Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 287
Filter
Add more filters

Publication year range
1.
Biomacromolecules ; 25(7): 4329-4343, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38833553

ABSTRACT

The development of nanotherapy targeting mitochondria to alleviate oxidative stress is a critical therapeutic strategy for vascular calcification (VC) in diabetes. In this study, we engineered mitochondria-targeted nanodrugs (T4O@TPP/PEG-PLGA) utilizing terpinen-4-ol (T4O) as a natural antioxidant and mitochondrial protector, PEG-PLGA as the nanocarrier, and triphenylphosphine (TPP) as the mitochondrial targeting ligand. In vitro assessments demonstrated enhanced cellular uptake of T4O@TPP/PEG-PLGA, with effective mitochondrial targeting. This nanodrug successfully reduced oxidative stress induced by high glucose levels in vascular smooth muscle cells. In vivo studies showed prolonged retention of the nanomaterials in the thoracic aorta for up to 24 h. Importantly, experiments in diabetic VC models underscored the potent antioxidant properties of T4O@TPP/PEG-PLGA, as evidenced by its ability to mitigate VC and restore mitochondrial morphology. These results suggest that these nanodrugs could be a promising strategy for managing diabetic VC.


Subject(s)
Antioxidants , Mitochondria , Oxidative Stress , Vascular Calcification , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Antioxidants/pharmacology , Antioxidants/chemistry , Vascular Calcification/drug therapy , Vascular Calcification/metabolism , Vascular Calcification/pathology , Oxidative Stress/drug effects , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Nanoparticles/chemistry , Mice , Male , Polyethylene Glycols/chemistry , Rats , Humans , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism
2.
Exp Cell Res ; 421(2): 113407, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36334793

ABSTRACT

Vascular calcification (VC) is closely related to higher cardiovascular mortality and morbidity, and vascular smooth muscle cell (VSMC) switching to osteogenic-like cells is crucial for VC. LncRNA LEF1-AS1 promotes atherosclerosis and dental pulp stem cells calcification, while its role in VC remains unknown. Visceral adipose tissue-derived serine protease inhibitor (vaspin) is an adipokine regulating bone metabolism. However, the relationship between vaspin and VC is still unclear. We aimed to explore the role of LEF1-AS1 on VSMC osteogenic transition, whether vaspin inhibited LEF1-AS1-mediated osteogenic differentiation of VSMCs, and the responsible mechanism. In this study, quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting analysis indicated that LEF1-AS1 overexpression significantly upregulated osteogenic marker Runt-related transcription factor-2 (RUNX2) level and downregulated VSMC contractile marker α-smooth muscle actin (α-SMA) level. Alizarin red staining, alkaline phosphatase (ALP) staining, ALP activity assay, and calcium content assay also suggested that LEF1-AS1 overexpression promoted calcium deposition in VSMCs. However, vaspin treatment abolished this phenomenon. Mechanistically, LEF1-AS1 markedly decreased phosphorylated YAP level, while vaspin reversed LEF1-AS1-induced phosphorylated YAP decline. Our results revealed that LEF1-AS1 accelerated the osteogenic differentiation of VSMCs by regulating the Hippo/YAP pathway, while vaspin eliminated the LEF1-AS1-meditated VSMCs osteogenic phenotype switch.


Subject(s)
RNA, Long Noncoding , Vascular Calcification , Humans , Muscle, Smooth, Vascular/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Osteogenesis/genetics , Calcium/metabolism , Myocytes, Smooth Muscle/metabolism , Vascular Calcification/chemically induced , Cell Differentiation/genetics , Signal Transduction , Cells, Cultured , Lymphoid Enhancer-Binding Factor 1
3.
Can J Physiol Pharmacol ; 100(3): 220-233, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34570985

ABSTRACT

Coronary artery disease (CAD) is currently a leading cause of death worldwide. In the history of percutaneous coronary intervention for the treatment of CAD, a drug-eluting stent (DES) is recognized as a revolutionary technology that has the unique ability to significantly reduce restenosis and provide both mechanical and biological solutions simultaneously to the target lesion. The aim of the research work was to design and fabricate DES coated with a nanoparticulate drug formulation. Sirolimus, an inhibitor of the smooth muscle cell (SMC) proliferation and migration, was encapsulated in polymeric nanoparticles (NPs). The NP formulation was characterized for various physicochemical parameters. Cell viability and cell uptake studies were performed using human coronary artery smooth muscle cells (HCASMCs). The developed NP formulation showed enhanced efficacy compared to plain drug solution and exhibited time-dependent uptake into the HCASMCs. The developed NP formulation was coated on the Flexinnium™ ultra-thin cobalt-chromium alloy coronary stent platform. The NP-coated stents were characterized for morphology and residual solvent analysis. In vitro drug release was also evaluated. Ex vivo arterial permeation was carried out to evaluate the NP uptake from the surface of the stents. The characterization studies together corroborated that the developed NP coated stent can be a promising replacement of the current DESs.


Subject(s)
Drug Compounding/methods , Drug Liberation , Drug-Eluting Stents , Nanoparticles , Percutaneous Coronary Intervention/methods , Sirolimus/administration & dosage , Cell Survival/drug effects , Cells, Cultured , Chemical Phenomena , Chromium Alloys , Coronary Vessels/cytology , Coronary Vessels/metabolism , Humans , In Vitro Techniques , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Sirolimus/pharmacokinetics , Sirolimus/pharmacology
4.
Int J Mol Sci ; 24(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36613563

ABSTRACT

Atherosclerosis is a major cause of mortality worldwide. The initial change in atherosclerosis is intimal thickening due to muscle cell proliferation and migration. A correlation has been observed between periodontal disease and atherosclerosis. Here, we investigated the proliferation and migration of human aortic smooth muscle cells (HASMCs) using Porphyromonas gingivalis-derived LPS (Pg-LPS). To elucidate intracellular signaling, toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88) of HASMCs were knocked down, and the role of these molecules in Pg-LPS-stimulated proliferation and migration was examined. The role of mitogen-activated protein kinase (MAPK) in HASMC proliferation and migration was further elucidated by MAPK inhibition. Pg-LPS stimulation increased the proliferation and migration of HASMCs and activated the TLR4/MyD88 pathway. TLR4 knockdown inhibited Pg-LPS stimulated HASMCs proliferation and migration. Pg-LPS stimulation led to the phosphorylation of P38 MAPK, JNK, and ERK, and MyD88 knockdown inhibited the phosphorylation of P38 MAPK and JNK but not ERK. P38 MAPK and SAPK/JNK inhibition did not suppress the proliferation of HASMCs upon Pg-LPS stimulation, but ERK inhibition significantly inhibited proliferation. SAPK/JNK and ERK inhibition suppressed Pg-LPS-stimulated migration of HASMCs. In conclusion, our findings suggest that Pg-LPS may promote atherosclerosis via the activation of MAPK through TLR4.


Subject(s)
Atherosclerosis , Myocytes, Smooth Muscle , Humans , Atherosclerosis/metabolism , Cell Proliferation , Lipopolysaccharides , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Myocytes, Smooth Muscle/cytology , p38 Mitogen-Activated Protein Kinases/metabolism , Porphyromonas gingivalis , Toll-Like Receptor 4/metabolism , Cell Movement
5.
J Cell Physiol ; 236(10): 6897-6906, 2021 10.
Article in English | MEDLINE | ID: mdl-33650160

ABSTRACT

Vascular stiffening, an early and common characteristic of cardiovascular diseases (CVDs), stimulates vascular smooth muscle cell (VSMC) proliferation which reciprocally accelerates the progression of CVDs. However, the mechanisms by which extracellular matrix stiffness accompanying vascular stiffening regulates VSMC proliferation remain largely unknown. In the present study, we examined the role of the intermediate-conductance Ca2+ -activated K+  (IKCa ) channel in the matrix stiffness regulation of VSMC proliferation by growing A7r5 cells on soft and stiff polydimethylsiloxane substrates with stiffness close to these of arteries under physiological and pathological conditions, respectively. Stiff substrates stimulated cell proliferation and upregulated the expression of the IKCa channel. Stiff substrate-induced cell proliferation was suppressed by pharmacological inhibition using TRAM34, an IKCa channel blocker, or genetic depletion of the IKCa channel. In addition, stiff substrate-induced cell proliferation was also suppressed by reducing extracellular Ca2+ concentration using EGTA or intracellular Ca2+ concentration using BAPTA-AM. Moreover, stiff substrate induced activation of extracellular signal-regulated kinases (ERKs), which was inhibited by treatment with TRAM34 or BAPTA-AM. Stiff substrate-induced cell proliferation was suppressed by treatment with PD98059, an ERK inhibitor. Taken together, these results show that substrates with pathologically relevant stiffness upregulate the IKCa channel expression to enhance intracellular Ca2+ signaling and subsequent activation of the ERK signal pathway to drive cell proliferation. These findings provide a novel mechanism by which vascular stiffening regulates VSMC function.


Subject(s)
Calcium Signaling , Cell Proliferation , Dimethylpolysiloxanes/chemistry , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Mechanotransduction, Cellular , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Cell Culture Techniques , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Rats
6.
Biochem Biophys Res Commun ; 526(3): 841-847, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32278550

ABSTRACT

BACKGROUND: In-stent restenosis (ISR) is a complex disease that occurs after coronary stenting procedures. The development of quality materials and improvement of our understanding on significant factors regulating ISR are essential for enhancing prognosis. Vascular smooth muscle cells (VSMCs) are the main constituent cells of blood vessel walls, and dysfunction of VMSCs can exacerbate ISR. Accordingly, in this study, we explored the influence of wrinkled material topography on the biological functions of VSMCs. METHODS: Polydimethylsiloxane with a wrinkled topography was synthesized using elastomer base and crosslinking and observed by atomic force microscopy. VSMC proliferation, apoptosis, and morphology were determined by Cell Counting Kit-8 assays, fluorescence-assisted cell sorting, and phalloidin staining. α-Smooth muscle actin (α-SMA), major histocompatibility complex (MHC), and calponin 1 (CNN-1) expression levels were measured by quantitative real-time polymerase chain reaction and western blotting. Moreover, p53 and cleaved caspase-3 expression levels were evaluated by western blotting in VSMCs to assess apoptotic induction. RESULTS: Surface topographies were not associated with a clear orientation or elongation of VSMCs. The number of cells was increased on wrinkled surfaces (0.7 µm in amplitude, and 3 µm in wavelength [W3]) compared with that on other surfaces, contributing to continuously increased cell proliferation. Moreover, interactions of VSMCs with the W3 surface suppressed phenotypic switching, resulting in ISR via regulation of α-SMA, calponin-1, and SM-MHC expression. The surface with an amplitude of 0.05 µm and a wavelength of 0.5 µm (W0.5) promoted apoptosis by inducing caspase 3 and p53 activities. CONCLUSION: Introduction of aligned topographies on biomaterial scaffolds could provide physical cues to modulate VSMC responses for engineering vascular constructs. Materials with wrinkled topographies could have applications in the development of stents to reduce ISR.


Subject(s)
Apoptosis , Dimethylpolysiloxanes/chemistry , Muscle, Smooth, Vascular/metabolism , Phenotype , Tissue Scaffolds/chemistry , Actins/genetics , Actins/metabolism , Biomechanical Phenomena , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Cell Proliferation , Cells, Cultured , Cross-Linking Reagents/chemistry , Gene Expression Regulation , Humans , Major Histocompatibility Complex/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microscopy, Atomic Force , Muscle, Smooth, Vascular/cytology , Surface Properties , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Calponins
7.
J Cardiovasc Pharmacol ; 75(6): 603-607, 2020 06.
Article in English | MEDLINE | ID: mdl-32168154

ABSTRACT

Adenoviral vectors are useful tools in manipulating a gene of interest in vitro and in vivo, including in the vascular system. The transduction efficiencies of adenoviral vectors in vascular cells such as endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are known to be lower than those in epithelial cell types. The effective entry for adenoviral vectors is primarily mediated through the coxsackievirus and adenovirus receptor (CAR), which has been shown to be expressed in both cell types. Cationic liposomes have been used to enhance adenovirus transduction efficiency in nonepithelial cells. Accordingly, the aim of this study is to obtain new information regarding differences in transduction efficiencies, cationic liposome sensitivity, and CAR expression between ECs and VSMCs. Using cultured rat aortic ECs and VSMCs, here, we have compared transduction efficiency of adenoviruses with or without inclusion of liposomes and CAR expression. A significant increase in basal transduction efficiency was observed in ECs compared with VSMCs. Cationic liposome polybrene enhanced transduction efficiency in VSMCs, whereas decreased efficiency was observed in ECs. Western blotting demonstrated expression of the CAR in ECs but not in VSMCs. Proteomic analysis and mouse aorta immunostaining further suggests significant expression of the CAR in ECs but not in VSMCs. In conclusion, adenoviruses can effectively transduce the gene of interest in aortic ECs likely because of abundant expression of the CAR, whereas cationic liposomes such as polybrene enhance the transduction efficiency in VSMCs lacking CAR expression.


Subject(s)
Adenoviridae/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Endothelial Cells/metabolism , Genetic Vectors , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transduction, Genetic , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Animals , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hexadimethrine Bromide/chemistry , Liposomes , Male , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism
8.
Ann Vasc Surg ; 64: 328-338, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31634610

ABSTRACT

BACKGROUND: Therapeutic neovascularization has some obstacles, such as it requires more than one proangiogenic factor, and these factors have short half-lives. To overcome these obstacles, combined delivery of granulocyte-colony stimulating factor (G-CSF), erythropoietin (EPO) and vascular endothelial growth factor (VEGF) using protein/dextran/poly (lactic-co-glycolic acid) (PLGA) sustained-release microspheres was proposed to promote neovascularization. METHODS: Dextran microparticles loaded with G-CSF, EPO or VEGF were prepared and encapsulated in PLGA microspheres to obtain protein-dextran-PLGA microspheres. The release behavior of microspheres was studied in vitro. The protein/dextran/PLGA microspheres were injected into the ischemic hindlimbs of rats. Neovascularization in ischemic muscle was measured. RESULTS: Microspheres released G-CSF, EPO and VEGF in vitro for more than 4 weeks. Combined therapy with VEGF, EPO and G-CSF promoted the expression of B-cell lymphoma-2 and stromal cell-derived factor 1, cellular proliferation and the incorporation of C-X-C chemokine receptor 4 positive cells. Capillary density and smooth muscle α-actin+ vessel density were higher in the combined treatment of VEGF, EPO and G-CSF than in the single factor treatment. CONCLUSIONS: The combined and sustained delivery of VEGF, EPO and G-CSF using dextran-PLGA microspheres had a more significant neovascularization effect than monotherapy with each factor alone. This combined therapy might be a promising treatment for ischemic vascular diseases.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Dextrans/chemistry , Drug Carriers , Erythropoietin/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Ischemia/drug therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Polyesters/chemistry , Vascular Endothelial Growth Factor A/administration & dosage , Angiogenesis Inducing Agents/chemistry , Animals , Cell Proliferation/drug effects , Delayed-Action Preparations , Disease Models, Animal , Drug Compounding , Drug Liberation , Erythropoietin/chemistry , Granulocyte Colony-Stimulating Factor/chemistry , Hindlimb , Injections, Intramuscular , Ischemia/pathology , Ischemia/physiopathology , Kinetics , Male , Microspheres , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Particle Size , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/chemistry
9.
Int J Mol Sci ; 21(1)2020 Jan 04.
Article in English | MEDLINE | ID: mdl-31947992

ABSTRACT

The bicarbonate ion has a fundamental role in vital systems. Impaired bicarbonate transport leads to various diseases, including immune disorders, cystic fibrosis, tumorigenesis, kidney diseases, brain dysfunction, tooth fracture, ischemic reperfusion injury, hypertension, impaired reproductive system, and systemic acidosis. Carbonic anhydrases are involved in the mechanism of bicarbonate movement and consist of complex of bicarbonate transport systems including bicarbonate transporters. This review focused on the convergent regulation of ion homeostasis through various ion transporters including bicarbonate transporters, their regulatory enzymes, such as carbonic anhydrases, pH regulatory role, and the expression pattern of ion transporters in non-secretory systems throughout the body. Understanding the correlation between these systems will be helpful in order to obtain new insights and design potential therapeutic strategies for the treatment of pH-related disorders. In this review, we have discussed the broad prospects and challenges that remain in elucidation of bicarbonate-transport-related biological and developmental systems.


Subject(s)
Bicarbonates/metabolism , Carbonic Anhydrases/metabolism , Bicarbonates/chemistry , Gastrointestinal Tract/metabolism , Humans , Hydrogen-Ion Concentration , Immune System/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ion Transport , Muscle, Smooth, Vascular/metabolism , Neoplasms/metabolism , Neoplasms/pathology
10.
Proc Natl Acad Sci U S A ; 113(40): 11190-11195, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647912

ABSTRACT

Mechanical compliance has been demonstrated to be a key determinant of cell behavior, directing processes such as spreading, migration, and differentiation. Durotaxis, directional migration from softer to more stiff regions of a substrate, has been observed for a variety of cell types. Recent stiffness mapping experiments have shown that local changes in tissue stiffness in disease are often accompanied by an altered ECM composition in vivo. However, the importance of ECM composition in durotaxis has not yet been explored. To address this question, we have developed and characterized a polyacrylamide hydrogel culture platform featuring highly tunable gradients in mechanical stiffness. This feature, together with the ability to control ECM composition, allows us to isolate the effects of mechanical and biological signals on cell migratory behavior. Using this system, we have tracked vascular smooth muscle cell migration in vitro and quantitatively analyzed differences in cell migration as a function of ECM composition. Our results show that vascular smooth muscle cells undergo durotaxis on mechanical gradients coated with fibronectin but not on those coated with laminin. These findings indicate that the composition of the adhesion ligand is a critical determinant of a cell's migratory response to mechanical gradients.


Subject(s)
Cell Culture Techniques/methods , Cell Movement/genetics , Extracellular Matrix/metabolism , Stress, Mechanical , Acrylic Resins/chemistry , Animals , Cattle , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Tracking/methods , Elasticity , Extracellular Matrix/genetics , Fibronectins/chemistry , Fibronectins/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Laminin/chemistry , Laminin/metabolism , Ligands , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Signal Transduction
11.
J Cell Physiol ; 233(6): 4759-4769, 2018 06.
Article in English | MEDLINE | ID: mdl-29150938

ABSTRACT

Porphyromonas (P.) gingivalis infection leading to the periodontitis has been associated with the development of systemic diseases, including cardiovascular diseases and diabetes. However, the effect of a high concentration of glucose (HG) on the invasion efficiency of P. gingivalis and the consequent modulation of pathogenesis in vascular cells, especially in the vascular smooth muscle cells (VSMCs), remains unclear. Hence, the aim of this study was to investigate whether treating P. gingivalis with HG could change its invasion capability and result in VSMC calcification and the underlying mechanism. Human aortic SMCs (HASMCs) and P. gingivalis strain CCUG25226 were used in this study. We found that HGPg infection of HASMCs could initiate the HASMC calcification by stimulating the autocrine regulation of bone morphogenetic protein (BMP) 4 in HASMCs. The upregulation of BMP4 expression in HASMCs was mediated by toll-like receptor 4 and ERK1/2-p38 signaling after P. gingivalis infection. Moreover, the autocrine action of BMP4 in HGPg infection-initiated HASMC calcification upregulated BMP4-specific downstream smad1/5/8-runx2 signaling to increase the expressions of bone-related matrix proteins, that is, osteopontin, osteocalcin, and alkaline phosphatase. This study elucidates the detailed mechanism of HGPg infection-initiated calcification of HASMCs and indicates a possible therapeutic role of BMP4 in P. gingivalis infection-associated vascular calcification.


Subject(s)
Aortic Diseases/microbiology , Bacteroidaceae Infections/microbiology , Glucose/pharmacology , Muscle, Smooth, Vascular/microbiology , Myocytes, Smooth Muscle/microbiology , Osteogenesis , Porphyromonas gingivalis/drug effects , Vascular Calcification/microbiology , Aorta/metabolism , Aorta/microbiology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Autocrine Communication , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/pathology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Osteogenesis/genetics , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/pathogenicity , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
12.
J Vasc Interv Radiol ; 29(7): 1041-1049.e3, 2018 07.
Article in English | MEDLINE | ID: mdl-29754850

ABSTRACT

PURPOSE: To compare the drug effect in treated vessels and downstream effects in distal skeletal muscle of drug-coated balloons (DCBs) and drug-eluting stents (DESs) in a healthy preclinical swine model. MATERIALS AND METHODS: Four groups of treated iliofemoral arteries (percutaneous transluminal angioplasty [PTA]+DES, DCB+DES, DCB+bare metal stent [BMS], and DCB alone) of 12 healthy swine were assessed, with euthanasia at 30 days. Biological drug effect was evaluated using smooth muscle cell (SMC) loss score according to both depth and circumference as well as a neointimal fibrin and medial proteoglycan scores which were compared between the 4 groups. Vascular and skeletal muscle changes in regions downstream from the treated site were also assessed histologically for evidence of emboli. RESULTS: DESs showed greater medial SMC loss in the treated arteries irrespective of preceding DCB or PTA treatment in terms of depth (DCB+DES vs PTA+DES vs DCB+BMS vs DCB alone; median, 4.0 mm vs 3.8 mm vs 3.0 mm vs 2.2 mm; P = .009) and circumference (4.0 mm vs 3.5 mm vs 2.0 mm vs 1.2 mm, respectively; P = .007). Sections of skeletal muscles downstream from the treated arteries showed arteriolar changes of fibrinoid necrosis consistent with paclitaxel effect exclusively in the DCB groups (DCB+BMS, 26.9% of sections; DCB+DES, 14.3%; DCB alone, 19.2%; PTA+DES, 0%; P = .02). CONCLUSIONS: In the treated arteries, irrespective of preceding DCB treatment or PTA, DES treatment showed maximum drug effects vs DCB alone or in combination with BMS placement, and there was no detrimental toxic effect in DCB-treated iliofemoral arteries before DES treatment compared with PTA before DES treatment. Downstream vascular changes were exclusively seen in groups treated with DCBs.


Subject(s)
Angioplasty, Balloon/instrumentation , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible , Drug-Eluting Stents , Femoral Artery/drug effects , Muscle, Skeletal/blood supply , Paclitaxel/administration & dosage , Popliteal Artery/drug effects , Vascular Access Devices , Angioplasty, Balloon/adverse effects , Animals , Cardiovascular Agents/toxicity , Femoral Artery/metabolism , Femoral Artery/pathology , Fibrin/metabolism , Models, Animal , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima , Paclitaxel/toxicity , Popliteal Artery/metabolism , Popliteal Artery/pathology , Proteoglycans/metabolism , Sus scrofa , Time Factors
13.
Basic Res Cardiol ; 112(4): 41, 2017 07.
Article in English | MEDLINE | ID: mdl-28540527

ABSTRACT

Ischemic heart disease is still the leading cause of death even with the advancement of pharmaceutical therapies and surgical procedures. Early vascularization in the ischemic heart is critical for a better outcome. Although stem cell therapy has great potential for cardiovascular regeneration, the ideal cell type and delivery method of cells have not been resolved. We tested a new approach of stem cell therapy by delivery of induced vascular progenitor cells (iVPCs) grown on polymer micro-bundle scaffolds in a rat model of myocardial infarction. iVPCs partially reprogrammed from vascular endothelial cells (ECs) had potent angiogenic potential and were able to simultaneously differentiate into vascular smooth muscle cells (SMCs) and ECs in 2D culture. Under hypoxic conditions, iVPCs also secreted angiogenic cytokines such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) as measured by enzyme-linked immunosorbent assay (ELISA). A longitudinal micro-scaffold made from poly(lactic-co-glycolic acid) was sufficient for the growth and delivery of iVPCs. Co-cultured ECs and SMCs aligned well on the micro-bundle scaffold similarly as in the vessels. 3D cell/polymer micro-bundles formed by iVPCs and micro-scaffolds were transplanted into the ischemic myocardium in a rat model of myocardial infarction (MI) with ligation of the left anterior descending artery. Our in vivo data showed that iVPCs on the micro-bundle scaffold had higher survival, and better retention and engraftment in the myocardium than free iVPCs. iVPCs on the micro-bundles promoted better cardiomyocyte survival than free iVPCs. Moreover, iVPCs and iVPC/polymer micro-bundles treatment improved cardiac function (ejection fraction and fractional shortening, endocardial systolic volume) measured by echocardiography, increased vessel density, and decreased infarction size [endocardial and epicardial infarct (scar) length] better than untreated controls at 8 weeks after MI. We conclude that iVPCs grown on a polymer micro-bundle scaffold are new promising approach for cell-based therapy designed for cardiovascular regeneration in ischemic heart disease.


Subject(s)
Endothelial Progenitor Cells/transplantation , Lactic Acid/chemistry , Muscle, Smooth, Vascular/transplantation , Myocardial Infarction/surgery , Myocardium/pathology , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , Polyglycolic Acid/chemistry , Tissue Engineering/methods , Tissue Scaffolds , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Endothelial Progenitor Cells/metabolism , Fibroblast Growth Factor 2/metabolism , Muscle, Smooth, Vascular/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocytes, Smooth Muscle/metabolism , Paracrine Communication , Phenotype , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/metabolism , Ventricular Remodeling
14.
J Vasc Surg ; 65(1): 207-217.e3, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27034112

ABSTRACT

OBJECTIVE: Lipid mediators derived from omega-3 polyunsaturated fatty acids such as resolvin D1 (RvD1) accelerate the resolution of inflammation and have potential as vascular therapeutics. The objective of this study was to evaluate local perivascular delivery of RvD1 as a means to attenuate neointimal hyperplasia in a rat model of arterial injury. METHODS: Smooth muscle cells were harvested from rat aortas to study the effects of RvD1 on rat arterial vascular smooth muscle cell responses in vitro, with focus on inflammation, proliferation, migration, cytoskeletal changes, and cytotoxicity. The safety and efficacy of perivascular delivery of RvD1 through thin biodegradable three-layered poly(lactic-co-glycolic acid) wraps or 25% Pluronic F127 gels were studied in a rat model of carotid angioplasty. A total of 200 ng of RvD1 was loaded into each construct for perivascular delivery after injury. Morphometric and histologic analyses were performed 3 and 14 days after injury. RESULTS: RvD1 attenuated rat arterial vascular smooth muscle cell inflammatory pathways, proliferation, migration, and mitogen-induced cytoskeletal changes in vitro, without evidence of cytotoxicity. RvD1-loaded wraps reduced neointimal formation after carotid angioplasty by 59% vs no-wrap controls (P = .001) and by 45% vs vehicle-wrap controls (P = .002). RvD1-loaded Pluronic gels similarly reduced neointimal formation by 49% vs no-gel controls (P = .02) and by 52% vs vehicle-gel controls (P = .02). No group was associated with infection, thrombosis, or negative vessel remodeling. Wraps were found to be easier to apply than gel constructs. Ki67 proliferation index was significantly lower in RvD1-loaded wrap-treated arteries compared with both no-wrap and vehicle-wrap controls at both 3 and 14 days after injury (65% vs no-wrap group and 70% vs vehicle-wrap group at day 3, 49% vs both control groups at day 14; P < .05). Similarly, oxidative stress (30% and 29%; P < .05) and nuclear factor κB activation (42% and 45%; P < .05) were significantly lower in the RvD1-loaded wrap group compared with both no-wrap and vehicle-wrap controls at 3 days after injury. CONCLUSIONS: Local perivascular delivery of RvD1 attenuates formation of neointimal hyperplasia without associated toxicity in a rat model of carotid angioplasty. This effect is likely due to attenuation of inflammatory pathways as well as decreased arterial smooth muscle cell proliferation and migration.


Subject(s)
Cardiovascular Agents/administration & dosage , Carotid Artery Diseases/drug therapy , Docosahexaenoic Acids/administration & dosage , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Angioplasty, Balloon/adverse effects , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Cardiovascular Agents/chemistry , Carotid Artery Diseases/etiology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/pathology , Disease Models, Animal , Docosahexaenoic Acids/chemistry , Drug Carriers , Drug Compounding , Hyperplasia , Inflammation Mediators/metabolism , Ki-67 Antigen/metabolism , Lactic Acid/chemistry , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Oxidative Stress/drug effects , Poloxamer/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Time Factors , Transcription Factor RelA/metabolism
15.
J Surg Res ; 219: 136-144, 2017 11.
Article in English | MEDLINE | ID: mdl-29078873

ABSTRACT

BACKGROUND: We have previously defined mechanisms of intimal hyperplasia that could be targets for molecular therapeutics aimed at vascular pathology. However, biocompatible nanocarriers are needed for effective delivery. Cationic liposomes (CLPs) have been demonstrated as effective nanocarriers in vitro. However, in vivo success has been hampered by cytotoxicity. Recently, neutral PEGylated liposomes (PLPs) have been modified with cell-penetrating peptides (CPPs) to enhance cellular uptake. We aim to establish CPP-modified neutral liposomes as viable molecular nanocarriers in vascular smooth muscle cells. METHODS: CLPs, PLPs, and CPP-modified PLPs (R8-PLPs) were assembled with short interfering RNA (siRNA) via ethanol injection. Characterization studies determined liposomal morphology, size, and charge. siRNA encapsulation efficiency was measured via RiboGreen assay. Vascular smooth muscle cells were exposed to equal lipid/siRNA across all groups. Rhodamine-labeled liposomes were used to quantify cell association via fluorometry, live/dead dual stain was used to measure cytotoxicity, and gene silencing was measured by quantitative polymerase chain reaction. RESULTS: R8-PLPs exhibited increased encapsulation efficiency equivalent to CLPs. PLPs and R8-PLP-5 mol% and R8-PLP-10 mol% had no cytotoxic effect. CLPs demonstrated significant cytotoxicity. R8-PLP-5 mol% and R8-PLP-10 mol% exhibited increased cell association versus PLPs. R8-PLP-10 mol% resulted in significant gene silencing, in a manner dependent on lipid-to-siRNA load capacity. CONCLUSIONS: The negligible cytotoxicity and enhanced cellular association and gene silencing capacity exhibited by R8-PLPs reveal this class of liposomes as a candidate for future applications. Further modifications for optimizing R8-PLPs are still warranted to improve efficacy, and in vivo studies are needed for translational development. However, this could prove to be an optimal nanocarrier for vascular gene therapeutics.


Subject(s)
Cell-Penetrating Peptides/metabolism , Genetic Therapy , Liposomes/chemistry , Myocytes, Smooth Muscle/metabolism , RNA, Small Interfering/administration & dosage , Humans , Muscle, Smooth, Vascular/metabolism , Nanoparticles/chemistry , Vascular Diseases/drug therapy
16.
Methods ; 99: 20-7, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26732049

ABSTRACT

Current techniques for tissue engineering blood vessels are not customizable for vascular size variation and vessel wall thickness. These critical parameters vary widely between the different arteries in the human body, and the ability to engineer vessels of varying sizes could increase capabilities for disease modeling and treatment options. We present an innovative method for producing customizable, tissue engineered, self-organizing vascular constructs by replicating a major structural component of blood vessels - the smooth muscle layer, or tunica media. We utilize a unique system combining 3D printed plate inserts to control construct size and shape, and cell sheets supported by a temporary fibrin hydrogel to encourage cellular self-organization into a tubular form resembling a natural artery. To form the vascular construct, 3D printed inserts are adhered to tissue culture plates, fibrin hydrogel is deposited around the inserts, and human aortic smooth muscle cells are then seeded atop the fibrin hydrogel. The gel, aided by the innate contractile properties of the smooth muscle cells, aggregates towards the center post insert, creating a tissue ring of smooth muscle cells. These rings are then stacked into the final tubular construct. Our methodology is robust, easily repeatable and allows for customization of cellular composition, vessel wall thickness, and length of the vessel construct merely by varying the size of the 3D printed inserts. This platform has potential for facilitating more accurate modeling of vascular pathology, serving as a drug discovery tool, or for vessel repair in disease treatment.


Subject(s)
Blood Vessel Prosthesis , Tissue Engineering , Biocompatible Materials/chemistry , Cells, Cultured , Gene Expression , Humans , Hydrogels/chemistry , Materials Testing , Muscle Contraction , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/physiology , Printing, Three-Dimensional , Tensile Strength
17.
BMC Genomics ; 17: 20, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728506

ABSTRACT

BACKGROUND: RNA interference (RNAi) is a powerful platform utilized to target transcription of specific genes and downregulate the protein product. To achieve effective silencing, RNAi is usually applied to cells or tissue with a transfection reagent to enhance entry into cells. A commonly used control is the same transfection reagent plus a "noncoding RNAi". However, this does not control for the genomic response to the transfection reagent alone or in combination with the noncoding RNAi. These control effects while not directly targeting the gene in question may influence expression of other genes that in turn alter expression of the target. The current study was prompted by our work focused on prevention of vascular bypass graft failure and our experience with gene silencing in human aortic smooth muscle cells (HAoSMCs) where we suspected that off target effects through this mechanism might be substantial. We have used Next Generation Sequencing (NGS) technology and bioinformatics analysis to examine the genomic response of HAoSMCs to the transfection reagent alone (polyethyleneimine (PEI)) or in combination with commercially obtained control small interfering RNA (siRNAs) (Dharmacon and Invitrogen). RESULTS: Compared to untreated cells, global gene expression of HAoSMcs after transfection either with PEI or in combination with control siRNAs displayed significant alterations in gene transcriptome after 24 h. HAoSMCs transfected by PEI alone revealed alterations of 213 genes mainly involved in inflammatory and immune responses. HAoSMCs transfected by PEI complexed with siRNA from either Dharmacon or Invitrogen showed substantial gene variation of 113 and 85 genes respectively. Transfection of cells with only PEI or with PEI and control siRNAs resulted in identification of 20 set of overlapping altered genes. Further, systems biology analysis revealed key master regulators in cells transfected with control siRNAs including the cytokine, Interleukin (IL)-1, transcription factor GATA Binding Protein (GATA)-4 and the methylation enzyme, Enhancer of zeste homolog 2 (EZH-2) a cytokine with an apical role in initiating the inflammatory response. CONCLUSIONS: Significant off-target effects in HAoSMCs transfected with PEI alone or in combination with control siRNAs may lead to misleading conclusions concerning the effectiveness of a targeted siRNA strategy. The lack of structural information about transfection reagents and "non coding" siRNA is a hindrance in the development of siRNA based therapeutics.


Subject(s)
Aorta/drug effects , Computational Biology , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/drug effects , Aorta/metabolism , Enhancer of Zeste Homolog 2 Protein , GATA4 Transcription Factor/biosynthesis , Gene Expression Regulation/genetics , Gene Silencing , High-Throughput Nucleotide Sequencing , Humans , Interleukin-1/biosynthesis , Muscle, Smooth, Vascular/metabolism , Polycomb Repressive Complex 2/biosynthesis , Polyethyleneimine/administration & dosage , RNA, Small Interfering/genetics , Transfection/methods
18.
J Vasc Surg ; 64(5): 1444-1449, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26432280

ABSTRACT

BACKGROUND: The production of growth factors from several experimental arterial conduits was determined. METHODS: We implanted 105 experimental arterial grafts that were 1 cm long in the abdominal aorta of Lewis rats (average weight, 250 g). Five different types of grafts were analyzed: arterial isografts, vein grafts, arterial allografts, and polytetrafluoroethylene (PTFE) grafts with normal or decreased compliance. Animals were killed humanely 4 weeks after surgery and the production of platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor-ß, tumor necrosis factor-α, and interleukin-1 was analyzed. RESULTS: Myointimal hyperplasia (MH) was evident in vein grafts, arterial allografts, and PTFE grafts, but not in arterial isografts. Growth factor production was increased for grafts prone to develop MH like vein, PTFE grafts, and arterial allografts. PDGF and bFGF were increased significantly for PTFE and vein grafts, but not for arterial allografts. The importance of bFGF and PGDF was confirmed by the capability of antibody to PDGF and to bFGF to reduce the mitogenic activity of smooth muscle cells, in vivo and in vitro, for PTFE and vein grafts, but not for arterial allografts, in which a predominant role was played by interleukin-1 and tumor necrosis factor-α. CONCLUSIONS: Agents able to neutralize this increased production of growth factors, either directly or by competition with their receptors, can prevent MH formation.


Subject(s)
Aorta, Abdominal/surgery , Arteries/transplantation , Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Intercellular Signaling Peptides and Proteins/metabolism , Veins/transplantation , Allografts , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Arteries/metabolism , Arteries/pathology , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned/metabolism , Fibroblast Growth Factor 2/metabolism , Hyperplasia , Interleukin-1/metabolism , Isografts , Models, Animal , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/transplantation , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/transplantation , Neointima , Platelet-Derived Growth Factor/metabolism , Polytetrafluoroethylene , Prosthesis Design , Rats, Inbred Lew , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Veins/metabolism , Veins/pathology
19.
Mol Pharm ; 13(5): 1575-86, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26985693

ABSTRACT

The effects of naked polyamidoamine (PAMAM) dendrimers on renin-angiotensin system (RAS) signaling via Angiotensin (Ang) II-mediated transactivation of the epidermal growth factor receptor (EGFR) and the closely related family member ErbB2 (HER2) were investigated. In primary aortic vascular smooth muscle cells, a cationic fifth-generation (G5) PAMAM dendrimer dose- and time-dependently inhibited Ang II/AT1 receptor-mediated transactivation of EGFR and ErbB2 as well as their downstream signaling via extracellular-regulated kinase 1/2 (ERK1/2). Inhibition even occurred at noncytotoxic concentrations at short (1 h) exposure times and was dependent on dendrimer generation (G7 > G6 > G5 > G4) and surface group chemistry (amino > carboxyl > hydroxyl). Mechanistically, the cationic G5 PAMAM dendrimer inhibited Ang II-mediated transactivation of EGFR and ErbB2 via inhibition of the nonreceptor tyrosine kinase Src. This novel, early onset, intrinsic biological action of PAMAM dendrimers as inhibitors of the Ang II/AT1/Src/EGFR-ErbB2/ERK1/2 signaling pathway could have important toxicological and pharmacological implications.


Subject(s)
Angiotensin II/pharmacology , Dendrimers/pharmacology , ErbB Receptors/metabolism , Polyamines/pharmacology , Polymers/pharmacology , Receptor, ErbB-2/metabolism , Transcriptional Activation/drug effects , Animals , Cell Line , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , src-Family Kinases/metabolism
20.
Am J Physiol Heart Circ Physiol ; 307(10): H1419-29, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25239800

ABSTRACT

Oral all-trans retinoic acid (atRA) has been shown to reduce the formation of neointimal hyperplasia; however, the dose required was 30 times the chemotherapeutic dose, which already has reported side effects. As neointimal formation is a localized process, new approaches to localized delivery are required. This study assessed whether atRA within a citrate-based polyester, poly(1,8 octanediolcitrate) (POC), perivascular membrane would prevent neointimal hyperplasia following arterial injury. atRA-POC membranes were prepared and characterized for atRA release via high-performance liquid chromatography with mass spectrometry detection. Rat adventitial fibroblasts (AF) and vascular smooth muscle cells (VSMC) were exposed to various concentrations of atRA; proliferation, apoptosis, and necrosis were assessed in vitro. The rat carotid artery balloon injury model was used to evaluate the impact of the atRA-POC membranes on neointimal formation, cell proliferation, apoptosis, macrophage infiltration, and vascular cell adhesion molecule 1 (VCAM-1) expression in vivo. atRA-POC membranes released 12 µg of atRA over 2 wk, with 92% of the release occurring in the first week. At 24 h, atRA (200 µmol/l) inhibited [(3)H]-thymidine incorporation into AF and VSMC by 78% and 72%, respectively (*P = 0.001), with negligible apoptosis or necrosis. Histomorphometry analysis showed that atRA-POC membranes inhibited neointimal formation after balloon injury, with a 56%, 57%, and 50% decrease in the intimal area, intima-to-media area ratio, and percent stenosis, respectively (P = 0.001). atRA-POC membranes had no appreciable effect on apoptosis or proliferation at 2 wk. Regarding biocompatibility, we found a 76% decrease in macrophage infiltration in the intima layer (P < 0.003) in animals treated with atRA-POC membranes, with a coinciding 53% reduction in VCAM-1 staining (P < 0.001). In conclusion, perivascular delivery of atRA inhibited neointimal formation and restenosis. These data suggest that atRA-POC membranes may be suitable as localized therapy to inhibit neointimal hyperplasia following open cardiovascular procedures.


Subject(s)
Adventitia/drug effects , Carotid Artery Injuries/therapy , Carotid Artery, Common/drug effects , Carotid Stenosis/therapy , Citrates/chemistry , Drug Carriers , Membranes, Artificial , Neointima , Polymers/chemistry , Tretinoin/administration & dosage , Adventitia/metabolism , Adventitia/pathology , Animals , Apoptosis/drug effects , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Hyperplasia , Macrophages/drug effects , Macrophages/pathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats, Sprague-Dawley , Recurrence , Time Factors , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL