Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(26): 13006-13015, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31189595

ABSTRACT

Abdominal aortic aneurysm (AAA) remains the second most frequent vascular disease with high mortality but has no approved medical therapy. We investigated the direct role of apelin (APLN) in AAA and identified a unique approach to enhance APLN action as a therapeutic intervention for this disease. Loss of APLN potentiated angiotensin II (Ang II)-induced AAA formation, aortic rupture, and reduced survival. Formation of AAA was driven by increased smooth muscle cell (SMC) apoptosis and oxidative stress in Apln-/y aorta and in APLN-deficient cultured murine and human aortic SMCs. Ang II-induced myogenic response and hypertension were greater in Apln-/y mice, however, an equivalent hypertension induced by phenylephrine, an α-adrenergic agonist, did not cause AAA or rupture in Apln-/y mice. We further identified Ang converting enzyme 2 (ACE2), the major negative regulator of the renin-Ang system (RAS), as an important target of APLN action in the vasculature. Using a combination of genetic, pharmacological, and modeling approaches, we identified neutral endopeptidase (NEP) that is up-regulated in human AAA tissue as a major enzyme that metabolizes and inactivates APLN-17 peptide. We designed and synthesized a potent APLN-17 analog, APLN-NMeLeu9-A2, that is resistant to NEP cleavage. This stable APLN analog ameliorated Ang II-mediated adverse aortic remodeling and AAA formation in an established model of AAA, high-fat diet (HFD) in Ldlr-/- mice. Our findings define a critical role of APLN in AAA formation through induction of ACE2 and protection of vascular SMCs, whereas stable APLN analogs provide an effective therapy for vascular diseases.


Subject(s)
Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/pathology , Apelin/metabolism , Neprilysin/metabolism , Aged , Aged, 80 and over , Angiotensin II/administration & dosage , Angiotensin-Converting Enzyme 2 , Animals , Aorta, Abdominal/cytology , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/etiology , Apelin/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cardiovascular Agents/chemistry , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Male , Mice, Transgenic , Middle Aged , Myocytes, Smooth Muscle , Neprilysin/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Peptidyl-Dipeptidase A/metabolism , Phenylephrine/administration & dosage , Primary Cell Culture , Proteolysis/drug effects , RNA, Small Interfering/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Vascular Remodeling/drug effects , Vascular Remodeling/genetics
2.
Lab Invest ; 100(12): 1564-1574, 2020 12.
Article in English | MEDLINE | ID: mdl-32612287

ABSTRACT

Abdominal aortic aneurysm (AAA) is a potential lethal disease that is defined by an irreversible dilatation (>50%) of the aorta. During AAA expansion, the aortic wall is often remodeled, which is featured by extracellular matrix (ECM) degeneration, medial and adventitial inflammation, depletion and phenotypic switching of vascular smooth muscle cells (SMCs). Recent studies have suggested microRNAs as vital regulators for vascular SMC function. Our earlier work demonstrated an anti-AAA role of miR-126-5p in ApoE-/- mice infused with angiotensin (Ang) II. The present study aimed to further elucidate its role in AAA pathogenesis with a focus on aortic SMC phenotypic switching. Ventricular zone expressed PH domain containing 1 (VEPH1) was identified as a novel negative regulator for vascular SMC differentiation by our group, and its expression was negatively correlated to miR-126-5p in mouse abdominal aortas based on the present microarray data. In vivo, in addition attenuating Ang II infusion-induced aortic dilation and elastin degradation, miR-126-5p agomirs also significantly reduced the expression of VEPH1. In vitro, to induce synthetic transition of human aortic smooth muscle cells (hAoSMCs), cells were stimulated with 1 µM Ang II for 24 h. Ectopic overexpression of miR-126-5p restored the differentiation of hAoSMCs-the expression of contractile/differentiated SMC markers, MYH11, and α-SMA, increased, whilst that of synthetic/dedifferentiated SMC markers, PCNA and Vimentin, decreased. Both mus and homo VEPH1 genes were validated as direct targets for miR-126-5p. VEPH1 re-expression impaired miR-126-5p-induced differentiation of hAoSMCs. In addition, Ang II-induced upregulation in matrix metalloproteinase (MMP)-9 and MMP2, two key proteases responsible for ECM degradation, in mouse aortas and hAoSMCs was reduced by miR-126-5p overexpression as well. Collectively, these results reveal an important, but previously unexplored, role of miR-126-5p in inhibiting AAA development-associated aortic SMC dedifferentiation.


Subject(s)
Angiotensin II/metabolism , Aorta, Abdominal , MicroRNAs , Muscle, Smooth, Vascular , Nerve Tissue Proteins/metabolism , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Cell Differentiation/genetics , Cells, Cultured , Male , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism
3.
Biochem Biophys Res Commun ; 511(2): 343-349, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30782482

ABSTRACT

Abdominal aortic aneurysm (AAA) is a multi-factorial progressive vascular disease with life-threatening complications. Increasing evidence suggests that smooth muscle cell (SMC) dysfunction and cell death contribute to dilatation and rupture of the aorta by inducing an inflammatory response. The exact mechanism of this response however, is incompletely understood. We here investigated in vitro the capacity of autologous necrotic cell debris (CD) to induce inflammasome components and inflammatory mediators in aortic SMC (AAA-SMC) isolated from patients with AAA undergoing surgical repair. AAA-SMCs were additionally primed with Interferon- γ (IFN-γ) before treatment with CD in order to mimic the proinflammatory status caused by higher IFN-γ concentrations that have been demonstrated in the wall of AAAs. Real-time RT-PCR revealed that CD significantly increased NLRP3 and IL1B mRNA expression in different SMC cultures within 6 h of exposure. Priming of the AAA-SMC with IFN-γ significantly increased expression of NLRP3, AIM2, IFI16 and CASP1 mRNAs, whereas IL1B mRNA was reduced. Additional exposure of IFN-γ-primed AAA-SMC to CD for 6-24 h, further augmented expression of AIM2, NLRP3, and Caspase-1 protein levels. Analysis of the SMC supernatants by ELISA revealed CD-induced release of the senescence-associated cytokines IL-6 and MCP-1 in native and IFN-γ-primed SMC, whereas no secretion of Interleukin-(IL) 1α and IL-1ß secretion were observed. Our results implicate a role of necrotic cell debris derived from dead neighboring cells in SMC dysfunction and in inflammatory response of AAA tissue.


Subject(s)
Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/pathology , Inflammasomes/immunology , Myocytes, Smooth Muscle/pathology , NF-kappa B/immunology , Aorta, Abdominal/cytology , Aorta, Abdominal/immunology , Aortic Aneurysm, Abdominal/complications , Aortic Aneurysm, Abdominal/immunology , Cells, Cultured , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/immunology , Necrosis/complications , Necrosis/immunology , Necrosis/pathology
4.
Mol Cell Biochem ; 459(1-2): 121-130, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31127491

ABSTRACT

The antihyperglycemic agent empagliflozin not only improves glycemic control but has also been associated with clinically meaningful reductions in cardiovascular events. Studies have shown that empagliflozin significantly reduces cardiovascular death and heart failure-associated hospitalizations. Given that endothelial dysfunction is closely linked with the pathogenesis of atherosclerotic cardiovascular disease, we hypothesized that the cardiovascular benefits observed with empagliflozin may be a result of its positive impact on the health of the endothelial glycocalyx (GCX), a critical component for the endothelium homeostasis. Human abdominal aortic endothelial cells (HAAECs) were either statically cultured or subjected to a steady wall shear stress of 10 dyne/cm2. Empagliflozin (50 µM, 24 h) restored heparinase III-mediated GCX disruption and the normal mechanotransduction responses in GCX-compromised HAAECs while reducing the attachment of all-trans retinoic acid-transformed NB4 cells to HAAECs. The current body of work suggests that the cardioprotective properties previously reported for empagliflozin may in part be due to the ability of empagliflozin to preserve and restore the structural integrity of the GCX, which in turn helps to maintain vascular health by promoting an anti-inflammatory endothelium, in the presence of a pro-inflammatory environment. Further studies are needed to fully understand the mechanisms underlying the cardiovascular benefits of empagliflozin.


Subject(s)
Aorta, Abdominal/metabolism , Benzhydryl Compounds/pharmacology , Cardiotonic Agents/pharmacology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Glucosides/pharmacology , Glycocalyx/metabolism , Aorta, Abdominal/cytology , Cell Line, Tumor , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Humans
5.
Bull Exp Biol Med ; 165(2): 264-268, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29926276

ABSTRACT

We have previously developed a polycaprolactone (PCL) vascular graft with incorporated vascular endothelial growth factor (VEGF). Functioning of the PCL/VEGF graft in rat circulatory system over 1, 3 and 6 months after implantation into abdominal aorta was tested. Graft patency and formation of vascular wall elements were assessed histologically and by immunofluorescence staining for von Willebrand factor, CD31, CD34, and collagens I and IV and DAPI staining. Local application of VEGF promoted endothelialization and improved patency of the graft. The wall of the PCL/VEGF graft underwent remodeling due to active cellular infiltration and the extracellular matrix deposition.


Subject(s)
Blood Vessel Prosthesis , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Polyesters/chemistry , Vascular Endothelial Growth Factors/pharmacology , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/drug effects , Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Male , Rats , Rats, Wistar , Vascular Grafting , Vascular Remodeling/drug effects
6.
J Biomech Eng ; 139(7)2017 07 01.
Article in English | MEDLINE | ID: mdl-28334369

ABSTRACT

The von Mises (VM) stress is a common stress measure for finite element models of tissue mechanics. The VM failure criterion, however, is inherently isotropic, and therefore may yield incorrect results for anisotropic tissues, and the relevance of the VM stress to anisotropic materials is not clear. We explored the application of a well-studied anisotropic failure criterion, the Tsai­Hill (TH) theory, to the mechanically anisotropic porcine aorta. Uniaxial dogbones were cut at different angles and stretched to failure. The tissue was anisotropic, with the circumferential failure stress nearly twice the axial (2.67 ± 0.67 MPa compared to 1.46 ± 0.59 MPa). The VM failure criterion did not capture the anisotropic tissue response, but the TH criterion fit the data well (R2 = 0.986). Shear lap samples were also tested to study the efficacy of each criterion in predicting tissue failure. Two-dimensional failure propagation simulations showed that the VM failure criterion did not capture the failure type, location, or propagation direction nearly as well as the TH criterion. Over the range of loading conditions and tissue geometries studied, we found that problematic results that arise when applying the VM failure criterion to an anisotropic tissue. In contrast, the TH failure criterion, though simplistic and clearly unable to capture all aspects of tissue failure, performed much better. Ultimately, isotropic failure criteria are not appropriate for anisotropic tissues, and the use of the VM stress as a metric of mechanical state should be reconsidered when dealing with anisotropic tissues.


Subject(s)
Stress, Mechanical , Animals , Anisotropy , Aorta, Abdominal/cytology , Biomechanical Phenomena , Finite Element Analysis , Materials Testing , Swine
7.
Clin Exp Pharmacol Physiol ; 42(5): 510-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25740656

ABSTRACT

Microgravity-induced vascular remodelling may play an important role in post-spaceflight orthostatic intolerance. In this study, we aimed to investigate the effects of simulated microgravity on monocyte adhesion to aortic endothelium in hindlimb unweighted rats and to elucidate the underlying mechanisms associated with this event. Sprague-Dawley rats were subjected to 4-week hindlimb unweighting to simulate microgravity. The recruitment of monocytes to the abdominal aorta was investigated by en face immunofluorescence staining and monocyte binding assays. The expression of the adhesion molecules E-selectin and vascular cell adhesion molecule-1 as well as the cytokine monocyte chemoattractant protein (MCP)-1 was evaluated by immunohistochemical staining, western blot, and quantitative reverse transcription polymerase chain reaction analyses. Additionally, nuclear factor-κB (NF-κB) activation and the messenger RNA expression levels of E-selectin, vascular cell adhesion molecule-1, and MCP-1 were assessed with the administration of an NF-κB inhibitor, pyrrolidine dithiocarbamate. Results showed that simulated microgravity significantly increased monocyte recruitment to the aortic endothelium, protein expression of E-selectin and MCP-1, and NF-κB activation in the abdominal aorta of rats. Pyrrolidine dithiocarbamate treatment not only significantly inhibited NF-κB activity but also reduced the messenger RNA levels of E-selectin, vascular cell adhesion molecule-1, and MCP-1 as well as monocyte recruitment in the abdominal aorta of hindlimb unweighted rats. These results suggest that simulated microgravity increases monocyte adhesion to rat aortic endothelium via the NF-κB-mediated expression of the adhesion molecule E-selectin and the cytokine MCP-1. Therefore, an NF-κB-mediated inflammatory response may be one of the cellular mechanisms responsible for arterial remodelling during exposure to microgravity.


Subject(s)
Aorta, Abdominal/cytology , Endothelium, Vascular/cytology , Monocytes/cytology , NF-kappa B/metabolism , Weightlessness Simulation , Active Transport, Cell Nucleus/drug effects , Animals , Cell Adhesion/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chemokine CCL2/genetics , E-Selectin/genetics , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Macrophages/cytology , Macrophages/drug effects , Male , Monocytes/drug effects , NF-kappa B/antagonists & inhibitors , Pyrrolidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Thiocarbamates/pharmacology , Vascular Cell Adhesion Molecule-1/genetics
8.
Proc Natl Acad Sci U S A ; 109(20): 7770-5, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22550179

ABSTRACT

Vascular endothelial cells (ECs) are constantly exposed to blood flow-induced shear stress, but the mechanism of force-specific activation of their signaling to modulate cellular function remains unclear. We have demonstrated that bone morphogenetic protein receptor (BMPR)-specific Smad1/5 can be force-specifically activated by oscillatory shear stress (OSS) in ECs to cause cell cycle progression. Smad1/5 is highly activated in ECs of atherosclerotic lesions in diseased human coronary arteries from patients with end-stage heart failure undergoing heart transplantation and from apolipoprotein E-deficient mice. Application of OSS (0.5 ± 4 dyn/cm(2)) causes the sustained activation of Smad1/5 in ECs through activations of mammalian target of rapamycin and p70S6 kinase, leading to up-regulation of cyclin A and down-regulations of p21(CIP1) and p27(KIP1) and, hence, EC cycle progression. En face examination of rat aortas reveals high levels of phospho-Smad1/5 in ECs of the inner, but not the outer, curvature of aortic arch, nor the straight segment of thoracic aorta [corrected]. Immunohistochemical and en face examinations of the experimentally stenosed abdominal aorta in rats show high levels of phospho-Smad1/5 in ECs at poststenotic sites, where OSS occurs. These OSS activations of EC Smad1/5 in vitro and in vivo are not inhibited by the BMP-specific antagonist Noggin and, hence, are independent of BMP ligand. Transfecting ECs with Smad1/5-specific small interfering RNAs inhibits the OSS-induced EC cycle progression. Our findings demonstrate the force-specificity of the activation of Smad1/5 and its contribution to cell cycle progression in ECs induced by disturbed flow.


Subject(s)
Atherosclerosis/physiopathology , Cell Cycle/physiology , Endothelial Cells/physiology , Gene Expression Regulation/physiology , Regional Blood Flow/physiology , Smad1 Protein/metabolism , Stress, Mechanical , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/pathology , Apolipoproteins E/genetics , Biomechanical Phenomena , Coronary Vessels/cytology , Coronary Vessels/pathology , Cyclin A/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Humans , Immunohistochemistry , Mice , Mice, Knockout , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Taiwan
9.
Biochem Biophys Res Commun ; 444(2): 205-11, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24440697

ABSTRACT

Abdominal aortic aneurysms (AAA) are progressive dilatations of infra-renal aorta causing structural weakening rendering the aorta prone to rupture. AAA can be potentially stabilized by inhibiting inflammatory enzymes such as matrix metalloproteinases (MMP); however, active regression of AAA is not possible without new elastic fiber regeneration. Here we report the elastogenic benefit of direct delivery of polyphenols such as pentagalloyl glucose (PGG), epigallocatechin gallate (EGCG), and catechin, to smooth muscle cells obtained either from healthy or from aneurysmal rat aorta. Addition of 10 µg/ml PGG and ECGC induce elastin synthesis, organization, and crosslinking while catechin does not. Our results indicate that polyphenols bind to monomeric tropoelastin and enhance coacervation, aid in crosslinking of elastin by increasing lysyl oxidase (LOX) synthesis, and by blocking MMP-2 activity. Thus, polyphenol treatments leads to increased mature elastin fibers synthesis without increasing the production of intracellular tropoelastin.


Subject(s)
Elastic Tissue/metabolism , Myocytes, Smooth Muscle/drug effects , Polyphenols/pharmacology , Tropoelastin/metabolism , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/metabolism , Aorta, Abdominal/physiology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/physiopathology , Catechin/analogs & derivatives , Catechin/metabolism , Catechin/pharmacology , Cells, Cultured , Elastic Tissue/ultrastructure , Elastin/metabolism , Elastin/ultrastructure , Fibrillins , Hydrolyzable Tannins/metabolism , Hydrolyzable Tannins/pharmacology , Kinetics , Male , Matrix Metalloproteinase 2/metabolism , Microfilament Proteins/metabolism , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Polyphenols/metabolism , Protein-Lysine 6-Oxidase/metabolism , Rats , Rats, Sprague-Dawley , Regeneration
10.
J Surg Res ; 176(2): 687-95, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22316675

ABSTRACT

BACKGROUND: In humans, there is a 4:1 male:female ratio in the incidence of abdominal aortic aneurysms (AAAs). c-Jun-N-terminal kinase (JNK) is an important upstream regulator of several enzymes involved in AAA formation, including the matrix metalloproteinases (MMPs). The purpose of this study was to determine if there is a gender difference between males and females in JNK during AAA formation. MATERIALS AND METHODS: Male and female C57/B6 mice underwent aortic perfusion with elastase or heat inactivated elastase with aortas harvested at d 3 and 14 for phenotype determination, RT-PCR, Western blot, and zymography. Additionally, in vitro experiments using siRNA were conducted to define JNK regulation of matrix metalloproteinases (MMPs). A t-test was used to compare between groups. RESULTS: Males formed larger AAAs at d 14 compared with females (P < 0.001), with significantly higher levels of JNK1 protein, proMMP9, proMMP2, and active MMP2. At d 3, males had more JNK1 mRNA, protein, and MMP activity. Knockdown of JNK 1 or 2 in vitro decreased MMP activity, while knockdown of JNK 1 and 2 together blocked all MMP activity. CONCLUSION: Alterations in JNK between genders is partially responsible for the differential rates of experimental AAA formation, likely through differential regulation of MMPs.


Subject(s)
Aorta, Abdominal/enzymology , Aortic Aneurysm, Abdominal/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Sex Characteristics , Animals , Aorta, Abdominal/cytology , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Pancreatic Elastase/pharmacology , RNA, Small Interfering/genetics
11.
J Surg Res ; 178(1): 55-62, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22482758

ABSTRACT

BACKGROUND: The purpose of the present study was to assess the differentiation potential of adipose-derived stem cells (ASCs) into smooth muscle cells (SMCs) and their potential for promoting regeneration of smooth muscle for ureteral tissue engineering. METHODS: ASCs were isolated, proliferated, and identified in vitro. SMC differentiation was induced using SMC induction medium. Gene expression was evaluated by quantitative polymerase chain reaction, immunofluorescence, and Western blotting. Vessel extracellular matrix was obtained by a decellularization process. The induced cells were seeded onto vessel extracellular matrix for ureter reconstitution. Grafts were obtained for evolutionary histologic studies. Renal function and ureteral patency was evaluated by intravenous urography at 16 wk. RESULTS: Flow cytometry demonstrated that the ASCs expressed CD90, but did not express CD45 or CD34. After 6 wk of induction, upregulation of α-smooth muscle actin expression was determined by quantitative polymerase chain reaction, and smooth muscle myosin heavy chain expression was confirmed by immunofluorescence and Western blotting in the induced cells. Vessel extracellular matrix exhibited a nontoxic and bioactive effect on the induced cells. Histologically, stratified urothelium and organized muscle bundles were observed in the grafts at 16 wk. Intravenous urography demonstrated no ureteral stricture or hydroureteronephrosis. CONCLUSIONS: These results have demonstrated that ASCs can be differentiated into SMCs and this potential promoted smooth muscle regeneration for ureteral tissue engineering.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/cytology , Regeneration/physiology , Tissue Engineering/methods , Uterus , Actins/genetics , Actins/metabolism , Adventitia/cytology , Animals , Aorta, Abdominal/cytology , Arabidopsis Proteins/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Extracellular Matrix/physiology , Female , Integrin beta1/genetics , Integrin beta1/metabolism , Intramolecular Transferases/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Rabbits , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Uterus/cytology , Uterus/physiology , Uterus/transplantation
12.
J Mater Sci Mater Med ; 23(5): 1235-45, 2012 May.
Article in English | MEDLINE | ID: mdl-22359212

ABSTRACT

A novel 316L type Cu-bearing stainless steel was developed in present work, aiming at reducing the occurrence of the in-stent restenosis after implantations of coronary stents, through trace amount of Cu release from surface of the steel in body fluid. It was found that there was a trace amount of Cu released from the Cu-bearing steel in a simulated body fluid, with no cytotoxicity. All the in vitro experimental results proved that this Cu-bearing steel could not only inhibit the proliferation of vascular smooth muscle cells, reducing the formation of thrombosis, which are the main reasons for happening of the in-stent restenosis, but also promote the proliferation of vascular endothelial cells needed for the revascularization, showing that this novel steel is prospective to be a new material for manufacturing coronary stents with function of reducing the in-stent restenosis.


Subject(s)
Blood Vessel Prosthesis , Copper/pharmacokinetics , Graft Occlusion, Vascular/prevention & control , Stainless Steel/pharmacokinetics , Stents , Animals , Aorta, Abdominal/cytology , Body Fluids/chemistry , Body Fluids/metabolism , Cells, Cultured , Copper/analysis , Copper/chemistry , Copper/physiology , Equipment Design , Equipment Failure Analysis , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/metabolism , Humans , Microtechnology/methods , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Osmolar Concentration , Rats , Stainless Steel/chemistry , Whole Blood Coagulation Time
13.
Microsurgery ; 31(2): 138-45, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268111

ABSTRACT

The objective of this preliminary study was to develop a reabsorbable vascular patch that did not require in vitro cell or biochemical preconditioning for vascular wall repair. Patches were composed only of hyaluronic acid (HA). Twenty male Wistar rats weighing 250-350 g were used. The abdominal aorta was exposed and isolated. A rectangular breach (1 mm × 5 mm) was made on vessel wall and arterial defect was repaired with HA made patch. Performance was assessed at 1, 2, 4, 8, and 16 weeks after surgery by histology and immunohistochemistry. Extracellular matrix components were evaluated by molecular biological methods. After 16 weeks, the biomaterial was almost completely degraded and replaced by a neoartery wall composed of endothelial cells, smooth muscle cells, collagen, and elastin fibers organized in layers. In conclusion, HA patches provide a provisional three-dimensional support to interact with cells for the control of their function, guiding the spatially and temporally multicellular processes of artery regeneration.


Subject(s)
Absorbable Implants , Guided Tissue Regeneration/instrumentation , Hyaluronic Acid , Tissue Scaffolds , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/injuries , Aorta, Abdominal/physiology , Aorta, Abdominal/surgery , Biomarkers/analysis , Gene Expression , Guided Tissue Regeneration/methods , Male , Pilot Projects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome , Wound Healing
14.
Zhonghua Xin Xue Guan Bing Za Zhi ; 39(7): 658-63, 2011 Jul.
Article in Zh | MEDLINE | ID: mdl-22088249

ABSTRACT

OBJECTIVE: To investigate the effects of telmisartan and pyridoxamine on vascular smooth muscle cells (VSMCs) proliferation and apoptosis as well as abdominal aorta vascular remodeling in spontaneously hypertensive rats (SHRs). METHODS: SHRs randomly received placebo, telmisartan (6 mg kg(-1) x d(-1)), pyridoxamine (200 mg x kg(-1) x d(-1)) or telmisartan (6 mg x kg(-1) x d(-1)) plus pyridoxamine (200 mg x kg(-1) x d(-1), n = 12 each) for 16 weeks. Wistar-Kyoto (WKY, n = 12) rats serve as normotensive control. The systolic blood pressure (SBP) of rat was measured before and weekly thereafter. The serum advanced glycation end-products (AGEs) were detected by competitive ELISA. The serum super oxide dismutase (SOD) and nitric oxide (NO) were measured. The abdominal aorta were assessed by image analysis in HE stained sections. The VSMCs apoptosis and proliferation in abdominal aorta were detected with in situ end labeling technique and proliferating cell nuclear antigen (PCNA) immunohistochemistry staining respectively. RESULTS: SBP were significantly lower in telmisartan and telmisartan plus pyridoxamine therapy group than in placebo treated hypertensive rats while not affected by pyridoxamine (P > 0.05). Activity of SOD and NO were significantly higher and AGEs significantly lower in telmisartan, pyridoxamine and combination therapy treated SHRs than in placebo treated hypertensive rats (P < 0.01). The telmisartan, pyridoxamine and combination therapy can significantly inhibit the PCNA expression and significantly enhance the apoptosis value in abdominal aorta (P < 0.01). The efficacy of combined treatment was significantly higher than telmisartan and pyridoxamine alone (P < 0.05). CONCLUSION: Telmisartan and pyridoxamine could attenuate abdominal aorta vascular remodeling via reducing oxidative stress and AGEs production as well as restoring the balance of VSMCs proliferation and apoptosis in SHRs abdominal aorta.


Subject(s)
Aorta, Abdominal/drug effects , Benzimidazoles/pharmacology , Benzoates/pharmacology , Cell Proliferation/drug effects , Pyridoxamine/pharmacology , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/metabolism , Blood Pressure , Glycation End Products, Advanced/blood , Male , Nitric Oxide/metabolism , Oxidative Stress , Random Allocation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Superoxide Dismutase/metabolism , Telmisartan
15.
J Neurosci ; 29(46): 14451-62, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19923279

ABSTRACT

Amyloid precursor protein (APP) is a ubiquitously expressed type 1 integral membrane protein. It has the ability to bind numerous extracellular matrix components and propagate signaling responses via its cytoplasmic phospho-tyrosine, (682)YENPTY(687), binding motif. We recently demonstrated increased protein levels of APP, phosphorylated APP (Tyr682), and beta-amyloid (Abeta) in brain vasculature of atherosclerotic and Alzheimer's disease (AD) tissue colocalizing primarily within the endothelial layer. This study demonstrates similar APP changes in peripheral vasculature from human and mouse apoE(-/-) aorta, suggesting that APP-related changes are not restricted to brain vasculature. Therefore, primary mouse aortic endothelial cells and human umbilical vein endothelial cells were used as a model system to examine the function of APP in endothelial cells. APP multimerization with an anti-N-terminal APP antibody, 22C11, to simulate ligand binding stimulated an Src kinase family-dependent increase in protein phospho-tyrosine levels, APP phosphorylation, and Abeta secretion. Furthermore, APP multimerization stimulated increased protein levels of the proinflammatory proteins, cyclooxygenase-2 and vascular cell adhesion molecule-1 also in an Src kinase family-dependent manner. Endothelial APP was also involved in mediating monocytic cell adhesion. Collectively, these data demonstrate that endothelial APP regulates immune cell adhesion and stimulates a tyrosine kinase-dependent response driving acquisition of a reactive endothelial phenotype. These APP-mediated events may serve as therapeutic targets for intervention in progressive vascular changes common to cerebrovascular disease and AD.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Endothelial Cells/enzymology , Protein-Tyrosine Kinases/metabolism , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/enzymology , Aorta, Abdominal/immunology , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/immunology , Enzyme Activation/immunology , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
16.
J Vasc Surg ; 51(5): 1248-59, 2010 May.
Article in English | MEDLINE | ID: mdl-20223627

ABSTRACT

OBJECTIVE: Isopropylamine NONOate (IPA/NO) is a nitroxyl (HNO) donor at physiologic pH. HNO is a positive inotrope and vasodilator, but little is known about its effect on neointimal hyperplasia. The aims of this study are to determine the effect of IPA/NO on endothelial and vascular smooth muscle cells (VSMC) in vitro and to determine if IPA/NO inhibits neointimal hyperplasia in vivo. METHODS: VSMC were harvested from the abdominal aortas of male Sprague Dawley rats, and human umbilical vein endothelial cells were purchased from ATCC. In vitro, cellular proliferation was assessed by (3)H-thymidine incorporation, cell migration was assessed using the scrape assay, and cell death was assessed using Guava personal cell analysis (PCA). Cell cycle analysis was performed using propidium iodide staining and flow cytometry analysis. Protein expression was assessed using Western blot analysis. Phosphorylated proteins were assessed using immunoprecipitation and Western blot analysis. In vivo, the carotid artery injury model was performed on male Sprague Dawley rats treated with (n = 12) or without (n = 6) periadventitial IPA/NO (10 mg). Arteries harvested at 2 weeks were assessed for morphometrics using ImageJ. Inflammation was assessed using immunohistochemistry. Endothelialization was assessed by Evans blue staining of carotid arteries harvested 7 days after balloon injury from rats treated with (n = 6) or without (n = 3) periadventitial IPA/NO (10 mg). RESULTS: In vitro, 1000 micromol/L IPA/NO inhibited both VSMC (38.7 +/- 4.5% inhibition vs control, P = .003) and endothelial cell proliferation (54.0 +/- 2.9% inhibition vs control, P < or = 0.001) without inducing cell death or inhibiting migration. In VSMC, this inhibition was associated with an S-phase cell cycle arrest and increased expression of cyclin A, cyclin D1, and the cyclin-dependent kinase inhibitor p21. No change was noted in the phosphorylation status of cdk2, cdk4, or cdk6 by IPA/NO. In rodents subjected to the carotid artery balloon injury model, IPA/NO caused significant reductions in neointimal area (298 +/- 20 vs 422 +/- 30, P < or = .001) and medial area (311 +/- 14 vs 449 +/- 16, P < or = .001) compared with injury alone, and reduced macrophage infiltration to 1.7 +/- 0.8 from 16.1 +/- 3.5 cells per high power field (P < or = .001). IPA/NO also prevented re-endothelialization compared with injury alone (55.9 +/- 0.5% nonendothelialized vs 21 +/- 4.4%, respectively, P = .001). Lastly, a 50% mortality rate was observed in the IPA/NO-treated groups. CONCLUSIONS: In summary, while IPA/NO modestly inhibited neointimal hyperplasia by inhibiting VSMC proliferation and macrophage infiltration, it also inhibited endothelial cell proliferation and induced significant mortality in our animal model. Since HNO is being investigated as a treatment for congestive heart failure, our results raise some concerns about the use of IPA/NO in the vasculature and suggest that further studies be conducted on the safety of HNO donors in the cardiovascular system.


Subject(s)
Carotid Artery Injuries/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hydrazines/metabolism , Muscle, Smooth, Vascular/metabolism , Tunica Intima/metabolism , Tunica Intima/pathology , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/drug effects , Blotting, Western , Carotid Artery Injuries/complications , Carotid Artery Injuries/pathology , Cell Death/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Humans , Hydrazines/administration & dosage , Hyperplasia/etiology , Hyperplasia/metabolism , Hyperplasia/pathology , Hyperplasia/prevention & control , Immunohistochemistry , Male , Muscle, Smooth, Vascular/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Tunica Intima/drug effects , Umbilical Veins/cytology , Umbilical Veins/drug effects
17.
Arterioscler Thromb Vasc Biol ; 29(11): 1902-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19696400

ABSTRACT

OBJECTIVE: Vascular endothelial cells (ECs) confer atheroprotection at locations of the arterial tree where pulsatile laminar flow (PS) exists with a high shear stress and a large net forward direction. We investigated whether the PS-induced expression of the transcription factor Krüppel-Like Factor 2 (KLF2) in cultured ECs and its expression in the mouse aorta is regulated by AMP-activated protein kinase (AMPK). METHODS AND RESULTS: AMPK inhibition by Compound C or siRNA had a significant blocking effect on the PS-induced KLF2 expression. The induction of KLF2 by PS led to the increase in eNOS and the suppression of ET-1, which could be reversed by KLF2 siRNA. In addition, PS induced the phosphorylation of ERK5 and MEF2 which are necessary for the KLF2 expression. These mechanotransduction events were abrogated by the blockade of AMPK. Furthermore, the phosphorylation levels of ERK5 and MEF2, as well as the expression of KLF2, were significantly reduced in the aorta of AMPKalpha2 knockout mice when compared with wild-type control mice. CONCLUSIONS: The flow-mediated AMPK activation is a newly defined KLF2 regulatory pathway in vascular endothelium that acts via ERK5/MEF2.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Endothelial Cells/enzymology , Kruppel-Like Transcription Factors/metabolism , Analysis of Variance , Animals , Aorta, Abdominal/cytology , Cells, Cultured , DNA, Complementary/metabolism , Endothelial Cells/cytology , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Phosphorylation , Probability , Pulsatile Flow/physiology , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stress, Mechanical , Transfection , Up-Regulation
18.
Acta Pharmacol Sin ; 31(4): 413-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20348945

ABSTRACT

AIM: To investigate whether adenosine A(3) receptors (A(3)AR) stimulation restore vascular reactivity after hemorrhagic shock through a ryanodine receptor (RyR)-mediated and large conductance calcium-activated potassium (BK(Ca)) channel-dependent pathway. METHODS: Rat hemorrhagic shock model (40 mmHg) and vascular smooth muscle cell (VSMC) hypoxic model were used. The expression of A(3)AR was determined by Western blot and RT-PCR. The effect of A(3)AR stimulation on RyR-mediated Ca(2+) release in VSMCs was analyzed by the Fura-3/AM loading Ca(2+) imaging. The modulation of vascular reactivity to norepinephrine (NE) by A(3)AR stimulation was monitored by an isolated organ tension instrument. RESULTS: Decrease of A(3)AR expression is consistent with the loss of vasoreactivity to NE in hemorrhagic shock rats. The stimulation of A(3)AR with a selective agonist, IB-MECA, could partly but significantly restore the vasoreactivity in the rats, and this restorative effect could be counteracted by MRS1523, a selective A(3)AR antagonist. In hypoxic VSMCs, RyR activation by caffeine significantly evoked the rise of [Ca(2+)] compared with the control cells, a phenomenon closely associated with the development of vascular hyporeactivity in hemorrhagic shock rats. The stimulation of A(3)AR with IB-MECA significantly blocked this over activation of RyR-mediated Ca(2+) release. RyR activation by caffeine and BK(Ca) channel activation by NS1619 attenuated the restoration of vasoreactivity to NE resulting from A(3)AR stimulation by IB-MECA after hemorrhagic shock; this attenuation effect could be antagonized by a selective BK(Ca) channel blocker. CONCLUSION: These findings suggest that A(3)AR is involved in the modulation of vasoreactivity after hemorrhagic shock and that stimulation of A(3)AR can restore the decreased vasoreactivity to NE through a RyR-mediated, BK(Ca) channel-dependent signal pathway.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth, Vascular/drug effects , Norepinephrine/pharmacology , Receptor, Adenosine A3/metabolism , Shock, Hemorrhagic/metabolism , Vasoconstrictor Agents/pharmacology , Animals , Aorta, Abdominal/cytology , Calcium/metabolism , Cell Hypoxia/drug effects , Cells, Cultured , Gene Expression , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Adenosine A3/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Vasoconstriction/drug effects
19.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 41(5): 784-8, 2010 Sep.
Article in Zh | MEDLINE | ID: mdl-21302441

ABSTRACT

OBJECTIVE: To investigate the effects of high glucose on expression of core binding factor alpha1 (cbfalpha-1) and osteocalcin (OC) in vascular smooth muscle cells (VSMCs), and discuss the mechanism of small vessels calcification induced by high glucose (GS) in vitro. METHODS: The primary cultured VSMCs from rats' aortic segments were divided into three groups, including normal control group (5 mmol/L D-glucose), high glucose group (25 mmol/L D-glucose) and mannitol group (5 mmol/L D-glucose plus 25 mmol/L mannitol). We measured quantitatively the calcium deposition in VSMCs and investigated the calcium extent of VSMCs by alizarin red stain in each group. The mRNA levels of cbfalpha-1 and OC were measured by real-time PCR, and the protein expression levels of cbfalpha-1 and OC were examined by Western blot. The activity of alkaline phosphatase was measured by alkaline phosphatase activity testing kit, and the protein level of alpha-smooth muscle actin (a-SMA) was detected by immunohistochemistry. RESULTS: When compared with the normal group and mannitol group, the high glucose group showed that the calcium deposition and calcium extent of VSMCs increased obviously, the mRNA and protein levels of cbfalpha-1 and OC also increased significantly (P < 0.05), while the protein level of alpha-SMA decreased (P < 0.05), which were in a dose-dependent manner. The level of alkaline phosphatase activity of VSMCs was approximately doubled in high glucose group. CONCLUSION: The mechanism of high glucose induced calcification in VSMCs may be due to the increased expression of cbfalpha-1 and OC. High glucose decrease the expression of alpha-SMA in VSMCs, which could induce the transdifferentiation from RVSMCs to osteoblast-like cells.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Glucose/pharmacology , Muscle, Smooth, Vascular/metabolism , Osteocalcin/metabolism , Animals , Aorta, Abdominal/cytology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Muscle, Smooth, Vascular/cytology , Osteocalcin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
20.
Aging (Albany NY) ; 12(13): 12684-12702, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32629426

ABSTRACT

Long non-coding RNAs (lncRNAs) have been largely reported to contribute to the development and progression of abdominal aortic aneurysm (AAA), a common vascular degenerative disease. The present study was set out with the aim to investigate the possible role of lncRNA Sox2ot in the development of AAA. In this study, we found that lncRNA Sox2ot and early growth response factor-1 (Egr1) were highly expressed, while microRNA (miR)-145 was poorly expressed in Ang II-induced AAA mice and oxidative stress-provoked vascular smooth muscle cell (VSMC) model. Egr1 was a potential target gene of miR-145, and lncRNA Sox2ot could competitively bind to miR-145 to upregulate Egr1 expression. Overexpression of miR-145-5p was found to attenuate oxidative stress and inflammation by inhibiting Egr1 both in vivo and in vitro, which was counteracted by lncRNA Sox2ot. Taken together, the present study provides evidence that downregulation of lncRNA Sox2ot suppressed the expression of Egr1 through regulating miR-145, thus inhibiting the development of AAA, highlighting a theoretical basis for AAA treatment.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Early Growth Response Protein 1/metabolism , MicroRNAs/metabolism , Oxidative Stress/genetics , RNA, Long Noncoding , Animals , Aorta, Abdominal/cytology , Aorta, Abdominal/metabolism , Apolipoproteins E/genetics , Cells, Cultured , Down-Regulation , Early Growth Response Protein 1/antagonists & inhibitors , Gene Silencing , Inflammation , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL