Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(7): e0132451, 2015.
Article in English | MEDLINE | ID: mdl-26167913

ABSTRACT

AIM: There is an unmet need to develop an innovative cardioprotective modality for acute myocardial infarction (AMI), for which the effectiveness of interventional reperfusion therapy is hampered by myocardial ischemia-reperfusion (IR) injury. Pretreatment with statins before ischemia is shown to reduce MI size in animals. However, no benefit was found in animals and patients with AMI when administered at the time of reperfusion, suggesting insufficient drug targeting into the IR myocardium. Here we tested the hypothesis that nanoparticle-mediated targeting of pitavastatin protects the heart from IR injury. METHODS AND RESULTS: In a rat IR model, poly(lactic acid/glycolic acid) (PLGA) nanoparticle incorporating FITC accumulated in the IR myocardium through enhanced vascular permeability, and in CD11b-positive leukocytes in the IR myocardium and peripheral blood after intravenous treatment. Intravenous treatment with PLGA nanoparticle containing pitavastatin (Pitavastatin-NP, 1 mg/kg) at reperfusion reduced MI size after 24 hours and ameliorated left ventricular dysfunction 4-week after reperfusion; by contrast, pitavastatin alone (as high as 10 mg/kg) showed no therapeutic effects. The therapeutic effects of Pitavastatin-NP were blunted by a PI3K inhibitor wortmannin, but not by a mitochondrial permeability transition pore inhibitor cyclosporine A. Pitavastatin-NP induced phosphorylation of Akt and GSK3ß, and inhibited inflammation and cardiomyocyte apoptosis in the IR myocardium. CONCLUSIONS: Nanoparticle-mediated targeting of pitavastatin induced cardioprotection from IR injury by activation of PI3K/Akt pathway and inhibition of inflammation and cardiomyocyte death in this model. This strategy can be developed as an innovative cardioprotective modality that may advance currently unsatisfactory reperfusion therapy for AMI.


Subject(s)
Cardiotonic Agents/therapeutic use , Drug Delivery Systems/methods , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Nanoparticles/therapeutic use , Quinolines/therapeutic use , Signal Transduction/drug effects , Animals , Blotting, Western , Capillary Permeability , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/analysis , Cardiotonic Agents/blood , Disease Models, Animal , Echocardiography , Flow Cytometry , Injections, Intravenous , Male , Myocardium/chemistry , Myocardium/pathology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Quinolines/administration & dosage , Quinolines/analysis , Quinolines/blood , Rats , Rats, Sprague-Dawley
2.
Arterioscler Thromb Vasc Biol ; 32(10): 2427-34, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22879581

ABSTRACT

OBJECTIVE: Critical limb ischemia is a severe form of peripheral artery disease (PAD) for which neither surgical revascularization nor endovascular therapy nor current medicinal therapy has sufficient therapeutic effects. Peroxisome proliferator activated receptor-γ agonists present angiogenic activity in vitro; however, systemic administration of peroxisome proliferator-activated receptor-γ agonists is hampered by its side effects, including heart failure. Here, we demonstrate that the nanoparticle (NP)-mediated delivery of the peroxisome proliferator activated receptor-γ agonist pioglitazone enhances its therapeutic efficacy on ischemia-induced neovascularization in a murine model. METHODS AND RESULTS: In a nondiabetic murine model of hindlimb ischemia, a single intramuscular injection of pioglitazone-incorporated NP (1 µg/kg) into ischemic muscles significantly improved the blood flow recovery in the ischemic limbs, significantly increasing the number of CD31-positive capillaries and α-smooth muscle actin-positive arterioles. The therapeutic effects of pioglitazone-incorporated NP were diminished by the peroxisome proliferator activated receptor-γ antagonist GW9662 and were not observed in endothelial NO synthase-deficient mice. Pioglitazone-incorporated NP induced endothelial NO synthase phosphorylation, as demonstrated by Western blot analysis, as well as expression of multiple angiogenic growth factors in vivo, including vascular endothelial growth factor-A, vascular endothelial growth factor-B, and fibroblast growth factor-1, as demonstrated by real-time polymerase chain reaction. Intramuscular injection of pioglitazone (1 µg/kg) was ineffective, and oral administration necessitated a >500 µg/kg per day dose to produce therapeutic effects equivalent to those of pioglitazone-incorporated NP. CONCLUSIONS: NP-mediated drug delivery is a novel modality that may enhance the effectiveness of therapeutic neovascularization, surpassing the effectiveness of current treatments for peripheral artery disease with critical limb ischemia.


Subject(s)
Hindlimb/blood supply , Ischemia/drug therapy , Nanoparticles , Neovascularization, Physiologic/drug effects , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Anilides/pharmacology , Animals , Fibroblast Growth Factor 1/metabolism , Humans , Injections, Intramuscular , Ischemia/physiopathology , Lactic Acid , Mice , Mice, Knockout , Models, Animal , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Pioglitazone , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Thiazolidinediones/administration & dosage , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor B/metabolism
3.
J Vasc Surg ; 52(2): 412-20, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20573471

ABSTRACT

OBJECTIVES: We recently demonstrated in a murine model that nanoparticle-mediated delivery of pitavastatin into vascular endothelial cells effectively increased therapeutic neovascularization. For the development of a clinically applicable approach, further investigations are necessary to assess whether this novel system can induce the development of collateral arteries (arteriogenesis) in a chronic ischemia setting in larger animals. METHODS: Chronic hind limb ischemia was induced in rabbits. They were administered single injections of nanoparticles loaded with pitavastatin (0.05, 0.15, and 0.5 mg/kg) into ischemic muscle. RESULTS: Treatment with pitavastatin nanoparticles (0.5 mg/kg), but not other nanoparticles, induced angiographically visible arteriogenesis. The effects of intramuscular injections of phosphate-buffered saline, fluorescein isothiocyanate (FITC)-loaded nanoparticles, pitavastatin (0.5 mg/kg), or pitavastatin (0.5 mg/kg) nanoparticles were examined. FITC nanoparticles were detected mainly in endothelial cells of the ischemic muscles for up to 4 weeks. Treatment with pitavastatin nanoparticles, but not other treatments, induced therapeutic arteriogenesis and ameliorated exercise-induced ischemia, suggesting the development of functional collateral arteries. Pretreatment with nanoparticles loaded with vatalanib, a vascular endothelial growth factor receptor (VEGF) tyrosine kinase inhibitor, abrogated the therapeutic effects of pitavastatin nanoparticles. Separate experiments with mice deficient for VEGF receptor tyrosine kinase demonstrated a crucial role of VEGF receptor signals in the therapeutic angiogenic effects. CONCLUSIONS: The nanotechnology platform assessed in this study (nanoparticle-mediated endothelial cell-selective delivery of pitavastatin) may be developed as a clinically feasible and promising strategy for therapeutic arteriogenesis in patients.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Collateral Circulation/drug effects , Drug Carriers , Endothelial Cells/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Ischemia/drug therapy , Muscle, Skeletal/blood supply , Nanoparticles , Neovascularization, Physiologic/drug effects , Quinolines/administration & dosage , Angiogenesis Inducing Agents/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Cells, Cultured , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Endothelial Cells/metabolism , Feasibility Studies , Hindlimb Suspension , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Injections, Intramuscular , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Oxygen/metabolism , Phthalazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Quinolines/metabolism , Rabbits , Regional Blood Flow/drug effects , Time Factors , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...