Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Mol Sci ; 22(16)2021 Aug 17.
Article in English | MEDLINE | ID: mdl-34445530

ABSTRACT

Inflammation is a major contributor to the development and progression of atherosclerosis. Interleukin (IL)-33 and IL-37, members of the IL-1 family, modulate inflammation, with IL-33 having a pro-inflammatory effect and IL-37 having anti-inflammatory properties. IL-37 is constitutively expressed at low levels but upregulated in inflammatory contexts. The aim of this study was to evaluate the effect of vitamin D on the expression of IL-33, IL-37, macrophages, and caspase-1 in the neointimal tissue of coronary artery in Yucatan microswine with vitamin D deficient, sufficient, and supplemented status. The intimal injury was induced by balloon angioplasty and stenting in the coronary artery, and tissues were harvested after 6 months. The expression of various proteins of interest was evaluated by immunostaining. Increased expression of IL-33 and IL-37 in the neointimal tissue of the vitamin D deficient, as compared to the sufficient and supplemented microswine, as revealed by histological evaluation and semi-quantitative analysis, suggested the immunomodulatory effect of vitamin D on the expression of IL-33 and IL-37. The minimal expression or absence of IL-33 and IL-37 expression in stented arteries is suggestive of an attenuated inflammatory response in stented arteries, compared to balloon angioplasty. The decreased IL-33 expression in the sufficient and supplemented microswine could be a potential mechanism for controlling the inflammatory process and neointima formation leading to attenuated luminal narrowing of the coronary artery. Overall, these results support supplementation of vitamin D to attenuate inflammation, neointima formation, and restenosis.


Subject(s)
Angioplasty, Balloon, Coronary/methods , Coronary Artery Disease/immunology , Hyperlipidemias/physiopathology , Interleukin-1/metabolism , Interleukin-33/metabolism , Neointima/immunology , Stents , Vitamin D/metabolism , Animals , Coronary Artery Disease/pathology , Coronary Artery Disease/therapy , Dietary Supplements , Immunomodulation , Neointima/pathology , Neointima/therapy , Swine
2.
Theranostics ; 10(23): 10712-10728, 2020.
Article in English | MEDLINE | ID: mdl-32929376

ABSTRACT

Rationale: For intravascular stent implantation to be successful, the processes of vascular tissue repair and therapy are considered to be critical. However, the mechanisms underlying the eventual fate of vascular smooth muscle cells (VSMCs) during vascular tissue repair remains elusive. In this study, we hypothesized that M2 macrophage-derived exosomes to mediate cell-to-cell crosstalk and induce dedifferentiation phenotypes in VSMCs. Methods:In vivo, 316L bare metal stents (BMS) were implanted from the left iliac artery into the abdominal aorta of 12-week-old male Sprague-Dawley (SD) rats for 7 and 28 days. Hematoxylin and eosin (HE) were used to stain the neointimal lesions. En-face immunofluorescence staining of smooth muscle 22 alpha (SM22α) and CD68 showed the rat aorta smooth muscle cells (RASMCs) and macrophages. Immunohistochemical staining of total galactose-specific lectin 3 (MAC-2) and total chitinase 3-like 3 (YM-1) showed the total macrophages and M2 macrophages. In vitro, exosomes derived from IL-4+IL-13-treated macrophages (M2Es) were isolated by ultracentrifugation and characterized based on their specific morphology. Ki-67 staining was conducted to assess the effects of the M2Es on the proliferation of RASMCs. An atomic force microscope (AFM) was used to detect the stiffness of the VSMCs. GW4869 was used to inhibit exosome release. RNA-seq was performed to determine the mRNA profiles of the RASMCs and M2Es-treated RASMCs. Quantitative real-time PCR (qRT-PCR) analysis was conducted to detect the expression levels of the mRNAs. Western blotting was used to detect the candidate protein expression levels. T-5224 was used to inhibit the DNA binding activity of AP-1 in RASMCs. Results: M2Es promote c-KIT expression and softening of nearby VSMCs, hence accelerating the vascular tissue repair process. VSMCs co-cultured in vitro with M2 macrophages presented an increased capacity for de-differentiation and softening, which was exosome dependent. In addition, the isolated M2Es helped to promote VSMC dedifferentiation and softening. Furthermore, the M2Es enhanced vascular tissue repair potency by upregulation of VSMCs c-KIT expression via activation of the c-Jun/activator protein 1 (AP-1) signaling pathway.Conclusions: The findings of this study emphasize the prominent role of M2Es during VSMC dedifferentiation and vascular tissue repair via activation of the c-Jun/AP-1 signaling pathway, which has a profound impact on the therapeutic strategies of coronary stenting techniques.


Subject(s)
Coronary Disease/surgery , Endovascular Procedures/instrumentation , Macrophages/metabolism , Muscle, Smooth, Vascular/immunology , Neointima/immunology , Animals , Cell Communication/immunology , Cell Differentiation/immunology , Cell Line , Coronary Disease/immunology , Disease Models, Animal , Exosomes/immunology , Exosomes/metabolism , Humans , Macrophages/immunology , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-kit/metabolism , RNA-Seq , Rats , Rats, Sprague-Dawley , Signal Transduction/immunology , Stents , Transcription Factor AP-1/metabolism
3.
J Interv Cardiol ; 2020: 1956015, 2020.
Article in English | MEDLINE | ID: mdl-32410915

ABSTRACT

OBJECTIVES: We evaluated the effect of the different carrier systems on early vascular response through histological analysis and scanning electron microscopy using a porcine model. BACKGROUND: Although Synergy™ and Promus PREMIER™ share an identical stent material and drug elution (everolimus), they use different drug carrier systems: biodegradable abluminal coating polymer or durable conformal coating polymer, respectively. However, data regarding the impact of the different coating systems on vessel healing are currently limited. METHODS: Twelve Synergy™ and Promus PREMIER™ were implanted in 12 swine. Histopathological analysis of the stented segments was performed on the 2nd and 14th days after implantation. Morphometric analysis of the inflammation and intimal fibrin content was also performed. RESULTS: On the 2nd day, neointimal thickness, percentage of neointimal area, and inflammatory and intimal fibrin content scores were not significantly different between the two groups. On the 14th day, the inflammatory and intimal fibrin content scores were significantly lower in Synergy™ versus those observed in Promus PREMIER™. In Synergy™, smooth muscle cells were found and the neointimal layers were smooth. In contrast, inflammatory cells were observed surrounding the struts of Promus PREMIER™. CONCLUSIONS: These results demonstrate that termination of reactive inflammation is accelerated after abluminal coating stent versus implantation of conformal coating stent.


Subject(s)
Coronary Vessels , Drug-Eluting Stents , Inflammation/prevention & control , Neointima/immunology , Stents/adverse effects , Vascular Grafting/instrumentation , Absorbable Implants , Animals , Coated Materials, Biocompatible/pharmacology , Coronary Vessels/immunology , Coronary Vessels/surgery , Drug Carriers/pharmacology , Everolimus/pharmacology , Inflammation/etiology , Models, Anatomic , Polymers/pharmacology , Swine
4.
Transplantation ; 102(10): 1674-1683, 2018 10.
Article in English | MEDLINE | ID: mdl-30247451

ABSTRACT

BACKGROUND: Transplant vasculopathy is a major cause of chronic rejection of transplanted organs. In the present study, we examined the effects of CX-5461, a novel selective inhibitor of RNA polymerase I, on development of transplant vasculopathy using a modified model of rat aortic transplantation. METHODS: The thoracic aortas from Fischer rats were transplanted into the abdominal cavity of Lewis rats. CX-5461 was mixed in pluronic gel and administered via perivascular release. RESULTS: Treatment with CX-5461 mitigated the development of neointimal hyperplasia and vascular inflammation. This effect was likely to be attributable in part to inhibition of macrophage-dependent innate immunity reactions. Specifically, CX-5461 exhibited potent inhibitory effects on macrophage migration and lipopolysaccharide-induced activation. Treatment with CX-5461 also prevented macrophage differentiation and maturation from primary bone marrow cells. In macrophages, CX-5461 did not alter the total amount of p53 protein, but significantly increased p53 phosphorylation, which was involved in regulating cytokine-stimulated macrophage proliferation. CONCLUSIONS: In conclusion, our results suggest that pharmacological inhibition of RNA polymerase I may be a novel strategy to treat transplantation-induced arterial remodeling.


Subject(s)
Aorta/transplantation , Aortic Diseases/prevention & control , Arteriosclerosis/prevention & control , Benzothiazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Heart Transplantation/adverse effects , Naphthyridines/therapeutic use , Neointima/prevention & control , Allografts/cytology , Allografts/drug effects , Allografts/immunology , Animals , Aorta/cytology , Aorta/drug effects , Aorta/immunology , Aortic Diseases/immunology , Arteriosclerosis/immunology , Benzothiazoles/pharmacology , Cell Movement/drug effects , Cell Movement/immunology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heart Transplantation/methods , Humans , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Naphthyridines/pharmacology , Neointima/immunology , Primary Cell Culture , RAW 264.7 Cells , RNA Polymerase I/antagonists & inhibitors , Rats , Rats, Inbred F344 , Treatment Outcome , Vascular Remodeling/drug effects , Vascular Remodeling/immunology
5.
Eur J Pharmacol ; 820: 222-228, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29269019

ABSTRACT

Accumulating evidence suggests that activated factor X (FXa), a key coagulation factor, plays an important role in the development of vascular inflammation through activation of many cell types. Here, we investigated whether pharmacological blockade of FXa attenuates neointima formation after wire-mediated vascular injury. Transluminal femoral artery injury was induced in C57BL/6 mice by inserting a straight wire. Rivaroxaban (5mg/kg/day), a direct FXa inhibitor, was administered from one week before surgery until killed. At four weeks after surgery, rivaroxaban significantly attenuated neointima formation in the injured arteries compared with control (P<0.01). Plasma lipid levels and blood pressure were similar between the rivaroxaban-treated group and non-treated group. Quantitative RT-PCR analyses demonstrated that rivaroxaban reduced the expression of inflammatory molecules (e.g., IL-1ß and TNF-α) in injured arteries at seven days after surgery (P<0.05, respectively). In vitro experiments using mouse peritoneal macrophages demonstrated that FXa increased the expression of inflammatory molecules (e.g., IL-1ß and TNF-α), which was blocked in the presence of rivaroxaban (P<0.05). Also, in vitro experiments using rat vascular smooth muscle cells (VSMC) demonstrated that FXa promoted both proliferation and migration of this cell type (P<0.05), which were blocked in the presence of rivaroxaban. Inhibition of FXa by rivaroxaban attenuates neointima formation after wire-mediated vascular injury through inhibition of inflammatory activation of macrophages and VSMC.


Subject(s)
Factor Xa Inhibitors/pharmacology , Factor Xa/metabolism , Neointima/drug therapy , Rivaroxaban/pharmacology , Vascular System Injuries/pathology , Animals , Factor Xa Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Hyperplasia/drug therapy , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Neointima/immunology , Neointima/metabolism , Rivaroxaban/therapeutic use , Signal Transduction/drug effects
6.
Biosci Rep ; 37(5)2017 Oct 31.
Article in English | MEDLINE | ID: mdl-28851839

ABSTRACT

Inflammatory cells play key roles in restenosis upon vascular surgical procedures such as bypass grafts, angioplasty and stent deployment but the molecular mechanisms by which these cells affect restenosis remain unclear. The p110δ isoform of phosphoinositide 3-kinase (PI3K) is mainly expressed in white blood cells. Here, we have investigated whether p110δ PI3K is involved in the pathogenesis of restenosis in a mouse model of carotid injury, which mimics the damage following arterial grafts. We used mice in which p110δ kinase activity has been disabled by a knockin (KI) point mutation in its ATP-binding site (p110δD910A/D910A PI3K mice). Wild-type (WT) and p110δD910A/D910A mice were subjected to longitudinal carotid injury. At 14 and 30 days after carotid injury, mice with inactive p110δ showed strongly decreased infiltration of inflammatory cells (including T lymphocytes and macrophages) and vascular smooth muscle cells (VSMCs), compared with WT mice. Likewise, PI-3065, a p110δ-selective PI3K inhibitor, almost completely prevented restenosis after artery injury. Our data showed that p110δ PI3K plays a main role in promoting neointimal thickening and inflammatory processes during vascular stenosis, with its inhibition providing significant reduction in restenosis following carotid injury. p110δ-selective inhibitors, recently approved for the treatment of human B-cell malignancies, therefore, present a new therapeutic opportunity to prevent the restenosis upon artery injury.


Subject(s)
Carotid Artery Injuries/enzymology , Carotid Stenosis/enzymology , Class I Phosphatidylinositol 3-Kinases/immunology , Inflammation/enzymology , Animals , Carotid Arteries/enzymology , Carotid Arteries/immunology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/immunology , Carotid Artery Injuries/pathology , Carotid Stenosis/genetics , Carotid Stenosis/immunology , Carotid Stenosis/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Disease Models, Animal , Gene Knock-In Techniques , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice, Inbred C57BL , Neointima/enzymology , Neointima/genetics , Neointima/immunology , Neointima/pathology , Point Mutation
7.
Cell Physiol Biochem ; 42(4): 1303-1312, 2017.
Article in English | MEDLINE | ID: mdl-28715799

ABSTRACT

BACKGROUND/AIMS: Neointimal formation following vascular injury remains a major mechanism of restenosis, whereas the precise sources of neointimal cells are still uncertain. We tested the hypothesis that both injured arterial cells and non-arterial cells contribute to intimal hyperplasia. METHODS: Following allograft transplantation of the balloon-injured carotid common artery (n = 3-6), the cellular composition of the transplant grafts and the origins of neointimal cells were measured by immunohistochemistry and immunofluorescence staining. RESULTS: Smooth muscle actin (SMA)-positive and CD68-positive cells were clearly observed 14 days later in the neointima after allograft transplantation of the balloon-injured carotid common artery, where re-endothelialization was not yet complete. Green fluorescent protein (GFP) and wild-type (WT) allograft transplantation revealed that the majority of the neointima cells were apparently from the recipient (≈85%) versus the donor (≈15%). Both monocyte chemotactic protein-1 (MCP-1)/CCR2 and stromal cell-derived factor-1 (SDF-1)/CXCR4 signaling were involved in intimal hyperplasia, with bone marrow-derived cells also playing a role. CONCLUSION: These data support the hypothesis that intimal hyperplasia could develop in our novel rat allograft transplantation model of arterial injury, where neointima is attributable not only to local arterial cells but also non-arterial cells including the bone marrow.


Subject(s)
Bone Marrow Cells/pathology , Carotid Artery Injuries/pathology , Carotid Artery, Common/pathology , Endothelial Cells/pathology , Neointima/pathology , Tissue Transplantation , Tunica Intima/injuries , Actins/genetics , Actins/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/immunology , Bone Marrow Cells/immunology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/immunology , Carotid Artery Injuries/surgery , Carotid Artery, Common/immunology , Carotid Artery, Common/surgery , Cell Lineage/immunology , Cell Tracking , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Endothelial Cells/immunology , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Neointima/genetics , Neointima/immunology , Neointima/surgery , Rats , Rats, Inbred Lew , Rats, Transgenic , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Signal Transduction , Transplantation, Homologous , Tunica Intima/immunology , Tunica Intima/surgery
8.
Immunol Cell Biol ; 95(4): 372-379, 2017 04.
Article in English | MEDLINE | ID: mdl-27808085

ABSTRACT

The leukocyte adhesion cascade involves multiple events that efficiently localize circulating leukocytes into the injured sites to mediate inflammatory responses. From rolling to firm adhesion, the interactions between adhesion molecules have pivotal roles in increasing the avidity of leukocytes to endothelial cells. Thrombomodulin (TM), an essential anticoagulant protein in the vasculature, is also expressed on leukocytes. We previously demonstrated that Lewisy (Ley), a specific ligand of TM, is upregulated in inflamed endothelium and is involved in leukocyte adhesion. The current study aimed to investigate whether leukocyte-expressed TM promotes cell adhesion by interacting with Ley. Using human monocytic THP-1 cells as an in vitro cell model, we showed that TM increases THP-1 cell adhesion to inflamed endothelium as well as to Ley-immobilized surface. When THP-1 adhered to activated endothelium and Ley-immobilized surface, the TM distribution became polarized. Addition of soluble Ley to a suspension of THP-1 cells with TM expression triggered an increase in the level of phosphorylated p38 mitogen-activated protein kinase (MAPK), which enabled THP-1 to adhere firmly to intercellular adhesion molecule (ICAM)-1 by activating ß2 integrins. In vivo, macrophage infiltration and neointima formation following arterial ligation-induced vascular injury were higher in wild-type TM (TMflox/flox) than in myeloid-specific TM-deficient (LysMcre/TMflox/flox) mice. Taken together, these results suggest a novel function for TM as an adhesion molecule in monocytes, where it enhances cell adhesion by binding Ley, leading to ß2 integrin activation via p38 MAPK.


Subject(s)
Endothelial Cells/immunology , Inflammation/immunology , Monocytes/immunology , Neointima/immunology , Thrombomodulin/metabolism , Animals , CD18 Antigens/metabolism , Cell Adhesion , Disease Models, Animal , Humans , Intercellular Adhesion Molecule-1/metabolism , Lewis Blood Group Antigens/metabolism , Ligands , Mice , Mice, Knockout , RNA, Small Interfering/genetics , Signal Transduction , THP-1 Cells , Thrombomodulin/agonists , Thrombomodulin/genetics , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Sci Rep ; 6: 35387, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759053

ABSTRACT

Percutaneous coronary intervention is widely adopted to treat patients with coronary artery disease. However, restenosis remains an unsolved clinical problem after vascular interventions. The role of the systemic and local immune response in the development of restenosis is not fully understood. Hence, the aim of the current study was to investigate the role of the human immune system on subsequent neointima formation elicited by vascular injury in a humanized mouse model. Immunodeficient NOD.Cg-PrkdcscidIL2rgtm1Wjl(NSG) mice were reconstituted with human (h)PBMCs immediately after both carotid wire and femoral cuff injury were induced in order to identify how differences in the severity of injury influenced endothelial regeneration, neointima formation, and homing of human inflammatory and progenitor cells. In contrast to non-reconstituted mice, hPBMC reconstitution reduced neointima formation after femoral cuff injury whereas hPBMCs promoted neointima formation after carotid wire injury 4 weeks after induction of injury. Neointimal endothelium and smooth muscle cells in the injured arteries were of mouse origin. Our results indicate that the immune system may differentially respond to arterial injury depending on the severity of injury, which may also be influenced by the intrinsic properties of the arteries themselves, resulting in either minimal or aggravated neointima formation.


Subject(s)
Carotid Artery Injuries/immunology , Femoral Artery/immunology , Graft Occlusion, Vascular/immunology , Leukocytes, Mononuclear/immunology , Vascular System Injuries/immunology , Animals , Carotid Artery Injuries/parasitology , Carotid Artery Injuries/therapy , Disease Models, Animal , Femoral Artery/injuries , Femoral Artery/transplantation , Graft Occlusion, Vascular/physiopathology , Humans , Leukocytes, Mononuclear/transplantation , Mice , Mice, SCID/immunology , Mice, SCID/injuries , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Neointima/immunology , Neointima/physiopathology , Vascular System Injuries/physiopathology , Vascular System Injuries/therapy
10.
Immunol Lett ; 176: 105-13, 2016 08.
Article in English | MEDLINE | ID: mdl-27317647

ABSTRACT

Renal transplantation is the treatment of choice for end-stage renal failure. Although acute rejection is not a major issue anymore, chronic rejection, especially vascular rejection, is still a major factor that might lead to allograft dysfunction on the long term. The role of the local immune-regulating cytokine interleukin-10 (IL-10) in chronic renal allograft is unclear. Many clinical observations showed that local IL-10 level was negatively related to kidney allograft function. It is unknown this negative relationship was the result of immunostimulatory property or insufficient immunosuppression property of local IL-10. We performed ex vivo transduction before transplantation through artery of the renal allograft using adeno-associated viral vectors carrying IL-10 gene. Twelve weeks after transplantation, we found intrarenal IL-10 gene transduction significantly inhibited arterial neointimal proliferation, the number of occluded intrarenal artery, interstitial fibrosis, peritubular capillary congestion and glomerular inflammation in renal allografts compared to control allografts receiving PBS or vectors carrying YFP. IL-10 transduction increased serum IL-10 level at 4 weeks but not at 8 and 12 weeks. Renal IL-10 level increased while serum creatinine decreased significantly in IL-10 group at 12 weeks compared to PBS or YFP controls. Immunohistochemical staining showed unchanged total T cells (CD3) and B cells (CD45R/B220), decreased cytotoxic T cells (CD8), macrophages (CD68) and increased CD4+ and FoxP3+ cells in IL-10 group. In summary, intrarenal IL-10 inhibited the allograft rejection while modulated immune response.


Subject(s)
Graft Rejection/prevention & control , Interleukin-10/metabolism , Kidney Transplantation , Kidney/physiology , Neointima/prevention & control , T-Lymphocytes, Regulatory/immunology , Vaccines, DNA/immunology , Adenoviridae/genetics , Administration, Intravenous , Animals , Cell Movement/drug effects , Cells, Cultured , Creatinine/blood , Graft Rejection/immunology , Immunomodulation , Interleukin-10/genetics , Interleukin-10/immunology , Male , Neointima/immunology , Rats , Rats, Wistar , Renal Artery/metabolism , Transduction, Genetic , Transplantation, Homologous
11.
J Nutr Biochem ; 32: 73-84, 2016 06.
Article in English | MEDLINE | ID: mdl-27142739

ABSTRACT

Sulforaphane (SFN), a dietary phase-2 enzyme inducer that mitigates cellular oxidative stress through nuclear factor erythroid 2-related factor 2 (Nrf2) activation, is known to exhibit beneficial effects in the vessel wall. For instance, it inhibits vascular smooth muscle cell (VSMC) proliferation, a major event in atherosclerosis and restenosis after angioplasty. In particular, SFN attenuates the mitogenic and pro-inflammatory actions of platelet-derived growth factor (PDGF) and tumor necrosis factor-α (TNFα), respectively, in VSMCs. Nevertheless, the vasoprotective role of SFN has not been examined in the setting of obesity characterized by hyperleptinemia and insulin resistance. Using the mouse model of western diet-induced obesity, the present study demonstrates for the first time that subcutaneous delivery of SFN (0.5mg/Kg/day) for~3weeks significantly attenuates neointima formation in the injured femoral artery [↓ (decrease) neointima/media ratio by~60%; n=5-8]. This was associated with significant improvements in metabolic parameters, including ↓ weight gain by~52%, ↓ plasma leptin by~42%, ↓ plasma insulin by~63%, insulin resistance [↓ homeostasis model assessment of insulin resistance (HOMA-IR) index by~73%], glucose tolerance (↓ AUCGTT by~24%), and plasma lipid profile (e.g., ↓ triglycerides). Under in vitro conditions, SFN significantly decreased leptin-induced VSMC proliferation by~23% (n=5) with associated diminutions in leptin-induced cyclin D1 expression and the phosphorylation of p70S6kinase and ribosomal S6 protein (n=3-4). The present findings reveal that, in addition to improving systemic metabolic parameters, SFN inhibits leptin-induced VSMC proliferative signaling that may contribute in part to the suppression of injury-induced neointima formation in diet-induced obesity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , Isothiocyanates/therapeutic use , Leptin/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Neointima/prevention & control , Obesity/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Antioxidants/administration & dosage , Antioxidants/pharmacology , Aorta , Cell Proliferation/drug effects , Cells, Cultured , Diet, Western/adverse effects , Femoral Artery/injuries , Humans , Injections, Subcutaneous , Insulin Resistance , Isothiocyanates/administration & dosage , Isothiocyanates/pharmacology , Leptin/metabolism , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Neointima/immunology , Neointima/metabolism , Neointima/pathology , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Oxidative Stress/drug effects , Sulfoxides , Weight Gain/drug effects
12.
Am J Physiol Heart Circ Physiol ; 310(6): H705-15, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26801304

ABSTRACT

Interleukin-8 (IL8) is highly expressed by injured arteries in a variety of diseases and is a chemoattractant for neutrophils which express IL8 receptors IL8RA and RB (IL8RA/B) on their membranes. Neutrophils interact with the damaged endothelium and initiate an inflammatory cascade at the site of injury. We have generated a novel translational targeted cell therapy for acute vascular injury using adenoviral vectors to overexpress IL8RA/B and green fluorescent protein (GFP) on the surface of endothelial cells (ECs) derived from human induced pluripotent stem cells (HiPS-IL8RA/B-ECs). We hypothesize that HiPS-IL8RA/B-ECs transfused intravenously into rats with balloon injury of the carotid artery will target to the injured site and compete with neutrophils, thus inhibiting inflammation and neointima formation. Young adult male Sprague-Dawley rats underwent balloon injury of the right carotid artery and received intravenous transfusion of saline vehicle, 1.5 × 10(6) HiPS-ECs, 1.5 × 10(6) HiPS-Null-ECs, or 1.5 × 10(6) HiPS-IL8RA/B-ECs immediately after endoluminal injury. Tissue distribution of HiPS-IL8RA/B-ECs was analyzed by a novel GFP DNA qPCR method. Cytokine and chemokine expression and leukocyte infiltration were measured in injured and uninjured arteries at 24 h postinjury by ELISA and immunohistochemistry, respectively. Neointimal, medial areas, and reendothelialization were measured 14 days postinjury. HiPS-IL8RA/B-ECs homed to injured arteries, inhibited inflammatory mediator expression and inflammatory cell infiltration, accelerated reendothelialization, and attenuated neointima formation after endoluminal injury while control HiPS-ECs and HiPS-Null-ECs did not. HiPS-IL8RA/B-ECs transfused into rats with endoluminal carotid artery injury target to the injured artery and provide a novel strategy to treat vascular injury.


Subject(s)
Carotid Arteries/pathology , Carotid Artery Injuries/therapy , Cell- and Tissue-Based Therapy/methods , Induced Pluripotent Stem Cells/transplantation , Neointima/prevention & control , Receptors, Interleukin-8/immunology , Animals , Carotid Arteries/immunology , Carotid Artery Injuries/immunology , Carotid Artery Injuries/pathology , Endothelial Cells , Enzyme-Linked Immunosorbent Assay , Green Fluorescent Proteins/genetics , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Inflammation , Male , Neointima/immunology , Neointima/pathology , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-8/genetics
13.
PLoS One ; 10(9): e0138459, 2015.
Article in English | MEDLINE | ID: mdl-26389595

ABSTRACT

BACKGROUND: The introduction of drug-eluting stents (DES) has dramatically reduced restenosis rates compared with bare metal stents, but in-stent thrombosis remains a safety concern, necessitating prolonged dual anti-platelet therapy. The drug 6-Mercaptopurine (6-MP) has been shown to have beneficial effects in a cell-specific fashion on smooth muscle cells (SMC), endothelial cells and macrophages. We generated and analyzed a novel bioresorbable polymer coated DES, releasing 6-MP into the vessel wall, to reduce restenosis by inhibiting SMC proliferation and decreasing inflammation, without negatively affecting endothelialization of the stent surface. METHODS: Stents spray-coated with a bioresorbable polymer containing 0, 30 or 300 µg 6-MP were implanted in the iliac arteries of 17 male New Zealand White rabbits. Animals were euthanized for stent harvest 1 week after implantation for evaluation of cellular stent coverage and after 4 weeks for morphometric analyses of the lesions. RESULTS: Four weeks after implantation, the high dose of 6-MP attenuated restenosis with 16% compared to controls. Reduced neointima formation could at least partly be explained by an almost 2-fold induction of the cell cycle inhibiting kinase p27Kip1. Additionally, inflammation score, the quantification of RAM11-positive cells in the vessel wall, was significantly reduced in the high dose group with 23% compared to the control group. Evaluation with scanning electron microscopy showed 6-MP did not inhibit strut coverage 1 week after implantation. CONCLUSION: We demonstrate that novel stents coated with a bioresorbable polymer coating eluting 6-MP inhibit restenosis and attenuate inflammation, while stimulating endothelial coverage. The 6-MP-eluting stents demonstrate that inhibition of restenosis without leaving uncovered metal is feasible, bringing stents without risk of late thrombosis one step closer to the patient.


Subject(s)
Drug-Eluting Stents/adverse effects , Iliac Artery/drug effects , Immunosuppressive Agents/administration & dosage , Inflammation/prevention & control , Mercaptopurine/administration & dosage , Neointima/prevention & control , Animals , Coated Materials, Biocompatible/chemistry , Iliac Artery/pathology , Iliac Artery/surgery , Immunosuppressive Agents/therapeutic use , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Male , Mercaptopurine/therapeutic use , Neointima/etiology , Neointima/immunology , Neointima/pathology , Polymers/chemistry , Rabbits
14.
Histochem Cell Biol ; 144(1): 59-66, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25787768

ABSTRACT

Neointimal hyperplasia, transplant rejection and thus immunogenicity of allografts are possible reasons for poorer patency rates in cryopreserved venous allografts for peripheral bypass surgery in comparison with autologous venous grafts. To expand the limited knowledge from human allografts, we histologically investigated allogeneic and autologous venous grafts in arterial location. Specimens of allogeneic and autologous venous graft stenosis, harvested 6 months after bypass implantation, were immunohistochemically characterized. Examination of the lesions showed a uniform morphological pattern. A continuous endothelial layer, tissue fibrosis and a thickened neointima with monocytes and dedifferentiated vascular smooth muscle cells were seen in both conduits with very low cell turnover and the absence of acute and chronic inflammation. Neoangiogenesis with CD34-positive endothelium was abundant in the vessel media. The morphological patterns of allogeneic and autologous neointima formation are similar. Consequently, neointimal hyperplasia in venous grafts may reflect a uniform physiological host response of non-immunological factors with the reasons for poorer clinical outcome of cryopreserved allografts yet to be elucidated.


Subject(s)
Neointima/pathology , Saphenous Vein/transplantation , Allografts , Autografts , Cryopreservation , Endothelium, Vascular/pathology , Humans , Hyperplasia , Muscle, Smooth, Vascular/pathology , Neointima/immunology , Neovascularization, Pathologic
15.
Immunobiology ; 220(5): 673-83, 2015 May.
Article in English | MEDLINE | ID: mdl-25476849

ABSTRACT

In chronic transplant dysfunction (CTD), persistent (allo)immune-mediated inflammation eventually leads to tissue remodeling including neointima formation in intragraft arteries. We previously showed that recipient-derived neointimal α-SMA(+) smooth muscle-like cells are present in human renal allografts with CTD. Human PBMC contain myeloid cells capable of differentiating into α-SMA(+) cells in vitro; the phenotype of the ancestral subset is as yet unknown. This study aimed to investigate whether monocyte subsets contain cells with smooth muscle-like cell differentiation capacity and whether CTD in renal transplant recipients is associated with a shift in these monocyte subsets. To accomplish this goal, monocyte subsets from healthy controls were sorted based on CD14 and CD16 expression to investigate gene expression levels of mesenchymal markers α-SMA and SM22α. CD14(+)/CD16(++) monocytes displayed increased α-SMA and SM22α mRNA expression compared with CD14(++)/CD16(-) monocytes, suggesting increased differentiation potential toward smooth muscle-like cells. Flow cytometry revealed that in non-CTD transplant recipients the percentage of CD14(+)/CD16(++) monocytes was reduced, with an even further reduction in patients with CTD. To determine a potential correlation between CD14(+)/CD16(++) monocytes and α-SMA(+) cell outgrowth potential in vitro, PBMC of healthy controls and transplant recipients with and without CTD were cultured under fibrotic culture conditions, and indeed a significant correlation (p=0.0002, r=0.62) was observed. Finally, double staining for α-SMA and CD16 revealed presence of α-SMA(+)CD16(+) cells in kidney explants from CTD patients, albeit at very low numbers. Our data represent evidence that, compared to CD14(++)CD16(-) monocytes, CD14(+)CD16(++) monocytes have an increased expression of smooth muscle cell-associated genes. This monocyte subpopulation is reduced in renal transplant patients with CTD, possibly due to selective migration into the allograft.


Subject(s)
Actins/metabolism , Allografts/immunology , Graft Rejection/immunology , Kidney Transplantation , Microfilament Proteins/metabolism , Monocytes/immunology , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/immunology , Neointima/immunology , Postoperative Complications/immunology , Actins/genetics , Allografts/blood supply , Cell Differentiation , Chronic Disease , Graft Rejection/etiology , Humans , Lipopolysaccharide Receptors/metabolism , Microfilament Proteins/genetics , Monitoring, Immunologic/methods , Muscle Proteins/genetics , Neointima/etiology , Receptors, IgG/metabolism
16.
J Am Heart Assoc ; 3(5): e001369, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25315347

ABSTRACT

BACKGROUND: Antiphospholipid syndrome patients have antiphospholipid antibodies (aPLs) that promote thrombosis, and they have increased cardiovascular disease risk. Although the basis for the thrombosis has been well delineated, it is not known why antiphospholipid syndrome patients also have an increased prevalence of nonthrombotic vascular occlusion. The aims of this work were to determine if aPLs directly promote medial hypertrophy or neointima formation in mice and to identify the underlying mechanisms. METHODS AND RESULTS: Medial hypertrophy and neointima formation invoked by carotid artery endothelial denudation were evaluated in mice administered normal human IgG or aPLs. While aPLs had no effect on medial hypertrophy, they caused exaggerated neointima development. This was related to an aPL-induced impairment in reendothelialization post denudation, and scratch assays in cell culture revealed that there are direct effects of aPLs on endothelium that retard cell migration. Further experiments showed that aPL antagonism of endothelial migration and repair is mediated by antibody recognition of ß2-glycoprotein I, apolipoprotein E receptor 2, and a decline in bioavailable NO. Consistent with these mechanisms, the adverse impacts of aPLs on reendothelialization and neointima formation were fully prevented by the NO donor molsidomine. CONCLUSIONS: APLs blunt endothelial repair, and there is related aPL-induced exaggeration in neointima formation after endothelial injury in mice. The initiating process entails NO deficiency mediated by ß2-glycoprotein I recognition by aPLs and apolipoprotein E receptor 2. The modulation of endothelial apolipoprotein E receptor 2 function or NO bioavailability may represent new interventions to prevent the nonthrombotic vascular occlusion and resulting cardiovascular disorders that afflict antiphospholipid syndrome patients.


Subject(s)
Antibodies, Antiphospholipid/metabolism , Carotid Artery Injuries/pathology , Cell Movement/physiology , Endothelium, Vascular/cytology , Neointima/pathology , Nitric Oxide/metabolism , Animals , Antibodies, Antiphospholipid/immunology , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/physiopathology , Biological Availability , Carotid Artery Injuries/physiopathology , Cell Movement/immunology , Cells, Cultured , Disease Models, Animal , Hypertrophy/pathology , Male , Mice , Mice, Inbred C57BL , Neointima/immunology , Random Allocation , Sensitivity and Specificity
17.
J Exp Med ; 211(9): 1779-92, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25073791

ABSTRACT

Interventional strategies to treat atherosclerosis, such as transluminal angioplasty and stent implantation, often cause vascular injury. This leads to intimal hyperplasia (IH) formation that induces inflammatory and fibroproliferative processes and ultimately restenosis. We show that phosphoinositide 3-kinase γ (PI3Kγ) is a key player in IH formation and is a valid therapeutic target in its prevention/treatment. PI3Kγ-deficient mice and mice expressing catalytically inactive PI3Kγ (PI3Kγ KD) showed reduced arterial occlusion and accumulation of monocytes and T cells around sites of vascular lesion. The transfer of PI3Kγ KD CD4(+) T cells into Rag2-deficient mice greatly reduced vascular occlusion compared with WT cells, clearly demonstrating the involvement of PI3Kγ in CD4(+) T cells during IH formation. In addition we found that IH is associated with increased levels of Th1 and Th17 cytokines. A specific decrease in the Th1 response was observed in the absence of PI3Kγ activity, leading to decreased CXCL10 and RANTES production by smooth muscle cells. Finally, we show that treatment with the PI3Kγ inhibitor AS-605240 is sufficient to decrease IH in both mouse and rat models, reinforcing the therapeutic potential of PI3Kγ inhibition. Altogether, these findings demonstrate a new role for PI3Kγ activity in Th1-controlled IH development.


Subject(s)
Neointima/enzymology , Neointima/immunology , Phosphoinositide-3 Kinase Inhibitors , Th1 Cells/drug effects , Th1 Cells/immunology , Animals , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/immunology , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Class Ib Phosphatidylinositol 3-Kinase/genetics , Enzyme Inhibitors/pharmacology , Femoral Artery/enzymology , Femoral Artery/immunology , Femoral Artery/injuries , Gene Targeting , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neointima/drug therapy , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Thiazolidinediones/pharmacology
18.
Angiol Sosud Khir ; 20(1): 21-6, 2014.
Article in Russian | MEDLINE | ID: mdl-24722017

ABSTRACT

The article deals with an analysis of the literature data concerning immunological mechanisms of the formation of restenoses after damage of the arterial wall, considering the participants of the early and late phases of inflammatory response initiated by endothelial damage. Also given is characteristics of the process of formation on the neointima, followed by description of the role of intercellular adhesion molecules in initiation and maintaining of the processes of acute and chronic inflammation.


Subject(s)
Cell Adhesion Molecules/metabolism , Endothelium, Vascular , Graft Occlusion, Vascular/immunology , Acute-Phase Reaction/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/injuries , Endothelium, Vascular/physiopathology , Graft Occlusion, Vascular/physiopathology , Humans , Neointima/immunology , Vascular Patency/immunology
19.
J Am Heart Assoc ; 2(3): e000155, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23702879

ABSTRACT

BACKGROUND: CD8(+) T-cell activation, characterized by increased CD28 expression, reduces neointima formation after arterial injury in mice. The CD8(+)CD28(hi) phenotype is associated with increased effector function. In this study, we used a mouse model that has CD8(+) but no CD4(+) T cells (CD4-/-) to assess the role of CD8(+) T cells and test the function of CD8(+)CD28(hi) T cells in modulating neointima formation after arterial injury. METHODS AND RESULTS: Neointima formation after pericarotid arterial cuff injury was significantly less in CD4-/- mice compared with wild-type (WT) mice. Negligible baseline lytic activity by splenic CD8(+) T cells from uninjured WT mice against target syngeneic smooth muscle cells was significantly increased 7 days after injury. Interestingly, CD8(+) T cells from uninjured CD4-/- mice had significant lytic activity at baseline that remained elevated 7 days after injury. CD8(+) T-cell lytic activity was significantly reduced by depletion of CD28(hi) cells. CD8(+)CD28(hi) T cells adoptively transferred into recipient Rag-1-/- mice significantly reduced neointima formation compared with CD8(+)CD28(+) T-cell recipient mice. CONCLUSIONS: CD8(+) T cells reduced neointima formation after arterial injury, attributed in part to increased function of the CD8(+)CD28(hi) phenotype.


Subject(s)
Arteries/injuries , CD8-Positive T-Lymphocytes/physiology , Neointima/immunology , Vascular System Injuries/complications , Animals , Male , Mice
20.
Curr Atheroscler Rep ; 15(5): 320, 2013 May.
Article in English | MEDLINE | ID: mdl-23512604

ABSTRACT

Atherosclerosis is a chronic inflammatory disease in which initial vascular damage leads to extensive macrophage and lymphocyte infiltration. Although acutely glucocorticoids suppress inflammation, chronic glucocorticoid excess worsens atherosclerosis, possibly by exacerbating systemic cardiovascular risk factors. However, glucocorticoid action within the lesion may reduce neointimal proliferation and inflammation. Glucocorticoid levels within cells do not necessarily reflect circulating levels due to pre-receptor metabolism by 11ß-hydroxysteroid dehydrogenases (11ß-HSDs). 11ß-HSD2 converts active glucocorticoids into inert 11-keto forms. 11ß-HSD1 catalyses the reverse reaction, regenerating active glucocorticoids. 11ß-HSD2-deficiency/inhibition causes hypertension, whereas deficiency/inhibition of 11ß-HSD1 generates a cardioprotective lipid profile and improves glycemic control. Importantly, 11ß-HSD1-deficiency/inhibition is atheroprotective, whereas 11ß-HSD2-deficiency accelerates atherosclerosis. These effects are largely independent of systemic risk factors, reflecting modulation of glucocorticoid action and inflammation within the vasculature. Here, we consider whether evidence linking the 11ß-HSDs to vascular inflammation suggests these isozymes are potential therapeutic targets in vascular injury and atherosclerosis.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/physiology , Atherosclerosis/immunology , Glucocorticoids/immunology , Vasculitis/immunology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/immunology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/immunology , Atherosclerosis/complications , Atherosclerosis/enzymology , Glucocorticoids/metabolism , Humans , Neointima/immunology , Vasculitis/complications , Vasculitis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...