Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 196
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2210-2219, 2017 09.
Article in English | MEDLINE | ID: mdl-28645655

ABSTRACT

Given the importance of leukotrienes in vascular inflammation induced by local tissue injury, this study investigated the role for 5-lipoxygenase (5-LO) in monocytes in the development of intimal hyperplasia. As a mechanistic study, the importance of monocyte 5-LO in monocyte-macrophage differentiation with subsequent infiltration in neointima was evaluated. In a mouse model of wire-injured femoral artery, intimal hyperplasia started as early as 2wks after injury, and luminal area and blood flow were reduced due to increased neointima formation. Time-dependent increases in macrophage infiltration were observed in neointima and showed a positive relationship with neointima volume. In 5-LO-deficient (KO) mice or wild-type (WT) mice treated with an inhibitor of 5-LO activating protein (MK886, 1 and 10mg/kg), intimal hyperplasia and macrophage infiltration into neointima were reduced, but monocyte adhesion to injured luminal surface was not inhibited, which suggested 5-LO participates in monocyte-macrophage differentiation. In an in vitro study, monocyte-macrophage differentiation was found to be increased by high mobility group box 1 protein (HMGB1), but this effect was attenuated in cells isolated from 5-LO-KO mice. Furthermore, macrophage infiltration and intimal hyperplasia were more prominent in 5-LO-KO mice transplanted with monocytes from WT mice than in 5-LO-KO mice transplanted with monocytes from 5-LO-KO mice. Taken together, it was suggested that 5-LO in monocytes played a pivotal role in monocyte-macrophage differentiation and subsequent infiltration of macrophage in neointima, leading to vascular remodeling after vascular injury.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Femoral Artery , Indoles/pharmacology , Lipoxygenase Inhibitors/pharmacology , Macrophages/enzymology , Monocytes/enzymology , Neointima , Animals , Arachidonate 5-Lipoxygenase/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Femoral Artery/enzymology , Femoral Artery/injuries , Femoral Artery/pathology , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Hyperplasia , Macrophages/pathology , Mice , Mice, Knockout , Monocytes/pathology , Neointima/drug therapy , Neointima/enzymology , Neointima/pathology , Tunica Intima/enzymology , Tunica Intima/pathology , Vascular Remodeling/drug effects , Vascular Remodeling/genetics
2.
Arch Biochem Biophys ; 598: 11-7, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27050934

ABSTRACT

The role of the Mas receptor in the activity of valsartan against intimal hyperplasia is unclear. Herein, we investigated the role of the angiotensin-converting enzyme 2 (ACE2)-angiotensin-(1-7)-Mas receptor axis on the activity of valsartan against intimal hyperplasiain balloon-injured rat aortic arteries. Wistar rats were randomized equally into the sham control group, injured group, and injured plus valsartan (20 mg/kg/d)-treated group. Valsartan significantly attenuated the vascular smooth muscle cell proliferation and intimal and medial thickening on days 14 and 28 after injury. The angiotensin-(1-7) levels as well as ACE2 and Mas receptor mRNA/protein expression were significantly decreased in the injured rats, compared to the uninjured rats; meanwhile, the angiotensin II level as well as the ACE and AT1 receptor mRNA/protein expression were increased (all P < 0.05 or < 0.01). Additionally, the p-ERK protein expression was increased (P < 0.01). Treatment with valsartan significantly increased the angiotensin-(1-7) levels as well as ACE2 and Mas receptor mRNA/protein expression but decreased the angiotensin II level, ACE and AT1 receptor mRNA/protein expression, as well as the p-ERK protein expression, compared to the injured group (all P < 0.05 or < 0.01). These results suggest that valsartan attenuates neointimal hyperplasiain balloon-injured rat aortic arteries through activation of the ACE2-angiotensin-(1-7)-Mas axis as well as inhibition of the ACE-angiotensin II-AT1 and p-ERK pathways.


Subject(s)
Aorta , Gene Expression Regulation, Enzymologic/drug effects , MAP Kinase Signaling System/drug effects , Peptidyl-Dipeptidase A/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Tunica Intima , Valsartan/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Aorta/enzymology , Aorta/injuries , Aorta/pathology , Hyperplasia , Male , Proto-Oncogene Mas , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Tunica Intima/enzymology , Tunica Intima/injuries , Tunica Intima/pathology
3.
Immunopharmacol Immunotoxicol ; 36(6): 397-403, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25204222

ABSTRACT

OBJECTIVE: To investigated whether CBS3830, a highly selectively inhibitor of p38MAPK, could ameliorate inflammation and intimal hyperplasia in arterialized vein grafts (AVGs). METHODS: Sixty male Sprague-Dawley rats underwent a reversed right jugular vein to common carotid artery interposition graft and were randomly treatment with vehicle (control) or single-dose (3 mg/kg, preoperative) or double-dose (3 mg/kg, preoperative and 4 d postoperative) CBS3830. Twenty rats underwent sham operation. The levels of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) were determined by ELISA. Vein grafts were analyzed by intimal/medial morphometry, proliferating cell nuclear antigen (PCNA) expression, and p38MAPK phosphorylation. RESULTS: TNF-α, IL-1ß, and IL-6 gradually increased then slowly decreased in AVG rats. However, at 4 d and 7 d, TNF-α levels decreased by 37.5% and 29.5% (p = 0.003, 0.05, respectively) in the single-dose CBS3830 group, and by 37.6% and 32.5%, respectively (both p = 0.003) in the double-dose group compared with those of control. IL-1ß levels significantly reduced at 4 d and 14 d in both dosage groups. IL-6 levels significantly reduced at 7 d in both groups. Intima and medial thickening were significantly reduced in both dosage treated groups at 7, 14, and 28 d (all p = 0.000) compared to the controls. Further study showed CBS3830 inhibited p38MAPK phosphorylation and decreased PCNA expression. CONCLUSIONS: CBS3830 significantly decreases inflammation and intimal hyperplasia in AVGs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Proliferation/drug effects , Dibenzocycloheptenes/pharmacology , Jugular Veins/transplantation , Tunica Intima/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Graft Occlusion, Vascular/enzymology , Graft Occlusion, Vascular/immunology , Graft Occlusion, Vascular/pathology , Graft Occlusion, Vascular/prevention & control , Hyperplasia , Immunity, Innate/drug effects , Jugular Veins/enzymology , Jugular Veins/immunology , Jugular Veins/pathology , Male , Proliferating Cell Nuclear Antigen/biosynthesis , Rats, Sprague-Dawley , Tunica Intima/enzymology , Tunica Intima/immunology , Tunica Intima/pathology
4.
Am J Physiol Heart Circ Physiol ; 304(7): H927-34, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23376828

ABSTRACT

We have previously reported that bradykinin relaxes the fetal ductus arteriosus via endothelium-derived hyperpolarizing factor (EDHF) when other naturally occurring relaxants (prostaglandin E2, nitric oxide, and carbon monoxide) are suppressed, but the identity of the agent could not be ascertained. Here, we have examined in the mouse whether hydrogen sulfide (H2S) is a relaxant of the ductus and, if so, whether it may also function as an EDHF. We found in the vessel transcripts for the H2S synthetic enzymes, cystathionine-γ-lyase (CSE) and cystathionine-ß-synthase (CBS), and the presence of these enzymes was confirmed by immunofluorescence microscopy. CSE and CBS were distributed across the vessel wall with the former prevailing in the intimal layer. Both enzymes occurred within the endoplasmic reticulum of endothelial and muscle cells, whereas only CSE was located also in the plasma membrane. The isolated ductus contracted to inhibitors of CSE (d,l-propargylglycine, PPG) and CBS (amino-oxyacetic acid), and PPG contraction was attenuated by removal of the endothelium. EDHF-mediated bradykinin relaxation was curtailed by both PPG and amino-oxyacetic acid, whereas the relaxation to sodium nitroprusside was not affected by either treatment. The H2S donor sodium hydrogen sulfide (NaHS) was also a potent, concentration-dependent relaxant. We conclude that the ductus is endowed with a H2S system exerting a tonic relaxation. In addition, H2S, possibly via an overriding CSE source, qualifies as an EDHF. These findings introduce a novel vasoregulatory mechanism into the ductus, with implications for antenatal patency of the vessel and its transitional adjustments at birth.


Subject(s)
Ductus Arteriosus/metabolism , Endothelium-Dependent Relaxing Factors/metabolism , Hydrogen Sulfide/metabolism , Vasodilation , Alkynes/pharmacology , Aminooxyacetic Acid/pharmacology , Animals , Bradykinin/pharmacology , Cell Membrane/metabolism , Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Ductus Arteriosus/enzymology , Ductus Arteriosus/physiology , Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Mice , Mice, Inbred C57BL , Muscle Cells/metabolism , Nitroprusside/pharmacology , RNA, Messenger/biosynthesis , Transcription, Genetic , Tunica Intima/cytology , Tunica Intima/enzymology , Tunica Intima/metabolism
5.
Circ Res ; 109(11): 1230-9, 2011 11 11.
Article in English | MEDLINE | ID: mdl-21980125

ABSTRACT

RATIONALE: Adenosine monophosphate-activated protein kinase (AMPK), a metabolic and redox sensor, is reported to suppress cell proliferation of nonmalignant and tumor cells. Whether AMPKα alters vascular neointima formation induced by vascular injury is unknown. OBJECTIVE: The aim of this study was to determine the roles of AMPKα in the development of vascular neointima hyperplasia and to elucidate the underlying mechanisms. METHODS AND RESULTS: Vascular smooth muscle cell (VSMC) proliferation and neointimal hyperplasia were evaluated in cultured VSMCs and wire-injured mouse carotid arteries from wild-type (WT, C57BL/6J), AMPKα2(-/-), and AMPKα1(-/-) mice. Mouse VSMCs derived from aortas of AMPKα2(-/-) mice exhibited increased proliferation compared with either WT or AMPKα1(-/-) VSMCs. Further, deletion of AMPKα2 but not AMPKα1 reduced the level of p27(Kip1), a cyclin-dependent kinase inhibitor, and increased the level of S-phase kinase-associated protein 2 (Skp2), a known E3 ubiquitin ligase for p27(Kip1), through activation of p52 nuclear factor kappa B (NF-κB)-2. Moreover, either pharmacological (ie, through compound C) or genetical (ie, through AMPKα2-specific siRNA) inhibition of AMPK decreased p27(Kip1) levels but increased the abundance of Skp2 in human VSMCs. Furthermore, gene silencing of Skp2 reversed the levels of p27(Kip1) and VSMCs proliferation. Finally, neointima formation after mechanical arterial injury was increased in AMPKα2(-/-) but not AMPKα1(-/-) mice. CONCLUSIONS: These findings indicate that deletion of AMPKα2 through p52-Skp2-mediated ubiquitination and degradation of p27(Kip1) accentuates neointimal hyperplasia in response to wire injury.


Subject(s)
AMP-Activated Protein Kinases/deficiency , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , S-Phase Kinase-Associated Proteins/physiology , Tunica Intima/injuries , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/physiology , Animals , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Cell Division , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Deletion , Hyperplasia , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , S-Phase Kinase-Associated Proteins/biosynthesis , S-Phase Kinase-Associated Proteins/genetics , Tunica Intima/enzymology , Tunica Intima/pathology , Ubiquitination , Up-Regulation
6.
Heart Lung Circ ; 22(9): 751-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23523564

ABSTRACT

OBJECTIVE: Following bypass surgery vein grafts undergo a remodelling process that can lead to restenosis and ultimately vein graft failure. Signalling through mitogen activated protein kinases (MAPKs) is a key mechanism involved in vein graft failure. Here, we investigated whether CBS3830 (c-a-i-r biosciences GmbH, Tübingen, Germany), a new highly selectively inhibitor of p38 MAPK, has a significant effect on inhibiting intimal, medial and adventitial hyperplasia. METHODS: Sixty specific pathogen free Sprague Dawley male rats were randomly divided into three groups. The control group with a reversed right jugular vein, which is common to carotid artery interposition graft, was compared with sham-operated, and CBS3830 treated animals. Intimal, medial and adventitia morphometric examinations and expression of proliferating cell nuclear antigen (PCNA) were analysed after one, two and four weeks for vein grafts. RESULTS: Intimal, medial and adventitia thickening in CBS3830 group were significantly lower than in the control group at each time point. Moreover, CBS3830 significantly reduced the phosphorylation of p38 MAPK and PCNA expression compared to the control. CONCLUSION: On the basis of the present work, intima, media and adventitia of saphenous vein grafts undergo vascular remodelling after surgery. The new, highly selective p38 MAPK inhibitor, CBS3830, ameliorates intimal, medial, and adventitial remodelling by varying degrees.


Subject(s)
Coronary Artery Bypass , Graft Occlusion, Vascular/prevention & control , Protein Kinase Inhibitors/pharmacology , Saphenous Vein/enzymology , Tunica Intima/enzymology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adventitia/enzymology , Adventitia/pathology , Adventitia/physiopathology , Animals , Gene Expression Regulation/drug effects , Graft Occlusion, Vascular/enzymology , Graft Occlusion, Vascular/pathology , Graft Occlusion, Vascular/physiopathology , Male , Phosphorylation/drug effects , Proliferating Cell Nuclear Antigen/biosynthesis , Rats , Rats, Sprague-Dawley , Saphenous Vein/pathology , Saphenous Vein/physiopathology , Tunica Intima/pathology , Tunica Intima/physiopathology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Circulation ; 123(6): 631-9, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282500

ABSTRACT

BACKGROUND: Microsomal (m) prostaglandin (PG) E2 synthase (S)-1 catalyzes the formation of PGE2 from PGH2, a cyclooxygenase product that is derived from arachidonic acid. Previous studies in mice suggest that targeting mPGES-1 may be less likely to cause hypertension or thrombosis than cyclooxygenase-2-selective inhibition or deletion in vivo. Indeed, deletion of mPGES-1 retards atherogenesis and angiotensin II-induced aortic aneurysm formation. The role of mPGES-1 in the response to vascular injury is unknown. METHODS AND RESULTS: Mice were subjected to wire injury of the femoral artery. Both neointimal area and vascular stenosis were significantly reduced 4 weeks after injury in mPGES-1 knockout mice compared with wild-type controls (65.6 ± 5.7 versus 37.7 ± 5.1 × 10³ pixel area and 70.5 ± 13.4% versus 47.7 ± 17.4%, respectively; P < 0.01). Induction of tenascin-C, a proproliferative and promigratory extracellular matrix protein, after injury was attenuated in the knockouts. Consistent with in vivo rediversion of PG biosynthesis, mPGES-1-deleted vascular smooth muscle cells generated less PGE2 but more PGI2 and expressed reduced tenascin-C compared with wild-type cells. Both suppression of PGE2 and augmentation of PGI2 attenuate tenascin-C expression and vascular smooth muscle cell proliferation and migration in vitro. CONCLUSIONS: Deletion of mPGES-1 in mice attenuates neointimal hyperplasia after vascular injury, in part by regulating tenascin-C expression. This raises for consideration the therapeutic potential of mPGES-1 inhibitors as adjuvant therapy for percutaneous coronary intervention.


Subject(s)
Femoral Artery/enzymology , Femoral Artery/injuries , Intramolecular Oxidoreductases/metabolism , Microsomes/enzymology , Animals , Cell Movement , Cell Proliferation , Constriction, Pathologic/enzymology , Constriction, Pathologic/pathology , Dinoprostone/biosynthesis , Epoprostenol/biosynthesis , Intramolecular Oxidoreductases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Prostaglandin H2/metabolism , Prostaglandin-E Synthases , Tenascin/metabolism , Tunica Intima/enzymology , Tunica Intima/injuries , Tunica Intima/pathology
8.
Circulation ; 123(6): 621-30, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282495

ABSTRACT

Background- The molecular mechanisms that determine the localized formation of thin-capped atheromata in the coronary arteries remain unknown. This study tested the hypothesis that low endothelial shear stress augments the expression of matrix-degrading proteases and thereby promotes the formation of thin-capped atheromata. Methods and Results- Intravascular ultrasound-based, geometrically correct 3-dimensional reconstruction of the coronary arteries of 12 swine was performed in vivo 23 weeks after initiation of diabetes mellitus and a hyperlipidemic diet. Local endothelial shear stress was calculated in plaque-free subsegments of interest (n=142) with computational fluid dynamics. At week 30, the coronary arteries (n=31) were harvested and the same subsegments were identified. The messenger RNA and protein expression and elastolytic activity of selected elastases and their endogenous inhibitors were assessed. Subsegments with low preceding endothelial shear stress at week 23 showed reduced endothelial coverage, enhanced lipid accumulation, and intense infiltration of activated inflammatory cells at week 30. These lesions showed increased expression of messenger RNAs encoding matrix metalloproteinase-2, -9, and -12, and cathepsins K and S relative to their endogenous inhibitors and increased elastolytic activity. Expression of these enzymes correlated positively with the severity of internal elastic lamina fragmentation. Thin-capped atheromata developed in regions with lower preceding endothelial shear stress and had reduced endothelial coverage, intense lipid and inflammatory cell accumulation, enhanced messenger RNA expression and elastolytic activity of MMPs and cathepsins, and severe internal elastic lamina fragmentation. Conclusions- Low endothelial shear stress induces endothelial discontinuity and accumulation of activated inflammatory cells, thereby augmenting the expression and activity of elastases in the intima and shifting the balance with their inhibitors toward matrix breakdown. Our results provide new insight into the mechanisms of regional formation of plaques with thin fibrous caps.


Subject(s)
Coronary Vessels/enzymology , Endothelium, Vascular/enzymology , Extracellular Matrix/metabolism , Matrix Metalloproteinases/biosynthesis , Plaque, Atherosclerotic/enzymology , Shear Strength , Animals , Cathepsin K/biosynthesis , Cathepsins/biosynthesis , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/enzymology , Coronary Artery Disease/physiopathology , Coronary Vessels/diagnostic imaging , Coronary Vessels/physiopathology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Dietary Fats/metabolism , Endothelium, Vascular/physiopathology , Enzyme Induction , Male , Stress, Mechanical , Swine , Tunica Intima/enzymology , Tunica Intima/physiopathology , Ultrasonography
9.
Circ Res ; 107(10): 1265-74, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20884880

ABSTRACT

RATIONALE: Mechanical stress plays an important role in proliferation of venous smooth muscle cells (SMCs) in neointima, a process of formation that contributes to failure of vein grafts. However, it is unknown what intracellular growth signal leads to proliferation of venous SMCs. OBJECTIVE: The objective of this study is to identify mechanisms of mechanical stretch on neointima formation. METHODS AND RESULTS: By a microarray analysis, we found that mechanical cyclic stretch (15% elongation) stimulated the transcription of SGK-1 (serum-, glucocorticoid-regulated kinase-1). Mechanical stretch-induced SGK-1 mRNA expression was blocked by actinomycin D. The mechanism for the SGK-1 expression involved MEK1 but not p38 or JNK signaling pathway. SGK-1 activation in response to stretch is blocked by insulin-like growth factor (IGF)-1 receptor inhibitor and mammalian target of rapamycin complex (mTORC)2 inhibitor (Ku-0063794) but not mTORC1 inhibitor (rapamycin). Mechanical stretch-induced bromodeoxyuridine incorporation was reduced by 83.5% in venous SMCs isolated from SGK-1 knockout mice. In contrast, inhibition of Akt, another downstream signal of PI3K resulted in only partial inhibition of mechanical stretch-induced proliferation of venous SMCs. Mechanical stretch also induced phosphorylation and nuclear exportation of p27(kip1), whereas knockout of SGK-1 attenuated this effect of mechanical stretch on p27(kip1). In vivo, we found that placement of a vein graft into artery increased SGK-1 expression. Knockout of SGK-1 effectively prevented neointima formation in vein graft. There is significant lower level of p27(kip1) located in the nucleus of neointima cells in SGK-1 knockout mice compared with that of wild-type vein graft. In addition, we also found that wire injury of artery or growth factors in vitro increased expression of SGK-1. CONCLUSIONS: These results suggest that SGK-1 is an injury-responsive kinase that could mediate mechanical stretch-induced proliferation of vascular cells in vein graft, leading to neointima formation.


Subject(s)
Cell Proliferation , Immediate-Early Proteins/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tunica Intima/enzymology , Animals , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Models, Animal , Enzyme Activation , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , MAP Kinase Kinase 1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Stress, Mechanical , Time Factors , Tissue Culture Techniques , Trans-Activators/metabolism , Transcription Factors , Transfection , Tunica Intima/drug effects , Tunica Intima/pathology , Veins/enzymology , Veins/pathology , Veins/transplantation
10.
J Surg Res ; 173(2): 224-31, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21777927

ABSTRACT

BACKGROUND: Hormone replacement therapy increases intimal hyperplasia (IH) following vascular intervention. Matrix metalloproteinases (MMPs) play a role in IH development. We have shown estrogen up-regulates MT1-MMP expression, a transmembrane protein that activates MMP-2, and increases vascular smooth muscle cell (VSMC) collagen invasion via increased MMP-2 activity. Here we hypothesize inhibition of MT1-MMP will prevent hormonally-stimulated increased MMP-2 activation and the downstream cellular processes of IH pathogenesis. METHODS: VSMCs from a postmenopausal donor were transfected with MT1-MMP or negative control siRNAs, treated with estrogen (Est), analyzed by q-PCR, Western blot, zymography, migration, invasion, and proliferation assays. RESULTS: Est treatment of MT1-MMP silenced cells still resulted in increased MT1-MMP expression (C = 41% ± 4%; Est = 52% ± 2%; P < 0.05). Silencing of MT1-MMP decreased basal MMP-2 activity (nonsilenced = 100%; MT1-silenced = 87% ± 3%; P < 0.05) but had no effect on basal invasion or proliferation. Est treatment of MT1-MMP silenced cells still resulted in increased MMP-2 activity (C = 87% ± 3%; Est = 101% ± 4%; P < 0.05) and invasion (C = 89% ± 6%; Est = 109% ± 3%; P < 0.05) compared with MT1-MMP silenced control cells. However, silencing of MT1-MMP did inhibit Est- and serum-stimulated proliferation (C = 106% ± 18%; Est = 104% ± 16%; FBS = 121% ± 24%; P = NS). CONCLUSION: Silencing of MT1-MMP in aged VSMCs results in impaired but not complete inhibition of basal and Est-stimulated increases in MMP-2 activity. Other mechanisms appear to be playing a role in hormonally-regulated cellular processes of IH pathogenesis. Future studies will target other signaling cascades, with the goal of identifying mechanisms responsible for hormonally-modulated unbalanced MMPs. In vivo manipulation of the expression patterns of MT1-MMP will be examined for the prevention of IH in animal models of vascular disease.


Subject(s)
Estrogens/metabolism , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Muscle, Smooth, Vascular/enzymology , Postmenopause/metabolism , Tunica Intima/enzymology , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen Type IV , Estrogen Replacement Therapy , Female , Humans , Hyperplasia/enzymology , Hyperplasia/etiology , Middle Aged , RNA Interference , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
11.
J Surg Res ; 178(1): 40-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-21962816

ABSTRACT

BACKGROUND: Intimal hyperplasia remains the principal lesion in the development of restenosis after vessel wall injury. G-protein coupled receptors are involved in smooth muscle cell proliferation but the role of Gßγ in arterial intimal hyperplasia has not been well defined. The aim of this study is to characterize the expression of Gßγ G-proteins in the developing intimal hyperplasia in a murine model and the impact of disruption of Gßγ signaling on intimal hyperplasia development. METHODS: The murine femoral wire injury model was employed. Specimens were perfusion-fixed and sections were stained with H&E and Movat's stains such that morphometry could be performed using an Image-Pro system. Additional specimens of femoral artery were also harvested and snap frozen for Western blotting for the Gßγ expression and for Western blotting and zymography to allow for the study of gelatinase and plasminogen activator expression and activation. Contralateral vessels were used as controls. Additional vessels were immersed in pluronic gel containing an adenovirus with the Gßγ inhibitor ßARK(CT). RESULTS: The injured femoral arteries developed intimal hyperplasia, while sham vessels did not produce such a response. Cell proliferation peaked at 3-5 d and cell migration at 7 d after injury. There was a marked time-dependent increase in Gßγ over the 28 d following injury. Inhibition of Gßγ with ßARK(CT) inhibited cell proliferation, cell migration and the development of intimal hyperplasia. Inhibition of Gßγ decreased peak uPA activity and expression without increasing early PAI-1 activity and expression. Inhibition of Gßγ reduced peak MMP-2 activity at d 1 but not at d 7 and also reduced peak MMP-9 activity at d 3. Protein expression for both MMP-2 and MMP-9 was also transiently decreased. There were no changes in TIMP-1 and TIMP-2 expression and activity. CONCLUSIONS: These data demonstrate a time-dependent increase in Gßγ G-protein expression following wire injury in the mouse. Inhibition of Gßγ alters cell proliferation and migration with associated changes in MMP-2, MMP-9, and uPA expression and activity.


Subject(s)
Femoral Artery/enzymology , Femoral Artery/injuries , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Peptide Hydrolases/metabolism , Animals , Apoptosis/physiology , Disease Models, Animal , Femoral Artery/pathology , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Hyperplasia/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Peptides/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/physiology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tunica Intima/enzymology , Tunica Intima/injuries , Tunica Intima/pathology
12.
Arterioscler Thromb Vasc Biol ; 31(1): 110-6, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20947825

ABSTRACT

OBJECTIVE: To determine whether increased 12/15-lipoxygenase (12/15LO) expression in vivo enhances neointimal formation in response to injury. METHODS AND RESULTS: 12/15LO expression in the vessel wall is increased in animal models of metabolic syndrome and diabetes mellitus. Increased expression of 12/15LO enhances cultured vascular smooth muscle cell (VSMC) proliferation, an effect mediated by the helix-loop-helix factor inhibitor of differentiation 3 (Id3). Carotid endothelial denudation was performed on apolipoprotein (Apo) E(-/-), ApoE(-/-)/12/15LO(-/-), C57BL/6, and 12/15LO-overexpressing transgenic mice. ApoE(-/-)/12/15LO(-/-) mice had attenuated and 12/15LO-overexpressing transgenic mice had enhanced neointimal formation compared with control mice. 12/15LO-overexpressing transgenic mice had greater postinjury carotid Id3 and Ki-67 expression, cell number, and fibronectin deposition compared with C57BL/6 mice. Loss of 12/15LO attenuated proliferation of cultured ApoE(-/-) VSMCs, whereas 12/15LO overexpression induced VSMC proliferation. Loss of Id3 enhanced immunoglobulin trascription factor (ITF)-2b binding to and activation of the p21(cip1) promoter and abrogated 12/15LO-induced VSMC proliferation. CONCLUSIONS: To our knowledge, these data are the first demonstration that increased expression of 12/15LO in the vessel wall enhances Id3-dependent cell proliferation, fibronectin deposition, and neointimal formation in response to injury. Results identify p21(cip1) as a potential target of the 12/15LO-Id3 pathway and suggest that modulation of this pathway may have therapeutic implications for targeting the increased risk of restenosis in patients with diabetes.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Carotid Artery Injuries/enzymology , Cell Proliferation , Fibronectins/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Tunica Intima/enzymology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites , Carotid Artery Injuries/pathology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Hyperplasia , Inhibitor of Differentiation Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Promoter Regions, Genetic , Time Factors , Transcription Factor 4 , Tunica Intima/pathology
13.
J Vasc Res ; 48(1): 43-51, 2011.
Article in English | MEDLINE | ID: mdl-20606470

ABSTRACT

BACKGROUND: Extracellular matrix deposition is the main factor inducing stenotic lesions in arterial grafts. Lysyl oxidases (LOX) play a key role in stabilizing collagen and elastin. OBJECTIVE: To examine the repair response to arterial allografts in terms of LOX expression and collagen/elastin deposition using LOX inhibitors. METHODS: Lewis/Fisher-344 rats were used as donors/recipients. Donor segments were grafted to the right iliac artery of recipients and retrieved 14/30 (short-term) or 90/180 days (long-term) after surgery. One group of animals was injected with a potent irreversible LOX inhibitor daily for 30 days. RESULTS: Intimal hyperplasia increased in thickness until 90/180 days postsurgery. Elastin showed great expression in the neointima at 14/30 days and in the media at 90/180 days. LOX/LOXL1 were similarly expressed in the arterial wall during the first month. In the long term, their overexpression was confined to neointimal layers. At 14 days, collagen types I/III were identified in the grafts. The neointima acquired collagen I over time. In the group of animal treated with the LOX inhibitor, intimal hyperplasia was significantly inhibited. CONCLUSION: LOX were overexpressed in late stages of intimal hyperplasia in the allografts. LOX inhibitors prevented the development of the neointimal layer, such that their modulation could reduce the excessive extracellular matrix deposition that leads to stenosis.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Iliac Artery/enzymology , Iliac Artery/transplantation , Neointima/enzymology , Animals , Collagen/metabolism , Extracellular Matrix/enzymology , Extracellular Matrix/pathology , Female , Graft Survival/physiology , Hyperplasia , Iliac Artery/pathology , Neointima/pathology , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Transplantation, Homologous , Tropoelastin/metabolism , Tunica Intima/enzymology , Tunica Intima/pathology
14.
Mol Cell Biochem ; 347(1-2): 209-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21052790

ABSTRACT

The development of hypercholesterolemia is a multifactorial process in which elevated plasma cholesterol levels play a central role. This study analyzed the variability of the expression and activity of myosin light chain kinase (MLCK) and endothelial permeability in the artery wall of rabbits after feeding the animals with a normal or a high-cholesterol diet. Hypercholesterolemia was induced by a high-cholesterol diet for 4 weeks. Aortas were removed and analyzed for endothelial permeability and MLCK expression. Samples of the arterial media were analyzed for MLCK activity and expression. A selective MLCK inhibitor 1-(5-iodonaphthalene-1-sulfonyl)-1H-hexahydro-1,4-diazepine hydrochloride (ML7) were used in hypercholesterolemia rabbit (1 mg/kg body weight). The aortas of high-cholesterol diet rabbits showed an increase in MLCK expression and activity (nearly threefold compare with control) as well as endothelial permeability. ML7 inhibit MLC phosphorylation and MLCK activity (nearly twofold compare with control) and endothelial permeability stimulated by cholesterol. These results indicate for the first time that hypercholesterolemia may be associated with MLCK expression and activity through which endothelial permeability is increased.


Subject(s)
Aorta/enzymology , Aorta/physiopathology , Capillary Permeability/physiology , Hypercholesterolemia/enzymology , Myosin-Light-Chain Kinase/metabolism , Tunica Intima/enzymology , Tunica Intima/physiopathology , Animals , Aorta/drug effects , Aorta/pathology , Azepines/pharmacology , Blotting, Western , Capillary Permeability/drug effects , Hypercholesterolemia/pathology , Hypercholesterolemia/physiopathology , Male , Naphthalenes/pharmacology , Phosphorylation/drug effects , Rabbits , Tunica Intima/drug effects , Tunica Intima/pathology
15.
Circ Res ; 105(10): 965-72, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19797175

ABSTRACT

RATIONALE: Fatty acid nitroalkenes are endogenously generated electrophilic byproducts of nitric oxide and nitrite-dependent oxidative inflammatory reactions. Existing evidence indicates nitroalkenes support posttranslational protein modifications and transcriptional activation that promote the resolution of inflammation. OBJECTIVE: The aim of this study was to assess whether in vivo administration of a synthetic nitroalkene could elicit antiinflammatory actions in vivo using a murine model of vascular injury. METHODS AND RESULTS: The in vivo administration (21 days) of nitro-oleic acid (OA-NO(2)) inhibited neointimal hyperplasia after wire injury of the femoral artery in a murine model (OA-NO(2) treatment resulted in reduced intimal area and intima to media ratio versus vehicle- or oleic acid (OA)-treated animals,P<0.0001). Increased heme oxygenase (HO)-1 expression accounted for much of the vascular protection induced by OA-NO(2) in both cultured aortic smooth muscle cells and in vivo. Inhibition of HO by Sn(IV)-protoporphyrin or HO-1 small interfering RNA reversed OA-NO(2)-induced inhibition of platelet-derived growth factor-stimulated rat aortic smooth muscle cell migration. The upregulation of HO-1 expression also accounted for the antistenotic actions of OA-NO(2) in vivo, because inhibition of neointimal hyperplasia following femoral artery injury was abolished in HO-1(-/-) mice (OA-NO(2)-treated wild-type versus HO-1(-/-) mice, P=0.016). CONCLUSIONS: In summary, electrophilic nitro-fatty acids induce salutary gene expression and cell functional responses that are manifested by a clinically significant outcome, inhibition of neointimal hyperplasia induced by arterial injury.


Subject(s)
Femoral Artery/enzymology , Femoral Artery/injuries , Heme Oxygenase (Decyclizing)/biosynthesis , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Tunica Intima/enzymology , Animals , Cell Movement/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/metabolism , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitro Compounds/metabolism , Oleic Acids/metabolism , Oxidation-Reduction/drug effects , Platelet-Derived Growth Factor/pharmacology , Rats , Up-Regulation/drug effects
16.
Circ Res ; 104(1): 69-78, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-19023129

ABSTRACT

Bcr is a serine/threonine kinase activated by platelet-derived growth factor that is highly expressed in the neointima after vascular injury. Here, we demonstrate that Bcr is an important mediator of angiotensin (Ang) II and platelet-derived growth factor-mediated inflammatory responses in vascular smooth muscle cells (VSMCs). Among transcription factors that might regulate Ang II-mediated inflammatory responses we found that ligand-mediated peroxisome proliferator-activated receptor (PPAR)gamma transcriptional activity was significantly decreased by Ang II. Ang II increased Bcr expression and kinase activity. Overexpression of Bcr significantly inhibited PPARgamma activity. In contrast, knockdown of Bcr using Bcr small interfering RNA and a dominant-negative form of Bcr (DN-Bcr) reversed Ang II-mediated inhibition of PPARgamma activity significantly, suggesting the critical role of Bcr in Ang II-mediated inhibition of PPARgamma activity. Point-mutation and in vitro kinase analyses showed that PPARgamma was phosphorylated by Bcr at serine 82. Overexpression of wild-type Bcr kinase did not inhibit ligand-mediated PPARgamma1 S82A mutant transcriptional activity, indicating that Bcr regulates PPARgamma activity via S82 phosphorylation. DN-Bcr and Bcr small interfering RNA inhibited Ang II-mediated nuclear factor kappaB activation in VSMCs. DN-PPARgamma reversed DN-Bcr-mediated inhibition of nuclear factor kappaB activation, suggesting that PPARgamma is downstream from Bcr. Intimal proliferation in low-flow carotid arteries was decreased in Bcr knockout mice compared with wild-type mice, suggesting the critical role of Bcr kinase in VSMC proliferation in vivo, at least in part, via regulating PPARgamma/nuclear factor kappaB transcriptional activity.


Subject(s)
Angiotensin II/physiology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , PPAR gamma/antagonists & inhibitors , Proto-Oncogene Proteins c-bcr/physiology , Angiotensin II/pharmacology , Animals , Enzyme Activation , Mice , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NF-kappa B/genetics , NF-kappa B/physiology , PPAR gamma/agonists , PPAR gamma/physiology , Phosphorylation , Phosphoserine/metabolism , Platelet-Derived Growth Factor/pharmacology , Point Mutation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcr/antagonists & inhibitors , Proto-Oncogene Proteins c-bcr/deficiency , Proto-Oncogene Proteins c-bcr/genetics , RNA, Small Interfering/pharmacology , Rats , Recombinant Fusion Proteins/physiology , Tunica Intima/enzymology , Tunica Intima/pathology , Vasculitis/physiopathology
17.
Circ Res ; 104(5): 688-98, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19168437

ABSTRACT

The migration of vascular smooth muscle cells (VSMCs) plays an essential role during the development of atherosclerosis and restenosis. Extensive studies have implicated the importance of extracellular matrix (ECM)-degrading proteinases in VSMC migration. A recently described family of proteinases, a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTs), is capable of degrading vascular ECM proteins. Here, we sought to determine whether ADAMTS-7 is involved in VSMC migration and neointima formation in response to vascular injury. ADAMTS-7 protein accumulated preferentially in neointima of the carotid artery wall after balloon injury. In primary VSMCs, ADAMTS-7 level was enhanced by the proinflammatory cytokine tumor necrosis factor alpha and growth factor platelet-derived growth factor-BB. ADAMTS-7 overexpression greatly accelerated and small interfering RNA knockdown markedly retarded VSMC migration/invasion in vitro. In addition, luminal delivery of ADAMTS-7 adenovirus to carotid arteries exacerbated intimal thickening nearly sixfold 7 days after injury. Conversely, perivascular administration of ADAMTS-7 small interfering RNA but not scramble small interfering RNA to injured arteries attenuated intimal thickening by 50% at 14 days after injury. Furthermore, ADAMTS-7 mediated degradation of the vascular ECM cartilage oligomeric matrix protein (COMP) in injured vessels. Replenishing COMP circumvented the promigratory effect of ADAMTS-7 on VSMCs. Enforced expression of COMP significantly suppressed VSMC migration and neointima formation postinjury, which indicates that ADAMTS-7 facilitated intimal hyperplasia through degradation of inhibitory matrix protein COMP. ADAMTS-7 may therefore serve as a novel therapeutic target for atherosclerosis and postangioplasty restenosis.


Subject(s)
ADAM Proteins/metabolism , Carotid Artery Injuries/enzymology , Cell Movement , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Tunica Intima/enzymology , ADAM Proteins/genetics , ADAMTS7 Protein , Animals , Becaplermin , Carotid Arteries/enzymology , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/pathology , Catheterization/adverse effects , Cells, Cultured , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Hyperplasia , Male , Matrilin Proteins , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Transcription Factor AP-1/metabolism , Transduction, Genetic , Tumor Necrosis Factor-alpha/metabolism , Tunica Intima/pathology
18.
Circ Res ; 104(2): 219-27, 5p following 227, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19038866

ABSTRACT

The role of apurinic/apyrimidinic endonuclease-1/redox factor-1 (Ref-1) in vascular smooth muscle cells has yet to be clearly elucidated. Therefore, we attempted to determine the roles of Ref-1 in the migration induced by platelet-derived growth factor (PDGF)-BB and in its signaling in rat aortic smooth muscle cells (RASMCs). Cellular migration, superoxide (O(2)(-*)) production, Rac-1 activity, and neointima formation were determined in cells transfected with adenoviruses encoding for Ref-1 (AdRef-1) and small interference RNA of Ref-1. Overexpression of Ref-1 induced by treatment with RASMCs coupled with AdRef-1 inhibited the migration induced by PDGF-BB. PDGF-BB also increased the phosphorylation of the PDGFbeta receptor, spleen tyrosine kinase (Syk), mitogen-activated protein kinase, and heat shock protein 27, but these increases were significantly inhibited by AdRef-1 treatment. PDGF-BB increased O(2)(-*) production and Rac-1 activity, and these were diminished in cells transfected with AdRef-1. In contrast, RASMC migration, phosphorylation of Syk and O(2)(-*) production in response to PDGF-BB were increased by the knock down of Ref-1 with small interference RNA. The phosphorylation of PDGFbeta receptor in response to PDGF-BB was inhibited completely by the Syk inhibitor and was partly attenuated by a NADPH oxidase inhibitor. PDGF-BB increased the sprout outgrowth of the aortic ring ex vivo, which was inhibited in the AdRef-1-infected RASMCs as compared with the controls. Balloon injury-induced neointimal formation was significantly attenuated by the gene transfer of AdRef-1. These results indicate that Ref-1 inhibits the PDGF-mediated migration signal via the inhibition of reactive oxygen species-mediated Syk activity in RASMCs.


Subject(s)
Cell Movement , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/enzymology , Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Superoxides/metabolism , Tunica Intima/enzymology , Adenoviridae/genetics , Animals , Aorta/enzymology , Becaplermin , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cell Proliferation , Cells, Cultured , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Genetic Vectors , Humans , Hyperplasia , Male , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neovascularization, Physiologic , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Syk Kinase , Transduction, Genetic , Transfection , Tunica Intima/pathology , rac1 GTP-Binding Protein
19.
Circ Res ; 104(7): 842-50, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19229058

ABSTRACT

Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are important pathogenic mechanisms in atherosclerosis and restenosis after vascular injury. In this study, we investigated the effects of beta-lapachone (betaL) (3,4-Dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione), which is a potent antitumor agent that stimulates NAD(P)H:quinone oxidoreductase (NQO)1 activity, on neointimal formation in animals given vascular injury and on the proliferation of VSMCs cultured in vitro. betaL significantly reduced the neointimal formation induced by balloon injury. betaL also dose-dependently inhibited the FCS- or platelet-derived growth factor-induced proliferation of VSMCs by inhibiting G(1)/S phase transition. betaL increased the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase 1 in rat and human VSMCs. Chemical inhibitors of AMPK or dominant-negative AMPK blocked the betaL-induced suppression of cell proliferation and the G(1) cell cycle arrest, in vitro and in vivo. The activation of AMPK in VSMCs by betaL is mediated by LKB1 in the presence of NQO1. Taken together, these results show that betaL inhibits VSMCs proliferation via the NQO1 and LKB1-dependent activation of AMPK. These observations provide the molecular basis that pharmacological stimulation of NQO1 activity is a new therapy for the treatment of vascular restenosis and/or atherosclerosis which are caused by proliferation of VSMCs.


Subject(s)
Carotid Artery Injuries/drug therapy , Carotid Stenosis/drug therapy , Cell Proliferation/drug effects , Enzyme Activators/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/pharmacology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Carotid Stenosis/enzymology , Carotid Stenosis/pathology , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Activators/toxicity , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Hyperplasia , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/genetics , Naphthoquinones/toxicity , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Retinoblastoma Protein/metabolism , Secondary Prevention , Time Factors , Tumor Suppressor Protein p53/metabolism , Tunica Intima/drug effects , Tunica Intima/enzymology , Tunica Intima/pathology
20.
J Vasc Interv Radiol ; 22(9): 1321-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21515073

ABSTRACT

PURPOSE: To evaluate the efficacy of an activin receptor-like kinase (ALK) 5 inhibitor, IN-1233, for the prevention of neointimal hyperplasia after bare stent placement in a rat common iliac artery (CIA) model. MATERIALS AND METHODS: All experiments were approved by the committee of animal research. A self-expanding metallic bare stent (2 mm × 6 mm) was inserted into the left CIA of 26 Sprague-Dawley male rats (300-360 g) under fluoroscopic guidance. IN-1233 was injected via the intraperitoneal route daily in 13 rats for 8 weeks after stent placement (group A); the other 13 rats underwent stent placement only (group B). Angiography was performed immediately and 4 weeks and 8 weeks after stent placement. Rats were sacrificed at 8 weeks after stent placement, and histologic findings were obtained. The neointimal area (NA), percentage of neointimal hyperplasia (%NH), and neointimal-to-medial area ratio (N/M) were assessed and compared between the two groups. RESULTS: Stent placement was technically successful. In 25 rats, arteries with stent placement were angiographically patent, whereas 1 rat in group B had an occlusion. The NA (0.31 mm(2) ± 0.09 vs 0.56 mm(2) ± 0.17; P < .001), the %NH (26.16% ± 8.75 vs 44.71% ± 17.75; P < .001) and the N/M (1.93 ± 0.77 vs 4.77 ± 2.26; P < .001) were significantly decreased in group A compared with group B. CONCLUSIONS: IN-1233 was shown in this study to be effective for the prevention of neointimal hyperplasia after bare metallic stent placement in a rat CIA model.


Subject(s)
Benzamides/pharmacology , Endovascular Procedures/instrumentation , Iliac Artery/drug effects , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Stents , Tunica Intima/drug effects , Animals , Benzamides/administration & dosage , Endovascular Procedures/adverse effects , Hyperplasia , Iliac Artery/diagnostic imaging , Iliac Artery/enzymology , Iliac Artery/pathology , Injections, Intraperitoneal , Male , Metals , Prosthesis Design , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Quinolines/administration & dosage , Radiography , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Time Factors , Tunica Intima/diagnostic imaging , Tunica Intima/enzymology , Tunica Intima/pathology
SELECTION OF CITATIONS
SEARCH DETAIL