Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Microvasc Res ; 139: 104266, 2022 01.
Article in English | MEDLINE | ID: mdl-34688627

ABSTRACT

The no-reflow phenomenon induced by ischemia-reperfusion (I/R) injury seriously limits the therapeutic value of coronary recanalization and leads to a poor prognosis. Previous studies have shown that luteolin (LUT) is a vasoprotective factor. However, whether LUT can be used to prevent the no-reflow phenomenon remains unknown. Positron emission tomography perfusion imaging, performed to detect the effects of LUT on the no-reflow phenomenon in vivo, revealed that LUT treatment was able to reduce the no-reflow area in rat I/R models. In vitro, LUT was shown to reduce the hypoxia-reoxygenation injury-induced endothelial permeability and apoptosis. The levels of malondialdehyde, reactive oxygen species and NADPH were also measured and the results indicated that LUT could inhibit the oxidative stress. Western blot analysis revealed that LUT protected endothelial cells from I/R injury by regulating the Wnt/ß-catenin pathway. Overall, we concluded that the use of LUT to minimize I/R induced microvascular damage is a feasible strategy to prevent the no-reflow phenomenon.


Subject(s)
Coronary Circulation/drug effects , Coronary Vessels/drug effects , Endothelial Cells/drug effects , Luteolin/pharmacology , Myocardial Reperfusion Injury/prevention & control , No-Reflow Phenomenon/prevention & control , Wnt Signaling Pathway/drug effects , Animals , Apoptosis/drug effects , Capillary Permeability/drug effects , Cells, Cultured , Coronary Vessels/diagnostic imaging , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Myocardial Perfusion Imaging , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , No-Reflow Phenomenon/diagnostic imaging , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/physiopathology , Oxidative Stress/drug effects , Positron-Emission Tomography , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
2.
Am J Physiol Heart Circ Physiol ; 321(6): H1030-H1041, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34623177

ABSTRACT

The "no reflow" phenomenon, where the coronary artery is patent after treatment of acute myocardial infarction (AMI) but tissue perfusion is not restored, is associated with worse outcome. The mechanism of no reflow is unknown. We hypothesized that pericytes contraction, in an attempt to maintain a constant capillary hydrostatic pressure during reduced coronary perfusion pressure, causes capillary constriction leading to no reflow and that this effect is mediated through the orphan receptor, GPR39, present in pericytes. We created AMI (coronary occlusion followed by reperfusion) in GPR39 knock out mice and littermate controls. In a separate set of experiments, we treated wild-type mice undergoing coronary occlusion with vehicle or VC43, a specific inhibitor of GPR39, before reperfusion. We found that no reflow zones were significantly smaller in the GPR39 knockouts compared with controls. Both no reflow and infarct size were also markedly smaller in animals treated with VC43 compared with vehicle. Immunohistochemistry revealed greater capillary density and larger capillary diameter at pericyte locations in the GPR39-knockout and VC43-treated mice compared with controls. We conclude that GPR39-mediated pericyte contraction during reduced coronary perfusion pressure causes capillary constriction resulting in no reflow during AMI and that smaller no reflow zones in GPR39-knockout and VC43-treated animals are associated with smaller infarct sizes. These results elucidate the mechanism of no reflow in AMI, as well as providing a therapeutic pathway for the condition.NEW & NOTEWORTHY The mechanism of "no reflow" phenomenon, where the coronary artery is patent after treatment of acute myocardial infarction but tissue perfusion is not restored, is unknown. This condition is associated with worse outcome. Here, we show that GPR39-mediated pericyte contraction during reduced coronary perfusion pressure causes capillary constriction resulting in no reflow. Smaller no-reflow zones in GPR39-knockout animals and those treated with a GPR39 inhibitor are associated with smaller infarct size. These results could have important therapeutic implications.


Subject(s)
Cardiovascular Agents/pharmacology , Coronary Circulation/drug effects , Coronary Vessels/drug effects , Myocardial Infarction/drug therapy , No-Reflow Phenomenon/prevention & control , Pericytes/drug effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , Vasoconstriction/drug effects , Animals , Calcium Signaling/drug effects , Cells, Cultured , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/physiopathology , Pericytes/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
3.
Oxid Med Cell Longev ; 2021: 6699516, 2021.
Article in English | MEDLINE | ID: mdl-33613824

ABSTRACT

Coronary artery no-reflow is a complex problem in the area of reperfusion therapy, and the molecular mechanisms underlying coronary artery no-reflow injury have not been fully elucidated. In the present study, we explored whether oxidative stress caused damage to coronary endothelial cells by inducing mitochondrial fission and activating the JNK pathway. The hypoxia/reoxygenation (H/R) model was induced in vitro to mimic coronary endothelial no-reflow injury, and mitochondrial fission, mitochondrial function, and endothelial cell viability were analyzed using western blotting, quantitative polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA), and immunofluorescence. Our data indicated that reactive oxygen species (ROS) were significantly induced upon H/R injury, and this was followed by decreased endothelial cell viability. Mitochondrial fission was induced and mitochondrial bioenergetics were impaired in cardiac endothelial cells after H/R injury. Neutralization of ROS reduced mitochondrial fission and protected mitochondrial function against H/R injury. Our results also demonstrated that ROS stimulated mitochondrial fission via JNK-mediated Drp1 phosphorylation. These findings indicate that the ROS-JNK-Drp1 signaling pathway may be one of the molecular mechanisms underlying endothelial cell damage during H/R injury. Novel treatments for coronary no-reflow injury may involve targeting mitochondrial fission and the JNK-Drp1 signaling pathway.


Subject(s)
Dynamins/metabolism , Endothelium, Vascular/pathology , MAP Kinase Signaling System , Mitochondrial Dynamics , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/pathology , Reactive Oxygen Species/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Mitochondria/metabolism , Oxidative Stress , Oxygen , Phosphorylation
4.
Pharmacol Res ; 156: 104771, 2020 06.
Article in English | MEDLINE | ID: mdl-32234339

ABSTRACT

Thrombolytic therapy and revascularization strategies create a complete recanalization of the occluded epicardial coronary artery in patients with myocardial infarction (MI). However, about 35 % of patients still experience an impaired myocardial reperfusion, which is termed a no-reflow phenomenon mainly caused by cardiac microvascular ischemia-reperfusion (I/R) injury. Mitochondria are essential for microvascular endothelial cells' survival, both because of their roles as metabolic energy producers and as regulators of programmed cell death. Mitochondrial structure and function are regulated by a mitochondrial quality control (MQC) system, a series of processes including mitochondrial biogenesis, mitochondrial dynamics/mitophagy, mitochondrial proteostasis, and mitochondria-mediated cell death. Our review discusses the MQC mechanisms and how they are linked to cardiac microvascular I/R injury. Additionally, we will summarize the molecular basis that results in defective MQC mechanisms and present potential therapeutic interventions for improving MQC in cardiac microvascular I/R injury.


Subject(s)
Mitochondria, Heart/pathology , Mitochondrial Dynamics , Mitophagy , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , No-Reflow Phenomenon/pathology , Organelle Biogenesis , Animals , Apoptosis , Cardiovascular Agents/therapeutic use , Coronary Circulation , Energy Metabolism , Humans , Microcirculation , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Necrosis , No-Reflow Phenomenon/drug therapy , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/physiopathology , Signal Transduction , Unfolded Protein Response
5.
Cardiovasc Res ; 115(7): 1143-1155, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30428011

ABSTRACT

The coronary circulation is both culprit and victim of acute myocardial infarction. The rupture of an epicardial atherosclerotic plaque with superimposed thrombosis causes coronary occlusion, and this occlusion must be removed to induce reperfusion. However, ischaemia and reperfusion cause damage not only in cardiomyocytes but also in the coronary circulation, including microembolization of debris and release of soluble factors from the culprit lesion, impairment of endothelial integrity with subsequently increased permeability and oedema formation, platelet activation and leucocyte adherence, erythrocyte stasis, a shift from vasodilation to vasoconstriction, and ultimately structural damage to the capillaries with eventual no-reflow, microvascular obstruction (MVO), and intramyocardial haemorrhage (IMH). Therefore, the coronary circulation is a valid target for cardioprotection, beyond protection of the cardiomyocyte. Virtually all of the above deleterious endpoints have been demonstrated to be favourably influenced by one or the other mechanical or pharmacological cardioprotective intervention. However, no-reflow is still a serious complication of reperfused myocardial infarction and carries, independently from infarct size, an unfavourable prognosis. MVO and IMH can be diagnosed by modern imaging technologies, but still await an effective therapy. The current review provides an overview of strategies to protect the coronary circulation from acute myocardial ischaemia/reperfusion injury. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.


Subject(s)
Cardiovascular Agents/therapeutic use , Coronary Circulation/drug effects , Ischemic Postconditioning , Ischemic Preconditioning , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , No-Reflow Phenomenon/prevention & control , Animals , Cardiovascular Agents/adverse effects , Collateral Circulation/drug effects , Humans , Ischemic Preconditioning/adverse effects , Ischemic Preconditioning, Myocardial , Microcirculation/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Neovascularization, Physiologic/drug effects , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/pathology , No-Reflow Phenomenon/physiopathology , Treatment Outcome
6.
Curr Pharm Des ; 24(25): 2934-2942, 2018.
Article in English | MEDLINE | ID: mdl-30205793

ABSTRACT

The no-reflow phenomenon refers to the post-percutaneous coronary intervention condition in which, despite re-establishing epicardial coronary vessel patency, the flow to the previously ischemic myocardium is markedly reduced. When it does occur, it attenuates the beneficial effect of reperfusion therapy and substantial regions of the myocardium fail to receive adequate perfusion. The pathophysiology of this phenomenon is not completely understood. The possible mechanisms could be related to alterations in the microvascular circulation. Various mechanisms such as activation of inflammatory pathways, vascular damage and hemorrhage, leukocyte infiltration, and cellular edema may be responsible. As the no-reflow phenomenon is associated with adverse clinical consequences, it is of great importance to identify exact responsible mechanisms and apply effective preventive and therapeutic strategies. In this review, we describe an updated overview of the pathophysiological mechanisms and the current preventive tools for no-reflow as well as therapeutic interventions in order to improve coronary blood flow and consequently the prognosis for these patients.


Subject(s)
Coronary Vessels/diagnostic imaging , Microcirculation , No-Reflow Phenomenon/diagnostic imaging , Coronary Vessels/metabolism , Humans , No-Reflow Phenomenon/metabolism
7.
Curr Pharm Des ; 24(25): 2927-2933, 2018.
Article in English | MEDLINE | ID: mdl-29962336

ABSTRACT

Timely delivered coronary revascularization with no residual anatomical stenosis does not always lead to prompt restoration of anterograde coronary flow and complete myocardial reperfusion. This condition is known as coronary no-reflow and is associated with major clinical adverse events and poor prognosis. The pathophysiology of no-reflow phenomenon is still poorly understood. Proposed mechanisms include distal microembolization of thrombus and plaque debris, ischemic injury, endothelial dysfunction and individual susceptibility to microvascular dysfunction/obstruction. Older age, diabetes, hypercholesterolemia, prolonged ischemic time, hemodynamic instability, high thrombus burden, complex angiographic lesions and multivessel disease are frequently reported to be associated with the no-reflow phenomenon. There is no general consensus on the correct prevention and management of no-reflow. Non-pharmacological measures such as distal embolic protection devices and manual thrombus aspiration did not result in improved flow or reduction of infarct size. Current preventive measures include reduction of time from symptoms onset to reperfusion therapy, and intracoronary administration of vasodilators such as adenosine, verapamil or nitroprusside.


Subject(s)
Adenosine/pharmacology , Coronary Vessels/drug effects , Microcirculation/drug effects , Nitroprusside/pharmacology , No-Reflow Phenomenon/drug therapy , Verapamil/pharmacology , Adenosine/administration & dosage , Coronary Vessels/metabolism , Humans , Nitroprusside/administration & dosage , No-Reflow Phenomenon/metabolism , Verapamil/administration & dosage
8.
Mol Med Rep ; 17(1): 580-585, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115615

ABSTRACT

Pradaxa is a novel oral anticoagulant, which was originally used to prevent thrombosis following joint replacement surgery. The aim of the current study was to investigate the effect dabigatran on acute myocardial infarction through regulating no­reflow phenomenon and oxidative stress, neutrophil intraplaque infiltration and apoptosis. In the present study, dabigatran significantly inhibited the infarct size, increased arterial pressure and reduced no­reflow phenomenon in acute myocardial infarction (AMI) vehicle rabbits. Treatment with dabigatran significantly inhibited the P65 of nuclear factor κB, tumor necrosis factor α, interleukin (IL)­1ß and IL­6 activities and significantly enhanced the catalase and superoxide dismutase activities in the AMI rabbits. In addition, dabigatran significantly suppressed inducible nitric oxide synthase (iNOS), collagen I, transforming growth factor ß1 (TGF­ß1), α­smooth muscle actin (α­SMA) and connective tissue growth factor (CTGF) protein expression in AMI rabbits. Taken together, these results suggest that the effects of dabigatran inhibit no­reflow phenomenon, infarct size and enhance arterial pressure in AMI through anti­inflammatory and anti­oxidative activity, and regulating iNOS, collagen I, TGF­ß1, α­SMA and CTGF protein expression in AMI rabbits.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Connective Tissue Growth Factor/metabolism , Dabigatran/pharmacology , Myocardial Infarction/metabolism , No-Reflow Phenomenon/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Blood Pressure/drug effects , Connective Tissue Growth Factor/genetics , Dabigatran/chemistry , Gene Expression , Mice , Myocardial Infarction/genetics , Myocardial Infarction/pathology , No-Reflow Phenomenon/drug therapy , Oxidative Stress/drug effects , Rabbits , Transforming Growth Factor beta1/metabolism
9.
Biosci Rep ; 37(4)2017 08 31.
Article in English | MEDLINE | ID: mdl-28667102

ABSTRACT

To determine the effect of adiponectin (APN) on the coronary no-reflow (NR) injury in rats with Type 2 diabetes mellitus (T2DM), 80 male Sprague-Dawley rats were fed with a high-sugar-high-fat diet to build a T2DM model. Rats received vehicle or APN in the last week and then were subjected to myocardial ischemia reperfusion (MI/R) injury. Endothelium-dependent vasorelaxation of the thoracic aorta was significantly decreased and serum levels of endothelin-1 (ET-1), intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) were noticably increased in T2DM rats compared with rats without T2DM. Serum APN was positively correlated with the endothelium-dependent vasorelaxation, but negatively correlated with the serum level of ET-1. Treatment with APN improved T2DM-induced endothelium-dependent vasorelaxation, recovered cardiac function, and decreased both NR size and the levels of ET-1, ICAM-1 and VCAM-1. Hypoadiponectinemia was associated with the aggravation of coronary NR in T2DM rats. APN could alleviate coronary NR injury in T2DM rats by protecting the endothelium and improving microcirculation.


Subject(s)
Adiponectin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Endothelium, Vascular/metabolism , No-Reflow Phenomenon/drug therapy , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Endothelin-1/metabolism , Endothelium, Vascular/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Male , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/physiopathology , Rats , Rats, Sprague-Dawley , Vascular Cell Adhesion Molecule-1/metabolism
10.
Mol Med Rep ; 13(1): 623-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26647773

ABSTRACT

The aim of the current study was to investigate the time course of the expression of growth differentiation factor­15 (GDF­15) in rat ischemic myocardium with increasing durations of reperfusion, and to elucidate its physiopathological role in the no­reflow phenomenon. Wistar rats were randomly divided into ischemia reperfusion (I/R) and sham groups, and myocardial I/R was established by ligation of the left anterior descending coronary artery for 1 h followed by reperfusion for 2, 4, 6, 12, 24 h and 7 days whilst rats in the sham group were subjected to a sham operation. The expression levels of GDF­15 and ICAM­1 were measured, in addition to myeloperoxidase (MPO) activity. The myocardial anatomical no­reflow and infarction areas were assessed. The area at risk was not significantly different following various periods of reperfusion, while the infarct area and no­reflow area were significantly greater following 6 h of reperfusion (P<0.05). The mRNA and protein expression levels of GDF­15 were increased during the onset and development of no­reflow, and peaked following 24 h of reperfusion. MPO activity was reduced with increasing reperfusion duration, while ICAM­1 levels were increased. Hematoxylin and eosin staining demonstrated that myocardial neutrophil infiltration was significantly increased by I/R injury, in particular following 2, 4 and 6 h of reperfusion. GDF­15 expression levels were negatively correlated with MPO activity (r=­0.55, P<0.001), and the MPO activity was negatively correlated with the area of no­reflow (r=­0.46, P<0.01). By contrast, GDF­15 was significantly positively correlated with ICAM­1 levels (r=0.52, P<0.01), which additionally were demonstrated to be significantly positively associated with the size of the no­reflow area (r=0.39, P<0.05). The current study demonstrated the time course effect of reperfusion on the expression of GDF­15 in the myocardial I/R rat model, with the shorter reperfusion times (6 h) resulting in significant no­reflow in ischemic myocardium. GDF­15 may protect the I/R myocardium from no­reflow by inhibiting the inflammatory­like response, which involves neutrophil infiltration and transendothelial migration.


Subject(s)
Cardiotonic Agents/metabolism , Growth Differentiation Factor 15/metabolism , Inflammation/pathology , Myocardium/pathology , Neutrophil Infiltration , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/pathology , Animals , Benzothiazoles , Enzyme-Linked Immunosorbent Assay , Inflammation/complications , Intercellular Adhesion Molecule-1/metabolism , Male , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Peroxidase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Thiazoles/metabolism , Time Factors
11.
Life Sci ; 142: 8-18, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26482204

ABSTRACT

AIMS: The effects of sodium tanshinone IIA sulfonate (STS) on coronary no-reflow (CNR) relevant to microvascular obstruction (MVO) remain unknown. Studies had shown that fibrinogen-like protein 2 (FGL2) expressed in microvascular endothelial cells (MECs) is a key mediator in MVO. Thus, we aimed to elucidate the roles of STS in CNR and relations between STS and FGL2. MAIN METHODS: Myocardial ischemia/reperfusion was selected to represent CNR model. The no-reflow zone and infarct area were assessed using Thioflavin S and TTC staining, and cardiac functional parameters were detected using echocardiography. Western blot was used to detected FGL2 level, fibrin level, protease-activated receptor-1 (PAR-1) activation and inflammation cells infiltration. FGL2 and inflammation cells were also identified by IHC. Microthrombus was detected by Carstairs' and MSB staining. We also detected the roles of STS on FGL2 expression, thrombin generation, phospho-Akt and NF-κB levels in MECs. KEY FINDINGS: Upon treatment with STS in CNR model, the no-reflow and infarct areas decreased significantly and cardiac function improved. The FGL2 expression was inhibited by STS in vivo as well as in vitro with thrombin generation inhibition. In addition, STS up-regulates Akt phosphorylation and suppressed NF-κB expression in activated MECs. Furthermore, fibrin deposition, PAR-1 activation and inflammatory response were inhibited with STS administration in CNR model. SIGNIFICANCE: Our results displayed a novel pharmacological action of STS on CNR. STS is able to ameliorate CNR through inhibition of FGL2 expression mediated by Akt and NF-κB pathways as well as prevention of MVO by suppressing fibrin deposition and inflammation.


Subject(s)
Coronary Circulation/drug effects , Down-Regulation/drug effects , Fibrinogen/biosynthesis , No-Reflow Phenomenon/metabolism , Phenanthrenes/pharmacology , Animals , Coronary Circulation/genetics , Disease Models, Animal , Down-Regulation/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibrin/metabolism , Fibrinogen/genetics , Male , No-Reflow Phenomenon/drug therapy , No-Reflow Phenomenon/genetics , No-Reflow Phenomenon/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction
12.
Basic Res Cardiol ; 110(2): 1, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25589055

ABSTRACT

A high proportion of primary percutaneous coronary interventions performed in the setting of acute myocardial infarction, concur with inadequate myocardial perfusion at the microvascular level. This phenomenon, known as "no-reflow" contributes to reperfusion injury, poor prognosis and to unfavorable clinical outcome. In this study, we evaluated the hypothesis that the synthetic 17ß-aminoestrogen Prolame, may confer cardioprotection and prevent against no-reflow. In an open-chest model of 30-min ischemia and 90-min reperfusion, male Wistar rats were randomly assigned to different groups: Control, Prolame, Prolame followed by the nitric oxide synthase inhibitor (L-NAME), and 17ß-estradiol. Areas of risk, infarct size and no-reflow were determined by planimetry with triphenyltetrazolium chloride and thioflavin-S stains. Structural damage of the vasculature was measured as capillary compression in clarified tissue after intra-atrial injection of Microfil. Hemodynamic function was obtained at the end of stabilization, ischemia and reperfusion; nitric oxide (NO·) content was determined indirectly using the Griess reaction. Activation of the eNOS signaling cascade was determined by western blot. Prolame reduced the infarcted area, decreased the zones of no-reflow and capillary compression by activating the PI3K/Akt/eNOS signaling pathway in correlation with NO· increase. Prolame also activated endothelial cells augmenting NO· production, which was inhibited by ICI182780 (a selective estrogen receptor down-regulator), supporting the notion that the cardioprotective effect of Prolame involves the preservation of endothelium through the activation of estrogen receptor downstream signaling. Our results provide evidence that Prolame has potential therapeutic application in patients with AMI, as it prevents from both vascular and cardiac tissue damage.


Subject(s)
Estrenes/pharmacology , Hemodynamics/drug effects , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/prevention & control , No-Reflow Phenomenon/prevention & control , Signal Transduction/drug effects , Animals , Blotting, Western , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Male , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Nitric Oxide Synthase Type III/metabolism , No-Reflow Phenomenon/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Umbilical Veins
13.
Clin Exp Pharmacol Physiol ; 42(2): 162-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25399887

ABSTRACT

No reflow after reperfusion therapy for myocardial infarction is a strong predictor of clinical outcome. Increased levels of inflammatory factors, including C-reactive protein (CRP), in patients with acute myocardial infarction (AMI) undergoing primary percutaneous coronary intervention (PCI) may affect myocardial perfusion. However, why the no-reflow phenomenon increases in inflammation stress after PCI is not clear. The aim of the present study was to determine the effects and molecular mechanisms underlying the effects of CRP on the expression of cyclo-oxygenase (COX) on the development of the no-reflow phenomenon. There was a significant increase in plasma levels of CRP and interleukin (IL)-6 in no-reflow patients, suggesting that inflammatory factors play an important role in the development of the no-reflow phenomenon. The mechanisms involved were further evaluated after reperfusion in a rat model mimicking the no-reflow phenomenon. Compared with normal reflow rats, there were significant increases in both COX-1 and COX-2 in cardiac tissue from no-reflow rats. The COX inhibitor indomethacin (5 mg/kg, i.p.) significantly reduced the no-reflow area. In another series of experiments, human coronary artery endothelial cells (HCAEC) were treated with CRP at clinically relevant concentrations (5-25 µg/mL). C-Reactive protein significantly increased COX-1 and COX-2 levels in a time- and concentration-dependent manner. In addition, extracellular signal-regulated kinase (ERK) and Jun N-terminal kinase (JNK) were activated in CRP (5, 10, 25 µg/mL)-treated HCAEC cultures. Furthermore, the ERK inhibitor pd98059 (30 µmol/L) and the JNK inhibitor sp600125 (10 µmol/L) blocked CRP-induced COX-1 and COX-2 expression for 12 h. Together, the findings of the present study suggest that CRP can promote the development of the no-reflow phenomenon by increasing COX-1 and COX-2 expression, which is regulated, in part, via ERK and JNK activity.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Inflammation/pathology , Myocardial Infarction/pathology , No-Reflow Phenomenon/pathology , Acute Disease , Animals , C-Reactive Protein/metabolism , Cyclooxygenase Inhibitors/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Indomethacin/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , Male , Middle Aged , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , No-Reflow Phenomenon/drug therapy , No-Reflow Phenomenon/metabolism , Rats , Rats, Sprague-Dawley
15.
Braz. j. med. biol. res ; 47(3): 245-251, 03/2014. graf
Article in English | LILACS | ID: lil-704619

ABSTRACT

The mechanisms of statins relieving the no-reflow phenomenon and the effects of single-dose statins on it are not well known. This study sought to investigate the effects of inflammation on the no-reflow phenomenon in a rabbit model of acute myocardial infarction and reperfusion (AMI/R) and to evaluate the effects of single-dose atorvastatin on inflammation and myocardial no-reflow. Twenty-four New Zealand white male rabbits (5-6 months old) were randomized to three groups of eight: a sham-operated group, an AMI/R group, and an atorvastatin-treated group (10 mg/kg). Animals in the latter two groups were subjected to 4 h of coronary occlusion followed by 2 h of reperfusion. Serum levels of interleukin (IL)-6 were measured by enzyme-linked immunosorbent assay. The expression of interferon gamma (IFN-γ) in normal and infarcted (reflow and no-reflow) myocardial tissue was determined by immunohistochemical methods. The area of no-reflow and necrosis was evaluated pathologically. Levels of serum IL-6 were significantly lower in the atorvastatin group than in the AMI/R group (P<0.01). Expression of IFN-γ in infarcted reflow and no-reflow myocardial tissue was also significantly lower in the atorvastatin group than in the AMI/R group. The mean area of no-reflow [47.01% of ligation area (LA)] was significantly smaller in the atorvastatin group than in the AMI/R group (85.67% of LA; P<0.01). The necrosis area was also significantly smaller in the atorvastatin group (85.94% of LA) than in the AMI/R group (96.56% of LA; P<0.01). In a secondary analysis, rabbits in the atorvastatin and AMI/R groups were divided into two groups based on necrosis area (90% of LA): a small group (<90% of LA) and a large group (>90% of LA). There was no significant difference in the area of no-reflow between the small (61.40% of LA) and large groups (69.87% of LA; P>0.05). Single-dose atorvastatin protected against inflammation and myocardial no-reflow and reduced infarct size during AMI/R in rabbits. No-reflow was not dependent on the reduction of infarct size.


Subject(s)
Animals , Male , Rabbits , Anticholesteremic Agents/administration & dosage , Heptanoic Acids/administration & dosage , Interferon-gamma/metabolism , /metabolism , Myocardial Infarction/drug therapy , Myocardial Reperfusion/methods , No-Reflow Phenomenon/drug therapy , Pyrroles/administration & dosage , Coronary Occlusion/drug therapy , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Inflammation , Ligation , Multivariate Analysis , Myocardial Infarction/metabolism , Myocardium/pathology , Necrosis , No-Reflow Phenomenon/metabolism , Random Allocation
16.
Braz J Med Biol Res ; 47(3): 245-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24554037

ABSTRACT

The mechanisms of statins relieving the no-reflow phenomenon and the effects of single-dose statins on it are not well known. This study sought to investigate the effects of inflammation on the no-reflow phenomenon in a rabbit model of acute myocardial infarction and reperfusion (AMI/R) and to evaluate the effects of single-dose atorvastatin on inflammation and myocardial no-reflow. Twenty-four New Zealand white male rabbits (5-6 months old) were randomized to three groups of eight: a sham-operated group, an AMI/R group, and an atorvastatin-treated group (10 mg/kg). Animals in the latter two groups were subjected to 4 h of coronary occlusion followed by 2 h of reperfusion. Serum levels of interleukin (IL)-6 were measured by enzyme-linked immunosorbent assay. The expression of interferon gamma (IFN-γ) in normal and infarcted (reflow and no-reflow) myocardial tissue was determined by immunohistochemical methods. The area of no-reflow and necrosis was evaluated pathologically. Levels of serum IL-6 were significantly lower in the atorvastatin group than in the AMI/R group (P<0.01). Expression of IFN-γ in infarcted reflow and no-reflow myocardial tissue was also significantly lower in the atorvastatin group than in the AMI/R group. The mean area of no-reflow [47.01% of ligation area (LA)] was significantly smaller in the atorvastatin group than in the AMI/R group (85.67% of LA; P<0.01). The necrosis area was also significantly smaller in the atorvastatin group (85.94% of LA) than in the AMI/R group (96.56% of LA; P<0.01). In a secondary analysis, rabbits in the atorvastatin and AMI/R groups were divided into two groups based on necrosis area (90% of LA): a small group (<90% of LA) and a large group (>90% of LA). There was no significant difference in the area of no-reflow between the small (61.40% of LA) and large groups (69.87% of LA; P>0.05). Single-dose atorvastatin protected against inflammation and myocardial no-reflow and reduced infarct size during AMI/R in rabbits. No-reflow was not dependent on the reduction of infarct size.


Subject(s)
Anticholesteremic Agents/administration & dosage , Heptanoic Acids/administration & dosage , Interferon-gamma/metabolism , Interleukin-6/metabolism , Myocardial Infarction/drug therapy , Myocardial Reperfusion/methods , No-Reflow Phenomenon/drug therapy , Pyrroles/administration & dosage , Animals , Atorvastatin , Coronary Occlusion/drug therapy , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Inflammation , Ligation , Male , Multivariate Analysis , Myocardial Infarction/metabolism , Myocardium/pathology , Necrosis , No-Reflow Phenomenon/metabolism , Rabbits , Random Allocation
17.
Isr Med Assoc J ; 15(12): 753-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24449979

ABSTRACT

BACKGROUND: Coronary slow flow phenomenon (CSFP) is a functional and structural disease that is diagnosed by coronary angiogram. OBJECTIVES: To evaluate the possible association between CSFP and small artery elasticity in an effort to understand the pathogenesis of CSFP. METHODS: The study population comprised 12 patients with normal coronary arteries and CSFP and 12 with normal coronary arteries without CSFP. We measured conjugated diene formation at 234 nm during low density lipoprotein (LDL) oxidation, as well as platelet aggregation. We estimated, noninvasively, arterial elasticity parameters. Mann-Whitney nonparametric test was used to compare differences between the groups. Data are presented as mean +/- standard deviation. RESULTS: Waist circumference was 99.2 +/- 8.8 cm and 114.9 +/- 10.5 cm in the normal flow and CSFP groups, respectively (P = 0.003). Four patients in the CSFP group and one in the normal flow group had type 2 diabetes. Area under the curve in the oral glucose tolerance test was 22% higher in the CSFP than in the normal group (P = 0.04). There was no difference in systolic and diastolic blood pressure, plasma concentrations of total cholesterol, triglycerides, high density lipoprotein, LDL and platelet aggregation parameters between the groups. Lag time required until initiation of LDL oxidation in the presence of CuSO4 was 17% longer (P = 0.02) and homocysteine fasting plasma concentration was 81% lower (P = 0.05) in the normal flow group. Large artery elasticity was the same in both groups. Small artery elasticity was 5 +/- 1.5 ml/mmHg x 100 in normal flow subjects and 6.1 +/- 1.9 ml/mmHg x 100 in the CSFP patients (P = 0.02). CONCLUSIONS: Patients with CSFP had more metabolic derangements. Arterial stiffness was not increased in CSFP.


Subject(s)
Coronary Vessels/physiopathology , No-Reflow Phenomenon , Obesity , Adult , Area Under Curve , Blood Pressure/physiology , Coronary Angiography/methods , Elasticity Imaging Techniques/methods , Female , Glucose Tolerance Test/methods , Humans , Lipoproteins, LDL/metabolism , Male , Middle Aged , No-Reflow Phenomenon/diagnosis , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/physiopathology , Obesity/metabolism , Obesity/physiopathology , Platelet Aggregation/physiology , Statistics as Topic
18.
Clin Appl Thromb Hemost ; 19(1): 19-28, 2013.
Article in English | MEDLINE | ID: mdl-22387586

ABSTRACT

No-reflow phenomenon due to cardiac microvascular dysfunction or disturbance aggravates clinic outcomes of a portion of patients with acute myocardial infarction undergoing percutaneous coronary intervention or thrombolytic therapy. Our working hypothesis was that cardiac microthrombosis would play an important role in the pathogenesis. We investigated that cardiac microthrombi were observed by Martius, Scarlet, Blue methocl (MSB) and Masson trichrome staining. Furthermore, we investigated the expression of fibrinogen-like protein 2 (fgl2) in rats with acute myocardial ischemia/reperfusion (MI/R) and its possible pathological and clinical significance. The fgl2 was highly expressed in myocardium of rats with acute MI/R and located at cardiac microvascular walls. We found that the expression of fgl2 in peripheral mononuclear cells of rats with acute MI/R significantly increased correspondingly with its cardiac expression. Expression of cardiac fgl2 was correlated with no-reflow size of rats with acute MI/R, which was detected and calculated by thioflavin S staining. No-reflow size was in line with cardiac diastolic dysfunction of rats with acute MI/R monitored by hemodynamics. Thus, microthrombosis is involved in cardiac microvascular dysfunction or disturbance of rats with acute MI/R as one cause, and fgl2 may emerge as a predictor of the occurrence of no-reflow phenomenon.


Subject(s)
Coronary Thrombosis/metabolism , Fibrinogen/biosynthesis , Gene Expression Regulation , Leukocytes, Mononuclear/metabolism , Myocardial Reperfusion Injury/metabolism , No-Reflow Phenomenon/metabolism , Acute Disease , Animals , Coronary Thrombosis/pathology , Coronary Thrombosis/physiopathology , Hemodynamics , Leukocytes, Mononuclear/pathology , Male , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , No-Reflow Phenomenon/pathology , No-Reflow Phenomenon/physiopathology , Rats , Rats, Sprague-Dawley
19.
Kardiol Pol ; 70(9): 903-9, 2012.
Article in English | MEDLINE | ID: mdl-22992998

ABSTRACT

BACKGROUND: Slow coronary flow (SCF) is an angiographic finding characterised by delayed opacification of epicardial coronary arteries without obstructive coronary disease. Epicardial adipose tissue (EAT), localised beneath the visceral pericardium, is a metabolically active endocrine and paracrine organ with possible interactions within the heart. EAT and low-grade inflammation play major roles in the atherosclerotic vascular processes and may be important in other coronary pathologies such as SCF. AIM: To investigate whether EAT and C-reactive protein (CRP) are increased in patients with isolated SCF compared to normal subjects. METHODS: The present study was cross-sectional and observational, consisting of 66 individuals who underwent coronary angiography with a suspicion of coronary artery disease and who had angiographically normal coronary arteries of varying coronary flow rates. The relationship between EAT, CRP and SCF phenomenon was investigated. Thirty-three patients with isolated SCF (mean age: 56 ± 10 years) and 33 age- and gender-matched control participants with normal coronary flow (NCF), but without SCF, (mean age: 55 ± 10 years) were included in the study. RESULTS: EAT thickness was significantly increased in the SCF group compared to the NCF group (7.1 ± 2.7 vs. 4.7 ± 1.9 mm, p < 0.001). Body mass index (BMI, p < 0.001) and the percentage of isolated SCF (p = 0.002) were significantly higher in patients with increased EAT thickness. CRP was not related to SCF. When we performed multiple logistic regression analysis, only increased EAT thickness was related to the presence of SCF (OR 1.720, 95% CI 1.175-2.516, p = 0.005) independent of BMI and CRP. CONCLUSIONS: This study revealed, for the first time, a significant increase in EAT thickness in patients with SCF compared to NCF. We believe that further studies are needed to clarify the role of adipose tissue in patients with SCF.


Subject(s)
Adipose Tissue/diagnostic imaging , No-Reflow Phenomenon/diagnostic imaging , Pericardium/diagnostic imaging , Adipose Tissue/metabolism , C-Reactive Protein/metabolism , Coronary Angiography , Cross-Sectional Studies , Echocardiography , Female , Humans , Male , Middle Aged , No-Reflow Phenomenon/metabolism , Pericardium/metabolism , Regression Analysis
20.
Microvasc Res ; 84(1): 44-54, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22542438

ABSTRACT

OBJECTIVE: To investigate whether ischemic preconditioning (IP) can reduce myocardial no-reflow by activating endothelial (e-) nitric oxide synthase (NOS) via the protein kinase A (PKA) pathway. METHODS AND RESULTS: In a 90-min ischemia and 3-h reperfusion model, minipigs were assigned into sham, ischemia-reperfusion (IR), IR+IP, IR+IP+L-NNA (an eNOS inhibitor, 10mg·kg(-1)), IR+IP+H-89 (a PKA inhibitor, 1.0µg·kg(-1)·min(-1)), IR+L-NNA, and IR+H-89 groups. IP pretreatment improved cardiac function and coronary blood flow, decreased the activities of creatine kinase by 36.6% after 90 min of ischemia and by 32.8% after 3 h of reperfusion (P<0.05), reduced the no-reflow areas from 49.9% to 11.0% (P<0.01), and attenuated the infarct size from 78.2% to 35.4% (P<0.01). IP stimulated myocardial PKA activities and the expression of PKA and Ser(133) phosphorylated (p-) cAMP response element-binding protein (CREB) in the reflow and no-reflow myocardium, and enhanced the activities of constitutive NOS and the phosphorylation of eNOS at Ser(1179) and Ser(635) in the no-reflow myocardium. IP suppressed the expression of tumor necrosis factor-α and P-selectin, and attenuated cardiomyocytes apoptosis by regulating the expression of Bcl-2 and caspase-3 in the reflow and no-reflow myocardium. The eNOS inhibitor L-NNA completely canceled these beneficial effects of IP without any influence on PKA activity, whereas the PKA inhibitor H-89 partially blocked the IP cardioprotective effects and eNOS phosphorylation at the same time. CONCLUSION: IP attenuates myocardial no-reflow and infarction after ischemia and reperfusion by activating the phosphorylation of eNOS at Ser(1179) and Ser(635) in a partly PKA-dependent manner.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Ischemic Preconditioning , Myocardial Reperfusion Injury/pathology , Myocardial Stunning/pathology , Nitric Oxide Synthase Type III/metabolism , No-Reflow Phenomenon/pathology , Animals , Apoptosis , Biomarkers/metabolism , Hemodynamics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardial Stunning/metabolism , Myocardial Stunning/physiopathology , Myocardium/enzymology , Myocardium/pathology , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/physiopathology , Phosphorylation , Swine , Swine, Miniature
SELECTION OF CITATIONS
SEARCH DETAIL
...