Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Signal ; 76: 109783, 2020 12.
Article in English | MEDLINE | ID: mdl-32956789

ABSTRACT

Pathological neointimal growth can develop in patients as a result of vascular injury following percutaneous coronary intervention and coronary artery bypass grafting using autologous saphenous vein, leading to arterial or vein graft occlusion. Neointima formation driven by intimal hyperplasia occurs as a result of a complex interplay between molecular and cellular processes involving different cell types including endothelial cells, vascular smooth muscle cells and various inflammatory cells. Therefore, understanding the intercellular communication mechanisms underlying this process remains of fundamental importance in order to develop therapeutic strategies to preserve endothelial integrity and vascular health post coronary interventions. Extracellular vesicles (EVs), including microvesicles and exosomes, are membrane-bound particles secreted by cells which mediate intercellular signalling in physiological and pathophysiological states, however their role in neointima formation is not fully understood. The purification and characterization techniques currently used in the field are associated with many limitations which significantly hinder the ability to comprehensively study the role of specific EV types and make direct functional comparisons between EV subpopulations. In this review, the current knowledge focusing on EV signalling in neointima formation post vascular injury is discussed.


Subject(s)
Endothelial Cells , Extracellular Vesicles/metabolism , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Neointima/metabolism , Vascular System Injuries , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
2.
Sci Rep ; 9(1): 3154, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816157

ABSTRACT

The counter-regulatory axis, Angiotensin Converting Enzyme 2, Angiotensin-(1-7), Mas receptor (ACE2/Ang-1-7/MasR), of the renin angiotensin system (RAS) is a potential therapeutic target in stroke, with Ang-(1-7) reported to have neuroprotective effects in pre-clinical stroke models. Here, an extensive investigation of the functional and mechanistic effects of Ang-(1-7) was performed in a rodent model of stroke. Using longitudinal magnetic resonance imaging (MRI) it was observed that central administration of Ang-(1-7) following transient middle cerebral artery occlusion (MCAO) increased the amount of tissue salvage compared to reperfusion alone. This protective effect was not due to early changes in blood brain barrier (BBB) permeability, microglia activation or inflammatory gene expression. However, increases in NADPH oxidase 1 (Nox1) mRNA expression were observed in the treatment group compared to control. In order to determine whether Ang-(1-7) has direct cerebrovascular effects, laser speckle contrast imaging (LSCI) was performed to measure dynamic changes in cortical perfusion following reperfusion. Delivery of Ang-(1-7) did not have any effect on cortical perfusion following reperfusion however; it showed an indication to prevent the 'steal phenomenon' within the contralateral hemisphere. The comprehensive series of studies have demonstrated a moderate protective effect of Ang-(1-7) when given alongside reperfusion to increase tissue salvage.


Subject(s)
Angiotensin I/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , NADPH Oxidase 1/genetics , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/genetics , Stroke/drug therapy , Angiotensin-Converting Enzyme 2 , Animals , Blood-Brain Barrier/drug effects , Contrast Media/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Infarction, Middle Cerebral Artery/diagnostic imaging , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/physiopathology , Inflammation/diagnostic imaging , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Magnetic Resonance Imaging , Microglia/drug effects , Microglia/pathology , Middle Cerebral Artery/diagnostic imaging , Middle Cerebral Artery/drug effects , Neuroprotective Agents , Proto-Oncogene Mas , RNA, Messenger/genetics , Rats , Renin-Angiotensin System/genetics , Reperfusion/methods , Stroke/diagnostic imaging , Stroke/genetics , Stroke/pathology
3.
Gene Ther ; 15(6): 443-51, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18004401

ABSTRACT

Targeted delivery of biological agents to atherosclerotic plaques may provide a novel treatment and/or useful tool for imaging of atherosclerosis in vivo. However, there are no known viral vectors that possess the desired tropism. Two plaque-targeting peptides, CAPGPSKSC (CAP) and CNHRYMQMC (CNH) were inserted into the capsid of adeno-associated virus 2 (AAV2) to assess vector retargeting. AAV2-CNH produced significantly higher levels of transduction than unmodified AAV2 in human, murine and rat endothelial cells, whereas transduction of nontarget HeLa cells was unaltered. Transduction studies and surface plasmon resonance suggest that AAV2-CNH uses membrane type 1 matrix metalloproteinase as a surface receptor. AAV2-CAP only produced higher levels of transduction in rat endothelial cells, possibly because the virus was found to be affected by proteasomal degradation. In vivo substantially higher levels of both peptide-modified AAV2 vectors was detected in the brachiocephalic artery (site of advanced atherosclerotic plaques) and aorta, whereas reduced levels were detected in all other organs examined. These results suggest that in the AAV2 platform the peptides are exposed on the capsid surface in a way that enables efficient receptor binding and so creates effective atherosclerotic plaque targeted vectors.


Subject(s)
Atherosclerosis/therapy , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Animals , Apolipoproteins E/genetics , Cell Line , Dependovirus/metabolism , Gene Targeting , Genetic Engineering , Genetic Vectors/genetics , Genetic Vectors/metabolism , Heparin/metabolism , Humans , Matrix Metalloproteinase 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/genetics , Protein Binding , Rats , Surface Plasmon Resonance , Transduction, Genetic/methods , Transgenes
4.
Gene Ther ; 11(16): 1296-300, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15292916

ABSTRACT

Adenovirus (Ad)-mediated gene delivery is a promising approach for genetic manipulation of the vasculature and is being used in both preclinical models and clinical trials. However, safety concerns relating to infection of nontarget tissue and the poor infectivity of vascular cells compared to other cell types necessitates Ad vector refinement. Here, we combine a transductional targeting approach to improve vascular cell infectivity through RGD peptide insertion into adenovirus fibers, combined with transcriptional targeting to endothelial cells using a approximately 1 kb fragment of the fms-like tyrosine kinase receptor-1 (FLT-1) promoter. Single- and double-modified vectors were characterized in human cell lines that either support or have silenced FLT-1 expression. In rat hepatocytes and endothelial cells, the double modification substantially shifted transduction profiles toward vascular endothelial cells. Furthermore, in intact aortae derived from spontaneously hypertensive rats that display enhanced alphav integrin expression on dysfunctional endothelium, enhanced levels of transduction were observed using the double-modified vector but not in aortae derived from normotensive control rats. Our data indicate that Ad-mediated transduction can be beneficially modified in vitro and in vivo by combining fiber modification and a cell-selective promoter within a single-component vector system.


Subject(s)
Adenoviridae Infections/genetics , Adenoviridae/genetics , Gene Targeting/methods , Animals , Aorta/physiopathology , Cell Line , Cells, Cultured , Endothelial Cells/physiology , Genetic Therapy/methods , Genetic Vectors/genetics , Hepatocytes/physiology , Humans , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor Protein-Tyrosine Kinases , Receptors, Virus/genetics , Transcription, Genetic/genetics , Transduction, Genetic/methods , Vascular Endothelial Growth Factor Receptor-1/genetics
5.
Mol Ther ; 4(6): 534-42, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11735337

ABSTRACT

Adenovirus type 5 (Ad) based vectors transduce vascular endothelial cells (EC) and have been widely used for vascular gene transfer. However, many cell types express the Ad receptor (cox-sackievirus adenovirus receptor; CAR), preventing selective EC infection and precluding clinical use. We previously isolated the human EC-binding peptides SIGYPLP and LSNFHSS by phage display and demonstrated by means of a bispecific antibody that SIGYPLP directs efficient, high-level, EC-selective Ad-mediated gene transfer. We now generate genetically modified Ad fiber proteins with selective EC tropism by engineering these peptides into the HI loop of the Ad fiber. SIGYPLP, but not LSNFHSS, enhanced EC selectivity, demonstrating maintenance of peptide-cell binding fidelity upon incorporation into virions. Combining fiber mutations that block CAR binding (detargeting) with SIGYPLP insertion (retargeting) generated a novel Ad vector, AdKO1SIG, in a single component system. AdKO1SIG demonstrated efficient and selective tropism for EC compared with control Ad vectors. This is the first demonstration of genetic incorporation of a novel, mammalian, cell-selective ligand that retains its targeting fidelity in the Ad fiber HI loop, in combination with point mutations that abolish fiber-CAR interaction. This study demonstrates the potential for improving the cell-selectivity and safety of adenoviral vectors.


Subject(s)
Adenoviridae/physiology , Endothelium, Vascular/virology , Genetic Therapy/methods , Peptide Fragments/genetics , Receptors, Virus/metabolism , Binding Sites , Binding, Competitive , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein , DNA Primers/chemistry , Flow Cytometry , Gene Targeting/methods , Genetic Vectors , Green Fluorescent Proteins , Humans , Immunoblotting , Luminescent Proteins/metabolism , Plasmids/genetics , Receptors, Virus/genetics , T-Lymphocytes/metabolism , Transduction, Genetic , Transfection
6.
Circulation ; 104(13): 1538-43, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11571249

ABSTRACT

BACKGROUND: Mitogenic stimuli present at the site of coronary arterial balloon injury contribute to the progression and development of a restenotic lesion, many signaling through a common pathway involving the small G protein p21(ras). Our aim was to demonstrate in biochemical studies that farnesyl protein transferase inhibitor III (FPTIII) is an inhibitor of p21(ras) processing and that when it is given locally in vivo at the site of coronary balloon injury in a porcine model, it can inhibit neointima formation. METHODS AND RESULTS: FPTIII (1 to 25 micromol/L) concentration-dependently reduced p21(ras) levels in porcine coronary artery smooth muscle cell membranes. FPTIII also prevented p42/p44 MAPK activation and DNA synthesis in response to platelet-derived growth factor in these cells at a concentration of 25 micromol/L. Application of 25 micromol/L FPTIII locally for 15 minutes to balloon-injured porcine coronary arteries in vivo prevented neointima formation assessed at 4 weeks, reduced proteoglycan deposition, and inhibited adventitial hypertrophy. Coronary arteries from FPTIII-treated pigs had no deterioration in contraction or in endothelium-dependent relaxation. CONCLUSIONS: The study demonstrates in the pig that short-term local delivery of inhibitors of p21(ras)-dependent mitogenic signal transduction prevents restenosis after balloon angioplasty.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Angioplasty, Balloon, Coronary/adverse effects , Enzyme Inhibitors/therapeutic use , Organophosphonates/therapeutic use , Postoperative Complications/prevention & control , Animals , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Organophosphonates/administration & dosage , Signal Transduction/drug effects , Swine , Tunica Intima/drug effects , Tunica Intima/pathology , Vascular Diseases/etiology , Vascular Diseases/prevention & control
7.
Eur J Pharmacol ; 345(3): 269-77, 1998 Mar 26.
Article in English | MEDLINE | ID: mdl-9592026

ABSTRACT

Previous studies from our laboratory have shown that nitric oxide (NO) can reduce the release of free radicals from activated leukocytes. The aim of this study was to assess the role of endothelium-derived nitric oxide and leukocyte-derived free radicals in the contractile response to non-preactivated leukocytes. Vessel tension studies were performed in rabbit endothelium-intact aortic vessel rings precontracted with 5-hydroxytryptamine (1 microM). Addition of leukocytes isolated from rabbit blood were added to the rings in increasing concentrations (10(3)-10(6) cell ml(-1)) under control conditions and in the presence of L-nitroarginine methyl ester (L-NAME 1 mM), D-NAME (1 mM), or superoxide dismutase (100 U ml(-1)). The responses to superoxide radical (generated by xanthine plus xanthine oxidase, X/XO), hydrogen peroxide, hypochlorite and peroxynitrite were also assessed. The nature of the free radicals released from non-activated isolated leukocytes, zymosan-stimulated leukocytes (in whole blood) and isolated vessel rings was assessed using luminol-enhanced chemiluminescence. Cumulative addition of leukocyte suspensions to aortic rings caused a concentration-dependent contractile response which was abolished by preincubation of the vessel ring with L-NAME. D-NAME and superoxide dismutase were without effect. All the free radicals tested produced a relaxation of the precontracted aortic ring. The response to X/XO was not affected by superoxide dismutase, but abolished by catalase. The responses to hydrogen peroxide and hypochlorite were both found to be dependent upon the presence of endothelium and NO. The response to peroxynitrite was endothelium-independent and was blocked by methylene blue. While the main free radical released from unstimulated leukocytes and vessel rings was superoxide, the main radical released from activated leukocytes was found to be hypochlorite. These results suggest that the vascular contraction seen in response to non-preactivated leukocytes is due to inhibition, by NO, of the release of free radicals from the leukocytes when activated by contact with the vascular endothelium, thus allowing co-released vasoconstrictor substances to exert their effect.


Subject(s)
Endothelium, Vascular/physiology , Leukocytes/physiology , Muscle, Smooth, Vascular/physiology , Nitric Oxide/physiology , Animals , Aorta, Thoracic/physiology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Free Radicals , Hydroxyl Radical , In Vitro Techniques , Luminescent Measurements , Male , Muscle Contraction/physiology , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/cytology , Rabbits , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/physiology , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...