Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Atherosclerosis ; 351: 9-17, 2022 06.
Article in English | MEDLINE | ID: mdl-35605369

ABSTRACT

BACKGROUND AND AIMS: Endothelial cell injury causes vascular barrier dysfunction and leukocyte recruitment to the underlying tissue. Bone morphogenetic protein 4 (BMP-4) is a transforming growth factor that exerts pro-inflammatory effects on the endothelium. Here, we investigated the effects of BMP-4 on endothelial cell (EC) migration following balloon injury in SD rats. METHODS: An intimal hyperplasia model was established using balloon injury. Hematoxylin-eosin staining (HE) and silver staining were used to detect the alteration of endothelial cells recovery after balloon injury. Serum BMP-4 levels were assessed by ELISA. Human umbilical vein endothelial cells (HUVECs) were cultured. MTT assay was used to measure cell viability. Protein expression was detected by Western blot. Intracellular reactive oxygen species (ROS) was detected by dichloro-dihydro-fluorescein diacetate (DCFH-DA). HUVECs migration was measured via transwell assay and scratch wound assay. RESULTS: The results indicated that BMP-4 inhibition significantly decreased total plasma activity of BMP-4 and reduced neointimal hyperplasia by stimulating endothelial cell migration, but did not affect the medial area following balloon injury. BMP-4 suppressed the formation of ROS via forkhead box O3 (FoXO-3)/superoxide dismutase 1 (SOD-1). In vitro, a high level of ROS induced by BMP-4 impeded HUVECs migration. CONCLUSIONS: The results suggest that BMP-4 inhibition is a potential means of preventing intimal hyperplasia formation after balloon injury.


Subject(s)
Bone Morphogenetic Protein 4 , Human Umbilical Vein Endothelial Cells , Animals , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/blood , Carotid Artery Injuries/blood , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Movement , Cells, Cultured , Forkhead Box Protein O3/biosynthesis , Forkhead Box Protein O3/blood , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hyperplasia , Neointima/blood , Neointima/metabolism , Neointima/pathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/blood , Reactive Oxygen Species/metabolism , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/blood
2.
Cardiovasc Res ; 117(10): 2275-2288, 2021 08 29.
Article in English | MEDLINE | ID: mdl-32931562

ABSTRACT

AIMS: Arterial thrombosis as a result of plaque rupture or erosion is a key event in acute cardiovascular events. Sirtuin 5 (SIRT5) belongs to the lifespan-regulating sirtuin superfamily and has been implicated in acute ischaemic stroke and cardiac hypertrophy. This project aims at investigating the role of SIRT5 in arterial thrombus formation. METHODS AND RESULTS: Sirt5 transgenic (Sirt5Tg/0) and knock-out (Sirt5-/-) mice underwent photochemically induced carotid endothelial injury to trigger arterial thrombosis. Primary human aortic endothelial cells (HAECs) were treated with SIRT5 silencing-RNA (si-SIRT5) as well as peripheral blood mononuclear cells from acute coronary syndrome (ACS) patients and non-ACS controls (case-control study, total n = 171) were used to increase the translational relevance of our data. Compared to wild-type controls, Sirt5Tg/0 mice displayed accelerated arterial thrombus formation following endothelial-specific damage. Conversely, in Sirt5-/- mice, arterial thrombosis was blunted. Platelet function was unaltered, as assessed by ex vivo collagen-induced aggregometry. Similarly, activation of the coagulation cascade as assessed by vascular and plasma tissue factor (TF) and TF pathway inhibitor expression was unaltered. Increased thrombus embolization episodes and circulating D-dimer levels suggested augmented activation of the fibrinolytic system in Sirt5-/- mice. Accordingly, Sirt5-/- mice showed reduced plasma and vascular expression of the fibrinolysis inhibitor plasminogen activator inhibitor (PAI)-1. In HAECs, SIRT5-silencing inhibited PAI-1 gene and protein expression in response to TNF-α. This effect was mediated by increased AMPK activation and reduced phosphorylation of the MAP kinase ERK 1/2, but not JNK and p38 as shown both in vivo and in vitro. Lastly, both PAI-1 and SIRT5 gene expressions are increased in ACS patients compared to non-ACS controls after adjustment for cardiovascular risk factors, while PAI-1 expression increased across tertiles of SIRT5. CONCLUSION: SIRT5 promotes arterial thrombosis by modulating fibrinolysis through endothelial PAI-1 expression. Hence, SIRT5 may be an interesting therapeutic target in the context of atherothrombotic events.


Subject(s)
Carotid Artery Injuries/enzymology , Carotid Artery Thrombosis/enzymology , Endothelial Cells/enzymology , Fibrinolysis , Sirtuins/metabolism , AMP-Activated Protein Kinases/metabolism , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/enzymology , Adult , Aged , Animals , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/genetics , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Sirtuins/genetics
3.
Arterioscler Thromb Vasc Biol ; 40(10): 2391-2403, 2020 10.
Article in English | MEDLINE | ID: mdl-32787521

ABSTRACT

OBJECTIVE: Reelin, a secreted glycoprotein, was originally identified in the central nervous system, where it plays an important role in brain development and maintenance. In the cardiovascular system, reelin plays a role in atherosclerosis by enhancing vascular inflammation and in arterial thrombosis by promoting platelet adhesion, activation, and thrombus formation via APP (amyloid precursor protein) and GP (glycoprotein) Ib. However, the role of reelin in hemostasis and arterial thrombosis is not fully understood to date. Approach and Results: In the present study, we analyzed the importance of reelin for cytoskeletal reorganization of platelets and thrombus formation in more detail. Platelets release reelin to amplify alphaIIb beta3 integrin outside-in signaling by promoting platelet adhesion, cytoskeletal reorganization, and clot retraction via activation of Rho GTPases RAC1 (Ras-related C3 botulinum toxin substrate) and RhoA (Ras homolog family member A). Reelin interacts with the collagen receptor GP (glycoprotein) VI with subnanomolar affinity, induces tyrosine phosphorylation in a GPVI-dependent manner, and supports platelet binding to collagen and GPVI-dependent RAC1 activation, PLC gamma 2 (1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2) phosphorylation, platelet activation, and aggregation. When GPVI was deleted from the platelet surface by antibody treatment in reelin-deficient mice, thrombus formation was completely abolished after injury of the carotid artery while being only reduced in either GPVI-depleted or reelin-deficient mice. CONCLUSIONS: Our study identified a novel signaling pathway that involves reelin-induced GPVI activation and alphaIIb beta3 integrin outside-in signaling in platelets. Loss of both, GPVI and reelin, completely prevents stable arterial thrombus formation in vivo suggesting that inhibiting reelin-platelet-interaction might represent a novel strategy to avoid arterial thrombosis in cardiovascular disease.


Subject(s)
Blood Platelets/enzymology , Carotid Artery Injuries/enzymology , Cell Adhesion Molecules, Neuronal/blood , Extracellular Matrix Proteins/blood , Nerve Tissue Proteins/blood , Neuropeptides/blood , Phospholipase C gamma/blood , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Serine Endopeptidases/blood , Thrombosis/enzymology , rac1 GTP-Binding Protein/blood , rhoA GTP-Binding Protein/blood , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood Coagulation , Carotid Artery Injuries/blood , Carotid Artery Injuries/etiology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Clot Retraction , Cytoskeleton/enzymology , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Platelet Activation , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/etiology
4.
Arterioscler Thromb Vasc Biol ; 40(10): e262-e272, 2020 10.
Article in English | MEDLINE | ID: mdl-32814440

ABSTRACT

OBJECTIVE: The risk of thrombosis in myeloproliferative neoplasms, such as primary myelofibrosis varies depending on the type of key driving mutation (JAK2 [janus kinase 2], CALR [calreticulin], and MPL [myeloproliferative leukemia protein or thrombopoietin receptor]) and the accompanying mutations in other genes. In the current study, we sought to examine the propensity for thrombosis, as well as platelet activation properties in a mouse model of primary myelofibrosis induced by JAK2V617F (janus kinase 2 with valine to phenylalanine substitution on codon 617) mutation. Approach and Results: Vav1-hJAK2V617F transgenic mice show hallmarks of primary myelofibrosis, including significant megakaryocytosis and bone marrow fibrosis, with a moderate increase in red blood cells and platelet number. This mouse model was used to study responses to 2 models of vascular injury and to investigate platelet properties. Platelets derived from the mutated mice have reduced aggregation in response to collagen, reduced thrombus formation and thrombus size, as demonstrated using laser-induced or FeCl3-induced vascular injury models, and increased bleeding time. Strikingly, the mutated platelets had a significantly reduced number of dense granules, which could explain impaired ADP secretion upon platelet activation, and a diminished second wave of activation. CONCLUSIONS: Together, our study highlights for the first time the influence of a hyperactive JAK2 on platelet activation-induced ADP secretion and dense granule homeostasis, with consequent effects on platelet activation properties.


Subject(s)
Blood Coagulation , Blood Platelets/enzymology , Carotid Artery Injuries/enzymology , Janus Kinase 2/blood , Megakaryocytes/enzymology , Platelet Activation , Primary Myelofibrosis/enzymology , Thrombosis/enzymology , Animals , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Disease Models, Animal , Janus Kinase 2/genetics , Mice, Transgenic , Mutation , Platelet Aggregation , Primary Myelofibrosis/blood , Primary Myelofibrosis/genetics , Thrombopoiesis , Thrombosis/blood , Thrombosis/genetics
5.
Mol Med ; 25(1): 33, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31307370

ABSTRACT

BACKGROUND: The beneficial effects of angiotensin II type 1 receptor blockers (ARBs) on atherosclerosis have been demonstrated in numerous studies. We investigated the effects of fimasartan on reducing neointimal formation and systemic inflammation after carotid artery (CA) injury in Apolipoprotein E knockout (ApoE KO) mice. METHODS: ApoE KO mice were randomly allocated to Group I (without CA injury), Group II (without CA injury + Fimasartan), Group III (CA injury), and Group IV (CA injury + Fimasartan). Fimasartan was orally administered everyday starting 3 days before iatrogenic left CA injury. RESULTS: At 28 days, neointimal hyperplasia and the inflammatory cytokines including TNFα, IL-6, ICAM, and MMP-9 in the peripheral blood were significantly reduced in Groups II and IV compared to Groups I and III, respectively. All fimasartan-administered groups revealed significant increases of CD4+CD25+Foxp3+ regulatory T (Treg) cells with increased plasma levels of IL-10 and TGFß. In addition, increased CD8+ T cells by fimasartan were correlated with reduced smooth muscle cell (SMC) proliferation in the neointima in Groups II and IV. Furthermore, the populations of Treg and CD8+ T cells in total splenocytes were increased in Groups II and IV compared to Groups I and III, respectively. The enlargement of spleens due to CA injury in the Group III was attenuated by fimasartan, as shown in the Group IV. These data indicate that fimasartan significantly reduced SMC proliferation in neointima and increased Treg cells in ApoE KO CA injury mice. CONCLUSIONS: This study suggests fimasartan could be an efficient strategy for reduction of atherosclerotic progression, with a decrease in immune response and systemic inflammation.


Subject(s)
Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/therapeutic use , Carotid Artery Injuries/blood , Carotid Artery Injuries/drug therapy , Inflammation/blood , Inflammation/drug therapy , Neointima/blood , Neointima/drug therapy , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Tetrazoles/pharmacokinetics , Tetrazoles/therapeutic use , Angiotensin Receptor Antagonists/therapeutic use , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Interleukin-6/blood , Male , Matrix Metalloproteinase 9/blood , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/drug effects , Tumor Necrosis Factor-alpha/blood
6.
J Am Heart Assoc ; 8(5): e011273, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30791801

ABSTRACT

Background Members of the regulator of G-protein signaling ( RGS ) family inhibit G-protein coupled receptor signaling by modulating G-protein activity. In platelets, there are 3 different RGS isoforms that are expressed at the protein level, including RGS 16. Recently, we have shown that CXCL 12 regulates platelet function via RGS 16. However, the role of RGS 16 in platelet function and thrombus formation is poorly defined. Methods and Results We used a genetic knockout mouse model approach to examine the role(s) of RGS 16 in platelet activation by using a host of in vitro and in vivo assays. We observed that agonist-induced platelet aggregation, secretion, and integrin activation were much more pronounced in platelets from the RGS 16 knockout ( Rgs16 -/-) mice relative to their wild type ( Rgs16 +/+) littermates. Furthermore, the Rgs16 -/- mice had a markedly shortened bleeding time and were more susceptible to vascular injury-associated thrombus formation than the controls. Conclusions These findings support a critical role for RGS 16 in regulating hemostatic and thrombotic functions of platelets in mice. Hence, RGS 16 represents a potential therapeutic target for modulating platelet function.


Subject(s)
Blood Platelets/metabolism , Carotid Artery Injuries/genetics , Hemostasis , Platelet Activation , RGS Proteins/blood , Thrombosis/blood , Animals , Carotid Artery Injuries/blood , Disease Models, Animal , Humans , Mice, Knockout , P-Selectin/blood , Phosphatidylserines/blood , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , RGS Proteins/deficiency , RGS Proteins/genetics , Signal Transduction , Thrombosis/genetics
7.
Ann Vasc Surg ; 57: 201-209, 2019 May.
Article in English | MEDLINE | ID: mdl-30684618

ABSTRACT

BACKGROUND: Intimal hyperplasia (IH) is the most common indicator for secondary intervention in peripheral vascular disease. Matrix metalloproteinases (MMPs) play a role in IH development due to their degradation of the extracellular matrix. Doxycycline (Doxy), a member of the tetracycline family of antibiotics, is a potent MMP inhibitor. We have previously shown that Doxy inhibits MMP activity and vascular smooth muscle cell migration in vitro. We hypothesized that Doxy would decrease MMP activity in vivo and inhibit the development of IH in a rodent model of vascular injury. METHODS AND RESULTS: Doxy (400 mg/pellet) was delivered by a slow-release pellet implanted 3 days prior to or at the time of balloon angioplasty (BA) of the common carotid artery in female rats. At 14 days post-BA, intima-to-media (I:M) ratios were 0.77 ± 0.21 and 1.04 ± 0.32 in the Doxy treated groups, respectively, compared to 1.25 ± 0.26 in the control group (P = not significant; n = 3). Additionally, the tested dose of Doxy in either group had no inhibitory effect on membrane type 1-MMP or MMP-2 tissue levels, as measured by immunohistochemistry, or on systemic levels of MMP, as measured by total MMP serum levels using enzyme-linked immunosorbent assay. At 14 days post-BA, VSMC proliferation in the injured artery was increased to Doxy treatment prior to and at the time of surgery (23.5 ± 3.4 and 27.2 ± 3.9%, respectively), compared to control (11.4 ± 0.4%; n = 3), as measured by proliferating cellular nuclear antigen immunostaining. CONCLUSIONS: In our in vivo model of vascular injury, systemic Doxy administration prior to or at the time of vascular injury does not significantly hinder the progression of IH development. Additional doses and routes of administration could be examined in order to correlate therapeutic serum levels of Doxy with effective MMP inhibition in serum and arterial tissue. However, alternative drug delivery systems are needed in order to optimize therapeutic administration of targeted MMP inhibitors for the prevention of IH development.


Subject(s)
Angioplasty, Balloon/adverse effects , Cardiovascular Agents/administration & dosage , Carotid Artery Injuries/drug therapy , Doxycycline/administration & dosage , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Animals , Carotid Artery Injuries/blood , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/enzymology , Carotid Artery, Common/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Female , Hyperplasia , Matrix Metalloproteinase 14/blood , Matrix Metalloproteinase 2/blood , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Rats, Sprague-Dawley
8.
Biomed Pharmacother ; 110: 887-894, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30572193

ABSTRACT

BACKGROUND: Intimal hyperplasia is the major therapeutic concern after percutaneous coronary intervention. The aim of this study is to investigate effects of 2,3,4',5-tetrahydroxystilbene-2-O-ß-D glucoside (TSG) on intimal hyperplasia and the underling mechanisms through attenuating the expressions of stromal cell-derived factor-1α (SDF-1α)/CXCR4, stem cell factor (SCF)/c-kit and fractalkine (FKN)/CX3CR1, and through promoting re-endothelialization with vascular endothelial growth factor (VEGF). METHOD: Rats were operated with carotid artery balloon injury. The treatment groups were gavaged with 50 and 100 mg/kg/d of TSG. After 10 days of treatment, carotid artery pathological changes were evaluated by histology. Serum levels of SDF-1α, SCF, FKN and VEGF were detected by enzyme linked immunosorbent assay. The protein expressions of the receptors c-kit, CXCR4, CX3CR1, as well as CD34 and proliferating cell nuclear antigen (PCNA) were detected by immunochemistry. RESULTS: TSG dose-dependently inhibited balloon injury-induced intimal hyperplasia, as evidenced by reducing neointima area (NIA), neointima area/media area (NIA/MA), neointima area/internal elastic area (NIA/IELA), and by decreasing the protein expression of PCNA. TSG reduced serum levels of SDF-1α, SCF and FKN, and it also decreased the expressions of the corresponding receptors c-kit, CXCR4, CX3CR1 in neointima. Importantly, the level of VEGF in peripheral blood and the expression of CD34 in vascular walls were increased to promote re-endothelialization. CONCLUSIONS: This study clearly demonstrated that TSG was effective in inhibiting intimal hyperplasia, and this effect was mediated, at least in part, through the SCF/c-kit, SDF-1α/CXCR4 and FKN/CX3CR1 axes. Importantly, TSG could increase VEGF and CD34 to promote endothelial repair.


Subject(s)
Carotid Artery Injuries/blood , Chemokine CX3CL1/blood , Chemokine CXCL12/blood , Glucosides/therapeutic use , Stem Cell Factor/blood , Stilbenes/therapeutic use , Vascular Endothelial Growth Factor A/blood , Angioplasty, Balloon/adverse effects , Animals , Carotid Artery Injuries/pathology , Carotid Artery Injuries/prevention & control , Carotid Artery, Common/drug effects , Carotid Artery, Common/pathology , Glucosides/pharmacology , Hyperplasia/blood , Hyperplasia/pathology , Hyperplasia/prevention & control , Male , Neointima/blood , Neointima/pathology , Neointima/prevention & control , Random Allocation , Rats , Rats, Sprague-Dawley , Stilbenes/pharmacology
9.
Int J Mol Med ; 42(2): 1074-1085, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29749466

ABSTRACT

Mono­unsaturated free fatty acids (FFAs) can serve as a predictive indicator of vascular restenosis following interventional therapy, particularly in individuals with high­fat diet­induced type 2 diabetes. However, the pathogenic mechanism remains to be fully elucidated. In the present study, the levels of tyrosine 3­monooxygenase/tryptophan 5­monooxygenase activation protein ß (YWHAB; also known as 14­3­3ß), in vascular smooth muscle cells (VSMCs) treated with different concentrations of oleic acid (OA) were examined by reverse transcription­quantitative polymerase chain reaction and western blot analyses. The migration of VSMCs was examined using wound­healing and Transwell migration assays. The protein distribution of B­cell lymphoma 2 (BCL­2)­associated death promoter (BAD) in VSMCs treated with OA was examined by immunofluorescence and western blot analyses. In in vivo experiments, the carotid artery morphology of rats in different groups was assessed at 14 days post­injury by non-invasive ultrasonographic imaging and confirmed by histological staining. The expression of YWHAB was upregulated by OA in a concentration­dependent manner in VSMCs. In the in vivo experiments, carotid stenosis was more serious among high­FFA diabetic rats. However, silencing of YWHAB significantly alleviated carotid neointimal hyperplasia among the diabetic rats with elevated FFA levels. In addition, YWHAB silencing alleviated the migration of OA­treated VSMCs and increased translocation of the BAD protein from the cytoplasm to the mitochondria. In conclusion, the results showed that FFA­induced upregulation of YWHAB was involved in neointimal hyperplasia by enhancing the migration of VSMCs following carotid artery injury. The inhibition of YWHAB may serve as a novel potential pharmacological target for preventing vascular restenosis following interventional therapy in diabetic individuals with high FFA levels.


Subject(s)
14-3-3 Proteins/genetics , Carotid Arteries/pathology , Carotid Artery Injuries/complications , Diabetes Mellitus, Experimental/complications , Fatty Acids/blood , Up-Regulation , 14-3-3 Proteins/metabolism , Animals , Carotid Arteries/metabolism , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Cell Movement , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Fatty Acids/metabolism , Gene Silencing , Male , Neointima/blood , Neointima/complications , Neointima/genetics , Neointima/metabolism , Rats, Sprague-Dawley
10.
J Am Heart Assoc ; 6(8)2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28751543

ABSTRACT

BACKGROUND: GPVI (Glycoprotein VI) is the essential platelet collagen receptor in atherothrombosis. Dimeric GPVI-Fc (Revacept) binds to GPVI binding sites on plaque collagen. As expected, it did not increase bleeding in clinical studies. GPVI-Fc is a potent inhibitor of atherosclerotic plaque-induced platelet aggregation at high shear flow, but its inhibition at low shear flow is limited. We sought to increase the platelet inhibitory potential by fusing GPVI-Fc to the ectonucleotidase CD39 (fusion protein GPVI-CD39), which inhibits local ADP accumulation at vascular plaques, and thus to create a lesion-directed dual antiplatelet therapy that is expected to lack systemic bleeding risks. METHODS AND RESULTS: GPVI-CD39 effectively stimulated local ADP degradation and, compared with GPVI-Fc alone, led to significantly increased inhibition of ADP-, collagen-, and human plaque-induced platelet aggregation in Multiplate aggregometry and plaque-induced platelet thrombus formation under arterial flow conditions. GPVI-CD39 did not increase bleeding time in an in vitro assay simulating primary hemostasis. In a mouse model of ferric chloride-induced arterial thrombosis, GPVI-CD39 effectively delayed vascular thrombosis but did not increase tail bleeding time in vivo. CONCLUSIONS: GPVI-CD39 is a novel approach to increase local antithrombotic activity at sites of atherosclerotic plaque rupture or injury. It enhances GPVI-Fc-mediated platelet inhibition and presents a potentially effective and safe molecule for the treatment of acute atherothrombotic events, with a favorable risk-benefit ratio.


Subject(s)
Antigens, CD/pharmacology , Apyrase/pharmacology , Carotid Artery Injuries/drug therapy , Fibrinolytic Agents/pharmacology , Glycoproteins/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/pharmacology , Thrombosis/prevention & control , Animals , Antigens, CD/toxicity , Apyrase/pharmacokinetics , Apyrase/toxicity , Carotid Artery Diseases/blood , Carotid Artery Diseases/pathology , Carotid Artery Injuries/blood , Carotid Artery Injuries/chemically induced , Carotid Artery Injuries/pathology , Chlorides , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Ferric Compounds , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/toxicity , Glycoproteins/pharmacokinetics , Glycoproteins/toxicity , Hemorrhage/chemically induced , Humans , Immunoglobulin Fc Fragments/toxicity , Male , Mice, Inbred C57BL , Plaque, Atherosclerotic , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/toxicity , Platelet Membrane Glycoproteins/pharmacokinetics , Platelet Membrane Glycoproteins/toxicity , Recombinant Fusion Proteins/pharmacology , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/pathology
11.
Thromb Haemost ; 117(9): 1782-1797, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28726976

ABSTRACT

Platelet degranulation at the site of vascular injury prevents bleeding and may affect the chronic vascular wound healing response. Transforming Growth Factor (TGF)-ß1 is a major component of platelet α-granules known to accumulating in thrombi. It was our aim to determine the role of TGFß1 released from activated platelets for neointima formation following arterial injury and thrombosis. Mice with platelet-specific deletion of TGFß1 (Plt.TGFß-KO) underwent carotid artery injury. Immunoassays confirmed the absence of active TGFß1 in platelet releasates and plasma of Plt.TGFß-KO mice. Whole blood analyses revealed similar haematological parameters, and tail cut assays excluded major bleeding defects. Platelet aggregation and the acute thrombotic response to injury in vivo did not differ between Plt.TGFß-KO and Plt.TGFß-WT mice. Morphometric analysis revealed that absence of TGFß1 in platelets resulted in a significant reduction of neointima formation with lower neointima area, intima-to-media ratio, and lumen stenosis. On the other hand, the media area was enlarged in mice lacking TGFß1 in platelets and contained increased amounts of proteases involved in latent TGFß activation, including MMP2, MMP9 and thrombin. Significantly increased numbers of proliferating cells and cells expressing the mesenchymal markers platelet-derived growth factor receptor-ß or fibroblast-specific protein-1, and the macrophage antigen F4/80, were observed in the media of Plt.TGFß-KO mice, whereas the medial smooth muscle-actin-immunopositive area and collagen content did not differ between genotypes. Our findings support an essential role for platelet-derived TGFß1 for the vascular remodelling response to arterial injury, apparently independent from the role of platelets in thrombosis or haemostasis.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Carotid Artery Injuries/blood , Carotid Artery, Common/metabolism , Neointima , Thrombosis/blood , Transforming Growth Factor beta1/blood , Vascular Remodeling , Animals , Biomarkers/metabolism , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Artery, Common/pathology , Disease Models, Animal , Down-Regulation , Female , Genetic Predisposition to Disease , Male , Mice, Knockout , Phenotype , Platelet Activation , Thrombosis/genetics , Thrombosis/pathology , Transforming Growth Factor beta1/genetics
12.
Arterioscler Thromb Vasc Biol ; 37(7): 1307-1314, 2017 07.
Article in English | MEDLINE | ID: mdl-28596377

ABSTRACT

OBJECTIVE: Currently prescribed antiplatelet drugs have 1 common side effect-an increased risk of hemorrhage and thrombocytopenia. On the contrary, bleeding defects associated with glycoprotein VI (GPVI) expression deficiency are usually slightly prolonged bleeding times. However, GPVI antagonists are lacking in clinic. APPROACH AND RESULTS: Using reverse-phase high-performance liquid chromatography and sequencing, we revealed the partial sequence of trowaglerix α subunit, a potent specific GPVI-targeting snaclec (snake venom C-type lectin protein). Hexapeptide (Troα6 [trowaglerix a chain hexapeptide, CKWMNV]) and decapeptide (Troα10) derived from trowaglerix specifically inhibited collagen-induced platelet aggregation through blocking platelet GPVI receptor. Computational peptide design helped to design a series of Troα6/Troα10 peptides. Protein docking studies on these decapeptides and GPVI suggest that Troα10 was bound at the lower surface of D1 domain and outer surface of D2 domain, which was at the different place of the collagen-binding site and the scFv (single-chain variable fragment) D2-binding site. The newly discovered site was confirmed by inhibitory effects of polyclonal antibodies on collagen-induced platelet aggregation. This indicates that D2 domain of GPVI is a novel and important binding epitope on GPVI-mediated platelet aggregation. Troα6/Troα10 displayed prominent inhibitory effect of thrombus formation in fluorescein sodium-induced platelet thrombus formation of mesenteric venules and ferric chloride-induced carotid artery injury thrombosis model without prolonging the in vivo bleeding time. CONCLUSIONS: We develop a novel antithrombotic peptides derived from trowaglerix that acts through GPVI antagonism with greater safety-no severe bleeding. The binding epitope of polypeptides on GPVI is novel and important. These hexa/decapeptides have therapeutic potential for developing ideal small-mass GPVI antagonists for arterial thrombogenic diseases.


Subject(s)
Blood Platelets/drug effects , Carotid Artery Injuries/drug therapy , Crotalid Venoms/pharmacology , Fibrinolytic Agents/pharmacology , Peptide Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Binding Sites , Blood Platelets/metabolism , Carotid Artery Injuries/blood , Carotid Artery Injuries/chemically induced , Chlorides , Computer-Aided Design , Crotalid Venoms/metabolism , Crotalid Venoms/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Ferric Compounds , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/toxicity , Fluorescein , Hemorrhage/chemically induced , Humans , Lectins, C-Type/metabolism , Male , Mice, Inbred ICR , Molecular Docking Simulation , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/toxicity , Platelet Membrane Glycoproteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction/drug effects , Thrombosis/blood , Thrombosis/chemically induced
13.
Cardiovasc Res ; 113(5): 498-507, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28199510

ABSTRACT

AIMS: The lectin-like oxLDL receptor-1 (LOX-1) promotes endothelial uptake of oxidized low-density lipoprotein (oxLDL) and plays an important role in atherosclerosis and acute coronary syndromes (ACS). However, its role in arterial thrombus formation remains unknown. We investigated whether LOX-1 plays a role in arterial thrombus formation in vivo at different levels of oxLDL using endothelial-specific LOX-1 transgenic mice (LOX-1TG) and a photochemical injury thrombosis model of the carotid artery. METHODS AND RESULTS: In mice fed a normal chow diet, time to arterial occlusion was unexpectedly prolonged in LOX-1TG as compared to WT. In line with this, tissue factor (TF) expression and activity in carotid arteries of LOX-1TG mice were reduced by half. This effect was mediated by activation of octamer transcription factor 1 (Oct-1) leading to upregulation of the mammalian deacetylase silent information regulator-two 1 (SIRT1) via binding to its promoter and subsequent inhibition of NF-κB signaling. In contrast, intravenous injection of oxLDL as well as high cholesterol diet for 6 weeks led to a switch from the Oct-1/SIRT1 signal transduction pathway to the ERK1/2 pathway and in turn to an enhanced thrombotic response with shortened occlusion time. CONCLUSIONS: Thus, LOX-1 differentially regulates thrombus formation in vivo depending on the degree of activation by oxLDL. At low oxLDL levels LOX-1 activates the protective Oct-1/SIRT1 pathway, while at higher levels of the lipoprotein switches to the thrombogenic ERK1/2 pathway. These findings may be important for arterial thrombus formation in ACS and suggest that SIRT1 may represent a novel therapeutic target in this context.


Subject(s)
Blood Coagulation , Carotid Arteries/enzymology , Carotid Artery Injuries/enzymology , Lipoproteins, LDL/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Octamer Transcription Factor-1/metabolism , Scavenger Receptors, Class E/metabolism , Sirtuin 1/metabolism , Thrombosis/enzymology , Animals , Binding Sites , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Cholesterol, Dietary , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Octamer Transcription Factor-1/genetics , Phenotype , Phosphorylation , Promoter Regions, Genetic , Scavenger Receptors, Class E/genetics , Signal Transduction , Sirtuin 1/genetics , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/genetics , Thrombosis/prevention & control , Time Factors
14.
Cardiovasc Res ; 113(1): 61-69, 2017 01.
Article in English | MEDLINE | ID: mdl-28028070

ABSTRACT

AIMS: The P2Y12 antagonist ticagrelor reduces mortality in patients with acute coronary syndrome (ACS), compared with clopidogrel, and the mechanisms underlying this effect are not clearly understood. Arterial thrombosis is the key event in ACS; however, direct vascular effects of either ticagrelor or clopidogrel with focus on arterial thrombosis and its key trigger tissue factor have not been previously investigated. METHODS AND RESULTS: Human aortic endothelial cells were treated with ticagrelor or clopidogrel active metabolite (CAM) and stimulated with tumour necrosis factor-alpha (TNF-α); effects on procoagulant tissue factor (TF) expression and activity, its counter-player TF pathway inhibitor (TFPI) and the underlying mechanisms were determined. Further, arterial thrombosis by photochemical injury of the common carotid artery, and TF expression in the murine endothelium were examined in C57BL/6 mice treated with ticagrelor or clopidogrel. Ticagrelor, but not CAM, reduced TNF-α-induced TF expression via proteasomal degradation and TF activity, independently of the P2Y12 receptor and the equilibrative nucleoside transporter 1 (ENT1), an additional target of ticagrelor. In C57BL/6 mice, ticagrelor prolonged time to arterial occlusion, compared with clopidogrel, despite comparable antiplatelet effects. In line with our in vitro results, ticagrelor, but not clopidogrel, reduced TF expression in the endothelium of murine arteries. CONCLUSION: Ticagrelor, unlike clopidogrel, exhibits endothelial-specific antithrombotic properties and blunts arterial thrombus formation. The additional antithrombotic properties displayed by ticagrelor may explain its greater efficacy in reducing thrombotic events in clinical trials. These findings may provide the basis for new indications for ticagrelor.


Subject(s)
Adenosine/analogs & derivatives , Blood Coagulation/drug effects , Carotid Artery Injuries/drug therapy , Endothelial Cells/drug effects , Fibrinolytic Agents/pharmacology , Thromboplastin/metabolism , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Adenosine/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Cells, Cultured , Clopidogrel , Disease Models, Animal , Down-Regulation , Endothelial Cells/metabolism , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Humans , Male , Mice, Inbred C57BL , Platelet Aggregation Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/metabolism , Thromboplastin/genetics , Thrombosis/blood , Thrombosis/genetics , Ticagrelor , Ticlopidine/pharmacology , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
15.
J Cardiovasc Pharmacol ; 69(2): 101-109, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27875385

ABSTRACT

Diabetic patients suffer an increased risk of restenosis and late stent thrombosis after angioplasty, complications which are related to a defective reendothelialization. Dipeptidyl peptidase-4 inhibitors have been suggested to exert a direct effect on endothelial and smooth muscle cells (SMCs). Therefore, the objective was to study if the dipeptidyl peptidase-4 inhibitor linagliptin could influence vascular repair and accelerate reendothelialization after arterial injury in healthy and diabetic animals. Diabetic Goto-Kakizaki and healthy Wistar rats were subjected to arterial injury and treated with linagliptin or vehicle. Vessel wall healing was monitored noninvasively using ultrasound, and on sacrifice, with Evans blue staining and immunohistochemistry. The effect of linagliptin on SMCs was also studied in vitro. We found that linagliptin reduced the proliferation and dedifferentiation of SMCs in vitro, and modulated the inflammatory response in the SMCs after arterial injury in vivo. However, these effects of linagliptin did not affect the neointima formation or the reendothelialization under normal and diabetic conditions. Although linagliptin did not influence vessel wall healing, it seems to possess a desirable antiproliferative influence on SMCs in vitro and an antiinflammatory effect in vivo. These pharmacological properties might carry a potential significance for favorable outcome after vascular interventions in diabetic patients.


Subject(s)
Carotid Artery Injuries/drug therapy , Carotid Artery, External/drug effects , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Linagliptin/therapeutic use , Wound Healing/drug effects , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Carotid Artery Injuries/blood , Carotid Artery, External/metabolism , Cell Proliferation/drug effects , Cell Proliferation/physiology , Diabetes Mellitus, Type 2/blood , Dose-Response Relationship, Drug , Hypoglycemic Agents/pharmacology , Linagliptin/pharmacology , Male , Random Allocation , Rats , Rats, Wistar , Treatment Outcome , Wound Healing/physiology
16.
Int J Hematol ; 105(1): 100-103, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27709451

ABSTRACT

Thrombopoietin receptor (TPO-R) agonists have been shown to be effective in refractory chronic immune thrombocytopenia (ITP); however, their efficacy in patients under critical care is not known. We report the case of a female patient with a newly diagnosed ITP who experienced severe bleeding from an external wound. The patient was administered the standard treatments for ITP, which are high-dose intravenous immunoglobulin (IVIg) and corticosteroids. However, following failure of these treatments, we administered romiplostim on day 6 after the onset of ITP. On day 6 after the initiation of romiplostim, there was improvement in platelet count and bleeding tendency. We were subsequently able to perform a splenectomy successfully. The efficacy of TPO-R agonists in ITP has been reported in several situations, including before surgery in an ITP patient; however, the use of TPO-R for arterial bleeding with shock has not been reported. To our knowledge, the present article is a rare case report of the use of a TPO-R agonist in a patient with critical artery injury. Our data suggest that the early use of romiplostim is effective in emergency cases of newly diagnosed ITP with life-threatening bleeding, which is refractory to standard treatment.


Subject(s)
Carotid Artery Injuries/complications , Hemorrhage/complications , Purpura, Thrombocytopenic, Idiopathic/complications , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Receptors, Fc/therapeutic use , Receptors, Thrombopoietin/agonists , Recombinant Fusion Proteins/therapeutic use , Thrombopoietin/therapeutic use , Aged , Carotid Artery Injuries/blood , Carotid Artery Injuries/drug therapy , Female , Hemorrhage/blood , Hemorrhage/drug therapy , Humans , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/blood
17.
Thromb Haemost ; 117(1): 83-89, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27761579

ABSTRACT

Despite public awareness of its deleterious effects, smoking remains a major cause of death. Indeed, it is a risk factor for atherothrombotic complications and in line with this, the introduction of smoking ban in public areas reduced smoking-associated cardiovascular complications. Nonetheless, smoking remains a major concern, and molecular mechanisms by which it causes cardiovascular disease are not known. Peripheral blood monocytes from healthy smokers displayed increased JNK2 and tissue factor (TF) gene expression compared to non-smokers (n=15, p<0.05). Similarly, human aortic endothelial cells exposed to cigarette smoke total particulate matter (CS-TPM) revealed increased TF expression mediated by JNK2 (n=4; p<0.05). Wild-type and JNK2-/- mice were exposed to cigarette smoke for two weeks after which arterial thrombosis was investigated. Wild-type mice exposed to smoke displayed reduced time to thrombotic arterial occlusion (n=8; p<0.05) and increased tissue factor activity (n=7; p<0.05) as compared to wild-type controls (n=6), while JNK2-/-mice exposed to smoke maintained an unaltered thrombotic potential (n=8; p=NS) and tissue factor activity (n=8) comparable to that of JNK2-/- and wild-type controls (n=6; p=NS). Smoking caused an increased production of reactive oxygen species (ROS) in wild-type but not in JNK2-/- mice (n=7; p<0.05 for wild-type mice and n=5-6; p=NS for JNK2-/- mice). In conclusion, the MAP kinase JNK2 mediates cigarette smoke-induced TF activation, arterial thrombosis and ROS production. These results underscore a major role of JNK2 in smoke-mediated thrombus formation and may offer an attractive target to prevent smoke-related thrombosis in those subjects which do not manage quitting.


Subject(s)
Arterial Occlusive Diseases/etiology , Blood Coagulation , Mitogen-Activated Protein Kinase 9/metabolism , Smoke/adverse effects , Smoking/adverse effects , Thrombosis/etiology , Animals , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/enzymology , Arterial Occlusive Diseases/genetics , Carotid Artery Injuries/blood , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/genetics , Cells, Cultured , Endothelial Cells/enzymology , Female , Genetic Predisposition to Disease , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction , Smoking/blood , Smoking/genetics , Thromboplastin/genetics , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/genetics
18.
J Biol Regul Homeost Agents ; 30(3): 795-799, 2016.
Article in English | MEDLINE | ID: mdl-27655500

ABSTRACT

The first choice of treatment for neck cancer is often radiotherapy. Therefore, we aimed to investigate the microinflammation after radiotherapy of the neck and the incidence of carotid stenosis. This study reports on patients treated with radiotherapy as part of the treatment for laryngeal cancer in the Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, P.R. China. Sixty-two males and nine females were treated with radiotherapy between 2006 and 3012. The carotid diameter was determined by measuring carotid intima-media thickness (IMT) in the common, external and internal carotid artery. Microinflammatory conditions were assessed by high-sensitivity C-reactive protein (hs-CRP), interleukin-6 (IL-6) and tumor necrosis factor–alpha (TNF-α). Other studied risk factors included age, treatment modalities, radiation dose and energy, the height of the radiation field, and follow-up time. Carotid stenosis was detected in all of the 71 patients. It was mainly clinically unsuspected; 19 patients had sustained a vascular event (14 TIA, 5 CVI) at a median of 3.11 years (range 2.3–5.6 years) following RT. In four of five CVI patients, CVI occurred on the side of the irradiation. Eleven patients who suffered vascular incident had severe stenosis of the carotid artery and 6 had moderate (31-49% of the lumen). Only two patients with mild stenosis on the irradiated side suffered TIAs. Serum hs-CRP levels in carotid stenosis were 9.4 (±SD=5.97) mg/ml, IL-6 = 12.8 (±SD=2.62) pg/ml and TNF-α = 15.4 (±SD=4.49) ng/ml. The clinical detection of asymptomatic carotid stenosis is challenging, and current recommendations regarding the follow-up period should be scrutinized.


Subject(s)
Carotid Artery Injuries/etiology , Carotid Stenosis/etiology , Head and Neck Neoplasms/radiotherapy , Radiation Injuries/etiology , Radiotherapy/adverse effects , Adult , Aged , Asymptomatic Diseases , Biomarkers , C-Reactive Protein/analysis , Carotid Artery Injuries/blood , Carotid Artery Injuries/epidemiology , Carotid Intima-Media Thickness , Carotid Stenosis/blood , Carotid Stenosis/epidemiology , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Radiation Injuries/blood , Radiation Injuries/epidemiology , Radiotherapy Dosage , Stroke/epidemiology , Stroke/etiology , Tumor Necrosis Factor-alpha/analysis
19.
Thromb Res ; 144: 136-43, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27337700

ABSTRACT

Activated platelets secrete different chemokines, among others CCL5, thereby triggering inflammatory cell recruitment into the vessel wall. Here, we investigated how CCL5 deficiency influences vascular remodeling processes. Experiments were performed in apolipoprotein E and CCL5 double deficient (ApoE(-/-)×CCL5(-/-)) mice, using ApoE(-/-)×CCL5(+/+) mice as controls. The ferric chloride model was applied to induce thrombosis at the site of carotid artery injury within minutes and the formation of a smooth muscle cell-rich neointima within 3weeks. In both groups, vascular injury resulted in thrombus formation. CCL5 deficiency did not alter thrombus resolution examined at day 7. Analysis at 21days revealed that CCL5 absence was associated with a significant reduction in the neointima area (p<0.05), neointima-to-media ratio (p<0.05) and lumen stenosis (p<0.05) compared to ApoE(-/-)×CCL5(+/+) mice. Immunohistochemical analysis of CCL5 receptors showed decreased CCR5 positive staining in ApoE(-/-)×CCL5(-/-) mice (p<0.01), whereas the amount of CCR1 (p=0.053) and Mac2-positive macrophages (p<0.05) was increased. The amount of SMA-positive smooth muscle cells was lower in ApoE(-/-) mice lacking CCL5 (p<0.05). Positive staining for Krüppel-like factor 4 (KLF4), an atheroprotective transcription factor, was increased in the neointima of ApoE(-/-)×CCL5(-/-) mice (p<0.05) and found to co-localize with smooth muscle cells. In summary, CCL5 deficiency resulted in reduced neointima formation after carotid artery injury and thrombosis. Hemodynamic and histochemical analyses suggested that this was not due to differences in thrombus formation or resolution. Possibly, the atheroprotective effect of CCL5 deficiency is mediated by KLF4 upregulation in smooth muscle cells.


Subject(s)
Apolipoproteins E/genetics , Carotid Arteries/pathology , Carotid Artery Injuries/complications , Chemokine CCL5/genetics , Neointima/etiology , Neointima/genetics , Thrombosis/complications , Animals , Carotid Arteries/metabolism , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Gene Deletion , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/analysis , Male , Mice, Inbred C57BL , Mice, Knockout , Neointima/blood , Neointima/pathology , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology
20.
J Surg Res ; 200(2): 722-31, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26490225

ABSTRACT

BACKGROUND: Little is known about how arterial injury, nitric oxide (NO), or the diabetic milieu impact microparticle (MP) levels in the vasculature. We hypothesized that MP levels would increase following local arterial injury, and that NO would modify MP levels differently based on the metabolic environment. MATERIALS AND METHODS: Type 1 diabetes was induced in male Lean Zucker (LZ) rats with streptozotocin, and type 2 diabetes was induced in male Zucker diabetic fatty rats through diet. Lean Zucker rats served as nondiabetic controls. The rat carotid balloon injury was performed ± NO (n > 4/group). Blood was obtained at intervals from baseline to 14 d after injury and analyzed for platelet MP (PMP), leukocyte MP (LMP), and endothelial MP (EMP) using fluorescence-activated cell sorting (FACS) analysis. RESULTS: At baseline, type 1 diabetic rats had the highest EMP levels (P < 0.05). After arterial injury, type 1 and type 2 diabetic rats had a transient increase in EMP levels (P < 0.05) before decreasing below baseline levels. Both LMP and PMP levels generally declined after injury in all three animal models but were the lowest in both type 1 and type 2 diabetic rats. NO therapy had little impact on MP levels in nondiabetic and type 1 diabetic rats after injury. Conversely, NO caused a dramatic increase in EMP, LMP, and PMP levels in type 2 diabetic animals at early time points after injury (P < 0.05). CONCLUSIONS: These data demonstrate that the diabetic milieu impacts MP levels at baseline, after arterial injury and with NO treatment.


Subject(s)
Carotid Artery Injuries/drug therapy , Cell-Derived Microparticles/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Free Radical Scavengers/therapeutic use , Nitric Oxide/therapeutic use , Animals , Biomarkers/blood , Carotid Artery Injuries/blood , Carotid Artery Injuries/complications , Carotid Artery, Common , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Flow Cytometry , Male , Rats , Rats, Zucker , Streptozocin , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...