Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 345
Filter
1.
Regen Biomater ; 11: rbae042, 2024.
Article in English | MEDLINE | ID: mdl-39027361

ABSTRACT

In-stent restenosis can be caused by the activation, proliferation and migration of vascular smooth muscle cells (VSMCs), which affects long-term efficacy of interventional therapy. Copper (Cu) has been proved to accelerate the endothelialization and reduce thrombosis formation, but little is known about its inhibition effect on the excessive proliferation of VSMCs. In this study, 316L-Cu stainless steel and L605-Cu cobalt-based alloy with varying Cu content were fabricated and their effects on surface property, blood compatibility and VSMCs were studied in vitro and in vivo. CCK-8 assay and EdU assay indicated that the Cu-bearing metals had obvious inhibitory effect on proliferation of VSMCs. Blood clotting and hemolysis tests showed that the Cu-bearing metals had good blood compatibility. The inhibition effect of the Cu-bearing metals on migration of cells was detected by Transwell assay. Further studies showed that Cu-bearing metals significantly decreased the mRNA expressions of bFGF, PDGF-B, HGF, Nrf2, GCLC, GCLM, NQO1 and HO1. The phosphorylation of AKT and Nrf2 protein expressions in VSMCs were significantly decreased by Cu-bearing metals. Furthermore, it was also found that SC79 and TBHQ treatments could recover the protein expressions of phospho-AKT and Nrf2, and their downstream proteins as well. Moreover, 316L-Cu stent proved its inhibitory action on the proliferation of VSMCs in vivo. In sum, the results demonstrated that the Cu-bearing metals possessed apparent inhibitory effect on proliferation and migration of VSMCs via regulating the AKT/Nrf2/ARE pathway, showing the Cu-bearing metals as promising stent materials for long-term efficacy of implantation.

2.
Mater Today Bio ; 27: 101133, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39027680

ABSTRACT

In-stent restenosis (ISR), caused by aggressive vascular smooth muscle cell (VSMC) proliferation, is a serious complication of stenting. Therefore, developing therapeutic approaches that target VSMC inhibition is imperative. Our previous study showed that VSMC hyperplasia was attenuated after iron stent degradation, and VSMC proliferation around the stented section was arrested. The corrosion products of the iron stents were primarily Fe3O4 particles. Therefore, we hypothesized that Fe3O4 particles generated by iron stents would prevent neointimal hyperplasia by inhibiting VSMC proliferation. To test this hypothesis, culture assays and flow cytometry were performed to investigate the proliferation of VSMC. Global gene sequencing and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to investigate the underlying mechanisms. Fe3O4-coated stents were implanted into rabbit carotid arteries to evaluate the inhibitory effects of Fe3O4 on neointimal hyperplasia. The major findings of the study were as follows: 1) Fe3O4 attenuated neointimal hyperplasia by preventing VSMC proliferation after stenting; 2) Fe3O4 exerted inhibitory effects on VSMCs by downregulating proliferative genes such as SOX9, EGR4, and TGFB1, but upregulated inhibitory genes such as DNMT1, TIMP3, and PCNA; 3) Fe3O4 inhibited VSMCs by preventing phenotypic transformation from the contractile to the synthetic phase; and 4) Fe3O4-coated stents achieved satisfactory hemocompatibility in a rabbit model. Our study highlights the additional benefits of Fe3O4 particles in inhibiting VSMC proliferation, indicating that Fe3O4 coated stent potentially served as an attractive therapeutic approach for ISR prevention.

3.
Comput Methods Programs Biomed ; 255: 108327, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39018788

ABSTRACT

BACKGROUND AND OBJECTIVE: Atherosclerotic lesions of coronary arteries (stenosis) are caused by the buildup of lipids and blood-borne substances within the artery wall. Their qualitative and rapid assessment is still a challenging task. The primary therapy for this pathology involves implanting coronary stents, which help to restore the blood flow in atherosclerosis-prone arteries. In-stent restenosis is a stenting-procedure complication detected in about 10-40% of patients. A numerical study using 2-way fluid-structure interaction (FSI) assesses the stenting procedure quality and can decrease the number of negative post-operative results. Nevertheless, boundary conditions (BCs) used in simulation play a crucial role in implementation of an adequate computational analysis. METHODS: Three CoCr stents designs were modelled with the suggested approach. A multi-layer structure describing the artery and plaque with anisotropic hyperelastic mechanical properties was adopted in this study. Two kinds of boundary conditions for a solid domain were examined - fixed support (FS) and remote displacement (RD) - to assess their impact on the hemodynamic parameters to predict restenosis. Additionally, the influence of artery elongation (short-artery model vs. long-artery model) on numerical results with the FS boundary condition was analyzed. RESULTS: The comparison of FS and RD boundary conditions demonstrated that the variation of hemodynamic parameters values did not exceed 2%. The analysis of short-artery and long-artery models revealed that the difference in hemodynamic parameters was less than 5.1%, and in most cases, it did not exceed 2.5%. The RD boundary conditions were found to reduce the computation time by up to 1.7-2.0 times compared to FS. Simple stent model was shown to be susceptible to restenosis development, with maximum WSS values equal to 183 Pa, compared to much lower values for other two stents. CONCLUSIONS: The study revealed that the stent design significantly affected the hemodynamic parameters as restenosis predictors. Moreover, the stress-strain state of the system artery-plaque-stent also depends on a proper choice of boundary conditions.

4.
Biomed Pharmacother ; 177: 117022, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38917756

ABSTRACT

BACKGROUND: The transplantation of endothelial progenitor cells (EPCs) has been shown to reduce neointimal hyperplasia following arterial injury. However, the efficacy of this approach is hampered by limited homing of EPCs to the injury site. Additionally, the in vivo recruitment and metabolic activity of transplanted EPCs have not been continuously monitored. METHODS: EPCs were labeled with indocyanine green (ICG)-conjugated superparamagnetic iron oxide nanoparticles (SPIONs) and subjected to external magnetic field targeting to enhance their delivery to a carotid balloon injury (BI) model in Sprague-Dawley rats. Magnetic particle imaging (MPI)/ fluorescence imaging (FLI) multimodal in vivo imaging, 3D MPI/CT imaging and MPI/FLI ex vivo imaging was performed after injury. Carotid arteries were collected and analyzed for pathology and immunofluorescence staining. The paracrine effects were analyzed by enzyme-linked immunosorbent assay. RESULTS: The application of a magnetic field significantly enhanced the localization and retention of SPIONs@PEG-ICG-EPCs at the site of arterial injury, as evidenced by both in vivo continuous monitoring and ex vivo by observation. This targeted delivery approach effectively inhibited neointimal hyperplasia and increased the presence of CD31-positive cells at the injury site. Moreover, serum levels of SDF-1α, VEGF, IGF-1, and TGF-ß1 were significantly elevated, indicating enhanced paracrine activity. CONCLUSIONS: Our findings demonstrate that external magnetic field-directed delivery of SPIONs@PEG-ICG-EPCs to areas of arterial injury can significantly enhance their therapeutic efficacy. This enhancement is likely mediated through increased paracrine signaling. These results underscore the potential of magnetically guided SPIONs@PEG-ICG-EPCs delivery as a promising strategy for treating arterial injuries.

5.
J Vasc Access ; : 11297298241256172, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836593

ABSTRACT

BACKGROUND: To investigate the technical advantages of a modified no-touch technique (MNTT) in constructing arteriovenous fistulas (AVF) compared to the conventional technique (CT) and assess its potential to reduce neointimal hyperplasia in the outflow vein. METHODS: Forty-seven New Zealand rabbits were randomly divided into three groups: control, CT, and MNTT. Rabbits in control group were observed using ultrasound and then euthanized to obtain external jugular vein (EJV) for Hematoxylin-eosin (H-E). We established common carotid artery (CCA)-EJV AVF using MNTT in the MNTT group and the CT in the CT group. AVF patency and complications were compared between the CT and MNTT groups. Rabbits with patent AVF in both groups were observed using ultrasound 2 weeks after surgery to evaluate changes in the vessel diameter and blood flow spectrum of the AVFs. H-E staining measured the intima thickness of EJV adjacent to the anastomosis and histologic characteristics of the AVF at 2 and 4 weeks after surgery. RESULTS: Five rabbits died after surgery with common symptoms of sneezing, coughing, runny nose, anorexia, and diarrhea; two in the MNTT group and three in the CT group. There were significant differences in the diameter (p = 0.010) and peak systolic velocities (PSV) (p = 0.001) of EJV between the CT and MNTT groups 2 weeks after surgery. Spiral laminar flow (SLF) was observed in CCA and EJV adjacent to anastomosis in the MNTT group. Additionally, histological observations showed less venous neointimal hyperplasia in the MNTT group than in the CT group 4 weeks after surgery. CONCLUSION: The rabbit model of CCA-EJV AVF established using MNTT demonstrated fewer complications, larger vein diameters, and reduced venous neointimal hyperplasia, indicating that this maybe an ideal animal model to further investigate the application of MNTT in AVF surgery.

6.
Nanomedicine ; 61: 102763, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38897395

ABSTRACT

The primary challenge in percutaneous coronary interventions for vascular restenosis is the occurrence of restenosis, which is defined by the excessive proliferation of neointimal tissue. Herein, our research team suggests that exosomes obtained from PSC, when paired with quercetin (Q@PSC-E), successfully reduce neointimal hyperplasia in a Sprague-Dawley rat model. Furthermore, the physical properties of the synthesized Q@PSC-E were examined using UV-vis, DLS, and FT-IR characterization techniques. The rats were subjected to balloon injury (BI) utilizing a 2-Fr Fogarty arterial embolectomy balloon catheter. Intimal hyperplasia and the degree of VSMC proliferation were evaluated using histological analysis in the rat groups that received a dosage of Q@PSC-E at 30 mg/kg/d. Significantly, Q@PSC-E inhibited cell proliferation through a pathway that does not include lipoxygenase, as demonstrated by [3H] thymidine incorporation, MTT, and flow cytometry studies. Additionally, the data indicate that Q@PSC-E hinders cell proliferation by targeting particular events that promote cell growth, including the activation of Akt and NF-κB, disruption of cell-cycle progression and also obstructs the ERK signaling pathway.

7.
ACS Appl Mater Interfaces ; 16(26): 33159-33168, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38912610

ABSTRACT

In the context of arteriovenous fistula (AVF) failure, local delivery enables the release of higher concentrations of drugs that can suppress neointimal hyperplasia (NIH) while reducing systemic adverse effects. However, the radiolucency of polymeric delivery systems hinders long-term in vivo surveillance of safety and efficacy. We hypothesize that using a radiopaque perivascular wrap to deliver anti-NIH drugs could enhance AVF maturation. Through electrospinning, we fabricated multifunctional perivascular polycaprolactone (PCL) wraps loaded with bismuth nanoparticles (BiNPs) for enhanced radiologic visibility and drugs that can attenuate NIH─rosuvastatin (Rosu) and rapamycin (Rapa). The following groups were tested on the AVFs of a total of 24 Sprague-Dawley rats with induced chronic kidney disease: control (i.e., without wrap), PCL-Bi (i.e., wrap with BiNPs), PCL-Bi-Rosu, and PCL-Bi-Rapa. We found that BiNPs significantly improved the wraps' radiopacity without affecting biocompatibility. The drug release profiles of Rosu (hydrophilic drug) and Rapa (hydrophobic drug) differed significantly. Rosu demonstrated a burst release followed by gradual tapering over 8 weeks, while Rapa demonstrated a gradual release similar to that of the hydrophobic BiNPs. In vivo investigations revealed that both drug-loaded wraps can reduce vascular stenosis on ultrasonography and histomorphometry, as well as reduce [18F]Fluorodeoxyglucose uptake on positron emission tomography. Immunohistochemical studies revealed that PCL-Bi-Rosu primarily attenuated endothelial dysfunction and hypoxia in the neointimal layer, while PCL-Bi-Rapa modulated hypoxia, inflammation, and cellular proliferation across the whole outflow vein. In summary, the controlled delivery of drugs with different properties and mechanisms of action against NIH through a multifunctional, radiopaque perivascular wrap can improve imaging and histologic parameters of AVF maturation.


Subject(s)
Bismuth , Rats, Sprague-Dawley , Rosuvastatin Calcium , Sirolimus , Animals , Rats , Sirolimus/chemistry , Sirolimus/pharmacology , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacology , Rosuvastatin Calcium/pharmacokinetics , Bismuth/chemistry , Bismuth/pharmacology , Polyesters/chemistry , Male , Arteriovenous Fistula/pathology , Metal Nanoparticles/chemistry , Neointima/pathology , Nanoparticles/chemistry , Humans , Drug Liberation
8.
Mol Cells ; 47(6): 100075, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38823606

ABSTRACT

Excessive blood vessel wall thickening, known as intimal hyperplasia, can result from injury or inflammation and increase the risk of vascular diseases. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) plays key roles in tumor surveillance, autoimmune diseases, and apoptosis; however, its role in vascular stenosis remains controversial. Treatment with recombinant isoleucine zipper hexamerization domain soluble TRAIL (ILz(6):TRAIL) significantly inhibited the progression of neointimal hyperplasia (NH) induced by anastomosis of the carotid artery and jugular vein dose dependently, and adenovirus expressing secretable ILz(6):TRAIL also inhibited NH induced by balloon injury in the femoral artery of rats. This study demonstrated the preventive and partial regressive effects of ILz(6):TRAIL on anastomosis of the carotid artery and jugular vein- or balloon-induced NH.


Subject(s)
Hyperplasia , Neointima , Rats, Sprague-Dawley , TNF-Related Apoptosis-Inducing Ligand , Animals , Neointima/pathology , Neointima/prevention & control , Rats , Male , TNF-Related Apoptosis-Inducing Ligand/metabolism , Carotid Arteries/pathology , Carotid Arteries/surgery , Jugular Veins/pathology , Femoral Artery/injuries , Femoral Artery/pathology , Femoral Artery/surgery
9.
Phytomedicine ; 130: 155704, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38759316

ABSTRACT

BACKGROUND: Dysregulation of vascular smooth muscle cell (VSMC) function leads to a variety of diseases such as atherosclerosis and hyperplasia after injury. However, antiproliferative drug targeting VSMC exhibits poor specificity. Therefore, there is an urgent to develop highly specific antiproliferative drugs to prevention and treatment VSMC dedifferentiation associated arteriosclerosis. Kanglexin (KLX), a new anthraquinone compound designed by our team, has potential to regulate VSMC phenotype according to the physicochemical properties. PURPOSE: This project aims to evaluate the therapeutic role of KLX in VSMC dedifferentiation and atherosclerosis, neointimal formation and illustrates the underlying molecular mechanism. METHODS: In vivo, the ApoE-/- mice were fed with high-fat diet (HFD) for a duration of 13 weeks to establish the atherosclerotic model. And rat carotid artery injury model was performed to establish the neointimal formation model. In vitro, PDGF-BB was used to induce VSMC dedifferentiation. RESULTS: We found that KLX ameliorated the atherosclerotic progression including atherosclerotic lesion formation, lipid deposition and collagen deposition in aorta and aortic sinus in atherosclerotic mouse model. In addition, The administration of KLX effectively ameliorated neointimal formation in the carotid artery following balloon injury in SD rats. The findings derived from molecular docking and surface plasmon resonance (SPR) experiments unequivocally demonstrate that KLX had potential to bind PDGFR-ß. Mechanism research work proved that KLX prevented VSMC proliferation, migration and dedifferentiation via activating the PDGFR-ß-MEK -ERK-ELK-1/KLF4 signaling pathway. CONCLUSION: Collectively, we demonstrated that KLX effectively attenuated the progression of atherosclerosis in ApoE-/- mice and carotid arterial neointimal formation in SD rats by inhibiting VSMC phenotypic conversion via PDGFR-ß-MEK-ERK-ELK-1/KLF4 signaling. KLX exhibits promising potential as a viable therapeutic agent for the treatment of VSMC phenotype conversion associated arteriosclerosis.


Subject(s)
Anthraquinones , Cell Dedifferentiation , Kruppel-Like Factor 4 , Muscle, Smooth, Vascular , Neointima , Animals , Male , Mice , Rats , Anthraquinones/pharmacology , Arteriosclerosis/drug therapy , Arteriosclerosis/prevention & control , Atherosclerosis/drug therapy , Becaplermin/pharmacology , Carotid Artery Injuries/drug therapy , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Diet, High-Fat , Disease Models, Animal , Kruppel-Like Transcription Factors/metabolism , Mice, Inbred C57BL , Molecular Docking Simulation , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima/drug therapy , Rats, Sprague-Dawley , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects
10.
Front Biosci (Landmark Ed) ; 29(5): 173, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38812305

ABSTRACT

BACKGROUND: Neointimal hyperplasia (NIH) is the pathological basis of vascular injury disease. Vascular cells are the dominant cells in the process of NIH, but the extent of heterogeneity amongst them is still unclear. METHODS: A mouse model of NIH was constructed by inducing carotid artery ligation. Single-cell sequencing was then used to analyze the transcriptional profile of vascular cells. Cluster features were determined by functional enrichment analysis, gene set scoring, pseudo-time analysis, and cell-cell communication analysis. Additionally, immunofluorescence staining was conducted on vascular tissues from fibroblast lineage-traced (PdgfraDreER-tdTomato) mice to validate the presence of Pecam1+Pdgfra+tdTomato+ cells. RESULTS: The left carotid arteries (ligation) were compared to right carotid arteries (sham) from ligation-induced NIH C57BL/6 mice. Integrative analyses revealed a high level of heterogeneity amongst vascular cells, including fourteen clusters and seven cell types. We focused on three dominant cell types: endothelial cells (ECs), vascular smooth muscle cells (vSMCs), and fibroblasts. The major findings were: (1) four subpopulations of ECs, including ECs4, mesenchymal-like ECs (ECs1 and ECs2), and fibro-like ECs (ECs3); (2) four subpopulations of fibroblasts, including pro-inflammatory Fibs-1, Sca1+ Fibs-2, collagen-producing Fibs-3, and mesenchymal-like Fibs-4; (3) four subpopulations of vSMCs, including vSMCs-1, vSMCs-2, vSMCs-3, and vSMCs-3-derived vSMCs; (4) ECs3 express genes related to extracellular matrix (ECM) remodeling and cell migration, and fibro-like vSMCs showed strong chemokine secretion and relatively high levels of proteases; (5) fibro-like vSMCs that secrete Vegfa interact with ECs mainly through vascular endothelial growth factor receptor 2 (Vegfr2). CONCLUSIONS: This study presents the dynamic cellular landscape within NIH arteries and reveals potential relationships between several clusters, with a specific focus on ECs3 and fibro-like vSMCs. These two subpopulations may represent potential target cells for the treatment of NIH.


Subject(s)
Gene Expression Profiling , Hyperplasia , Mice, Inbred C57BL , Muscle, Smooth, Vascular , Neointima , Single-Cell Analysis , Animals , Neointima/pathology , Neointima/metabolism , Neointima/genetics , Single-Cell Analysis/methods , Hyperplasia/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/cytology , Mice , Endothelial Cells/metabolism , Endothelial Cells/pathology , Carotid Arteries/pathology , Carotid Arteries/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Male , Fibroblasts/metabolism , Fibroblasts/pathology , Disease Models, Animal , Single-Cell Gene Expression Analysis
11.
Am J Physiol Heart Circ Physiol ; 326(6): H1446-H1461, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38578237

ABSTRACT

Clinical failure of arteriovenous neointimal hyperplasia (NIH) fistulae (AVF) is frequently due to juxta-anastomotic NIH (JANIH). Although the mouse AVF model recapitulates human AVF maturation, previous studies focused on the outflow vein distal to the anastomosis. We hypothesized that the juxta-anastomotic area (JAA) has increased NIH compared with the outflow vein. AVF was created in C57BL/6 mice without or with chronic kidney disease (CKD). Temporal and spatial changes of the JAA were examined using histology and immunofluorescence. Computational techniques were used to model the AVF. RNA-seq and bioinformatic analyses were performed to compare the JAA with the outflow vein. The jugular vein to carotid artery AVF model was created in Wistar rats. The neointima in the JAA shows increased volume compared with the outflow vein. Computational modeling shows an increased volume of disturbed flow at the JAA compared with the outflow vein. Endothelial cells are immediately lost from the wall contralateral to the fistula exit, followed by thrombus formation and JANIH. Gene Ontology (GO) enrichment analysis of the 1,862 differentially expressed genes (DEG) between the JANIH and the outflow vein identified 525 overexpressed genes. The rat jugular vein to carotid artery AVF showed changes similar to the mouse AVF. Disturbed flow through the JAA correlates with rapid endothelial cell loss, thrombus formation, and JANIH; late endothelialization of the JAA channel correlates with late AVF patency. Early thrombus formation in the JAA may influence the later development of JANIH.NEW & NOTEWORTHY Disturbed flow and focal endothelial cell loss in the juxta-anastomotic area of the mouse AVF colocalizes with acute thrombus formation followed by late neointimal hyperplasia. Differential flow patterns between the juxta-anastomotic area and the outflow vein correlate with differential expression of genes regulating coagulation, proliferation, collagen metabolism, and the immune response. The rat jugular vein to carotid artery AVF model shows changes similar to the mouse AVF model.


Subject(s)
Arteriovenous Shunt, Surgical , Hyperplasia , Jugular Veins , Mice, Inbred C57BL , Neointima , Rats, Wistar , Thrombosis , Animals , Thrombosis/physiopathology , Thrombosis/pathology , Thrombosis/genetics , Thrombosis/etiology , Thrombosis/metabolism , Male , Jugular Veins/metabolism , Jugular Veins/pathology , Jugular Veins/physiopathology , Disease Models, Animal , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Carotid Arteries/metabolism , Carotid Arteries/surgery , Mice , Rats , Regional Blood Flow , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Endothelium, Vascular/pathology , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology
12.
J Heart Lung Transplant ; 43(8): 1336-1347, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38670297

ABSTRACT

BACKGROUND: Cardiac allograft vasculopathy (CAV) remains the leading cause of long-term graft failure and mortality after heart transplantation. Effective preventive and treatment options are not available to date, largely because underlying mechanisms remain poorly understood. We studied the potential role of leukotriene B4 (LTB4), an inflammatory lipid mediator, in the development of CAV. METHODS: We used an established preclinical rat CAV model to study the role of LTB4 in CAV. We performed syngeneic and allogeneic orthotopic aortic transplantation, after which neointimal proliferation was quantified. Animals were then treated with Bestatin, an inhibitor of LTB4 synthesis, or vehicle control for 30 days post-transplant, and evidence of graft CAV was determined by histology. We also measured serial LTB4 levels in a cohort of 28 human heart transplant recipients with CAV, 17 matched transplant controls without CAV, and 20 healthy nontransplant controls. RESULTS: We showed that infiltration of the arterial wall with macrophages leads to neointimal thickening and a rise in serum LTB4 levels in our rat model of CAV. Inhibition of LTB4 production with the drug Bestatin prevents development of neointimal hyperplasia, suggesting that Bestatin may be effective therapy for CAV prevention. In a parallel study of heart transplant recipients, we found nonsignificantly elevated plasma LTB4 levels in patients with CAV, compared to patients without CAV and healthy, nontransplant controls. CONCLUSIONS: This study provides key evidence supporting the role of the inflammatory cytokine LTB4 as an important mediator of CAV development and provides preliminary data suggesting the clinical benefit of Bestatin for CAV prevention.


Subject(s)
Biomarkers , Heart Transplantation , Leukotriene B4 , Animals , Heart Transplantation/adverse effects , Leukotriene B4/blood , Leukotriene B4/metabolism , Rats , Male , Biomarkers/metabolism , Biomarkers/blood , Humans , Disease Models, Animal , Allografts , Middle Aged , Rats, Inbred Lew , Female , Neointima/pathology
13.
Microvasc Res ; 154: 104688, 2024 07.
Article in English | MEDLINE | ID: mdl-38640999

ABSTRACT

Arteriovenous fistula (AVF) failure often involves venous neointimal hyperplasia (VNH) driven by elevated hypoxia-inducible factor-1 alpha (HIF-1α) in the venous wall. Omentin, known for its anti-inflammatory and anti-hyperplasia properties, has an uncertain role in early AVF failure. This study investigates omentin's impact on VNH using a chronic renal failure (CRF) rabbit model. The CRF rabbit model of AVF received omentin-expressing adenoviral vector or control ß-gal vector to assess omentin's effects on VNH. Human vascular smooth muscle cells (HVSMCs), stimulated with tumor necrosis factor-α (TNF-α), were exposed to recombinant human omentin (Rh-OMT) to study its influence on cell proliferation and migration. The AMP-activated protein kinase (AMPK) inhibitor compound C and the mammalian target of rapamycin (mTOR) activator MHY1485 were employed to explore omentin's mechanisms in VNH reduction through HIF-1α inhibition. Omentin treatment reduced VNH in CRF rabbits, concomitant with HIF-1α down-regulation and the suppression of downstream factors, including vascular endothelial growth factor and matrix metalloproteinases. Rh-OMT inhibited TNF-α-induced HVSMC proliferation and migration by modulating both cell cycle and cell adhesion proteins. Additionally, omentin reduced HIF-1α expression through the AMPK/mTOR pathway activation. Notably, the blockade of AMPK/mTOR signaling reversed omentin-mediated inhibition of VNH, cell proliferation, and migration, both in vivo and in vitro. In conclusion, omentin mitigates VNH post-AVF creation by restraining HIF-1α via AMPK/mTOR signaling. Strategies boosting circulating omentin levels may offer promise in averting AVF failure.


Subject(s)
AMP-Activated Protein Kinases , Arteriovenous Shunt, Surgical , Cell Movement , Cell Proliferation , Cytokines , Disease Models, Animal , GPI-Linked Proteins , Hyperplasia , Hypoxia-Inducible Factor 1, alpha Subunit , Lectins , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Neointima , Signal Transduction , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cytokines/metabolism , Rabbits , Humans , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/pharmacology , GPI-Linked Proteins/genetics , Cell Proliferation/drug effects , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Lectins/pharmacology , Lectins/metabolism , Cell Movement/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , AMP-Activated Protein Kinases/metabolism , Cells, Cultured , Arteriovenous Shunt, Surgical/adverse effects , Male , Kidney Failure, Chronic/pathology , TOR Serine-Threonine Kinases/metabolism , Graft Occlusion, Vascular/pathology , Graft Occlusion, Vascular/prevention & control , Graft Occlusion, Vascular/metabolism , Graft Occlusion, Vascular/physiopathology , Jugular Veins/pathology , Jugular Veins/metabolism , Jugular Veins/transplantation
14.
Cancers (Basel) ; 16(7)2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38611112

ABSTRACT

TNF-α functions as a master regulator of inflammation, and it plays a prominent role in several immunological diseases. By promoting important cellular mechanisms, such as cell proliferation, migration, and phenotype switch, TNF-α induces its exacerbating effects, which are the underlying cause of many proliferative diseases such as cancer and cardiovascular disease. TNF-α primarily alters the immune component of the disease, which subsequently affects normal functioning of the cells. Monoclonal antibodies and synthetic drugs that can target TNF-α and impair its effects have been developed and are currently used in the treatment of a few select human diseases. Vascular restenosis is a proliferative disorder that is initiated by immunological mechanisms. In this review, the role of TNF-α in exacerbating restenosis resulting from neointimal hyperplasia, as well as molecular mechanisms and cellular processes affected or induced by TNF-α, are discussed. As TNF-α-targeting drugs are currently not approved for the treatment of restenosis, the summation of the topics discussed here is anticipated to provide information that can emphasize on the use of TNF-α-targeting drug candidates to prevent vascular restenosis.

15.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167099, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38428686

ABSTRACT

The abnormal proliferation, migration, and inflammation of vascular smooth muscle cells (VSMCs) play crucial roles in the development of neointimal hyperplasia and restenosis. Exposure to inflammatory cytokines such as platelet-derived growth factor (PDGF)-BB and tumour necrosis factor-alpha (TNF-α) induces the transformation of contractile VSMCs into abnormal synthetic VSMCs. Isoxanthohumol (IXN) has significant anti-inflammatory, antiproliferative, and antimigratory effects. This study aimed to explore the therapeutic impact and regulatory mechanism of IXN in treating neointimal hyperplasia. The present findings indicate that IXN effectively hinders the abnormal proliferation, migration, and inflammation of VSMCs triggered by PDGF or TNF-α. This inhibition is primarily achieved through the modulation of the apelin/AKT or AKT pathway, respectively. In an in vivo model, IXN effectively reduced neointimal hyperplasia in denuded femoral arteries. These results suggest that IXN holds promise as a potential and innovative therapeutic candidate for the treatment of restenosis.


Subject(s)
Proto-Oncogene Proteins c-akt , Tumor Necrosis Factor-alpha , Xanthones , Humans , Hyperplasia/drug therapy , Cell Proliferation , Proto-Oncogene Proteins c-akt/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Apelin , Cell Movement , Becaplermin/pharmacology , Neointima/drug therapy , Neointima/metabolism , Inflammation
16.
Atherosclerosis ; 391: 117480, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38447436

ABSTRACT

BACKGROUND AND AIMS: Vascular injury-induced endothelium-denudation and profound vascular smooth muscle cells (VSMCs) proliferation and dis-regulated apoptosis lead to post-angioplasty restenosis. Coptisine (CTS), an isoquinoline alkaloid, has multiple beneficial effects on the cardiovascular system. Recent studies identified it selectively inhibits VSMCs proliferation. However, its effects on neointimal hyperplasia, re-endothelialization, and the underlying mechanisms are still unclear. METHODS: Cell viability was assayed by 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and cell counting kit-8 (CCK-8). Cell proliferation and apoptosis were measured by flow cytometry and immunofluorescence of Ki67 and TUNEL. Quantitative phosphoproteomics (QPP) was employed to screen CTS-responsive phosphor-sites in the key regulators of cell proliferation and apoptosis. Neointimal hyperplasia was induced by balloon injury of rat left carotid artery (LCA). Adenoviral gene transfer was conducted in both cultured cells and LCA. Re-endothelialization was evaluated by Evan's blue staining of LCA. RESULTS: 1) CTS had strong anti-proliferative and pro-apoptotic effects in cultured rat VSMCs, with the EC50 4∼10-folds lower than that in endothelial cells (ECs). 2) Rats administered with CTS, either locally to LCA's periadventitial space or orally, demonstrated a potently inhibited balloon injury-induced neointimal hyperplasia, but had no delaying effect on re-endothelialization. 3) The QPP results revealed that the phosphorylation levels of Pak1S144/S203, Pak2S20/S197, Erk1T202/Y204, Erk2T185/Y187, and BadS136 were significantly decreased in VSMCs by CTS. 4) Adenoviral expression of phosphomimetic mutants Pak1D144/D203/Pak2D20/D197 enhanced Pak1/2 activities, stimulated the downstream pErk1T202/Y204/pErk2T185/Y187/pErk3S189/pBadS136, attenuated CTS-mediated inhibition of VSMCs proliferation and promotion of apoptosis in vitro, and potentiated neointimal hyperplasia in vivo. 5) Adenoviral expression of phosphoresistant mutants Pak1A144/A203/Pak2A20/A197 inactivated Pak1/2 and totally simulated the inhibitory effects of CTS on platelet-derived growth factor (PDGF)-stimulated VSMCs proliferation and PDGF-inhibited apoptosis in vitro and neointimal hyperplasia in vivo. 6) LCA injury significantly enhanced the endogenous phosphorylation levels of all but pBadS136. CTS markedly attenuated all the enhanced levels. CONCLUSIONS: These results indicate that CTS is a promising medicine for prevention of post-angioplasty restenosis without adverse impact on re-endothelialization. CTS-directed suppression of pPak1S144/S203/pPak2S20/S197 and the subsequent effects on downstream pErk1T202/Y204/pErk2T185/Y187/pErk3S189 and pBadS136 underline its mechanisms of inhibition of VSMCs proliferation and stimulation of apoptosis. Therefore, the phosphor-sites of Pak1S144/S203/Pak2S20/S197 constitute a potential drug-screening target for fighting neointimal hyperplasia restenosis.


Subject(s)
Berberine/analogs & derivatives , Carotid Artery Injuries , Muscle, Smooth, Vascular , Rats , Animals , Hyperplasia/pathology , Muscle, Smooth, Vascular/pathology , Endothelial Cells/metabolism , Cell Proliferation , Neointima/metabolism , Carotid Artery Injuries/pathology , Cells, Cultured , Myocytes, Smooth Muscle/pathology , Cell Movement
17.
JVS Vasc Sci ; 5: 100191, 2024.
Article in English | MEDLINE | ID: mdl-38510938

ABSTRACT

Objective: A central arteriovenous fistula (AVF) has been proposed as a potential novel solution to treat patients with refractory hypertension. We hypothesized that venous remodeling after AVF creation in the hypertensive environment reduces systemic blood pressure but results in increased AVF wall thickness compared with remodeling in the normotensive environment. Methods: A central AVF was performed in C57BL6/J mice previously made hypertensive with angiotensin II (Ang II); mice were sacrificed on postoperative day 7 or 21. Results: In mice treated with Ang II alone, the mean systolic blood pressure increased from 90 ± 5 mmHg to 160 ± 5 mmHg at day 21; however, in mice treated with both Ang II and an AVF, the blood pressure decreased with creation of an AVF. There were significantly more PCNA-positive cells, SM22α/PCNA-positive cells, collagen I deposition, and increased Krüppel-like Factor 2 immunoreactivity in hypertensive mice with an AVF compared with normotensive mice with an AVF. Conclusions: These data show that a central AVF decreases systemic hypertension as well as induces local alterations in venous remodeling.

18.
J Vasc Res ; 61(2): 89-98, 2024.
Article in French | MEDLINE | ID: mdl-38368869

ABSTRACT

INTRODUCTION: Vascular prosthetic grafts are widely used in vascular surgery; however, graft infection remains a major concern. Silver-coated vascular grafts have demonstrated anti-infection properties in clinical settings; however, whether the silver irons influence foreign body reaction or neointimal hyperplasia remains unclear. METHODS: Sodium alginate and hyaluronic acid (SA/HA) hydrogel patches loaded with rhodamine, with or without silver, were fabricated. Patches were implanted in the subcutaneous or abdominal cavity and inferior vena cava of rats. Samples were harvested on day 14 and examined via immunohistochemical and immunofluorescence analyses. RESULTS: Silver hydrogel was found to decrease the foreign body reaction; after subcutaneous and abdominal cavity implantation in rats, the capsule was found to be thinner in the silver hydrogel group than in the control hydrogel group. The silver hydrogel group had fewer CD68-positive cells and proliferating cell nuclear antigen and interleukin-33 (IL-33) dual-positive cells than the control hydrogel group. Additionally, the silver hydrogel patch reduced the neointimal thickness after patch venoplasty in rats, and the number of IL-33- and IL-1ß-positive cells was lower than that in the control patch. CONCLUSION: Silver-loaded SA/HA hydrogel patches decreased the foreign body reaction and venous neointimal hyperplasia in rats by the inhibition of IL-33 expression.


Subject(s)
Interleukin-33 , Silver , Rats , Animals , Hyperplasia , Neointima , Foreign-Body Reaction/etiology , Foreign-Body Reaction/prevention & control , Hydrogels
19.
Article in English | MEDLINE | ID: mdl-38409474

ABSTRACT

Following the placement of endovascular implants, perivascular adipose tissue (PVAT) becomes an early sensor of vascular injury to which it responds by undergoing phenotypic changes characterized by reduction in the secretion of adipocyte-derived relaxing factors and a shift to a proinflammatory and pro-contractile state. Thus, activated PVAT loses its anti-inflammatory function, secretes proinflammatory cytokines and chemokines, and generates reactive oxygen species, which are accompanied by differentiation of fibroblasts into myofibroblasts and proliferation of smooth muscle cells. These subsequently migrate into the intima, leading to intimal growth. In addition, periadventitial vasa vasorum undergoes neovascularization and functions as a portal for extravasation of inflammatory infiltrates and mobilization of PVAT resident stem/progenitor cells into the intima. This review focuses on the response of PVAT to endovascular intervention-induced injury and discusses potential therapeutic targets to suppress the PVAT-initiated pathways that mediate the formation of neointima.

20.
Heart Lung Circ ; 33(4): 486-492, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38423849

ABSTRACT

AIM: Angiotensin receptor blockers (ARBs) have been shown to inhibit restenosis in vitro and in vivo, but the evidence found in humans is inconsistent. This study aimed to evaluate the effectiveness of ARBs in preventing in-stent restenosis after percutaneous coronary intervention (PCI). METHOD: Databases including the Cochrane Library, MEDLINE, Web of Science, EMBASE, and CNKI were searched to collect randomised controlled trials on ARBs inhibiting restenosis that were published before October 2022. A total of 1,056 patients enrolled in eight trials were included in the study. RESULTS: The ARBs group showed lower target lesion revascularisation than the control group (RR 0.54; 95% CI 0.34-0.86; p=0.01), but the restenosis incidence between these two groups was not statistically significant (RR 0.85; 95% CI 0.65-1.11; p>0.05). CONCLUSION: This study found that ARBs might have a potential effect on reducing target lesion revascularisation after PCI in coronary heart disease patients but has no impact on angiographic restenosis.


Subject(s)
Angiotensin Receptor Antagonists , Coronary Restenosis , Percutaneous Coronary Intervention , Humans , Coronary Restenosis/prevention & control , Angiotensin Receptor Antagonists/therapeutic use , Percutaneous Coronary Intervention/methods , Stents/adverse effects , Graft Occlusion, Vascular/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...