Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
Mol Med Rep ; 21(4): 1959-1964, 2020 04.
Article in English | MEDLINE | ID: mdl-32319637

ABSTRACT

Intimal hyperplasia (IH) is a pathologic process that leads to restenosis after treatment for peripheral arterial disease. Heat shock protein 90 (HSP90) is a molecular chaperone that regulates protein maturation. Activation of HSP90 results in increased cell migration and proliferation. 17­N­allylamino­17­demethoxygeldanamycin (17­AAG) and 17­dimethylaminoethylamino­17­demethoxygeldanamycin (17­DMAG) are low toxicity Food and Drug Association approved HSP90 inhibitors. The current study hypothesized that HSP90 inhibition was predicted to reduce vascular smooth muscle cell (VSMC) migration and proliferation. In addition, localized HSP90 inhibition may inhibit post­angioplasty IH formation. For proliferation, VSMCs were treated with serum­free media (SFM), 17­DMAG or 17­AAG. The selected proliferative agents were SFM, platelet derived growth factor (PDGF) or fibronectin. After three days, proliferation was measured. For migration, VSMCs were treated with SFM, 17­AAG or 17­DMAG with SFM, PDGF or fibronectin as chemoattractants. Balloon injury to the carotid artery was performed in rats. The groups included in the present study were the control, saline control, 17­DMAG in 20% pluronic gel delivered topically to the adventitia or intraluminal delivery of 17­DMAG. After 14 days, arteries were fixed and sectioned for morphometric analysis. Data was analyzed using ANOVA or a student's t­test. P<0.05 was considered to indicate a statistically significant difference. The results revealed that 17­AAG and 17­DMAG had no effect on cell viability. PDGF and fibronectin also increased VSMC proliferation and migration. Furthermore, both 17­AAG and 17­DMAG decreased cell migration and proliferation in all agonists. Topical adventitial treatment with 17­DMAG after balloon arterial injury reduced IH. HSP90 inhibitors suppressed VSMC proliferation and migration without affecting cell viability. Topical treatment with a HSP90 inhibitor (DMAG) decreased IH formation after arterial injury. It was concluded that 17­DMAG may be utilized as an effective therapy to prevent restenosis after revascularization.


Subject(s)
Angioplasty/adverse effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Tunica Intima/pathology , Animals , Benzoquinones/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Fibronectins/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Humans , Hyperplasia , Lactams, Macrocyclic/pharmacology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Platelet-Derived Growth Factor/pharmacology , Rats, Sprague-Dawley
2.
J Cell Biochem ; 121(10): 4154-4165, 2020 10.
Article in English | MEDLINE | ID: mdl-32348623

ABSTRACT

The thrombospondins (TSPs), multifunctional matricellular proteins, are known mediators of endothelial cell (EC) angiogenesis and apoptosis. TSP-1, an antiangiogenic molecule, is important in the progression of vascular disease, in part by inducing EC apoptosis. TSP-2, although less studied, also induces EC apoptosis and inhibits angiogenesis. The effects of TSP-5 are largely unexplored in ECs, but TSP-5 is believed to be protective against arterial disease. Statin drugs have been shown to have beneficial pleiotropic effects, including decreasing EC apoptosis, increasing angiogenesis, and blocking TSP signaling. We hypothesized TSP-5 will be proangiogenic and antiapoptotic, and statin pretreatment would reverse the proapoptotic and antiangiogenic phenotype of TSP-1 and TSP-2. ECs were exposed to serum-free medium, TSP-1, TSP-2, or TSP-5 with or without fluvastatin pretreatment. Quantitative real-time polymerase chain reaction was performed on 96 apoptosis and 96 angiogenesis-related genes using microfluidic card assays. Angiogenesis was measured using Matrigel assays, while apoptosis was measured by fluorescent caspase assay. TSP-5 suppressed apoptotic genes and had a mixed effect on the angiogenic genes; however, TSP-5 did not alter apoptois but was proangiogenic. Pretreatment with fluvastatin downregulated proapoptotic genes and apoptosis and upregulated proangiogenic genes and angiogenesis. Findings indicate TSP-5 and fluvastatin have a protective effect on ECs, being proangiogenic and reversing the antiangiogenic effects of TSP-1 and TSP-2. In conclusion, TSP-5 and fluvastatin may be beneficial for inducing angiogenesis in the setting of ischemia.


Subject(s)
Apoptosis/drug effects , Cartilage Oligomeric Matrix Protein/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fluvastatin/pharmacology , Neovascularization, Physiologic/drug effects , Protective Agents/pharmacology , Aorta/cytology , Apoptosis/genetics , Cells, Cultured , Down-Regulation/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Physiologic/genetics , Thrombospondin 1/pharmacology , Thrombospondins/pharmacology , Up-Regulation/drug effects
3.
Vasc Endovascular Surg ; 53(5): 379-386, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30982448

ABSTRACT

INTRODUCTION: Oral statins reduce intimal hyperplasia (IH) after arterial injury by only ∼25%. Alternative drug delivery systems have gained attention as carriers for hydrophobic drugs. We studied the effects of simvastatin (free vs hyaluronic acid-tagged polysialic acid-polycaprolactone micelles) on vascular smooth muscle cell (VSMC) migration, VSMC proliferation and intimal hyperplasia. We hypothesized both free and micelle containing simvastatin would inhibit VSMC chemotaxis and proliferation, and local statin treatment would be more effective than oral in reducing IH in rats following carotid balloon injury. METHODS: VSMCs pretreated with free simvastatin (20 minutes or 20 hours) or simvastatin-loaded micelles underwent chemotaxis and proliferation to platelet-derived growth factor. Next, rats that underwent balloon injury of the common carotid artery received statin therapy-intraluminal simvastatin-loaded micelles prior to injury, periadventitial pluronic gel following injury, or combinations of gel, micelle, and oral simvastatin. After 14 days, morphometric analysis determined the -intimal to medial ratio. Findings were compared to controls receiving oral simvastatin or no statin therapy. Statistical analysis was by analysis of variance for the in vitro experiments and a factorial general linear model for the in vivo experiments. RESULTS: The simvastatin-loaded micelles and free simvastatin inhibited VSMC chemotaxis (54%-60%). IH was induced in all injured vessels. Simvastatin in pluronic gel or micelles reduced IH compared to untreated controls (0.208 ± 0.04 or 0.160 ± 0.03 vs 0.350 ± 0.03, respectively); however, neither gel nor simvastatin-loaded micelles were superior to oral statins (0.261 ± 0.03). Addition of oral statins or combining both local therapies did not provide additional benefit. Micelles were the single greatest contributing factor in IH attenuation. CONCLUSIONS: Intraluminally or topically delivered statins reduced IH. The efficacy of single-dose, locally delivered statin alone may lead to novel treatments to prevent IH. The different routes of administration may allow for treatment during endovascular procedures, without the need for systemic therapy.


Subject(s)
Carotid Artery Injuries/drug therapy , Carotid Artery, Common/drug effects , Drug Carriers , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Neointima , Polymers/chemistry , Simvastatin/administration & dosage , Tunica Intima/drug effects , Vascular Remodeling/drug effects , Administration, Oral , Animals , Caproates/chemistry , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery Injuries/physiopathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Carotid Artery, Common/physiopathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Drug Compounding , Humans , Hyaluronic Acid/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Lactones/chemistry , Micelles , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Rats, Sprague-Dawley , Sialic Acids/chemistry , Simvastatin/chemistry , Tunica Intima/metabolism , Tunica Intima/pathology , Tunica Intima/physiopathology
4.
J Surg Res ; 210: 1-7, 2017 04.
Article in English | MEDLINE | ID: mdl-28457315

ABSTRACT

BACKGROUND: Thrombospondin-1 (TSP-1) is functionally important to intimal hyperplasia (IH) development. Statin drugs have beneficial pleiotropic effects, including reduced IH; however, the effect of statins on IH in a TSP-1-independent setting is unknown. HYPOTHESIS: Statins will be less effective in attenuating IH after vascular injury in TSP-1-null (Thbs1-/-) mice compared with wild-type (WT) mice. MATERIALS AND METHODS: Carotid artery ligation was performed on WT and Thbs1-/- mice. Each strain was divided into two groups: no statin control or standard chow containing fluvastatin (10 or 40 mg/kg/d). After 28 d, analysis included morphometric analysis and real-time quantitative reverse transcription polymerase chain reaction on the arteries and enzyme-linked immunosorbent assay on plasma (TSP-1 WT, TSP-2 WT, and Thbs1-/-). Comparisons were made by analysis of variance, with P < 0.05 considered significant. RESULTS: In no statin controls, WT mice had more IH than Thbs1-/- mice (0.46 ± 0.09 versus 0.15 ± 0.04). Fluvastatin reduced IH in the WT (0.46 ± 0.09 versus 0.23 ± 0.06), but not in Thbs1-/- groups (0.15 ± 0.04 versus 0.22 ± 0.07). No difference in IH existed between Thbs1-/- no statin controls and fluvastatin WT and Thbs1-/- groups. Statin dose did not affect IH. TSP-1 plasma levels were increased in fluvastatin WT. TSP-2 levels were decreased in fluvastatin WT and elevated in fluvastatin Thbs1-/-. Fluvastatin had no effect on tissue Thbs1 or Thbs2 gene expression. CONCLUSIONS: TSP-1 is necessary for robust IH after arterial injury. Because fluvastatin had no effect on IH in Thbs1-/-, the data suggest that the statin effect on IH may be largely TSP-1 dependent. Both statins and the presence of TSP-1 affect TSP-1 and TSP-2 plasma levels.


Subject(s)
Carotid Arteries/pathology , Fatty Acids, Monounsaturated/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperplasia/prevention & control , Indoles/therapeutic use , Thrombospondin 1/metabolism , Tunica Intima/pathology , Animals , Biomarkers/metabolism , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hyperplasia/metabolism , Indoles/pharmacology , Male , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/deficiency , Tunica Intima/drug effects , Tunica Intima/metabolism
5.
Vasc Endovascular Surg ; 50(2): 107-18, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26983667

ABSTRACT

Dyslipidemia, more specifically, high-serum low-density lipoproteins and low-serum high-density lipoproteins, are known risk factors for cardiovascular disease. The current clinical treatment of dyslipidemia represents the outcome of a large body of fundamental basic science research on lipids, lipid metabolism, and the effects of different lipids on cellular components of the artery, inflammatory cells, and platelets. In general, lower density lipids activate intracellular pathways to increase local and systemic inflammation, monocyte adhesion, endothelial cell dysfunction and apoptosis, and smooth muscle cell proliferation, resulting in foam cell formation and genesis of atherosclerotic plaque. In contrast, higher density lipids prevent or attenuate atherosclerosis. This article is part 1 of a 2-part review, with part 1 focusing on lipid metabolism and the downstream effects of lipids on the development of atherosclerosis, and part 2 on the clinical treatment of dyslipidemia and the role of these drugs for patients with arterial disease exclusive of the coronary arteries.


Subject(s)
Arteries/metabolism , Atherosclerosis/metabolism , Dyslipidemias/metabolism , Lipid Metabolism , Lipids/blood , Animals , Arteries/pathology , Arteries/physiopathology , Atherosclerosis/etiology , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Biomarkers/blood , Dyslipidemias/complications , Dyslipidemias/pathology , Dyslipidemias/physiopathology , Humans , Inflammation Mediators/blood , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Plaque, Atherosclerotic , Risk Factors , Signal Transduction
7.
Mol Cell Biochem ; 412(1-2): 111-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26728995

ABSTRACT

Thrombospondin-1 (TSP-1) is an important regulator of vascular smooth muscle cell (VSMC) physiology and gene expression. MicroRNAs (microRNA), small molecules that regulate protein translation, have emerged as potent regulators of cell function. MicroRNAs have been shown to be involved in intimal hyperplasia, atherosclerosis, and upregulated in the vasculature in diabetes. The purpose of this study was to identify microRNAs regulated by TSP-1 in vascular smooth muscle cells (VSMCs). Human VSMCs were treated for 6 h with basal media or TSP-1 both supplemented with 0.2% FBS. Cells were then snap frozen and RNA extracted. An Affymetrix GeneChip microRNA array analysis was performed in triplicate on three separate collections. Confirmatory qrtPCR was performed. Data were analyzed by ANOVA or t test, with significance set at p < 0.05. MicroRNAs identified were subjected to KEGG pathway analysis using the DIANA tools miRPath online tool. TSP-1 upregulated 22 microRNAs and downregulated 18 microRNAs in VSMCs (p < 0.05). The most upregulated microRNA was miR-512-3p (45.12 fold). The microRNA most downregulated by TSP-1 was miR-25-5p, which was decreased by 9.61. Of note, five members of the mir-17-92 cluster were downregulated. KEGG analysis revealed that thirty-three cellular signaling pathways were impacted by these microRNAs and that nine pathways were relevant to vascular disease. MicroRNAs regulate protein expression at the level of translation and may represent a significant mechanism by which TSP-1 regulates VSMC function. Several of the microRNAs identified have a role in vascular function. The miR-17-92 cluster family, which was found to exhibit reduced expression in this study, is known to be involved in angiogenesis and vascular function. TSP-1 regulates multiple microRNAs in VSMCs adding a new layer of complexity to TSP-1 regulation of VSMC function.


Subject(s)
MicroRNAs/physiology , Muscle, Smooth, Vascular/metabolism , Thrombospondin 1/physiology , Cells, Cultured , Humans , Muscle, Smooth, Vascular/cytology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
8.
Mol Cell Biochem ; 410(1-2): 85-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26350564

ABSTRACT

UNLABELLED: Dyslipidemia is a risk factor for intimal hyperplasia (IH). Key to IH is vascular smooth muscle cell (VSMC) migration. Thrombospondin-1 (TSP-1) is a matricellular protein that stimulates VSMC migration. HYPOTHESIS: HDL will inhibit and LDL will augment TSP-1-induced VSMC chemotaxis. VSMC chemotaxis will be inhibited by the HDL moiety, S1P, through the S1PR1 receptor, and augmented by the LDL component, LPA, through the LPAR1 receptor. The goal of this study was to determine the effect of HDL and LDL and their receptors on TSP-1-induced VSMC chemotaxis. For VSMC chemotaxis to TSP-1 cells received the following pretreatments: low (25 µg/ml) or optimal (75 µg/ml) concentration of HDL, S1P, optimal (75 µg/ml) or high (175 µg/ml) concentration of LDL, or LPA. For the receptor studies, VSMCs were transfected with siRNA to S1PR1, S1PR3, LPAR1, LPAR2, LPAR3, or a S1PR2 receptor antagonist. The TSP-1-induced chemotaxis results were (1) HDL (25 µg/ml) or LDL (75 µg/ml) exhibited no effect on chemotaxis; (2) HDL (75 µg/ml) inhibited chemotaxis by 50.9 ± 8 % and S1P by 43.4 ± 11.6 %; (3) LDL (175 µg/ml) augmented chemotaxis by 30 ± 10.4 % and LPA by 25.6 ± 12.3 %; (4) S1PR1 and S1PR3 knockdown and S1PR2 antagonist-treated cells augmented chemotaxis; and (5) LPAR1 and LPAR2 knockdown inhibited and LPAR3 knockdown had no effect on chemotaxis. In conclusion, HDL/S1P inhibits, while LDL/LPA stimulates TSP-1-induced VSMC chemotaxis. The HDL/S1P effect is mediated by the S1PR1-3 receptors. The LDL/LPA effects are mediated by the LPAR1 and LPAR2 receptors, but not LPAR3. Therefore, lipids have significant effects on TSP-1-induced VSMC chemotaxis.


Subject(s)
Chemotaxis/drug effects , Dyslipidemias/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Thrombospondin 1/pharmacology , Cells, Cultured , Dyslipidemias/genetics , Dyslipidemias/pathology , Humans , Lipoproteins, HDL/pharmacology , Lipoproteins, LDL/pharmacology , Lysophospholipids/pharmacology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , RNA Interference , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Time Factors , Transfection
9.
Biochem Biophys Res Commun ; 464(4): 1022-1027, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26168731

ABSTRACT

INTRODUCTION: The thrombospondins (TSPs) are matricellular proteins that exert multifunctional effects by binding cytokines, cell-surface receptors and other proteins. TSPs play important roles in vascular pathobiology and are all expressed in arterial lesions. The differential effects of TSP-1, -2, and -5 represent a gap in knowledge in vascular smooth muscle cell (VSMC) physiology. Our objective is to determine if structural differences of the TSPs imparted different effects on VSMC functions critical to the formation of neointimal hyperplasia. We hypothesize that TSP-1 and -2 induce similar patterns of migration, proliferation and gene expression, while the effects of TSP-5 are different. METHODS: Human aortic VSMC chemotaxis was tested for TSP-2 and TSP-5 (1-40 µg/mL), and compared to TSP-1 and serum-free media (SFM) using a modified Boyden chamber. Next, VSMCs were exposed to TSP-1, TSP-2 or TSP-5 (0.2-40 µg/mL). Proliferation was assessed by MTS assay. Finally, VSMCs were exposed to TSP-1, TSP-2, TSP-5 or SFM for 3, 6 or 24 h. Quantitative real-time PCR was performed on 96 genes using a microfluidic card. Statistical analysis was performed by ANOVA or t-test, with p < 0.05 being significant. RESULTS: TSP-1, TSP-2 and TSP-5 at 20 µg/mL all induce chemotaxis 3.1 fold compared to serum-free media. TSP-1 and TSP-2 induced proliferation 53% and 54% respectively, whereas TSP-5 did not. In the gene analysis, overall, cardiovascular system development and function is the canonical pathway most influenced by TSP treatment, and includes multiple growth factors, cytokines and proteases implicated in cellular migration, proliferation, vasculogenesis, apoptosis and inflammation pathways. CONCLUSIONS AND RELEVANCE: The results of this study indicate TSP-1, -2, and -5 play active roles in VSMC physiology and gene expression. Similarly to TSP-1, VSMC chemotaxis to TSP-2 and -5 is dose-dependent. TSP-1 and -2 induces VSMC proliferation, but TSP-5 does not, likely due conservation of N-terminal domains in TSP-1 and -2. In addition, TSP-1, -2 and -5 significantly affect VSMC gene expression; however, little overlap exists in the specific genes altered. This study further delineates TSP-1, -2 and -5's contributions to processes related to VSMC physiology.


Subject(s)
Myocytes, Smooth Muscle/physiology , Thrombospondin 1/physiology , Thrombospondins/physiology , Cartilage Oligomeric Matrix Protein/genetics , Cartilage Oligomeric Matrix Protein/pharmacology , Cartilage Oligomeric Matrix Protein/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/physiology , Gene Expression/drug effects , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Neointima/etiology , Thrombospondin 1/genetics , Thrombospondin 1/pharmacology , Thrombospondins/genetics , Thrombospondins/pharmacology
10.
Vasc Endovascular Surg ; 48(7-8): 470-5, 2014.
Article in English | MEDLINE | ID: mdl-25361959

ABSTRACT

BACKGROUND: Thrombospondin 1 (TSP-1) induces vascular smooth muscle cell (VSMC) migration and intimal hyperplasia. Statins and nitric oxide (NO) donors decrease intimal hyperplasia. We previously showed that statins (long-term exposure) and NO donors inhibit TSP-1-induced VSMC chemotaxis. HYPOTHESES: (1) Pretreatment with short-term statin will inhibit TSP-1-induced VSMC chemotaxis and (2) NO donors will enhance statin inhibition of TSP-1-induced or platelet-derived growth factor (PDGF)-induced VSMC chemotaxis. METHODS: We examined these treatment effects on TSP-1-induced VSMC chemotaxis: (1) long-term (20 hours) versus short-term (20 minutes) pravastatin, (2) diethylenetriamine NONOate (DETA/NO) or S-nitroso-N-acetylpenicillamine (SNAP) in combination with pravastatin, and (3) comparison of TSP-1 to PDGF as a chemoattractant. RESULTS: Pravastatin (long term or short term) inhibited TSP-1-induced chemotaxis. Diethylenetriamine NONOate and SNAP impeded statin inhibition of TSP-1-induced chemotaxis. Platelet-derived growth factor and TSP-1 had opposite effects on DETA/NO-pravastatin treatment. CONCLUSION: Short-term statin pretreatment inhibited TSP-1-induced VSMC chemotaxis, suggesting a pleiotropic effect. High-dose NO reversed statin inhibition of TSP-1-induced chemotaxis, suggesting NO and statin combination therapies warrant further study.


Subject(s)
Chemotaxis/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Pravastatin/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Thrombospondin 1/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Nitroso Compounds/metabolism , Platelet-Derived Growth Factor/pharmacology , S-Nitroso-N-Acetylpenicillamine/metabolism , Time Factors
11.
Surgery ; 155(2): 228-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24314882

ABSTRACT

OBJECTIVES: Thrombospondin-1 (TSP-1) is a matricellular glycoprotein released from platelets at sites of arterial injury and is important in neointima development after balloon angioplasty. MicroRNAs are small noncoding RNAs that function by binding target gene mRNA and inhibiting protein translation. MicroRNA-21 (miR-21) is up-regulated after angioplasty, and inhibition of miR-21 leads to decreased intimal hyperplasia. In this study, we examined the effects of miR-21 inhibition on vascular smooth muscle cell (VSMC) processes. METHODS: VSMCs were exposed to TSP-1 and miR-21 inhibitor for 20 minutes. TSP-1-induced migration was assessed with a modified Boyden microchemotaxis chamber and proliferation with calcein-AM fluorescence. Phosphorylated extracellular signaling kinase (ERK) 1/2 expression was determined by Western Blot and densitometry. Quantitative real-time polymerase chain reaction for TSP-1, hyaluronic acid synthase 2 (HAS2), and transforming growth factor beta 2 (TGFß2) was performed. Statistical analysis was performed with analysis of variance (P < .05). RESULTS: Inhibition of miR-21 blocked TSP-1-induced VSMC migration, proliferation, and ERK 1/2 phosphorylation (P < .05) and had no effect on TSP-1-stimulated expression of genes for TSP-1, HAS2, or TGFß2 (P > .05). CONCLUSION: Acute inhibition of miR-21 led to a decrease in VSMC migration and proliferation caused by TSP-1. The decrease in TSP-1's activation of ERK 1/2 after acute miR-21 inhibition indicates an active role for miR-21 in TSP-1's cell signaling cascade. No effect on TSP-1-induced expression of the pro-stenotic genes thbs1, tgfb2, or has2, occurred after acute miR-21 inhibition. These data indicate that miR-21 directly modulates cell function and signaling pathways in ways other than inhibition of protein translation.


Subject(s)
Cell Movement/physiology , Cell Proliferation , Glucuronosyltransferase/metabolism , MicroRNAs/physiology , Myocytes, Smooth Muscle/metabolism , Thrombospondin 1/physiology , Transforming Growth Factor beta2/metabolism , Cells, Cultured , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/metabolism , Humans , Hyaluronan Synthases , MicroRNAs/antagonists & inhibitors , Phosphorylation
12.
PLoS One ; 8(12): e82482, 2013.
Article in English | MEDLINE | ID: mdl-24324797

ABSTRACT

The present study examined whether 20-hydroxyeicosatetraenoic acid (HETE) contributes to the vasoconstrictor effect of angiotensin II (ANG II) in renal microvessels by preventing activation of the large conductance Ca(2+)-activated K(+) channel (KCa) in vascular smooth muscle (VSM) cells. ANG II increased the production of 20-HETE in rat renal microvessels. This response was attenuated by the 20-HETE synthesis inhibitors, 17-ODYA and HET0016, a phospholipase A2 inhibitor AACOF3, and the AT1 receptor blocker, Losartan, but not by the AT2 receptor blocker, PD123319. ANG II (10(-11) to 10(-6) M) dose-dependently decreased the diameter of renal microvessels by 41 ± 5%. This effect was blocked by 17-ODYA. ANG II (10(-7) M) did not alter KCa channel activity recorded from cell-attached patches on renal VSM cells under control conditions. However, it did reduce the NPo of the KCa channel by 93.4 ± 3.1% after the channels were activated by increasing intracellular calcium levels with ionomycin. The inhibitory effect of ANG II on KCa channel activity in the presence of ionomycin was attenuated by 17-ODYA, AACOF3, and the phospholipase C (PLC) inhibitor U-73122. ANG II induced a peak followed by a steady-state increase in intracellular calcium concentration in renal VSM cells. 17-ODYA (10(-5) M) had no effect on the peak response, but it blocked the steady-state increase. These results indicate that ANG II stimulates the formation of 20-HETE in rat renal microvessels via the AT1 receptor activation and that 20-HETE contributes to the vasoconstrictor response to ANG II by blocking activation of KCa channel and facilitating calcium entry.


Subject(s)
Angiotensin II/pharmacology , Hydroxyeicosatetraenoic Acids/metabolism , Microvessels/drug effects , Microvessels/metabolism , Potassium Channels/metabolism , Renal Circulation/drug effects , Renal Circulation/physiology , Animals , Calcium/metabolism , Gene Expression , Ionomycin/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phospholipases A2/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/metabolism , Rats , Receptors, Angiotensin/genetics , Receptors, Angiotensin/metabolism , Type C Phospholipases/metabolism , Vasoconstrictor Agents/pharmacology
13.
Mol Cell Biochem ; 384(1-2): 181-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24104451

ABSTRACT

Angioplasty causes local vascular injury, leading to the release of thrombospondin-1 (TSP-1), which stimulates vascular smooth muscle cell (VSMC) migration and proliferation, important steps in the development of intimal hyperplasia. Transforming growth factor beta 2 (TGF-ß2) and hyaluronic acid synthase (HAS) are two pro-stenotic genes upregulated in VSMCs by TSP-1. We hypothesized that inhibition of TGF-ß2 or HAS would inhibit TSP-1-induced VSMC migration, proliferation, and TSP-1 signaling. Our data demonstrate that Inhibition of either TGF-ß2 or HAS inhibited TSP-1-induced VSMC migration and proliferation. Activation of ERK 1 was decreased by TGF-ß2 inhibition and unaffected by HAS inhibition. TGF-ß2 and HAS are not implicated in TSP-1-induced thbs1 expression, while they are each implicated in TSP-1-induced expression of their own gene. In summary, TSP-1-induced VSMC migration and proliferation rely on intact TGF-ß2 signaling and HAS function. TSP-1 activation of ERK 1 is dependent on TGF-ß2. These data further expand our understanding of the complexity of TSP-1 cellular signaling and the involvement of TGF-ß2 and HAS.


Subject(s)
Chemotaxis/physiology , Glucuronosyltransferase/metabolism , Myocytes, Smooth Muscle/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta2/metabolism , Angioplasty, Balloon/adverse effects , Cell Degranulation , Cell Proliferation , Enzyme Activation , Glucuronosyltransferase/antagonists & inhibitors , Humans , Hyaluronan Synthases , Hymecromone/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Signal Transduction , Transforming Growth Factor beta2/antagonists & inhibitors , Vascular System Injuries/metabolism
14.
J Surg Res ; 173(2): e63-72, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22104612

ABSTRACT

Atherosclerosis is a chronic inflammatory condition of the blood vessel wall that can lead to arterial narrowing and subsequent vascular compromise. Although there are a variety of open and endovascular procedures used to alleviate the obstructions caused by atherosclerotic plaque, blood vessel instrumentation itself can lead to renarrowing of the vessel lumen through intimal hyperplasia, wound contracture, or a combination of the two. While the cell types involved in both atherosclerosis and vessel renarrowing after surgical intervention are largely characterized, current research has shown that components of the extracellular matrix are also important in the pathogenesis of the aforementioned processes. One such component is hyaluronic acid (HA). The objective of this review, therefore, is to examine the involvement of HA in these pathologic processes. Literature on the structure and function of HA was reviewed, with particular attention given to the role of HA in the processes of atherogenesis, intimal hyperplasia, and wound contracture after blood vessel instrumentation. HA interacts with vascular smooth muscle cells (VSMCs), endothelial cells (ECs), and platelets to promote atherogenesis. In particular, VSMCs manufacture large amounts of HA that form "cable-like" structures important for leukocyte adhesion and rolling. Additionally, transmigration of leukocytes across the EC layer is mediated by HA. Platelets cleave large molecules of HA into fragments that up-regulate leukocyte production of chemokines and cytokines. HA also has a role in both intimal hyperplasia and wound contracture, the two processes most responsible for vessel renarrowing after vascular instrumentation. HA has a complex, and sometimes conflicting, role in the pathologic processes of atherogenesis and vessel wall renarrowing after surgical intervention.


Subject(s)
Atherosclerosis/etiology , Coronary Restenosis/etiology , Hyaluronic Acid/physiology , Animals , Atherosclerosis/therapy , Hyaluronan Receptors/metabolism , Hyaluronic Acid/biosynthesis , Hyaluronic Acid/chemistry , Hyperplasia/etiology , Tunica Intima/pathology
15.
Am J Surg ; 202(5): 545-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21944292

ABSTRACT

BACKGROUND: Vascular smooth muscle cell (VSMC) migration is an important process in many vascular disorders. Nicotine, thrombospondin-1 (TSP-1) and fibronectin (Fn) separately induce VSMC migration. The hypothesis of this study was that nicotine treatment of vascular cells would augment TSP-1-induced and Fn-induced VSMC migration. METHODS: VSMCs or endothelial cells (ECs) were treated with serum-free medium or nicotine. Migration of VSMCs was assessed using a modified Boyden chemotaxis chamber to serum-free medium, TSP-1, Fn, EC basal medium, and conditioned EC medium or nicotine-treated conditioned EC medium alone or with supplemented TSP-1 or Fn. RESULTS: Nicotine treatment increased VSMC chemotaxis to serum-free medium, but TSP-1 or Fn had no further effect on chemotaxis. Conditioned EC and nicotine-treated conditioned EC enhanced VSMC chemotaxis, which was further augmented by Fn supplementation. CONCLUSIONS: Nicotine-stimulated EC derived factors induce VSMC migration, which is augmented by the addition of Fn.


Subject(s)
Cell Movement/drug effects , Chemotaxis/drug effects , Endothelial Cells/drug effects , Ganglionic Stimulants/pharmacology , Myocytes, Smooth Muscle/drug effects , Nicotine/pharmacology , Cells, Cultured , Endothelial Cells/physiology , Fibronectins/physiology , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Thrombospondin 1/physiology
16.
Am J Surg ; 202(5): 553-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21906719

ABSTRACT

BACKGROUND: Thrombospondin-1 (TSP-1) stimulates vascular smooth muscle cell (VSMC) migration via defined intracellular signaling pathways. The aim of this study was to examine the signaling pathways whereby TSP-1 folded domains (amino-terminal [NH(2)], procollagen homology [PCH], all 3 type 1 repeats [3TSR], and a single recombinant protein containing the 3rd type 2 repeat, the type 3 repeats, and the carboxyl-terminal [E3T3C1]) induce VSMC migration. METHODS: Quiescent VSMCs were pretreated with serum-free media or inhibitors: PP2 (c-Src), LY294002 (phosphatidylinositol 3-kinase), FPT (Ras), Y27632 (Rho kinase), SB202190 (p38 kinase), and PD98059 (extracellular signal-regulated kinase). Migration induced by serum-free media, TSP-1, NH(2), PCH, 3TSR, and E3T3C1 was assessed using a modified Boyden chamber. RESULTS: TSP-1, NH(2), 3TSR, and E3T3C1 induced VSMC chemotaxis (P < .05), but PCH did not (P > .05). PP2, FPT, SB202190, and PD98059 attenuated chemotaxis stimulated by TSP-1, NH(2), 3TSR, and E3T3C1 (P < .05). LY294002 inhibited TSP-1-induced and E3T3C1-induced (P < .05) but not NH(2)-induced or 3TSR-induced (P > .05) chemotaxis. Y27632 inhibited NH(2)-induced, 3TSR-induced, and E3T3C1-induced (P < .05) but not TSP-1-induced (P > .05) induced chemotaxis. CONCLUSIONS: TSP-1 folded domains are differentially dependent on intracellular signaling pathways to induce migration.


Subject(s)
Cell Movement/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Signal Transduction/physiology , Thrombospondin 1/physiology , Amides/pharmacology , Cells, Cultured , Chemotaxis/physiology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Imidazoles/pharmacology , Morpholines/pharmacology , Organophosphonates/pharmacology , Protein Folding , Pyridines/pharmacology , Pyrimidines/pharmacology
17.
Vasc Endovascular Surg ; 45(1): 55-62, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21193465

ABSTRACT

BACKGROUND: Thrombospondin 1 (TSP-1), fibronectin (Fn), and vitronectin (Vn) promote vascular smooth muscle cell (VSMC) chemotaxis through a variety of second messenger systems, including Ras, ERK1/2, and p38. HYPOTHESIS: Ras, ERK1/2, and p38 differentially affect TSP-1-, Fn-, and Vn-induced VSMC chemotaxis. METHODS: Bovine VSMCs were transfected with Ras N17 or treated with the following inhibitors: a farnesyl protein transferase (FPT) inhibitor, PD098059 (ERK1/2 inhibitor), or SB202190 (p38 inhibitor). Thrombospondin 1, Fn, and Vn were used as chemoattractants. Results were analyzed by analysis of variance (ANOVA) with post hoc testing (P < .05). RESULTS: Ras N17 transfection or FPT inhibitor treatment inhibited TSP-1-, Fn-, and Vn-induced chemotaxis. PD098059 or SB202190 resulted in more inhibition of VSMC migration to TSP-1 than to Fn or Vn. CONCLUSIONS: Ras appears equally relevant in the signal transduction pathways of TSP-1-, Fn-, and Vn-induced VSMC chemotaxis. Thrombospondin 1-induced migration is more dependent upon ERK1/2 and p38 than Fn- or Vn-included migration.


Subject(s)
Chemotaxis , Fibronectins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Thrombospondin 1/metabolism , Vitronectin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Analysis of Variance , Animals , Cattle , Cells, Cultured , Chemotaxis/drug effects , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , ras Proteins/genetics
18.
J Surg Res ; 166(1): e59-69, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193206

ABSTRACT

BACKGROUND: Although many sepsis treatments have shown efficacy in acute animal models, at present only activated protein C is effective in humans. The likely reason for this discrepancy is that most of the animal models used for preclinical testing do not accurately replicate the complex pathogenesis of human sepsis. Our objective in this study was to develop a clinically applicable model of severe sepsis and gut ischemia/reperfusion (I/R) that would cause multiple organ injury over a period of 48 h. MATERIALS AND METHODS: Anesthetized, instrumented, and ventilated pigs were subjected to a "two-hit" injury by placement of a fecal clot through a laparotomy and by clamping the superior mesenteric artery (SMA) for 30 min. The animals were monitored for 48 h. Wide spectrum antibiotics and intravenous fluids were given to maintain hemodynamic status. FiO(2) was increased in response to oxygen desaturation. Twelve hours following injury, a drain was placed in the laparotomy wound. Extensive hemodynamic, lung, kidney, liver, and renal function measurements and serial measurements of arterial and mixed venous blood gases were made. Bladder pressure was measured as a surrogate for intra-peritoneal pressure to identify the development of the abdominal compartment syndrome (ACS). Plasma and peritoneal ascites cytokine concentration were measured at regular intervals. Tissues were harvested and fixed at necropsy for detailed morphometric analysis. RESULTS: Polymicrobial sepsis developed in all animals. There was a progressive deterioration of organ function over the 48 h. The lung, kidney, liver, and intestine all demonstrated clinical and histopathologic injury. Acute lung injury (ALI) and ACS developed by consensus definitions. Increases in multiple cytokines in serum and peritoneal fluid paralleled the dysfunction found in major organs. CONCLUSION: This animal model of Sepsis+I/R replicates the systemic inflammation and dysfunction of the major organ systems that is typically seen in human sepsis and trauma patients. The model should be useful in deciphering the complex pathophysiology of septic shock as it transitions to end-organ injury thus allowing sophisticated preclinical studies on potential treatments.


Subject(s)
Disease Models, Animal , Multiple Organ Failure/physiopathology , Reperfusion Injury/physiopathology , Shock, Septic/physiopathology , Sus scrofa , Animals , Blood Gas Analysis , Blood Pressure/physiology , Cytokines/blood , Electrolytes/blood , Female , Heart Rate/physiology , Humans , Kaplan-Meier Estimate , Kidney/physiology , Multiple Organ Failure/mortality , Multiple Organ Failure/therapy , Pulmonary Wedge Pressure/physiology , Reperfusion Injury/mortality , Reperfusion Injury/therapy , Shock, Septic/mortality , Shock, Septic/therapy
19.
J Surg Res ; 168(1): 149-54, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-20338582

ABSTRACT

BACKGROUND: Thrombospondin-1 (TSP-1) induces vascular smooth muscle cell (VSMC) migration and is important in the development of intimal hyperplasia. HMG-CoA reductase inhibitors, such as lovastatin, reduce the incidence of vascular restenosis after angioplasty by both cholesterol lowering and pleiotropic effects. Inhibition of the mevalonate pathway is largely responsible for these pleiotropic properties. This inhibition prevents isoprenylation of the small G proteins, Rho and Ras, by geranylgeranyl and farnesyl pyrophosphate, respectively. Isoprenylation is required for Ras and Rho activation, which is relevant for cell migration. HYPOTHESIS: Lovastatin inhibits TSP-1-induced VSMC chemotaxis by inhibiting small G proteins via the mevalonate pathway. METHODS: Chemotaxis was assessed using a modified Boyden chamber. Quiescent VSMCs were pretreated with serum free media (SFM), lovastatin with or without mevalonate farnesyl (FTI), geranylgeranyl transferase inhibitors (GGTI), farnesyl transferase inhibitor (FPT), or the Rho kinase inhibitor (Y-27632). Chemoattractants were SFM or TSP-1. Comparisons were made by ANOVA followed by post-hoc testing (P<0.05). The effect of lovastatin on Ras activation was evaluated using cells pretreated with SFM or lovastatin, with or without mevalonate prior to TSP-1 exposure. Western blot for Ras activation was performed. RESULTS: Lovastatin dose-dependently inhibited TSP-1-induced chemotaxis, which was reversed by mevalonate. Mevalonate did not induce chemotaxis independently. FTI and FPT, but not GGTI or Y-27632, inhibited TSP-1-induced Ras activation and TSP-1-induced chemotaxis. Lovastatin inhibition of Ras activation was reversed with mevalonate. CONCLUSION: Ras, not Rho, is relevant for TSP-1-induced VSMC chemotaxis. These data suggest that lovastatin suppresses TSP-1-induced chemotaxis by inhibition of Ras.


Subject(s)
Chemotaxis/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Thrombospondin 1/metabolism , Animals , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mevalonic Acid/pharmacology , Models, Animal , Monomeric GTP-Binding Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Signal Transduction/drug effects , Tunica Intima/cytology , Tunica Intima/drug effects , Tunica Intima/metabolism , Tunica Media/cytology , Tunica Media/drug effects , Tunica Media/metabolism , ras Proteins/metabolism
20.
Am J Surg ; 200(5): 615-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21056139

ABSTRACT

BACKGROUND: Neointimal hyperplasia involves the migration of medial vascular smooth muscle cells (VSMCs) in response to arterial injury. Thrombospondin-1 (TSP1), platelet-derived growth factor (PDGF), and fibronectin (Fn) induce VSMC migration. Nitric oxide (NO) limits VSMC migration. The hypothesis of this study is that NO would dose dependently inhibit TSP1-induced, PDGF-induced, and Fn-induced VSMC chemotaxis. METHODS: VSMCs were pretreated with serum free media or the NO donors diethylenetriamine NONOate or S-nitroso-N-acetyl-D,L-penicillamine. Chemotaxis to TSP1, PDGF, or Fn was determined. Analysis of variance with post hoc testing was done. P values < .05 were considered significant. RESULTS: PDGF, TSP1, and Fn induced VSMC chemotaxis. NO donors inhibited chemotaxis of VSMCs to PDGF in a concentration-dependent manner. NO donors had a variable effect on TSP1-induced chemotaxis. NO donors did not inhibit Fn-induced chemotaxis. CONCLUSION: The complex interactions of these proteins in vivo will need to be considered when developing NO-dependent therapies for neointimal hyperplasia.


Subject(s)
Fibronectins/pharmacology , Muscle, Smooth, Vascular/cytology , Nitric Oxide/pharmacology , Platelet-Derived Growth Factor/pharmacology , Thrombospondin 1/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL