08/30/2017] [Indexed: 10/18/2022]
|
12
|
Yu B, Liu Z, Fu Y, Wang Y, Zhang L, Cai Z, Yu F, Wang X, Zhou J, Kong W. CYLD Deubiquitinates Nicotinamide Adenine Dinucleotide Phosphate Oxidase 4 Contributing to Adventitial Remodeling. Arterioscler Thromb Vasc Biol 2017; 37:1698-1709. [PMID: 28751569 DOI: 10.1161/atvbaha.117.309859] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Transdifferentiation of adventitial fibroblasts (AFs) into myofibroblasts plays a critical role during the vascular remodeling that occurs during atherosclerosis, restenosis, and aortic aneurysm. The ubiquitination/deubiquitination regulatory system is essential for the quality control of proteins. The involvement of ubiquitination/deubiquitination during AF transdifferentiation remains largely unknown. In this study, we determined the role of cylindromatosis (CYLD), a deubiquitinase, in the process of AF differentiation and activation in vitro and in vivo. APPROACH AND RESULTS Transforming growth factor-β1 and homocysteine, 2 known inducers of AF transdifferentiation, greatly upregulated CYLD expression in a time- and dose-dependent manner. The silencing of CYLD significantly inhibited AF transdifferentiation and activation as evidenced by the expression of contractile proteins, the production of the proinflammatory cytokines MCP-1 (monocyte chemotactic protein 1) and IL-6 (interleukin-6), the deposition of extracellular matrix, and cell migration. We further asked whether CYLD mediates AF activation via the regulation of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) as it is an essential factor during AF transdifferentiation. Indeed, the silencing of CYLD repressed transforming growth factor-β1-induced and homocysteine-induced Nox4 upregulation and reactive oxygen species production, whereas Nox4 overexpression greatly rescued the inhibitory effect on AF activation by CYLD silencing. Most interestingly, transforming growth factor-β1 and homocysteine repressed Nox4 ubiquitination and prolonged the half-life of Nox4. Moreover, Nox4 was deubiquitinated via a direct interaction with the ubiquitin-specific protease domain of CYLD. In accordance, hyperhomocysteinemia significantly increased adventitial CYLD and Nox4 expression, promoted AF transdifferentiation, and aggravated CaPO4-induced abdominal aortic aneurysm in mice. These effects were abolished in CYLD-/- mice. CONCLUSIONS CYLD contributes to the transdifferentiation of AFs via deubiquitinating Nox4 and may play a role in vascular remodeling.
Collapse
Affiliation(s)
- Bing Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Ziyi Liu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yi Fu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yingbao Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Lu Zhang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Zeyu Cai
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Fang Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Jun Zhou
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| | - Wei Kong
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| |
Collapse
|
13
|
Roles of Cells from the Arterial Vessel Wall in Atherosclerosis. Mediators Inflamm 2017; 2017:8135934. [PMID: 28680196 PMCID: PMC5478858 DOI: 10.1155/2017/8135934] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/26/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis has been identified as a chronic inflammatory disease of the arterial vessel wall. Accumulating evidence indicates that different cells from the tunica intima, media, adventitia, and perivascular adipose tissue not only comprise the intact and normal arterial vessel wall but also participate all in the inflammatory response of atherosclerosis via multiple intricate pathways. For instance, endothelial dysfunction has historically been considered to be the initiator of the development of atherosclerosis. The migration and proliferation of smooth muscle cells also play a pivotal role in the progression of atherosclerosis. Additionally, the fibroblasts from the adventitia and adipocytes from perivascular adipose tissue have received considerable attention given their special functions that contribute to atherosclerosis. In addition, numerous types of cytokines produced by different cells from the arterial vessel wall, including endothelium-derived relaxing factors, endothelium-derived contracting factors, tumor necrosis factors, interleukin, adhesion molecules, interferon, and adventitium-derived relaxing factors, have been implicated in atherosclerosis. Herein, we summarize the possible roles of different cells from the entire arterial vessel wall in the pathogenesis of atherosclerosis.
Collapse
|
14
|
Zhou L, Yang M, Zuo S, Guan X, Wang J, Chen Q, Zuo X, Jia S, Guo R. Dlitiazem inhibits the oxidative stress induced by angiotensin II through growth hormone secretagogue receptor type 1a in human umbilicus vein endothelial cells. Biomed Pharmacother 2017; 89:76-82. [DOI: 10.1016/j.biopha.2017.02.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 01/24/2017] [Accepted: 02/07/2017] [Indexed: 12/01/2022] Open
|
15
|
Barman SA, Fulton D. Adventitial Fibroblast Nox4 Expression and ROS Signaling in Pulmonary Arterial Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:1-11. [PMID: 29047077 DOI: 10.1007/978-3-319-63245-2_1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease arising from remodeling and narrowing of pulmonary arteries (PA) resulting in high pulmonary arterial blood pressure and ultimately right ventricular failure. Elevated production of reactive oxygen species (ROS) by NADPH oxidase 4 (Nox4), a constitutively active enzyme, has been associated with oxygen sensing, vasomotor control, cellular proliferation, differentiation, migration, apoptosis, senescence, fibrosis, and angiogenesis. Further, elevated expression of Nox4 has been reported in a number of cardiovascular diseases, including atherosclerosis, hypertension, cardiac failure, ischemic stroke, and PAH. However, the cellular location of Nox4 and its contribution to aberrant vascular remodeling in PAH remains poorly understood. The goal of this review is to summarize the recent literature on the enzymatic regulation of Nox4 in the production of ROS in PAH. In the vascular wall, Nox4 is present in fibroblasts, a primary cell of the adventitia, and matches the adventitial location of ROS production in PAH. Further, in adventitial fibroblasts, Nox4 overexpression stimulates migration and proliferation as well as matrix gene expression. Collectively, reports indicate that Nox4 contributes to altered fibroblast behavior, ROS production leading to hypertensive vascular remodeling and the development of PAH. Finally, we address the functional significance of Nox4 in fibroblasts, and also suggest an "outside in" (adventitial) process of vascular remodeling that is mediated by Nox4, which although has physiological roles in the intimal layer (i.e., endothelium), may also have pathologic importance in the adventitial layer of the vascular wall through signaling in fibroblasts.
Collapse
Affiliation(s)
- Scott A Barman
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, 30912, USA.
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA
| |
Collapse
|
16
|
Sangartit W, Pakdeechote P, Kukongviriyapan V, Donpunha W, Shibahara S, Kukongviriyapan U. Tetrahydrocurcumin in combination with deferiprone attenuates hypertension, vascular dysfunction, baroreflex dysfunction, and oxidative stress in iron-overloaded mice. Vascul Pharmacol 2016; 87:199-208. [PMID: 27713040 DOI: 10.1016/j.vph.2016.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/01/2016] [Accepted: 10/01/2016] [Indexed: 12/13/2022]
|
17
|
Ardanaz N, Pagano PJ. Hydrogen Peroxide as a Paracrine Vascular Mediator: Regulation and Signaling Leading to Dysfunction. Exp Biol Med (Maywood) 2016; 231:237-51. [PMID: 16514169 DOI: 10.1177/153537020623100302] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Numerous studies have demonstrated the ability of a variety of vascular cells, including endothelial cells, smooth muscle cells, and fibroblasts, to produce reactive oxygen species (ROS). Until recently, major emphasis was placed on the production of superoxide anion (O2–) in the vasculature as a result of its ability to directly attenuate the biological activity of endothelium-derived nitric oxide (NO). The short half-life and radius of diffusion of O2– drastically limit the role of this ROS as an important paracrine hormone in vascular biology. On the contrary, in recent years, the O2– metabolite hydrogen peroxide (H2O2) has increasingly been viewed as an important cellular signaling agent in its own right, capable of modulating both contractile and growth-promoting pathways with more far-reaching effects. In this review, we will assess the vascular production of H2O2, its regulation by endogenous scavenger systems, and its ability to activate a variety of vascular signaling pathways, thereby leading to vascular contraction and growth. This discussion will include the ability of H2O2 to (i) Initiate calcium flux as well as (ii) stimulate pathways leading to sensitization of contractile elements to calcium. The latter involves a variety of protein kinases that have also been strongly implicated in vascular hypertrophy. Previous Intensive study has emphasized the ability of NADPH oxidase-derived O2– and H2O2 to activate these pathways in cultured smooth muscle cells. However, growing evidence indicates a considerably more complex array of unique oxidase systems in the endothelium, media, and adventitia that appear to participate in these deleterious effects in a sequential and temporal manner. Taken together, these findings seem consistent with a paracrine effect of H2O2 across the vascular wall.
Collapse
Affiliation(s)
- Noelia Ardanaz
- Hypertension and Vascular Research Division, RM 7044, E&R Building, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202-2689, USA
| | | |
Collapse
|
18
|
Early regression of coronary artery remodeling with esmolol and DDAH/ADMA pathway in hypertensive rats. Hypertens Res 2016; 39:692-700. [PMID: 27250567 DOI: 10.1038/hr.2016.57] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 02/18/2016] [Accepted: 03/24/2016] [Indexed: 12/22/2022]
Abstract
Our preclinical study demonstrated that esmolol produces early regression of left ventricular hypertrophy in arterial hypertension. The aim of this study was to assess the effects of short-term esmolol therapy on the regression of left anterior descending artery remodeling in spontaneously hypertensive rats (SHRs), and to determine whether the asymmetric dimethylarginine (ADMA)/dimethylarginine dimethylaminohydrolase (DDAH) pathway, a regulator of nitric oxide (NO) bioavailability, accounted for this regression. Fourteen-month-old male SHRs were treated intravenously with vehicle (SHR, n=15) or esmolol (SHR-E, n=20) (300 μg kg-1 min-1). Age-matched, vehicle-treated male Wistar-Kyoto rats (WKY, n=15) served as controls. SHRs were also treated with nitroglycerin (SHR-N, n=5). After 48 h, the left anterior descending artery structure and morphology were assessed, and dose-response curves for 5-hydroxytryptamine (5-HT, 10-9-3 × 10-5 mol l-1) were constructed. ADMA concentrations in plasma and left ventricle and DDAH activity in tissue were analyzed. Wall thickness and cross-sectional area were significantly lower after treatment with esmolol in SHR-E than in SHR. Media thickness and smooth muscle cell count were lower in SHR-E than in SHR. Esmolol induced a significant reduction in adventitial cell count in SHR-E. The area under the concentration-response curves was significantly higher in SHR than in SHR-E, as were the esmolol normalized coronary artery contracting responses to 5-HT. We found significantly lower ADMA levels and significantly higher DDAH activity in the ventricle in SHR-E than in SHR. The protective effect of esmolol on the regression of left anterior descending artery remodeling may be related to the reduction in ADMA levels.
Collapse
|
19
|
Khovaeva YB, Batalova AA, Golovskoy BV, Gerasimova АV. [The status of large vessels in overweight and obesity according to ultrasound findings]. TERAPEVT ARKH 2016; 88:29-34. [PMID: 27070160 DOI: 10.17116/terarkh201688429-34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM To investigate the impact of overweight and obesity on the structure and function of large arteries in apparently healthy individuals; to estimate the magnitude of atherosclerotic lesions of the vessels. MATERIALS AND METHODS Sixty-one apparently healthy individuals with varying weights were randomly examined. All the examinees were divided into 3 groups according to their body mass index (BMI) and blood pressure (BP). They all underwent ultrasonography of the brachiocephalic arteries at the extracranial level, as well as the abdominal aorta and femoral arteries. RESULTS The overweight and obese persons have increased diameters and thickness of the intima-media complex of the common carotid arteries and aorta. These changes are maximally pronounced in obesity in conjunction with high normal BP (systolic BP, 130-139 mm Hg; diastolic BP, 85-89 mm Hg). At the same time, there is a reduction in blood flow velocities and an increase in arterial wall stiffness. More marked structural and functional changes are observed in persons with abdominal obesity. The femoral artery belonging to muscular ones exhibited no structural changes, but displayed reduced blood flow velocities in obesity concurrent with high normal BP. In all the groups, the spread of atherosclerotic plaques is equal; the latter are recorded in one third of the examinees. However, the degree and incidence of vascular atherosclerosis are higher in overweight and obesity concurrent with high normal BP. CONCLUSION The overweight and obese persons without cardiovascular diseases were found to have eccentric remodeling of elastic and muscular-and-elastic vessels: the carotid arteries and aorta in conjunction with increased intima-media thickness, reduced blood flow velocities, and atherosclerotic vascular changes. These changes are most pronounced in the obese persons with high normal BP. In all the groups, atherosclerotic changes are encountered mainly in the brachiocephalic arteries at the extracranial level.
Collapse
Affiliation(s)
- Ya B Khovaeva
- Acad. E.A. Wagner Perm State Medical University, Ministry of Health of Russia, Perm, Russia
| | - A A Batalova
- Acad. E.A. Wagner Perm State Medical University, Ministry of Health of Russia, Perm, Russia
| | - B V Golovskoy
- Acad. E.A. Wagner Perm State Medical University, Ministry of Health of Russia, Perm, Russia
| | - А V Gerasimova
- Acad. E.A. Wagner Perm State Medical University, Ministry of Health of Russia, Perm, Russia
| |
Collapse
|
20
|
Sun M, Ji J, Guo X, Liu W, Wang Y, Ma S, Hu W, Wang J, Jiang F. Early adventitial activation characterized by NADPH oxidase expression and neovascularization in an aortic transplantation model. Exp Mol Pathol 2015; 100:67-73. [PMID: 26655438 DOI: 10.1016/j.yexmp.2015.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/02/2015] [Indexed: 12/16/2022]
Abstract
Increasing evidence has suggested that arterial adventitia may contribute to pathological vessel remodeling by producing reactive oxygen species and promoting neovascularization. However, these processes have not been studied yet in transplantation-induced vascular pathologies. We characterized the dynamic changes in NADPH oxidase expression and adventitial angiogenic response in a model of allograft aortic transplantation. The thoracic aorta from Fischer 344 rats were transplanted into the abdominal aorta of Lewis rats. Graft specimens were collected on days 0.5, 3, 7, and 14 for morphometry, immunohistochemistry, immunofluorescence staining, and quantitative PCR tests. Following transplantation, adventitial thickening was found as early as day 3, while neointima was observed from day 7. As compared to normal adventitial tissue, the expression levels of NADPH oxidase subunits gp91phox and p47phox in graft adventitia were elevated from day 3 and further increased up to day 14. Immunohistochemistry staining showed that infiltrating macrophages appeared to be a major source of NADPH oxidase expression. Increases in NADPH oxidase expression were also detected in fibroblasts isolated from the graft adventitia. Gene silencing of p47phox significantly suppressed proliferation and migration of the graft fibroblast cells. We also showed that adventitial thickening was accompanied by increased adventitial neovascularization; at day 14, there was a positive correlation between the density of adventitial microvessels and the neointimal thickness. Transplantation injury induces NADPH oxidase expression and neovascularization in the adventitia, raising the possibility that the activated adventitia may represent a target site for prevention of transplantation-induced transplant vasculopathy.
Collapse
Affiliation(s)
- Mengyao Sun
- Institute of Pathology and Pathophysiology, School of Medicine, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Jian Ji
- Institute of Pathology and Pathophysiology, School of Medicine, Shandong University, Jinan, Shandong Province, 250012, China; Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Xiaotong Guo
- Institute of Pathology and Pathophysiology, School of Medicine, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Wenjun Liu
- Institute of Pathology and Pathophysiology, School of Medicine, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Yanyan Wang
- Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Siqin Ma
- School of Stomatology, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Weicheng Hu
- Institute of Pathology and Pathophysiology, School of Medicine, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Jianli Wang
- Institute of Pathology and Pathophysiology, School of Medicine, Shandong University, Jinan, Shandong Province, 250012, China.
| | - Fan Jiang
- Institute of Pathology and Pathophysiology, School of Medicine, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
21
|
Bushueva OY, Bulgakova IV, Ivanov VP, Polonikov AV. Association of Flavin Monooxygenase Gene E158K Polymorphism with Chronic Heart Disease Risk. Bull Exp Biol Med 2015; 159:776-8. [PMID: 26519273 DOI: 10.1007/s10517-015-3073-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Indexed: 10/22/2022]
Abstract
We studied the relationship between the risk of chronic heart disease and FMO3 gene polymorphism E158K analyzed by PCR and restriction fragment length polymorphism (RFLP) analysis. The homozygous 158KK genotype of FMO3 gene is associated with high risk of chronic heart disease in women, but not in men. FMO3 gene polymorphism E158K is a significant predictor of predisposition to chronic heart disease in women.
Collapse
Affiliation(s)
- O Yu Bushueva
- Department of Biology, Medical Genetics, and Ecology, Kursk State Medical University, Kursk, Russia.
| | - I V Bulgakova
- Department of Biology, Medical Genetics, and Ecology, Kursk State Medical University, Kursk, Russia
| | - V P Ivanov
- Department of Biology, Medical Genetics, and Ecology, Kursk State Medical University, Kursk, Russia
| | - A V Polonikov
- Department of Biology, Medical Genetics, and Ecology, Kursk State Medical University, Kursk, Russia
| |
Collapse
|
22
|
Vascular nitric oxide: Beyond eNOS. J Pharmacol Sci 2015; 129:83-94. [PMID: 26499181 DOI: 10.1016/j.jphs.2015.09.002] [Citation(s) in RCA: 515] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/11/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
As the first discovered gaseous signaling molecule, nitric oxide (NO) affects a number of cellular processes, including those involving vascular cells. This brief review summarizes the contribution of NO to the regulation of vascular tone and its sources in the blood vessel wall. NO regulates the degree of contraction of vascular smooth muscle cells mainly by stimulating soluble guanylyl cyclase (sGC) to produce cyclic guanosine monophosphate (cGMP), although cGMP-independent signaling [S-nitrosylation of target proteins, activation of sarco/endoplasmic reticulum calcium ATPase (SERCA) or production of cyclic inosine monophosphate (cIMP)] also can be involved. In the blood vessel wall, NO is produced mainly from l-arginine by the enzyme endothelial nitric oxide synthase (eNOS) but it can also be released non-enzymatically from S-nitrosothiols or from nitrate/nitrite. Dysfunction in the production and/or the bioavailability of NO characterizes endothelial dysfunction, which is associated with cardiovascular diseases such as hypertension and atherosclerosis.
Collapse
|
23
|
Wang G, Jacquet L, Karamariti E, Xu Q. Origin and differentiation of vascular smooth muscle cells. J Physiol 2015; 593:3013-30. [PMID: 25952975 PMCID: PMC4532522 DOI: 10.1113/jp270033] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/19/2015] [Indexed: 12/18/2022] Open
Abstract
Vascular smooth muscle cells (SMCs), a major structural component of the vessel wall, not only play a key role in maintaining vascular structure but also perform various functions. During embryogenesis, SMC recruitment from their progenitors is an important step in the formation of the embryonic vascular system. SMCs in the arterial wall are mostly quiescent but can display a contractile phenotype in adults. Under pathophysiological conditions, i.e. vascular remodelling after endothelial dysfunction or damage, contractile SMCs found in the media switch to a secretory type, which will facilitate their ability to migrate to the intima and proliferate to contribute to neointimal lesions. However, recent evidence suggests that the mobilization and recruitment of abundant stem/progenitor cells present in the vessel wall are largely responsible for SMC accumulation in the intima during vascular remodelling such as neointimal hyperplasia and arteriosclerosis. Therefore, understanding the regulatory mechanisms that control SMC differentiation from vascular progenitors is essential for exploring therapeutic targets for potential clinical applications. In this article, we review the origin and differentiation of SMCs from stem/progenitor cells during cardiovascular development and in the adult, highlighting the environmental cues and signalling pathways that control phenotypic modulation within the vasculature.
![]()
Collapse
Affiliation(s)
- Gang Wang
- Department of Emergency Medicine, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Laureen Jacquet
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Eirini Karamariti
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
24
|
Konradi J, Mollenhauer M, Baldus S, Klinke A. Redox-sensitive mechanisms underlying vascular dysfunction in heart failure. Free Radic Res 2015; 49:721-42. [DOI: 10.3109/10715762.2015.1027200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
25
|
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: 300] [Impact Index Per Article: 27.3] [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
|
26
|
Pugliese SC, Poth JM, Fini MA, Olschewski A, El Kasmi KC, Stenmark KR. The role of inflammation in hypoxic pulmonary hypertension: from cellular mechanisms to clinical phenotypes. Am J Physiol Lung Cell Mol Physiol 2014; 308:L229-52. [PMID: 25416383 DOI: 10.1152/ajplung.00238.2014] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypoxic pulmonary hypertension (PH) comprises a heterogeneous group of diseases sharing the common feature of chronic hypoxia-induced pulmonary vascular remodeling. The disease is usually characterized by mild to moderate pulmonary vascular remodeling that is largely thought to be reversible compared with the progressive irreversible disease seen in World Health Organization (WHO) group I disease. However, in these patients, the presence of PH significantly worsens morbidity and mortality. In addition, a small subset of patients with hypoxic PH develop "out-of-proportion" severe pulmonary hypertension characterized by pulmonary vascular remodeling that is irreversible and similar to that in WHO group I disease. In all cases of hypoxia-related vascular remodeling and PH, inflammation, particularly persistent inflammation, is thought to play a role. This review focuses on the effects of hypoxia on pulmonary vascular cells and the signaling pathways involved in the initiation and perpetuation of vascular inflammation, especially as they relate to vascular remodeling and transition to chronic irreversible PH. We hypothesize that the combination of hypoxia and local tissue factors/cytokines ("second hit") antagonizes tissue homeostatic cellular interactions between mesenchymal cells (fibroblasts and/or smooth muscle cells) and macrophages and arrests these cells in an epigenetically locked and permanently activated proremodeling and proinflammatory phenotype. This aberrant cellular cross-talk between mesenchymal cells and macrophages promotes transition to chronic nonresolving inflammation and vascular remodeling, perpetuating PH. A better understanding of these signaling pathways may lead to the development of specific therapeutic targets, as none are currently available for WHO group III disease.
Collapse
Affiliation(s)
- Steven C Pugliese
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado;
| | - Jens M Poth
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Mehdi A Fini
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; and
| | - Karim C El Kasmi
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, University of Colorado Denver, School of Medicine, Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
27
|
Manickam N, Patel M, Griendling KK, Gorin Y, Barnes JL. RhoA/Rho kinase mediates TGF-β1-induced kidney myofibroblast activation through Poldip2/Nox4-derived reactive oxygen species. Am J Physiol Renal Physiol 2014; 307:F159-71. [PMID: 24872317 PMCID: PMC4101629 DOI: 10.1152/ajprenal.00546.2013] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 05/18/2014] [Indexed: 02/07/2023] Open
Abstract
The small G proteins Rac1 and RhoA regulate actin cytoskeleton, cell shape, adhesion, migration, and proliferation. Recent studies in our laboratory have shown that NADPH oxidase Nox4-derived ROS are involved in transforming growth factor (TGF)-β1-induced rat kidney myofibroblast differentiation assessed by the acquisition of an α-smooth muscle actin (α-SMA) phenotype and expression of an alternatively spliced fibronectin variant (Fn-EIIIA). Rac1 and RhoA are essential in signaling by some Nox homologs, but their role as effectors of Nox4 in kidney myofibroblast differentiation is not known. In the present study, we explored a link among Rac1 and RhoA and Nox4-dependent ROS generation in TGF-β1-induced kidney myofibroblast activation. TGF-β1 stimulated an increase in Nox4 protein expression, NADPH oxidase activity, and abundant α-SMA and Fn-EIIIA expression. RhoA but not Rac1 was involved in TGF-β1 induction of Nox4 signaling of kidney myofibroblast activation. TGF-β1 stimulated active RhoA-GTP and increased Rho kinase (ROCK). Inhibition of RhoA with small interfering RNA and ROCK using Y-27632 significantly reduced TGF-β1-induced stimulation of Nox4 protein, NADPH oxidase activity, and α-SMA and Fn-EIIIA expression. Treatment with diphenyleneiodonium, an inhibitor of NADPH oxidase, did not decrease RhoA activation but inhibited TGF-β1-induced α-SMA and Fn-EIIIA expression, indicating that RhoA is upstream of ROS generation. RhoA/ROCK also regulated polymerase (DNA-directed) δ-interacting protein 2 (Poldip2), a newly discovered Nox4 enhancer protein. Collectively, these data indicate that RhoA/ROCK is upstream of Poldip2-dependent Nox4 regulation and ROS production and induces redox signaling of kidney myofibroblast activation and may broader implications in the pathophysiology of renal fibrosis.
Collapse
Affiliation(s)
- Nagaraj Manickam
- The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| | - Mandakini Patel
- The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| | - Kathy K Griendling
- The Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Yves Gorin
- The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| | - Jeffrey L Barnes
- The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| |
Collapse
|
28
|
Barman SA, Chen F, Su Y, Dimitropoulou C, Wang Y, Catravas JD, Han W, Orfi L, Szantai-Kis C, Keri G, Szabadkai I, Barabutis N, Rafikova O, Rafikov R, Black SM, Jonigk D, Giannis A, Asmis R, Stepp DW, Ramesh G, Fulton DJR. NADPH oxidase 4 is expressed in pulmonary artery adventitia and contributes to hypertensive vascular remodeling. Arterioscler Thromb Vasc Biol 2014; 34:1704-15. [PMID: 24947524 DOI: 10.1161/atvbaha.114.303848] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is a progressive disease arising from remodeling and narrowing of pulmonary arteries (PAs) resulting in high pulmonary blood pressure and ultimately right ventricular failure. Elevated production of reactive oxygen species by NADPH oxidase 4 (Nox4) is associated with increased pressure in PH. However, the cellular location of Nox4 and its contribution to aberrant vascular remodeling in PH remains poorly understood. Therefore, we sought to identify the vascular cells expressing Nox4 in PAs and determine the functional relevance of Nox4 in PH. APPROACH AND RESULTS Elevated expression of Nox4 was detected in hypertensive PAs from 3 rat PH models and human PH using qualititative real-time reverse transcription polymerase chain reaction, Western blot, and immunofluorescence. In the vascular wall, Nox4 was detected in both endothelium and adventitia, and perivascular staining was prominently increased in hypertensive lung sections, colocalizing with cells expressing fibroblast and monocyte markers and matching the adventitial location of reactive oxygen species production. Small-molecule inhibitors of Nox4 reduced adventitial reactive oxygen species generation and vascular remodeling as well as ameliorating right ventricular hypertrophy and noninvasive indices of PA stiffness in monocrotaline-treated rats as determined by morphometric analysis and high-resolution digital ultrasound. Nox4 inhibitors improved PH in both prevention and reversal protocols and reduced the expression of fibroblast markers in isolated PAs. In fibroblasts, Nox4 overexpression stimulated migration and proliferation and was necessary for matrix gene expression. CONCLUSION These findings indicate that Nox4 is prominently expressed in the adventitia and contributes to altered fibroblast behavior, hypertensive vascular remodeling, and development of PH.
Collapse
Affiliation(s)
- Scott A Barman
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary.
| | - Feng Chen
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary.
| | - Yunchao Su
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Christiana Dimitropoulou
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Yusi Wang
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - John D Catravas
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Weihong Han
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Laszlo Orfi
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Csaba Szantai-Kis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Gyorgy Keri
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Istvan Szabadkai
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Nektarios Barabutis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Olga Rafikova
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Ruslan Rafikov
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Stephen M Black
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Danny Jonigk
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Athanassios Giannis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Reto Asmis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - David W Stepp
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Ganesan Ramesh
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - David J R Fulton
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary.
| |
Collapse
|
29
|
Abstract
The author summarizes the structure of the connective tissues, the increasing motion of the constituents, which determine the role in establishing the structure and function of that. The structure and function of the connective tissue are related to each other in the resting as well as inflammatory states. It is emphasized that cellular events in the connective tissue are part of the defence of the organism, the localisation of the damage and, if possible, the maintenance of restitutio ad integrum. The organism responds to damage with inflammation, the non specific immune response, as well as specific, adaptive immunity. These processes are located in the connective tissue. Sterile and pathogenic inflammation are relatively similar processes, but inevitable differences are present, too. Sialic acids and glycoproteins containing sialic acids have important roles, and the role of Siglecs is also highlighted. Also, similarities and differences in damages caused by pathogens and sterile agents are briefly summarized. In addition, the roles of adhesion molecules linked to each other, and the whole event of inflammatory processes are presented. When considering practical consequences it is stressed that the structure (building up) of the organism and the defending function of inflammation both have fundamental importance. Inflammation has a crucial role in maintaining the integrity and the unimpaired somato-psychological state of the organism. Thus, inflammation serves as a tool of organism identical with the natural immune response, inseparably connected with the specific, adaptive immune response. The main events of the inflammatory processes take place in the connective tissue.
Collapse
Affiliation(s)
- Lajos Jakab
- Semmelweis Egyetem, Általános Orvostudományi Kar III. Belgyógyászati Klinika Budapest Kútvölgyi út 4. 1125
| |
Collapse
|
30
|
Owens CD, Gasper WJ, Walker JP, Alley HF, Conte MS, Grenon SM. Safety and feasibility of adjunctive dexamethasone infusion into the adventitia of the femoropopliteal artery following endovascular revascularization. J Vasc Surg 2014; 59:1016-24. [PMID: 24423476 DOI: 10.1016/j.jvs.2013.10.051] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 10/04/2013] [Accepted: 10/04/2013] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Restenosis following endovascular treatment of the femoropopliteal segment is associated with the inflammatory response produced in the artery wall at the time of the procedure. Although local drug delivery to the superficial femoral and popliteal arteries promises improved patency, data are currently limited. We hypothesized that improved percutaneous delivery of an anti-inflammatory compound into the adventitia of the femoropopliteal at the time of endovascular treatment would be safe, feasible, and decrease the inflammatory response. METHODS This was a prospective, investigator-initiated, phase I, first-in-man study testing the safety and feasibility of percutaneous adventitial delivery of dexamethasone. Following successful intervention, an adventitial microinfusion catheter was advanced over a 0.014-inch wire to the treated segment. Its microneedle (0.9 mm long × 140-μm diameter) was deployed into the adventitia to deliver dexamethasone (4 mg/mL) mixed with contrast agent (80:20 ratio), providing fluoroscopic visualization. The primary safety outcome measure was freedom from vessel dissection, thrombosis, or extravasation while the primary efficacy outcome was duplex-determined binary restenosis defined as a peak systolic velocity ratio >2.5. RESULTS Twenty patients with Rutherford clinical category 2-5 enrolled in this study. The mean age was 66, and 55% had diabetes mellitus. Treated lesion length was 8.9 ± 5.3 cm, and 50% were chronic total occlusions. Eighty percent of treated lesions were in the distal superficial femoral or popliteal arteries. All lesions were treated by balloon angioplasty with provisional stenting (n = 6) for suboptimal result. Three patients were treated with atherectomy as well. A mean of 1.6 ± 1.1 mg (0.5 ± 0.3 mL) of dexamethasone sodium phosphate was injected per centimeter of lesion length. In total, a mean of 12.1 ± 6.1 mg of dexamethasone was injected per patient. The mean number of injections required per lesion was 3.0 ± 1.3 cm, minimum one and maximum six injections. There was 100% technical success of drug delivery and no procedural or drug-related adverse events. The mean Rutherford score decreased from 3.1 ± .7 (median, 3.0) preoperatively to .5 ± .7 at 6 months (median, 0.0; P < .00001). Over this same time interval, the index leg ankle-brachial index increased from .68 ± .15 to .89 ± .19 (P = .0003). The preoperative C-reactive protein in this study was 6.9 ± 8.5 indicating severe baseline inflammation, which increased to 14.0 ± 23.1 mg/L (103% increase) at 24 hours following the procedure. However, this increase did not reach statistical significance of P = .14. Two patients met the primary efficacy end point of loss of primary patency by reoccluding their treated segment of the index lesion during the follow-up period. CONCLUSIONS Adventitial drug delivery via a microinfusion catheter is a safe and feasible alternative to intimal-based methods for adjunctive treatment in the femoropopliteal segment. The 6-month preliminary results suggest perivascular dexamethasone treatment may improve outcomes following angioplasty to the femoral and popliteal arteries, and support further clinical investigation of this approach.
Collapse
Affiliation(s)
- Christopher D Owens
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif.
| | - Warren J Gasper
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| | - Joy P Walker
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| | - Hugh F Alley
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| | - Michael S Conte
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| | - S Marlene Grenon
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| |
Collapse
|
31
|
Abstract
The cellular metabolism of oxygen generates potentially deleterious reactive oxygen species, including superoxide anion, hydrogen peroxide and hydroxyl radical. Under normal physiologic conditions, the rate and magnitude of oxidant formation is balanced by the rate of oxidant elimination. However, an imbalance between pro-oxidants and antioxidants results in oxidative stress, which is the pathogenic outcome of the overproduction of oxidants that overwhelms the cellular antioxidant capacity. There is growing evidence that increased oxidative stress and associated oxidative damage are mediators of vascular injury in cardiovascular pathologies, including hypertension, atherosclerosis and ischemia-reperfusion. This development has evoked considerable interest because of the possibilities that therapies targeted against reactive oxygen intermediates by decreasing the generation of reactive oxygen species and/or by increasing availability of antioxidants may be useful in minimizing vascular injury. This review focuses on the vascular actions of reactive oxygen species, the role of oxidative stress in vascular damage in hypertension and the therapeutic potential of modulating oxygen radical bioavailability in hypertension. In particular, the following topics will be highlighted: chemistry and sources of reactive oxygen species, antioxidant defense mechanisms, signaling events mediated by reactive oxygen species, role of reactive oxygen species in hypertension and the putative therapeutic role of antioxidants in cardiovascular disease.
Collapse
Affiliation(s)
- Rhian M Touyz
- Clinical Research Institute of Montreal, Quebec, Canada.
| |
Collapse
|
32
|
Wang HD, Chapman A. Essential Role of Adventitial Reactive Oxygen Species (ROS) in Vascular Function. SYSTEMS BIOLOGY OF FREE RADICALS AND ANTIOXIDANTS 2014:1287-1303. [DOI: 10.1007/978-3-642-30018-9_54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Stenmark KR, Nozik-Grayck E, Gerasimovskaya E, Anwar A, Li M, Riddle S, Frid M. The adventitia: Essential role in pulmonary vascular remodeling. Compr Physiol 2013; 1:141-61. [PMID: 23737168 DOI: 10.1002/cphy.c090017] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A rapidly emerging concept is that the vascular adventitia acts as a biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. It is the most complex compartment of the vessel wall and comprises a variety of cells including fibroblasts, immunomodulatory cells, resident progenitor cells, vasa vasorum endothelial cells, and adrenergic nerves. In response to vascular stress or injury, resident adventitial cells are often the first to be activated and reprogrammed to then influence tone and structure of the vessel wall. Experimental data indicate that the adventitial fibroblast, the most abundant cellular constituent of adventitia, is a critical regulator of vascular wall function. In response to vascular stresses such as overdistension, hypoxia, or infection, the adventitial fibroblast is activated and undergoes phenotypic changes that include proliferation, differentiation, and production of extracellular matrix proteins and adhesion molecules, release of reactive oxygen species, chemokines, cytokines, growth factors, and metalloproteinases that, collectively, affect medial smooth muscle cell tone and growth directly and that stimulate recruitment and retention of circulating inflammatory and progenitor cells to the vessel wall. Resident dendritic cells also participate in "sensing" vascular stress and actively communicate with fibroblasts and progenitor cells to simulate repair processes that involve expansion of the vasa vasorum, which acts as a conduit for further delivery of inflammatory/progenitor cells. This review presents the current evidence demonstrating that the adventitia acts as a key regulator of pulmonary vascular wall function and structure from the "outside in."
Collapse
Affiliation(s)
- Kurt R Stenmark
- University of Colorado Denver - Pediatric Critical Care, Aurora, Colorado, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Owens CD, Gasper WJ, Rahman AS, Conte MS. Vein graft failure. J Vasc Surg 2013; 61:203-16. [PMID: 24095042 DOI: 10.1016/j.jvs.2013.08.019] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/12/2013] [Accepted: 08/14/2013] [Indexed: 02/06/2023]
Abstract
After the creation of an autogenous lower extremity bypass graft, the vein must undergo a series of dynamic structural changes to stabilize the arterial hemodynamic forces. These changes, which are commonly referred to as remodeling, include an inflammatory response, the development of a neointima, matrix turnover, and cellular proliferation and apoptosis. The sum total of these processes results in dramatic alterations in the physical and biomechanical attributes of the arterialized vein. The most clinically obvious and easily measured of these is lumen remodeling of the graft. However, although somewhat less precise, wall thickness, matrix composition, and endothelial changes can be measured in vivo within the healing vein graft. Recent translational work has demonstrated the clinical relevance of remodeling as it relates to vein graft patency and the systemic factors influencing it. By correlating histologic and molecular changes in the vein, insights into potential therapeutic strategies to prevent bypass failure and areas for future investigation are explored.
Collapse
Affiliation(s)
- Christopher D Owens
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif.
| | - Warren J Gasper
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| | - Amreen S Rahman
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| | - Michael S Conte
- Division of Vascular and Endovascular Surgery, University of California San Francisco Medical Center, San Francisco, Calif
| |
Collapse
|
35
|
Abstract
The acute and chronic constitutional reactions of the organism elicited by sterile causes and pathogenic structures threatening the soundness of the organism are surveyed by the author. It is emphasized that depending on causes which can be very different, there are various syndromes occurring in the clinical practice. On the basis of multitudiness of pathogenic factors and individual differences, the infammatory reactions are clinically, pathologically and pathobiochemically can be hugely variable. The acute inflammatory response may be sterile. It is often difficult to recognize in these processes whether the inflammation is harmful or beneficial for the organism as a whole. It is possible that the inflammatory response itself is the defending resource of the individual. The non-sterile acute inflammation is evoked by pathogenic microorganisms. The variety of clinical syndromes are explained by the high diversity of pathogenic microbes, the individualities of the defending organisms, and the natural and adaptive immunity of the organism which may be intact or possibly defective. In the latter case the inflammation itself is the disease, as a consequence of a pathological process conducted by the cortico-hypothalamo-adernal axis. The acute inflammation is a defending, preventing and repairing process, constituting an important part of the natural innate immune response. It is inseparable from the natural innate immune response, which is in close cooperation with the adaptive, specific immune response with mutual effects on each of the other. The conductor and the response reactions of the two immune responses are also the same. There are alterations in serum proteins/glycoproteins synthesized mostly by the hepatocytes. Because the concentration of almost all proteins/glycoproteins may change, the use of the discriminative term “acute phase reactant” is hardly relevant. For example, the HDL molecule is a negative “acute phase reactant”. On the gound of clinical, pathological and biochemical caracteristics, the chronic sterile inflammation is a very different entity. It has been established that atherosclerosis is one of the ab origine chronic inflammatory syndrome. It is a long-lasting pathological entity progressing, rather than resolving with different celerity, namely a unique vasculitis syndrome. We are speaking about risk factors instead of causes, which constitute larger or smaller groups to elicite the preventing reaction of the host. The propagations and final outcomes are quite different from that of the acute process. The disadvantages or benefits for the organism are scarcely predictable, albeit the chronic process may have roles in its prolonged nature. Orv. Hetil., 2013, 154, 1247–1255.
Collapse
Affiliation(s)
- Lajos Jakab
- Semmelweis Egyetem, Általános Orvostudományi Kar III. Belgyógyászati Klinika Budapest Kútvölgyi út 4. 1125
| |
Collapse
|
36
|
Oleic, linoleic and linolenic acids increase ros production by fibroblasts via NADPH oxidase activation. PLoS One 2013; 8:e58626. [PMID: 23579616 PMCID: PMC3620266 DOI: 10.1371/journal.pone.0058626] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/05/2013] [Indexed: 12/13/2022] Open
Abstract
The effect of oleic, linoleic and γ-linolenic acids on ROS production by 3T3 Swiss and Rat 1 fibroblasts was investigated. Using lucigenin-amplified chemiluminescence, a dose-dependent increase in extracellular superoxide levels was observed during the treatment of fibroblasts with oleic, linoleic and γ-linolenic acids. ROS production was dependent on the addition of β-NADH or NADPH to the medium. Diphenyleneiodonium inhibited the effect of oleic, linoleic and γ-linolenic acids on fibroblast superoxide release by 79%, 92% and 82%, respectively. Increased levels of p47phox phosphorylation due to fatty acid treatment were detected by Western blotting analyses of fibroblast proteins. Increased p47phox mRNA expression was observed using real-time PCR. The rank order for the fatty acid stimulation of the fibroblast oxidative burst was as follows: γ-linolenic > linoleic > oleic. In conclusion, oleic, linoleic and γ-linolenic acids stimulated ROS production via activation of the NADPH oxidase enzyme complex in fibroblasts.
Collapse
|
37
|
Mechanical stretch changes coronary artery fibroblasts function by upregulating HSF1 protein expression. Int J Biol Macromol 2013; 59:105-10. [PMID: 23541557 DOI: 10.1016/j.ijbiomac.2013.03.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 02/19/2013] [Accepted: 03/19/2013] [Indexed: 01/13/2023]
Abstract
The study is designed to investigate effect of mechanical stretch on the function of fibroblast cells. Human coronary artery fibroblasts were cultured. They were divided into two groups: stretch group (stretch for 24h) and no-stretch group (did not stretch). ELISA analysis was used for detection of collagen secretion. CCK-8 method was used for detection of cells proliferation. RT-PCR method was used for detection of MMP, TIMP, IL-6, alpha-SMA, HSF1 and HSP70 mRNA expression. Western-blotting method was used for detection of HSF1 protein expression. Results showed that cells proliferation in stretch group was stronger than that in no-stretch group. Hydroxyproline secretion in stretch group was more than that in no-stretch group. MMP-9/TIMP, alpha-SMA, IL-6, HSF1 and HSP70 in stretch group was higher than those in no-stretch group. Western-blotting analysis showed that HSF1 protein expression was upregulated in stretch group. It can be concluded that mechanical stretch changed human coronary artery fibroblasts cells proliferation, collagen formation, the secretion of inflammatory factor possibly by upregulating HSF1 protein expression.
Collapse
|
38
|
Grant CA, Twigg PC. Pseudostatic and dynamic nanomechanics of the tunica adventitia in elastic arteries using atomic force microscopy. ACS NANO 2013; 7:456-64. [PMID: 23241059 DOI: 10.1021/nn304508x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Tunica adventitia, the outer layer of blood vessels, is an important structural feature, predominantly consisting of collagen fibrils. This study uses pseudostatic atomic force microscopy (AFM) nanoindentation at physiological conditions to show that the distribution of indentation modulus and viscous creep for the tunica adventitia of porcine aorta and pulmonary artery are distinct. Dynamic nanoindentation demonstrates that the viscous dissipation of the tunica adventitia of the aorta is greater than the pulmonary artery. We suggest that this mechanical property of the aortic adventitia is functionally advantageous due to the higher blood pressure within this vessel during the cardiac cycle. The effects on pulsatile deformation and dissipative energy losses are discussed.
Collapse
Affiliation(s)
- Colin A Grant
- Advanced Materials Engineering RKT Centre, School of Engineering, Design and Technology, University of Bradford, Bradford, Yorkshire BD7 1DP, UK
| | | |
Collapse
|
39
|
Stenmark KR, Yeager ME, El Kasmi KC, Nozik-Grayck E, Gerasimovskaya EV, Li M, Riddle SR, Frid MG. The adventitia: essential regulator of vascular wall structure and function. Annu Rev Physiol 2012; 75:23-47. [PMID: 23216413 PMCID: PMC3762248 DOI: 10.1146/annurev-physiol-030212-183802] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The vascular adventitia acts as a biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. It is the most complex compartment of the vessel wall and is composed of a variety of cells, including fibroblasts, immunomodulatory cells (dendritic cells and macrophages), progenitor cells, vasa vasorum endothelial cells and pericytes, and adrenergic nerves. In response to vascular stress or injury, resident adventitial cells are often the first to be activated and reprogrammed to influence the tone and structure of the vessel wall; to initiate and perpetuate chronic vascular inflammation; and to stimulate expansion of the vasa vasorum, which can act as a conduit for continued inflammatory and progenitor cell delivery to the vessel wall. This review presents the current evidence demonstrating that the adventitia acts as a key regulator of vascular wall function and structure from the outside in.
Collapse
Affiliation(s)
- Kurt R. Stenmark
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Michael E. Yeager
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Karim C. El Kasmi
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Eva Nozik-Grayck
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | | | - Min Li
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Suzette R. Riddle
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Maria G. Frid
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| |
Collapse
|
40
|
Kivelä A, Hartikainen J, Ylä-Herttuala S. Dotted collar placed around carotid artery induces asymmetric neointimal lesion formation in rabbits without intravascular manipulations. BMC Cardiovasc Disord 2012; 12:91. [PMID: 23075120 PMCID: PMC3485168 DOI: 10.1186/1471-2261-12-91] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 10/09/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neointimal formation in atherosclerosis has been subject for intense research. However, good animal models mimicking asymmetrical lesion formation in human subjects have been difficult to establish. The aim of this study was to develop a model which would lead to the formation of eccentric lesions under macroscopically intact non-denuded endothelium. METHODS We have developed a new collar model where we placed two cushions or dots inside the collar. Arterial lesions were characterized using histology and ultrasound methods. RESULTS When this dotted collar was placed around carotid and femoral arteries it produced asymmetrical pressure on adventitia and a mild flow disturbance, and hence a change in shear stress. Our hypothesis was that this simple procedure would reproducibly produce asymmetrical lesions without any intraluminal manipulations. Intima/media ratio increased towards the distal end of the collar with the direction of blood flow under macroscopically intact endothelium. Macrophages preferentially accumulated in areas of the thickest neointima thus resembling early steps in human atherosclerotic plaque formation. Proliferating cells in these lesions and underlying media were scarce at eight weeks time point. CONCLUSION The improved dotted collar model produces asymmetrical human-like atherosclerotic lesions in rabbits. This model should be useful in studies regarding the pathogenesis and formation of eccentric atherosclerotic lesions.
Collapse
Affiliation(s)
- Antti Kivelä
- A.I.Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | | |
Collapse
|
41
|
Pathophysiological Mechanisms of Carotid Plaque Vulnerability: Impact on Ischemic Stroke. Arch Immunol Ther Exp (Warsz) 2012; 60:431-42. [DOI: 10.1007/s00005-012-0192-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 08/06/2012] [Indexed: 10/27/2022]
|
42
|
Common carotid artery diameter and cardiovascular risk factors in overweight or obese postmenopausal women. Int J Vasc Med 2012; 2012:169323. [PMID: 22957258 PMCID: PMC3432379 DOI: 10.1155/2012/169323] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 06/01/2012] [Indexed: 12/24/2022] Open
Abstract
Arterial diameter is an underutilized indicator of vascular health. We hypothesized that interadventitial and lumen diameter of the common carotid artery would be better indicators of vascular health than carotid plaque or intima media thickness (IMT). Participants were 491 overweight or obese, postmenopausal women who were former or current hormone therapy (HT) users, 52-62 years, with waist circumference >80 cm. We evaluated cross-sectional associations of cardiovascular risk factors with carotid measures, by HT status. Former HT users had a worse cardiovascular profile than current HT users: larger adventitial (6.94 mm versus 6.79 mm) and lumen diameter (5.44 mm versus 5.31 mm, both P < 0.01) independent of cardiovascular risk factors; IMT and plaque were similar. Larger diameters were best explained by former HT use, higher pulse pressure, and greater weight. Independent of potential confounders, overweight and obese postmenopausal former HT users had larger carotid diameters than current HT users. Carotid diameter should be considered in studies of HT.
Collapse
|
43
|
Nugent HM, Ng YS, White D, Groothius A, Kanner G, Edelman ER. Ultrasound-guided percutaneous delivery of tissue-engineered endothelial cells to the adventitia of stented arteries controls the response to vascular injury in a porcine model. J Vasc Surg 2012; 56:1078-88. [PMID: 22796118 DOI: 10.1016/j.jvs.2012.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 02/19/2012] [Accepted: 03/02/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE High restenosis rates are a limitation of peripheral vascular interventions. Previous studies have shown that surgical implantation of a tissue-engineered endothelium onto the adventitia surface of injured vessels regulates vascular repair. In the present study, we developed a particulate formulation of tissue-engineered endothelium and a method to deliver the formulation perivascular to injured blood vessels using a percutaneous, minimally invasive technique. METHODS Stainless steel stents were implanted in 18 balloon-injured femoral arteries of nine domestic swine, followed by ultrasound-guided percutaneous perivascular injection of gelatin particles containing cultured allogeneic porcine aortic endothelial cells (PAE). Controls received injections of empty particles (matrix) or no perivascular injection (sham) after stent deployment. Animals were sacrificed after 90 days. RESULTS Angiographic analysis revealed a significantly greater lumen diameter in the stented segments of arteries treated with PAE/matrix (4.72 ± 0.12 mm) compared with matrix (4.01 ± 0.20 mm) or sham (4.03 ± 0.16 mm) controls (P < .05). Similarly, histologic analysis revealed that PAE/matrix-treated arteries had the greatest lumen area (20.4 ± 0.7 mm(2); P < .05) compared with controls (16.1 ± 0.9 mm(2) and 17.1 ± 1.0 mm(2) for sham and matrix controls, respectively) and the smallest intimal area (3.3 ± 0.4 mm(2); P < .05) compared with controls (6.2 ± 0.5 mm(2) and 4.4 ± 0.5 mm(2) for sham and matrix controls, respectively). Overall, PAE-treated arteries had a 33% to 50% decrease in percent occlusion (P < .05) compared with controls. Histopathological analysis revealed fewer leukocytes present in the intima in the PAE/matrix group compared with control groups, suggesting that the biological effects were in part due to inhibition of the inflammatory phase of the vascular response to injury. CONCLUSIONS Minimally invasive, perivascular delivery of PAE/matrix to stented arteries was performed safely using ultrasound-guided percutaneous injections and significantly decreased stenosis. Application at the time of or subsequent to peripheral interventions may decrease clinical restenosis rates.
Collapse
|
44
|
Extracellular Matrix Remodeling in Takayasu's Arteritis: Role of Matrix Metalloproteinases and Adventitial Inflammation. Arch Med Res 2012; 43:406-10. [DOI: 10.1016/j.arcmed.2012.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 07/16/2012] [Indexed: 11/23/2022]
|
45
|
Majesky MW, Dong XR, Hoglund V, Daum G, Mahoney WM. The adventitia: a progenitor cell niche for the vessel wall. Cells Tissues Organs 2011; 195:73-81. [PMID: 22005572 DOI: 10.1159/000331413] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent observations suggest that the adventitial layer of blood vessels exhibits properties resembling a stem/progenitor cell niche. Progenitor cells have been isolated from the adventitia of both murine and human blood vessels with the potential to form endothelial cells, mural cells, osteogenic cells, and adipocytes. These progenitors appear to cluster at or near the border zone between the outer media and inner adventitia. In the mouse, this border zone region corresponds to a localized site of sonic hedgehog signaling in the artery wall. This brief review will discuss the emerging evidence that the tunica adventitia may provide a niche-like signaling environment for resident progenitor cells and will address the role of the adventitia in growth, remodeling, and repair of the artery wall.
Collapse
Affiliation(s)
- Mark W Majesky
- Seattle Children's Research Institute, University of Washington, Seattle, Wash., USA.
| | | | | | | | | |
Collapse
|
46
|
Hu Y, Xu Q. Adventitial biology: differentiation and function. Arterioscler Thromb Vasc Biol 2011; 31:1523-9. [PMID: 21677295 DOI: 10.1161/atvbaha.110.221176] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent evidence indicates that stem/progenitor cells are present in the adventitia and participate in vascular repair and the formation of neointimal lesions in severely damaged vessels. Data have also demonstrated that these resident stem/progenitor cells could differentiate into endothelial or smooth muscle cells in response to different stimuli. Under pathological conditions, adventitial inflammation results in releasing a panel of cytokines, such as stromal cell-derived factor-1 and tumor necrosis factor-α, that may lead to local stem/progenitor mobilization and differentiation. Overall, these data support the impact of the adventitial progenitors in pathophysiological processes of lesion development in the arterial wall. In the present review, we aim to summarize the data concerning the presence of the resident stem cells and discuss the pathological impact of the adventitia in vascular diseases. We will also discuss the possible signal pathways orchestrating stem cell differentiation toward vascular lineage and highlight controversial issues related to the role of adventitial progenitors.
Collapse
Affiliation(s)
- Yanhua Hu
- Cardiovascular Division, King's College London British Heart Foundation, Centre, London, United Kingdom
| | | |
Collapse
|
47
|
Majesky MW, Dong XR, Hoglund V, Mahoney WM, Daum G. The adventitia: a dynamic interface containing resident progenitor cells. Arterioscler Thromb Vasc Biol 2011; 31:1530-9. [PMID: 21677296 DOI: 10.1161/atvbaha.110.221549] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Conventional views of the tunica adventitia as a poorly organized layer of vessel wall composed of fibroblasts, connective tissue, and perivascular nerves are undergoing revision. Recent studies suggest that the adventitia has properties of a stem/progenitor cell niche in the artery wall that may be poised to respond to arterial injury. It is also a major site of immune surveillance and inflammatory cell trafficking and harbors a dynamic microvasculature, the vasa vasorum, that maintains the medial layer and provides an important gateway for macrophage and leukocyte migration into the intima. In addition, the adventitia is in contact with tissue that surrounds the vessel and may actively participate in exchange of signals and cells between the vessel wall and the tissue in which it resides. This brief review highlights recent advances in our understanding of the adventitia and its resident progenitor cells and discusses progress toward an integrated view of adventitial function in vascular development, repair, and disease.
Collapse
Affiliation(s)
- Mark W Majesky
- Seattle Children’s Research Institute, Departments of Pediatric, Center for Cardiovascular Biology, and the Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98101, USA.
| | | | | | | | | |
Collapse
|
48
|
Norton CE, Jernigan NL, Kanagy NL, Walker BR, Resta TC. Intermittent hypoxia augments pulmonary vascular smooth muscle reactivity to NO: regulation by reactive oxygen species. J Appl Physiol (1985) 2011; 111:980-8. [PMID: 21757577 DOI: 10.1152/japplphysiol.01286.2010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Intermittent hypoxia (IH) resulting from sleep apnea can lead to pulmonary hypertension. IH causes oxidative stress that may limit bioavailability of the endothelium-derived vasodilator nitric oxide (NO) and thus contribute to this hypertensive response. We therefore hypothesized that increased vascular superoxide anion (O(2)(-)) generation reduces NO-dependent pulmonary vasodilation following IH. To test this hypothesis, we examined effects of the O(2)(-) scavenger tiron on vasodilatory responses to the endothelium-dependent vasodilator ionomycin and the NO donor S-nitroso-N-acetylpenicillamine in isolated lungs from hypocapnic-IH (H-IH; 3 min cycles of 5% O(2)/air flush, 7 h/day, 4 wk), eucapnic-IH (E-IH; cycles of 5% O(2), 5% CO(2)/air flush), and sham-treated (air/air cycled) rats. Next, we assessed effects of endogenous O(2)(-) on NO- and cGMP-dependent vasoreactivity and measured O(2)(-) levels using the fluorescent indicator dihydroethidium (DHE) in isolated, endothelium-disrupted small pulmonary arteries from each group. Both E-IH and H-IH augmented NO-dependent vasodilation; however, enhanced vascular smooth muscle (VSM) reactivity to NO following H-IH was masked by an effect of endogenous O(2)(-). Furthermore, H-IH and E-IH similarly increased VSM sensitivity to cGMP, but this response was independent of either O(2)(-) generation or altered arterial protein kinase G expression. Finally, both H-IH and E-IH increased arterial O(2)(-) levels, although this response was more pronounced following H-IH, and H-IH exposure resulted in greater protein tyrosine nitration indicative of increased NO scavenging by O(2)(-). We conclude that IH increases pulmonary VSM sensitivity to NO and cGMP. Furthermore, endogenous O(2)(-) limits NO-dependent vasodilation following H-IH through an apparent reduction in bioavailable NO.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131-0001, USA.
| | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Al Ghouleh I, Pagano PJ. Endosomal ClC-3 and Nox1: moving marksmen of redox signaling? Arterioscler Thromb Vasc Biol 2011; 31:240-2. [PMID: 21248280 DOI: 10.1161/atvbaha.110.220053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|