Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Am J Physiol Heart Circ Physiol ; 321(6): H1014-H1029, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34623184

ABSTRACT

Heart failure is the one of the leading causes of death in the United States. Heart failure is a complex syndrome caused by numerous diseases, including severe myocardial infarction (MI). MI occurs after an occlusion of a cardiac artery causing downstream ischemia. MI is followed by cardiac remodeling involving extensive remodeling and fibrosis, which, if the original insult is severe or prolonged, can ultimately progress into heart failure. There is no "cure" for heart failure because therapies to regenerate dead tissue are not yet available. Previous studies have shown that in both post-MI and post-ischemia-reperfusion (I/R) models of heart failure, administration of cortical bone stem cell (CBSC) treatment leads to a reduction in scar size and improved cardiac function. Our first study investigated the ability of mouse CBSC-derived exosomes (mCBSC-dEXO) to recapitulate mouse CBSCs (mCBSC) therapeutic effects in a 24-h post-I/R model. This study showed that injection of mCBSCs and mCBSC-dEXOs into the ischemic region of an infarct had a protective effect against I/R injury. mCBSC-dEXOs recapitulated the effects of CBSC treatment post-I/R, indicating exosomes are partly responsible for CBSC's beneficial effects. To examine if exosomes decrease fibrotic activation, adult rat ventricular fibroblasts (ARVFs) and adult human cardiac fibroblasts (NHCFs) were treated with transforming growth factor ß (TGFß) to activate fibrotic signaling before treatment with mCBSC- and human CBSC (hCBSC)-dEXOs. hCBSC-dEXOs caused a 100-fold decrease in human fibroblast activation. To further understand the signaling mechanisms regulating the protective decrease in fibrosis, we performed RNA sequencing on the NHCFs after hCBSC-dEXO treatment. The group treated with both TGFß and exosomes showed a decrease in small nucleolar RNA (snoRNA), known to be involved with ribosome stability.NEW & NOTEWORTHY Our work is noteworthy due to the identification of factors within stem cell-derived exosomes (dEXOs) that alter fibroblast activation through the hereto-unknown mechanism of decreasing small nucleolar RNA (snoRNA) signaling within cardiac fibroblasts. The study also shows that the injection of stem cells or a stem-cell-derived exosome therapy at the onset of reperfusion elicits cardioprotection, emphasizing the importance of early treatment in the post-ischemia-reperfusion (I/R) wounded heart.


Subject(s)
Cortical Bone/cytology , Exosomes/transplantation , Fibroblasts/pathology , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/surgery , Myocardium/pathology , Stem Cell Transplantation , Ventricular Remodeling , Animals , Cells, Cultured , Disease Models, Animal , Exosomes/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Humans , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Rats , Signal Transduction , Transforming Growth Factor beta/pharmacology
2.
J Heart Lung Transplant ; 40(11): 1396-1407, 2021 11.
Article in English | MEDLINE | ID: mdl-34509349

ABSTRACT

BACKGROUND: Use of cardiac grafts obtained with donation after circulatory death (DCD) could significantly improve donor heart availability. As DCD hearts undergo potentially deleterious warm ischemia and reperfusion, clinical protocols require optimization to ensure graft quality. Thus, we investigated effects of alternative preservation conditions on endothelial and/or vascular and contractile function in comparison with the current clinical standard. METHODS: Using a rat DCD model, we compared currently used graft preservation conditions, St. Thomas n°2 (St. T) at 4°C, with potentially more suitable conditions for DCD hearts, adenosine-lidocaine preservation solution (A-L) at 4°C or 22°C. Following general anesthesia and diaphragm transection, hearts underwent either 0 or 18 min of in-situ warm ischemia, were explanted, flushed and stored for 15 min with either St. T at 4°C or A-L at 4°C or 22°C, and then reperfused under normothermic, aerobic conditions. Endothelial integrity and contractile function were determined. RESULTS: Compared to 4°C preservation, 22°C A-L significantly increased endothelial nitric oxide synthase (eNOS) dimerization and reduced oxidative tissue damage (p < 0.05 for all). Furthermore, A-L at 22°C better preserved the endothelial glycocalyx and coronary flow compared with St. T, tended to reduce tissue calcium overload, and stimulated pro-survival signaling. No significant differences were observed in cardiac function among ischemic groups. CONCLUSIONS: Twenty-two-degree Celsius A-L solution better preserves the coronary endothelium compared to 4°C St. T, which likely results from greater eNOS dimerization, reduced oxidative stress, and activation of the reperfusion injury salvage kinase (RISK) pathway. Improving heart preservation conditions immediately following warm ischemia constitutes a promising approach for the optimization of clinical protocols in DCD heart transplantation.


Subject(s)
Endothelium, Vascular/transplantation , Heart Transplantation/methods , Myocardial Reperfusion Injury/surgery , Recovery of Function , Tissue and Organ Procurement/methods , Vasodilation/physiology , Ventricular Function/physiology , Animals , Coronary Vessels/cytology , Coronary Vessels/transplantation , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Male , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Rats , Rats, Wistar
3.
J Cardiovasc Pharmacol Ther ; 26(1): 88-99, 2021 01.
Article in English | MEDLINE | ID: mdl-32677460

ABSTRACT

BACKGROUND: Allogeneic cardiosphere-derived cells (CDCs) exert cardioprotective effects when administered intracoronarily after reperfusion in animal models of acute myocardial infarction (AMI). The "no-reflow" phenomenon develops rapidly post-reperfusion and may undermine the efficacy of cell therapy, due to poor cell delivery in areas of microvascular obstruction (MVO). We hypothesized that CDC-induced cardioprotection would be enhanced by cell administration prior to reperfusion, when microvasculature is still relatively intact, to facilitate widespread cell delivery within the ischemic area. METHODS AND RESULTS: We studied 81 farm pigs; 55 completed the specified protocols. A dose-optimization study in infarcted pigs demonstrated that the doses of 5 million and 10 million CDCs are the maximum safe doses that can be administered intracoronarily at 5 minutes prior to and at 5 minutes post-reperfusion, respectively, without aggravating MVO. Quantification of acute cell retention by polymerase chain reaction demonstrated that cell delivery prior to reperfusion resulted in higher cardiac cell retention compared to delivery post-reperfusion. We then performed a randomized, placebo-controlled study to assess the long-term efficacy of intracoronary infusion of 5 million allogeneic CDCs, delivered at 5 minutes prior to reperfusion, in a porcine model of AMI. The CDC therapy resulted in decreased scar size, improved regional systolic function, and attenuation of adverse cardiac remodeling (manifested as preserved global systolic function, preserved end-systolic volume, and decreased interstitial fibrosis) compared to placebo at 30 days post-MI. CONCLUSIONS: Dose-optimized intracoronary infusion of allogeneic CDCs prior to reperfusion in a porcine model of AMI is feasible, safe and confers long-term benefits.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Myocardium , Myocytes, Cardiac , Ventricular Function, Left , Ventricular Remodeling , Animals , Male , Cells, Cultured , Disease Models, Animal , Fibrosis , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/surgery , Myocardium/pathology , Myocytes, Cardiac/transplantation , Sus scrofa , Transplantation, Homologous
5.
Int J Mol Sci ; 21(5)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155697

ABSTRACT

Pharmacological concentrations of melatonin reduce reperfusion arrhythmias, but less is known about the antiarrhythmic protection of the physiological circadian rhythm of melatonin. Bilateral surgical removal of the superior cervical ganglia irreversibly suppresses melatonin rhythmicity. This study aimed to analyze the cardiac electrophysiological effects of the loss of melatonin circadian oscillation and the role played by myocardial melatonin membrane receptors, SERCA2A, TNFα, nitrotyrosine, TGFß, KATP channels, and connexin 43. Three weeks after bilateral removal of the superior cervical ganglia or sham surgery, the hearts were isolated and submitted to ten minutes of regional ischemia followed by ten minutes of reperfusion. Arrhythmias, mainly ventricular tachycardia, increased during reperfusion in the ganglionectomy group. These hearts also suffered an epicardial electrical activation delay that increased during ischemia, action potential alternants, triggered activity, and dispersion of action potential duration. Hearts from ganglionectomized rats showed a reduction of the cardioprotective MT2 receptors, the MT1 receptors, and SERCA2A. Markers of nitroxidative stress (nitrotyrosine), inflammation (TNFα), and fibrosis (TGFß and vimentin) did not change between groups. Connexin 43 lateralization and the pore-forming subunit (Kir6.1) of KATP channels increased in the experimental group. We conclude that the loss of the circadian rhythm of melatonin predisposes the heart to suffer cardiac arrhythmias, mainly ventricular tachycardia, due to conduction disorders and changes in repolarization.


Subject(s)
Arrhythmias, Cardiac/pathology , Ganglionectomy/adverse effects , Heart/physiopathology , Myocardial Reperfusion Injury/surgery , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Circadian Rhythm , Connexin 43/genetics , Connexin 43/metabolism , Male , Melatonin/metabolism , Rats , Rats, Wistar , Receptors, Melatonin/genetics , Receptors, Melatonin/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
6.
Am J Physiol Heart Circ Physiol ; 317(4): H820-H829, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31441690

ABSTRACT

Ischemic heart diseases such as myocardial infarction (MI) are the largest contributors to cardiovascular disease worldwide. The resulting cardiac cell death impairs function of the heart and can lead to heart failure and death. Reperfusion of the ischemic tissue is necessary but causes damage to the surrounding tissue by reperfusion injury. Cortical bone stem cells (CBSCs) have been shown to increase pump function and decrease scar size in a large animal swine model of MI. To investigate the potential mechanism for these changes, we hypothesized that CBSCs were altering cardiac cell death after reperfusion. To test this, we performed TUNEL staining for apoptosis and antibody-based immunohistochemistry on tissue from Göttingen miniswine that underwent 90 min of lateral anterior descending coronary artery ischemia followed by 3 or 7 days of reperfusion to assess changes in cardiomyocyte and noncardiomyocyte cell death. Our findings indicate that although myocyte apoptosis is present 3 days after ischemia and is lower in CBSC-treated animals, myocyte apoptosis accounts for <2% of all apoptosis in the reperfused heart. In addition, nonmyocyte apoptosis trends toward decreased in CBSC-treated hearts, and although CBSCs increase macrophage and T-cell populations in the infarct region, the occurrence of apoptosis in CD45+ cells in the myocardium is not different between groups. From these data, we conclude that CBSCs may be influencing cardiomyocyte and noncardiomyocyte cell death and immune cell recruitment dynamics in the heart after MI, and these changes may account for some of the beneficial effects conferred by CBSC treatment.NEW & NOTEWORTHY The following research explores aspects of cell death and inflammation that have not been previously studied in a large animal model. In addition, apoptosis and cell death have not been studied in the context of cell therapy and myocardial infarction. In this article, we describe interactions between cell therapy and inflammation and the potential implications for cardiac wound healing.


Subject(s)
Apoptosis , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/surgery , Myocytes, Cardiac/pathology , Stem Cell Transplantation , Stem Cells , Tibia/cytology , Animals , Cells, Cultured , Disease Models, Animal , Female , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Leukocyte Common Antigens/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/immunology , Swine , Swine, Miniature , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
7.
Bioelectrochemistry ; 129: 170-178, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31181439

ABSTRACT

Our aim was to investigate if the cardioplegic solution HTK can be improved by the addition of the ROS scavenger melatonin. 158 guinea pig hearts without (UI80) or with HTK protection (HTK80) were investigated in ischemia/reperfusion experiments. Ischemia lasted 80 min at 30 °C. Melatonin was given before ischemia (UI80 + M1, HTK80 + M1) or before and after ischemia (UI80 + M2, HTK80 + M2). We measured the left ventricular developed pressure (LVDP), diastolic pressure (LVPmin), cardiac rhythm (VC-RR), time of electrical cell uncoupling (t-in) and recovery (t-ret), intracellular Ca++ [Ca++], and postischemic ROS. After 45 min reperfusion, LVDP in UI80 was significantly higher than in HTK80 (p < .01). Compared to UI80, the postischemic ROS burst was slightly smaller in HTK80 and significantly smaller in HTK80 + M1 and HTK80 + M2 (p < .05). Melatonin had no effect on LVPmin, t-in, t-ret, [Ca++], and on LVDP in groups UI80 + M1 and HTK80 + M1, improved slightly VC-RR (n. s.) but significantly decreased LVDP in the groups UI80 + M2 and HTK80 + M2 (p < .01). With melatonin we were able to attenuate the postischemic ROS burst, but the tissue damage by ROS seemed to be less important for the chosen ischemia condition because melatonin was unable to improve the functional recovery during reperfusion of HTK protected hearts.


Subject(s)
Cardioplegic Solutions/therapeutic use , Free Radical Scavengers/therapeutic use , Heart Arrest, Induced/methods , Heart/drug effects , Melatonin/therapeutic use , Myocardial Reperfusion Injury/therapy , Animals , Female , Guinea Pigs , Heart/physiopathology , Heart Rate/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/surgery
8.
Cardiovasc Res ; 115(13): 1918-1932, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31062860

ABSTRACT

AIMS: Adequate microcirculatory perfusion, and not just opening of occluded arteries, is critical to salvage heart tissue following myocardial infarction. However, the degree of microvascular perfusion taking place is not known, limited primarily by an inability to directly image coronary microcirculation in a beating heart in vivo. Haematopoietic stem/progenitor cells (HSPCs) offer a potential therapy but little is known about their homing dynamics at a cellular level and whether they protect coronary microvessels. This study used intravital microscopy to image the anaesthetized mouse beating heart microcirculation following stabilization. METHODS AND RESULTS: A 3D-printed stabilizer was attached to the ischaemia-reperfusion injured (IRI) beating heart. The kinetics of neutrophil, platelet and HSPC recruitment, as well as functional capillary density (FCD), was imaged post-reperfusion. Laser speckle contrast imaging (LSCI) was used for the first time to monitor ventricular blood flow in beating hearts. Sustained hyperaemic responses were measured throughout reperfusion, initially indicating adequate flow resumption. Intravital microscopy confirmed large vessel perfusion but demonstrated poor transmission of flow to downstream coronary microvessels. Significant neutrophil adhesion and microthrombus formation occurred within capillaries with the latter occluding them, resulting in patchy perfusion and reduced FCD. Interestingly, 'patrolling' neutrophils were also observed in capillaries. Haematopoietic stem/progenitor cells readily trafficked through the heart but local retention was poor. Despite this, remarkable anti-thromboinflammatory effects were observed, consequently improving microvascular perfusion. CONCLUSION: We present a novel approach for imaging multiple microcirculatory perturbations in the beating heart with LSCI assessment of blood flow. Despite deceptive hyperaemic responses, increased microcirculatory flow heterogeneity was seen, with non-perfused areas interspersed with perfused areas. Microthrombi, rather than neutrophils, appeared to be the major causative factor. We further applied this technique to demonstrate local stem cell presence is not a pre-requisite to confer vasculoprotection. This is the first detailed in vivo characterization of coronary microcirculatory responses post-reperfusion injury.


Subject(s)
Cell Tracking , Coronary Thrombosis/surgery , Coronary Vessels/diagnostic imaging , Hematopoietic Stem Cell Transplantation , Intravital Microscopy , Microvessels/diagnostic imaging , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/surgery , Animals , Cell Line , Coronary Circulation , Coronary Thrombosis/diagnostic imaging , Coronary Thrombosis/pathology , Coronary Thrombosis/physiopathology , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Disease Models, Animal , Hyperemia/diagnostic imaging , Hyperemia/physiopathology , Kinetics , Male , Mice, Inbred C57BL , Microcirculation , Microvessels/pathology , Microvessels/physiopathology , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Neutrophils/pathology
9.
Am J Physiol Heart Circ Physiol ; 317(2): H345-H356, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31125261

ABSTRACT

Intracoronary cardiosphere-derived cells (icCDCs) infused into the infarct-related artery reduce scar volume but do not improve left ventricular (LV) ejection fraction (LVEF). We tested the hypothesis that this reflects the inability of regional delivery to prevent myocyte death or promote myocyte proliferation in viable myocardium remote from the infarct. Swine (n = 23) pretreated with oral cyclosporine (200 mg/day) underwent a 1-h left anterior descending coronary artery (LAD) occlusion, which reduced LVEF from 61.6 ± 1.0 to 45.3 ± 1.5% 30 min after reperfusion. At that time, animals received global infusion of allogeneic icCDCs (n = 8), regional infusion of icCDCs restricted to the LAD using the stop-flow technique (n = 8), or vehicle (n = 7). After 1 mo, global icCDCs increased LVEF from 44.8 ± 1.9 to 60.8 ± 3.8% (P < 0.05) with no significant change after LAD stop-flow icCDCs (44.8 ± 3.6 to 50.9 ± 3.1%) or vehicle (46.5 ± 2.5 to 47.7 ± 2.6%). In contrast, global icCDCs did not alter infarct volume (%LV mass) assessed at 2 days (11.2 ± 2.3 vs. 12.6 ± 2.3%), whereas it was reduced after LAD stop-flow icCDCs (7.1 ± 1.1%, P < 0.05). Histopathological analysis of remote myocardium after global icCDCs demonstrated a significant increase in myocyte proliferation (147 ± 32 vs. 14 ± 10 nuclei/106 myocytes, P < 0.05) and a reduction in myocyte apoptosis (15 ± 9 vs. 46 ± 10 nuclei/106 myocytes, P < 0.05) that increased myocyte nuclear density (1,264 ± 39 vs. 1,157 ± 33 nuclei/mm2, P < 0.05) and decreased myocyte diameter (13.2 ± 0.2 vs. 14.5 ± 0.3 µm, P < 0.05) compared with vehicle-treated controls. In contrast, remote zone changes after regional LAD icCDCs were no different from vehicle. These data indicate that changes in global LVEF after icCDCs are dependent upon preventing myocyte loss and hypertrophy in myocardium remote from the infarct. These arise from stimulating myocyte proliferation and reducing myocyte apoptosis indicating the importance of directing cell therapy to viable remote regions.NEW & NOTEWORTHY Administration of allogeneic cardiosphere-derived cells to the entire heart via global intracoronary infusion shortly after myocardial infarction favorably influenced left ventricular ejection fraction by preventing myocyte death and promoting myocyte proliferation in remote, noninfarcted myocardium in swine. In contrast, regional intracoronary cell infusion did not significantly affect remote zone myocyte remodeling. Global cell administration targeting viable myocardium remote from the infarct may be an effective approach to prevent adverse ventricular remodeling after myocardial infarction.


Subject(s)
Myocardial Infarction/surgery , Myocardial Reperfusion Injury/surgery , Myocardium/pathology , Myocytes, Cardiac/transplantation , Regeneration , Spheroids, Cellular/transplantation , Stroke Volume , Ventricular Function, Left , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Male , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Recovery of Function , Sus scrofa , Time Factors , Tissue Survival , Transplantation, Homologous
10.
Nat Commun ; 10(1): 2312, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31127113

ABSTRACT

Cardioprotection by salvage of the infarct-affected myocardium is an unmet yet highly desired therapeutic goal. To develop new dedicated therapies, experimental myocardial ischemia/reperfusion (I/R) injury would require methods to simultaneously characterize extent and localization of the damage and the ensuing inflammatory responses in whole hearts over time. Here we present a three-dimensional (3D), simultaneous quantitative investigation of key I/R injury-components by combining bleaching-augmented solvent-based non-toxic clearing (BALANCE) using ethyl cinnamate (ECi) with light sheet fluorescence microscopy. This allows structural analyses of fluorescence-labeled I/R hearts with exceptional detail. We discover and 3D-quantify distinguishable acute and late vascular I/R damage zones. These contain highly localized and spatially structured neutrophil infiltrates that are modulated upon cardiac healing. Our model demonstrates that these characteristic I/R injury patterns can detect the extent of damage even days after the ischemic index event hence allowing the investigation of long-term recovery and remodeling processes.


Subject(s)
Heart/diagnostic imaging , Imaging, Three-Dimensional/methods , Myocardial Reperfusion Injury/diagnostic imaging , Myocardium/pathology , Animals , Biopsy , Cinnamates/chemistry , Coronary Artery Bypass , Disease Models, Animal , Humans , Luminescent Agents/chemistry , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence/methods , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/surgery , Myocardium/cytology , Myocardium/immunology , Neutrophils/immunology , Red Fluorescent Protein
11.
Biotechnol Appl Biochem ; 66(4): 555-563, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30994947

ABSTRACT

Although cilostazol was proved to have antitumor biological effects, its function in myocardial ischemia and reperfusion (I/R) injury and the underlying mechanisms were not fully illustrated yet. In this study, a rat model of I/R injury was constructed and quantitative real-time PCR, Western blot, and immunofluorescence (IF) assay were performed. Our results showed that cilostazol increased LC3 II/LC3 I ratio, reduced p62 abundance, and promoted the expressions of LAMP1, LAMP2, cathepsin B, and cathepsin D, indicating that cilostazol could activate autophagy and elevated lysosome activation. Following analysis showed that cilostazol enhanced nuclear protein expression of transcription factor EB (TFEB), an important regulator of autophagy-lysosome pathway. Furthermore, CCI-779, an inhibitor of TFEB, could reverse the effects of cilostazol on autophagic activity and lysosome activation. Importantly, cilostazol suppressed I/R injury-induced apoptosis by decreasing the cleavage of caspase 3 and PARP. Enzyme-linked immunosorbent assay showed that cilostazol reduced the serum levels of CTn1 and CK-MB and decreased infract size caused by I/R injuries. Altogether this study suggested that cilostazol protects against I/R injury by regulating autophagy, lysosome, and apoptosis in a rat model of I/R injury. The protective mechanism of cilostazol was partially through increasing the transcriptional activity of TFEB.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Cilostazol/pharmacology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Animals , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Disease Models, Animal , Lysosomes/drug effects , Myocardial Reperfusion Injury/surgery , Rats , Rats, Sprague-Dawley , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
12.
Bioelectrochemistry ; 125: 58-69, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30265864

ABSTRACT

We investigated isolated guinea pig hearts (n = 121) in an ischemia/ reperfusion model with the aim to compare the efficiency of the cardioplegic solution HTK with its novel replacement HTKN. Following consolidation with Tyrode's solution, ischemia started either immediately or after preceding cardioplegia with HTK, HTKN, or modified HTK enriched with Ca. Ischemia lasted either 80 min at 30 °C, or 360 min at 5 °C, or 81 min at 30 °C with intermittent cardioplegic perfusion. During ischemia we measured intracellular calcium (iCa++) and the time of gap junction uncoupling (t-in). During reperfusion we measured the reestablishment of cell coupling (t-ret), left ventricular developed pressure (LVDP), and heart rhythm (VC-RR). In 5 °C groups, iCa++ at t-in was significantly higher than before ischemia, and longest t-in, shortest t-ret, and best VC-RR were observed after HTK-protection. Of all 30 °C groups, the intermittent group with modified HTK showed shortest t-ret, best VC-RR, and the highest LVDP. At 5 °C, HTK groups had higher LVDP than HTK-N groups, but not at 30 °C. The data suggest that the higher calcium level in the HTK-N solution improves reperfusion after short ischemia at 30 °C but for long lasting ischemia at 5 °C it is beneficial to use the HTK solution.


Subject(s)
Cardioplegic Solutions/therapeutic use , Heart Arrest, Induced/methods , Myocardial Reperfusion Injury/therapy , Organ Preservation Solutions/therapeutic use , Animals , Calcium/metabolism , Female , Gap Junctions/metabolism , Gap Junctions/pathology , Glucose/therapeutic use , Guinea Pigs , Mannitol/therapeutic use , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/surgery , Myocardium/metabolism , Myocardium/pathology , Potassium Chloride/therapeutic use , Procaine/therapeutic use
13.
PLoS One ; 13(8): e0203083, 2018.
Article in English | MEDLINE | ID: mdl-30153281

ABSTRACT

Cardiac sympathetic tone overdrive is a key mechanism of arrhythmia. Cardiac sympathetic nerves denervation, such as LSG ablation or renal sympathetic denervation, suppressed both the prevalence of VAs and the incidence of SCD. Accumulating evidence demonstrates the ligament of Marshall (LOM) is a key component of the sympathetic conduit between the left stellate ganglion (LSG) and the ventricles. The present study aimed to investigate the roles of the distal segment of LOM (LOMLSPV) denervation in ischemia and reperfusion (IR)-induced VAs, and compared that LSG denervation. Thirty-three canines were randomly divided into group 1 (IR group, n = 11), group 2 (LOMLSPV Denervation + IR, n = 9), and group 3 (LSG Denervation + IR, n = 13). Hematoxylin-Eosin (HE) and Immunohistochemistry staining revealed that LOMLSPV contained bundles of sympathetic but not parasympathetic nerves. IR increased the cardiac sympathetic tone [serum concentrations of noradrenaline (NE) and epinephrine (E)] and induced the prevalence of VAs [ventricular premature beat (VPB), salvo of VPB, ventricular tachycardia (VT), VT duration (VTD) and ventricular fibrillation (VF)]. Both LOMLSPV denervation and LSG denervation could reduce the cardiac sympathetic tone in Baseline (BS) [heart rate variability (HRV)]. Compared with group 1, LOMLSPV denervation and LSG denervation similarly reduced sympathetic tone [NE (1.39±0.068 ng/ml in group 2, 1.29±0.081 ng/ml in group 3 vs 2.32±0.17 ng/ml in group 1, P<0.05) and E (114.64±9.22 pg/ml in group 2, 112.60±9.69 pg/ml in group 3 vs 166.18±15.78 pg/ml in group 1, P<0.05),] and VAs [VT (0±3.00 in group 2, 0±1.75 in group 3 vs 8.00±11.00 in group 1, P<0.05) and VTD (0 ± 4 s in group 2, 0±0.88s in group 3 vs 10.0 ± 22.00s in group 1, P<0.05)] after 2h reperfusion. These findings indicated LOMLSPV denervation reduced the prevalence of VT by suppressing SNS activity. These effects are comparable to those of LSG denervation. In myocardial IR, the anti-arrhythmic effects of LOMLSPV Denervation may be related to the inhibition of the expression of NE and E.


Subject(s)
Ablation Techniques , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/surgery , Myocardial Ischemia/surgery , Myocardial Reperfusion Injury/surgery , Pericardium/surgery , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Denervation , Disease Models, Animal , Dogs , Epinephrine/blood , Male , Myocardial Ischemia/complications , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/complications , Norepinephrine/blood , Parasympathetic Nervous System/metabolism , Parasympathetic Nervous System/pathology , Pericardium/metabolism , Pericardium/pathology , Random Allocation , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology
14.
Int J Mol Med ; 42(1): 589-596, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29693157

ABSTRACT

Myocardial ischemia­reperfusion (I/R) injury is a major cause of cardiovascular disease worldwide, and microRNAs have been implicated in the regulation of pathological and physiological processes in myocardial I/R injury. The present study aimed to investigate the role of microRNA (miR)­221­3p in myocardial I/R injury. Cell death and lactate dehydrogenase (LDH) activity were increased in hydrogen peroxide (H2O2)­treated H9c2 cells, as measured by flow cytometry and an LDH detection kit. The expression of miR­221­3p was elevated in H2O2­incubated cells and in remote areas of the rat I/R model, examined using reverse transcription­quantitative polymerase chain reaction analysis. The overexpression of miR­221­3p enhanced the number of propidium iodide (PI)+ cells and the activity of LDH in H2O2­treated cells. In I/R­induced rats, the overexpression of miR­221­3p promoted the number of myosin+ cells and inhibited the fractional shortening of left ventricular diameter (FSLVD%). The results showed that the expression of p57 at the gene and protein levels was decreased in H9c2 cells incubated with H2O2 and in rats subjected to I/R surgery; the expression of p57 decreased following the overexpression of miR­221­3p. Subsequently, the hypothesis that p57 was the direct target of miR­221­3p was confirmed by performing a dual­luciferase reporter assay. Finally, to examine the function of p57 in myocardial impairment, p57 was transfected into H9c2 cells and administered to the rats prior to undergoing H2O2 treatment and I/R surgery, respectively. The results indicated that p57 attenuated the number of PI+ cells and the activity of LDH in H2O2­treated cells, whereas p57 downregulated the number of myosin+ cells and upregulated FSLVD% in the I/R­treated rats. Therefore, these findings suggested that miR­221­3p exacerbated the H2O2­induced myocardial damage in H9c2 cells and myocardial I/R injury in the rat model by modulating p57.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/metabolism , Hydrogen Peroxide/pharmacology , MicroRNAs/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Base Sequence , Cell Line , Disease Models, Animal , Male , MicroRNAs/genetics , Myocardial Reperfusion Injury/surgery , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Rats, Sprague-Dawley , Up-Regulation/genetics
15.
Cardiovasc Res ; 114(7): 992-1005, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29518183

ABSTRACT

Aims: Cell therapy trials using cardiac-resident progenitor cells (CPCs) and bone marrow-derived mesenchymal stem/progenitor cells (BMCs) in patients after myocardial infarction have provided encouraging results. Exosomes, nanosized extracellular vesicles of endosomal origin, figure prominently in the bioactivities of these cells. However, a head-to-head comparison of exosomes from the two cell types has not been performed yet. Methods and results: CPCs and BMCs were derived from cardiac atrial appendage specimens and sternal bone marrow, respectively, from patients (n = 20; age, 69.9 ± 10.9) undergoing heart surgery for aortic valve disease and/or coronary artery disease. Vesicles were purified from cell conditioned media by centrifugation/filtration and ultracentrifugation. Vesicle preparations were predominantly composed of exosomes based on particle size and marker expression (CD9, CD63, CD81, Alix, and TSG-101). CPC-secreted exosomes prevented staurosporine-induced cardiomyocyte apoptosis more effectively than BMC-secreted exosomes. In vivo, CPC-secreted exosomes reduced scar size and improved ventricular function after permanent coronary occlusion in rats more efficiently than BMC-secreted exosomes. Both types of exosomes stimulated blood vessel formation. CPC-secreted exosomes, but not BMC-derived exosomes, enhanced ventricular function after ischaemia/reperfusion. Proteomics profiling identified pregnancy-associated plasma protein-A (PAPP-A) as one of the most highly enriched proteins in CPC vs. BMC exosomes. The active form of PAPP-A was detected on CPC exosome surfaces. These vesicles released insulin-like growth factor-1 (IGF-1) via proteolytic cleavage of IGF-binding protein-4 (IGFBP-4), resulting in IGF-1 receptor activation, intracellular Akt and ERK1/2 phosphorylation, decreased caspase activation, and reduced cardiomyocyte apoptosis. PAPP-A knockdown prevented CPC exosome-mediated cardioprotection both in vitro and in vivo. Conclusion: These results suggest that CPC-secreted exosomes may be more cardioprotective than BMC-secreted exosomes, and that PAPP-A-mediated IGF-1 release may explain the benefit. They illustrate a general mechanism whereby exosomes may function via an active protease on their surface, which releases a ligand in proximity to the transmembrane receptor bound by the ligand.


Subject(s)
Exosomes/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Myocardial Ischemia/surgery , Myocardial Reperfusion Injury/surgery , Myocytes, Cardiac/transplantation , Pregnancy-Associated Plasma Protein-A/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis , Atrial Appendage/cytology , Cell Line , Culture Media, Conditioned/metabolism , Exosomes/metabolism , Female , Humans , Insulin-Like Growth Factor I/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice , Middle Aged , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Phenotype , Pregnancy-Associated Plasma Protein-A/genetics , Rats, Wistar , Recovery of Function , Signal Transduction , Ventricular Function, Left
16.
J Am Heart Assoc ; 7(2)2018 01 13.
Article in English | MEDLINE | ID: mdl-29331956

ABSTRACT

BACKGROUND: During myocardial ischemia/reperfusion (MI/R) injury, there is extensive release of immunogenic metabolites that activate cells of the innate immune system. These include ATP and AMP, which upregulate chemotaxis, migration, and effector function of early infiltrating inflammatory cells. These cells subsequently drive further tissue devitalization. Mesenchymal stromal cells (MSCs) are a potential treatment modality for MI/R because of their powerful anti-inflammatory capabilities; however, the manner in which they regulate the acute inflammatory milieu requires further elucidation. CD73, an ecto-5'-nucleotidase, may be critical in regulating inflammation by converting pro-inflammatory AMP to anti-inflammatory adenosine. We hypothesized that MSC-mediated conversion of AMP into adenosine reduces inflammation in early MI/R, favoring a micro-environment that attenuates excessive innate immune cell activation and facilitates earlier cardiac recovery. METHODS AND RESULTS: Adult rats were subjected to 30 minutes of MI/R injury. MSCs were encapsulated within a hydrogel vehicle and implanted onto the myocardium. A subset of MSCs were pretreated with the CD73 inhibitor, α,ß-methylene adenosine diphosphate, before implantation. Using liquid chromatography/mass spectrometry, we found that MSCs increase myocardial adenosine availability following injury via CD73 activity. MSCs also reduce innate immune cell infiltration as measured by flow cytometry, and hydrogen peroxide formation as measured by Amplex Red assay. These effects were dependent on MSC-mediated CD73 activity. Finally, through echocardiography we found that CD73 activity on MSCs was critical to optimal protection of cardiac function following MI/R injury. CONCLUSIONS: MSC-mediated conversion of AMP to adenosine by CD73 exerts a powerful anti-inflammatory effect critical for cardiac recovery following MI/R injury.


Subject(s)
Adenosine/metabolism , Immunity, Innate , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/surgery , Myocardium/metabolism , Tissue Scaffolds , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/metabolism , Animals , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Enzyme Inhibitors/pharmacology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Hydrogen Peroxide/metabolism , Male , Mesenchymal Stem Cells/drug effects , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/immunology , Myocardium/pathology , Neutrophils/immunology , Neutrophils/metabolism , Rats, Sprague-Dawley , Recovery of Function , Stem Cell Niche
17.
J Am Heart Assoc ; 7(2)2018 01 22.
Article in English | MEDLINE | ID: mdl-29358198

ABSTRACT

BACKGROUND: Transplantation of adventitial pericytes (APCs) promotes cardiac repair in murine models of myocardial infarction. The aim of present study was to confirm the benefit of APC therapy in a large animal model. METHODS AND RESULTS: We performed a blind, randomized, placebo-controlled APC therapy trial in a swine model of reperfused myocardial infarction. A first study used human APCs (hAPCs) from patients undergoing coronary artery bypass graft surgery. A second study used allogeneic swine APCs (sAPCs). Primary end points were (1) ejection fraction as assessed by cardiac magnetic resonance imaging and (2) myocardial vascularization and fibrosis as determined by immunohistochemistry. Transplantation of hAPCs reduced fibrosis but failed to improve the other efficacy end points. Incompatibility of the xenogeneic model was suggested by the occurrence of a cytotoxic response following in vitro challenge of hAPCs with swine spleen lymphocytes and the failure to retrieve hAPCs in transplanted hearts. We next considered sAPCs as an alternative. Flow cytometry, immunocytochemistry, and functional/cytotoxic assays indicate that sAPCs are a surrogate of hAPCs. Transplantation of allogeneic sAPCs benefited capillary density and fibrosis but did not improve cardiac magnetic resonance imaging indices of contractility. Transplanted cells were detected in the border zone. CONCLUSIONS: Immunologic barriers limit the applicability of a xenogeneic swine model to assess hAPC efficacy. On the other hand, we newly show that transplantation of allogeneic sAPCs is feasible, safe, and immunologically acceptable. The approach induces proangiogenic and antifibrotic benefits, though these effects were not enough to result in functional improvements.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/surgery , Myocardium/pathology , Neovascularization, Physiologic , Pericytes/transplantation , Ventricular Function, Left , Ventricular Remodeling , Aged , Aged, 80 and over , Allogeneic Cells , Animals , Cells, Cultured , Disease Models, Animal , Female , Fibrosis , Heterografts , Humans , Male , Middle Aged , Myocardial Contraction , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Recovery of Function , Stroke Volume , Sus scrofa , Transplantation, Homologous
18.
Circ Res ; 121(11): 1263-1278, 2017 Nov 10.
Article in English | MEDLINE | ID: mdl-28912121

ABSTRACT

RATIONALE: Cortical bone stem cells (CBSCs) have been shown to reduce ventricular remodeling and improve cardiac function in a murine myocardial infarction (MI) model. These effects were superior to other stem cell types that have been used in recent early-stage clinical trials. However, CBSC efficacy has not been tested in a preclinical large animal model using approaches that could be applied to patients. OBJECTIVE: To determine whether post-MI transendocardial injection of allogeneic CBSCs reduces pathological structural and functional remodeling and prevents the development of heart failure in a swine MI model. METHODS AND RESULTS: Female Göttingen swine underwent left anterior descending coronary artery occlusion, followed by reperfusion (ischemia-reperfusion MI). Animals received, in a randomized, blinded manner, 1:1 ratio, CBSCs (n=9; 2×107 cells total) or placebo (vehicle; n=9) through NOGA-guided transendocardial injections. 5-ethynyl-2'deoxyuridine (EdU)-a thymidine analog-containing minipumps were inserted at the time of MI induction. At 72 hours (n=8), initial injury and cell retention were assessed. At 3 months post-MI, cardiac structure and function were evaluated by serial echocardiography and terminal invasive hemodynamics. CBSCs were present in the MI border zone and proliferating at 72 hours post-MI but had no effect on initial cardiac injury or structure. At 3 months, CBSC-treated hearts had significantly reduced scar size, smaller myocytes, and increased myocyte nuclear density. Noninvasive echocardiographic measurements showed that left ventricular volumes and ejection fraction were significantly more preserved in CBSC-treated hearts, and invasive hemodynamic measurements documented improved cardiac structure and functional reserve. The number of EdU+ cardiac myocytes was increased in CBSC- versus vehicle- treated animals. CONCLUSIONS: CBSC administration into the MI border zone reduces pathological cardiac structural and functional remodeling and improves left ventricular functional reserve. These effects reduce those processes that can lead to heart failure with reduced ejection fraction.


Subject(s)
Cortical Bone/cytology , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/surgery , Myocardium/pathology , Stem Cells/physiology , Ventricular Function, Left , Ventricular Remodeling , Animals , Apoptosis , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Hemodynamics , Myocardial Contraction , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Phenotype , Stroke Volume , Sus scrofa , Time Factors
19.
Int J Cardiol ; 243: 485-491, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28602551

ABSTRACT

BACKGROUND: Ischemia-reperfusion (I/R) injury in ST-segment elevation myocardial infarction (STEMI) significantly contributes to overall myocardial damage. As a consequence of I/R injury in the heart, the high-temperature requirement protein A2 (HtrA2) is released from the mitochondrial intermembrane space of cardiomyocytes to the cytoplasm, whereupon it induces apoptosis. METHODS: Serum was obtained from STEMI (n=37), non-ST-segment elevation myocardial infarction (NSTEMI) (n=20), stable coronary artery disease (CAD) (n=17) and patients with CAD excluded (n=9). In STEMI, I/R injury was assessed via measurement of ST-segment resolution. RESULTS: HtrA2 was significantly increased in STEMI compared to NSTEMI, stable CAD and patients with CAD excluded (981.3 (IQR: 543.5-1526.2)pg/mL vs. 494.5 (IQR: 413.8-607)pg/mL vs. 291 (IQR: 239-458.5)pg/mL vs. 692.2 (IQR: 276.6-964.7)pg/mL; p≤0.0001). STEMI patients with HtrA2 level of at least the median or above had a higher peak creatine kinase (CK) (p=0.0002) and cardiac troponin T levels (cTnT) (p=0.0019). Significantly more STEMI patients with HtrA2 levels of at least the median or above were identified as I/R injury (87% vs. 42%; p<0.0001). Serum HtrA2 demonstrated a superior area under a curve in a receiver operating characteristic analysis for predicting I/R injury compared to CK, creatine kinase myocardial-band (CK-MB) and cTnT levels (AUC=0.7105 vs. AUC=0.5632 vs. AUC=0.5660 vs. AUC=0.5407 respectively). CONCLUSION: HtrA2 shows promise as a novel potential biomarker for mitochondrial-induced cardiomyocyte apoptosis and may help to identify I/R injury after STEMI.


Subject(s)
High-Temperature Requirement A Serine Peptidase 2/blood , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/blood , Myocytes, Cardiac/metabolism , ST Elevation Myocardial Infarction/blood , Aged , Aged, 80 and over , Biomarkers/blood , Female , Humans , Male , Middle Aged , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/surgery , Percutaneous Coronary Intervention/methods , Prospective Studies , Retrospective Studies , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/surgery
20.
Int J Cardiol ; 241: 138-141, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28501350

ABSTRACT

AIMS: Pressure-controlled Intermittent Coronary Sinus Occlusion (PICSO) intermittently increases the pressure in the cardiac venous outflow tract using a balloon-tipped catheter introduced percutaneously into the coronary sinus. It aims to improve microvascular perfusion in STEMI patients during PCI, thus improving infarct healing. Its successful administration was associated with an improvement in myocardial recovery four months after primary-PCI as compared to control. However, it has never been used in other settings or for a prolonged period. The aim of this study was to report on the feasibility and efficacy of prolonged PICSO to treat refractory LV dysfunction and ischemia. METHODS AND RESULTS: Two patients with refractory LV dysfunction and ongoing ischemia with patent epicardial coronary artery and suspected underlying microvascular dysfunction were treated with prolonged off-label PICSO utilization. A medium of 23,990mmHg PICSO quantity (a marker of PICSO therapy performance) was achieved: it was almost fifty times the PICSO quantity previously reported in in short-term PICSO utilization. After PICSO placement, both patients showed significant improvement of myocardial ischemia and recovery of LV systolic function. CONCLUSION: Prolonged PICSO utilization was feasible and effective in two patients. These cases highlight a novel application of PICSO technology: redistribution of venous blood and improvement in microvascular perfusion that might be a new target in cases of refractory LV dysfunction and ongoing ischemia in the setting of patent epicardial coronary arteries.


Subject(s)
Cardiac Catheterization/methods , Coronary Sinus/surgery , Myocardial Ischemia/surgery , Percutaneous Coronary Intervention/methods , Ventricular Dysfunction, Left/surgery , Adult , Aged , Aortic Valve Insufficiency/diagnostic imaging , Aortic Valve Insufficiency/surgery , Coronary Sinus/diagnostic imaging , Coronary Vessels/diagnostic imaging , Coronary Vessels/surgery , Female , Humans , Male , Myocardial Ischemia/diagnostic imaging , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/surgery , Pressure , Treatment Outcome , Ventricular Dysfunction, Left/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...