key: cord-0019120-lrf5gsjp authors: Jiang, Xiaosi; Wu, Dan; Jiang, Zichao; Ling, Weiwei; Qian, Geng title: Protective Effect of Nicorandil on Cardiac Microvascular Injury: Role of Mitochondrial Integrity date: 2021-07-03 journal: Oxid Med Cell Longev DOI: 10.1155/2021/4665632 sha: f08cfa637dcd929d71a8b0f8dfe0524bf936a2b1 doc_id: 19120 cord_uid: lrf5gsjp A major shortcoming of postischemic therapy for myocardial infarction is the no-reflow phenomenon due to impaired cardiac microvascular function including microcirculatory barrier function, loss of endothelial activity, local inflammatory cell accumulation, and increased oxidative stress. Consequently, inadequate reperfusion of the microcirculation causes secondary ischemia, aggravating the myocardial reperfusion injury. ATP-sensitive potassium ion (K(ATP)) channels regulate the coronary blood flow and protect cardiomyocytes from ischemia-reperfusion injury. Studies in animal models of myocardial ischemia-reperfusion have illustrated that the opening of mitochondrial KATP (mito-K(ATP)) channels alleviates endothelial dysfunction and reduces myocardial necrosis. By contrast, blocking mito-K(ATP) channels aggravates microvascular necrosis and no-reflow phenomenon following ischemia-reperfusion injury. Nicorandil, as an antianginal drug, has been used for ischemic preconditioning (IPC) due to its mito-K(ATP) channel-opening effect, thereby limiting infarct size and subsequent severe ischemic insult. In this review, we analyze the protective actions of nicorandil against microcirculation reperfusion injury with a focus on improving mitochondrial integrity. In addition, we discuss the function of mitochondria in the pathogenesis of myocardial ischemia. Acute myocardial infarction (AMI), as a coronary artery disease, is increasingly becoming a leading cause of death worldwide. In clinical practice, primary percutaneous coronary intervention (PCI) is a standard therapeutic strategy to open blocked vessels in patients with ST-segment elevation myocardial infarction (STEMI), since it shortens the total ischemic time and reduces the mortality rate. However, a few patients still suffer from myocardial postischemic injury, which is termed as the no-reflow phenomenon during or after PCI [1] [2] [3] [4] . The potential mechanism of no-reflow phenomenon involves coronary microvascular dysfunction (CMD) which has been considered as an independent risk factor for rehospitalization and 30-day mortality in AMI patients during or after PCI therapy [5] . CMD is often responsible for angina and poor prognosis after PCI in AMI patients [6] . Recently, several researches have confirmed that mitochondrial dysfunction is a key factor in the pathogenesis of AMI and no-reflow phenomenon [7, 8] . Mitochondria maintain microcirculation function by modifying the postischemic injury signals-clearing the damaged mitochondria via mitochondrial autophagy (mitophagy), transmitting the extracellular signals to endothelial cells, and controlling endothelial apoptosis or survival [9] [10] [11] . Thus, mitochondrial dysfunction is a characteristic feature of myocardial ischemia-reperfusion injury (IRI) [12] [13] [14] [15] . Nicorandil is an antianginal drug with nitrate-like effects. It exerts its cardiomyocyte protection effects by directly opening the mitochondrial ATP-sensitive potassium ion (KATP) channels, increasing the K + influx, depolarizing the mitochondrial membrane, blocking mitochondrial Ca 2+ uptake, restoring mitochondrial function, promoting ATP generation, alleviating ischemic damage, and preventing cardiomyocyte apoptosis [16, 17] (Table 1) . Nicorandil's core property of maintaining mitochondrial integrity makes it a suitable candidate for inhibiting microcirculatory reperfusion injury. Microcirculatory Reperfusion Injury and Impaired Mitochondrial Integrity 2.1. Microcirculatory Reperfusion Injury. The pathogenesis of IRI involves microcirculatory injury-related no-reflow. At the molecular levels, reperfusion-triggered intracellular calcium overload, local accumulation of inflammatory cells due to blood flow restoration, excessive production of reactive oxygen species (ROS), and deficiency of high-energy phosphate compounds contribute to the development of microvascular injury [7, [18] [19] [20] [21] [22] . Structurally, microcirculatory disturbances are characterized by endothelial swelling, microvascular spasm, and increased capillary resistance, 2 Oxidative Medicine and Cellular Longevity consequently hindering or interrupting the communication between cardiomyocyte and fresh blood flow, with an effect that is followed by increased apoptosis of myocardial microvascular endothelial cells [23] [24] [25] [26] . Of note, both the structure and function of microvascular endothelium determine microvascular reperfusion and blood supply to cardiomyocytes [27] [28] [29] . No-reflow has been clinically observed in approximately 10% to 50% of patients with IRI. Reperfusion causes the platelets to bind neutrophils, promotes the retention of inflammatory cells in the microcirculation [30] [31] [32] , and therefore interferes with the diastolic function of the microcirculation, resulting into decreased blood flow to reperfused myocardium [33] [34] [35] . In addition, reperfusion-induced endothelial damage exposes the subendothelial collagen, allowing rapid binding of platelets to the surface of the microvascular endothelium through surface adhesion factors [36] [37] [38] . This cascade of events activates local platelets by releasing platelet factors, leading to microthrombosis and ultimately blocking the blood flow [39] [40] [41] . Although the blow flow through the epicardial large vessels is smooth, small vessels remain insufficiently perfused, causing secondary ischemia, local blood hypoperfusion, impaired energy metabolism, and consumption of high-energy phosphate compounds [9, 42] . Inadequate energy production limits cellular calcium recycling and contributes to intracellular calcium overload. Under physiological conditions, an appropriate increase in mitochondrial Ca 2+ is associated with the augmented tricarboxylic acid cycle and ATP production [43] [44] [45] . Similarly, appropriate cytoplasmic Ca 2+ levels increase cardiomyocyte contractility through calcium sparks. Besides, a moderate elevation in baseline Ca 2+ levels improves cytoskeletal tension and endothelial motility [36, 46] . However, excessive Ca 2+ accumulation can directly activate calcium-dependent protein kinases to trigger the endothelial apoptotic pathway. In addition, increased Ca 2+ concentrations activate calciumdependent xanthine oxidase (XO), inducing oxidative stress in endothelial cells [29] . The cytoplasmic calcium overload of endothelial cells, accompanied by mitochondrial calcium overload, induces cytoskeletal disintegration and impairs filamentous pseudopod formation through the IP3R-Ca 2+ -VDAC signaling pathway. These events cause impaired migration of endothelial cells and their reduced ability to revascularize after AMI [23, [47] [48] [49] , ultimately causing myocardial remodeling and heart failure [41, [50] [51] [52] . Thus, abnormal calcium signals aggravate oxidative stress, trigger mitochondrial damage, destroy endothelial motility and chemotaxis, promote apoptosis, and induce endothelial reperfusion injury and microcirculation dysfunction ( Table 1) . The endothelial barrier function is highly dependent on the expression of VE-cadherin that participates in endothelial filtration and resistance to substances in the blood through local gap junctions [53, 54] . IRI is characterized by reduced VE-cadherin expression, causing leakage of inflammatory cells into the subendothelial myocardium [55] [56] [57] . Although reperfusion-induced moderate inflammatory response helps to remove necrotic tissue and thus promote the reconstruction of the infarcted myocardium, excessive inflammation induces residual myocyte dysfunction, exacer-bating the myocardial injury and inducing oxidative stress in the myocardium and endothelial cells [37, 58, 59] . Microcirculation damage is closely associated with augmented IRI via multiple pathophysiological processes. Oxidative stress is one of the primary features of MIRI. First, abnormal redox biology substantially consumes the levels of reduced hydrogen [60] [61] [62] . Second, excessive ROS accumulation within the cytoplasm induces oxidation of lipid components in the mitochondrial membrane, especially cardiolipin, altering the mitochondrial membrane permeability, inducing changes in mitochondrial membrane potential, and disrupting energy metabolism [63] [64] [65] . Third, overproduced ROS disrupts the endoplasmic reticulum (ER) membrane structure and interferes with the modification of proteins, consequently increasing the level of unfolded proteins and causing ER stress. Fourth, excessive oxidative stress induces the oxidation of sarco/endoplasmic reticulum Ca2 + -ATPase (SERCA), a calcium recycling protein, in the ER [29, 66] , thus, reducing ER calcium recycling capacity and inducing cellular calcium overload and endothelial cell apoptosis [67] (Table 1 ). Mitochondria are membrane-bound eukaryotic organelles that synthesize ATP, maintain Ca 2+ steady-state, generate ROS, and regulate apoptosis, mitophagy, and the opening of mitochondrial permeability transition pore (mPTP) in cardiomyocytes [68, 69] . Mitochondria play a necessary role in myocardial metabolism through affecting mitochondrial dynamics, mitochondrial biogenesis, Ca 2+ homeostasis, and redox biology [70, 71] . Myocardial ischemia is followed by mitochondrial injury, which aggravates the IRI since Ca 2+ overload will in turn disrupt mitochondrial structure and function. Endothelial cells contain fewer mitochondria than cardiomyocytes; they fulfill 75% of their energy demands from glycolysis rather than mitochondrial oxidative phosphorylation. Instead of serving as traditional energy centers, mitochondria in endothelial cells participate in various signal transduction as well as endothelial stress responses [63, [72] [73] [74] . Recent studies have implicated mitochondrial dynamics (mitochondrial fission, fusion, and autophagy) in maintaining mitochondrial integrity [75] . During IRI, high levels of Na + activate reverse Na + /Ca 2+ exchange, causing massive Ca 2+ influx into the cytoplasm. In addition, insufficient ATP production and restoration of the mitochondrial membrane potential reduce the activity of the Ca 2+ pump and consequently activate the activity of mitochondrial calcium uniporter (MCU), resulting into intracellular Ca 2+ overload and exacerbating mitochondrial damage [76] [77] [78] . The initial phase of myocardial ischemia and reperfusion is characterized by an explosive increase in ROS, causing oxidative stress, lipid peroxidation, and mitochondrial damage [79] [80] [81] . Although mitophagy is able to clear the damaged cardiomyocytes and protects the adjacent normal myocardial tissues under physiological conditions [82] , massive production of ROS and Ca 2+ overload not only inhibit the activity of mitophagy but also delay the opening of mPTP, an effect that is followed by increased permeability of the mitochondrial outer membrane. The delayed 3 Oxidative Medicine and Cellular Longevity opening of mPTP also induces mitochondrial swelling and membrane rupture, promotes cytochrome C (Cyt C) release into the cytoplasm [83, 84] , increases Bax expression, and decreases Bcl2 expression, ultimately activating the mitochondria-dependent apoptotic cascade in reperfused heart tissues [85, 86] (Figure 1, Table 1 ). Mitochondrial Integrity 3.1. Dilation of Coronary Arteries. Nicorandil exerts its antiischemic effect primarily through dilating the coronary arteries and reducing myocardial oxygen demand. The opening of K ATP channels in vascular smooth muscle cells by nicorandil hyperpolarizes the membrane, closing voltage-sensitive calcium channels and reducing the calcium inflow, ultimately reducing the vascular resistance and therefore promoting the dilation of the blood vessels [87, 88] . Furthermore, its nitrate effect generates NO free radicals, which directly activate the guanylate cyclase and increase cGMP synthesis in myocardial microcirculation. Elevated cGMP levels, in turn, target cGMP-dependent kinases and cyclic nucleotide-gated ion channel effectors [89] [90] [91] . Reduced intracellular free Ca 2+ and desensitization of smooth muscle cell contractile proteins to Ca 2+ result in vasodilation, decreased vascular resistance, and widening of blood vessels [92] . Repeated transient coronary occlusion will increase the myocardial tolerance to prolonged ischemia and reduce infarct size after myocardial infarction, which is termed by ischemic preconditioning (IPC) [93, 94] . The mitochondrial K ATP (mito-K ATP ) channel is the terminal effector of IPC. Shortterm ischemia can increase the resistance of the myocardium to subsequent long-term ischemia and protect the heart by reducing oxidative stress during and after IRI [95] [96] [97] . Cardiomyocyte mito-K ATP is closed at the basic state and opens at the stage of ischemia, whereas nicorandil is capable to improve myocardial resistance to ischemia challenge by opening mito-K ATP channels, which enhance the cardioprotective actions of IPC. During myocardial ischemia, intracellular Ca 2+ increases, mitochondrial matrix contracts, respiratory function is impaired, ATP production decreases, and myocardial cells become apoptotic. Nicorandil, as a K ATP channel agonist, directly opens the mito-K ATP channels, increases mitochondrial K + inward flow, decreases the transmembrane potential difference, depolarizes the mitochondrial membrane, reduces Ca 2+ inward flow dynamics, inhibits Ca 2+ inward flow, and effectively prevents calcium overload in mitochondria, leading to mitochondrial relaxation, enhanced respiratory function, and increased ATP production. These regulatory effects finally protect the reperfused heart through reducing cardiomyocyte apoptosis and mitigating cardiomyocyte injury and apoptosis [16, 17, 98] . Inflammatory response after AMI promotes cardiac healing during IRI, whereas prolonged inflammation enhances postischemic injury and adverse cardiac remodeling [99] . Su et al. [99] constructed a coronary microembolization (CME) rat model and demonstrated that nicorandil inhibited myocardial inflammation, alleviated myocardial injury, and improved cardiac function primarily by inhibiting Toll-like receptor 4-(TLR4-) induced myeloid differentiation primary response protein 88-(MyD88-) dependent activation of nuclear factor-kappa B (NF-κB) signaling pathway. These alterations reduced the release of proinflammatory cytokines, such as tumor necrosis factor-(TNF-) α and interleukin-(IL-) 1β, from ischemic cardiomyocytes. Similarly, Zhang et al. [100] showed that nicorandil maintained the macrophage M1/M2 status in M1 and M2 cell models by inhibiting the differentiation of monocytes into mature macrophages, suppressing M1 phenotype transition, and promoting the transition to the M2 phenotype to exert anti-inflammatory effects. In addition, both of their data demonstrated that NF-κb and its target gene controlled the macrophage phenotype (Table 1) . Oxidative stress or redox imbalance resulting from the excessive generation of ROS during reperfusion is one of the major contributing factors to IRI pathogenesis. Some of its detrimental effects include macromolecule oxidation, membrane lipid peroxidation, membrane dysfunction, altered calcium homeostasis, neutrophil migration, DNA lesions, mitochondria-triggered apoptosis, and mPTP opening [69] . Nicorandil, as K ATP channel agonists, slightly lowers the mitochondrial membrane potential and prevents ROS formation without compromising the cellular energetics during hypoxia-reoxygenation attack as well as IRI [101, 102] . Nicorandil alleviated mitochondrial Ca 2+ overload and depolarized the mitochondrial membrane in rat ventricular cells [103] . In addition, the nicotinamide moiety of nicorandil exerted appreciable hydroxyl radical scavenging activity via a mechanism independent from its K ATP channel opening effect [81, 104] (Figure 2 , Table 1 ). Pretreatment with low doses of nicorandil reduced IRI in rats by selectively opening the mito-K ATP channels [105] and protected cardiomyocytes against IRI biochemical changes and ventricular arrhythmias. The use of 5-hydroxydecanoate, a mito-K ATP channel selective blocker, inhibited the protective effects of nicorandil on the ventricular myocardium, confirming the function of mito-K ATP on improving coronary microcirculation and protecting the myocardium [105] . Therefore, the mito-K ATP channel serves as a terminal effector of myocardial IPC-short-term ischemia that increases the myocardial resistance to subsequent long-term ischemia and protects the heart by reducing oxidative stress during IRI [95, 96] . Murry et al. [96] showed that four 5minute episodes of ischemia followed by a sustained 40minute coronary occlusion caused less necrosis compared with a single 40-minute occlusion in the canine heart. However, this protective effect disappeared when sustained blood loss was prolonged to 3 h [88, 106] . Interestingly, nicorandil opens the mito-K ATP channels to mimic or enhance myocardial IPC and therefore improve myocardial ischemic resistance ( Table 1) . The glycolytic pathway serves as the primary energy production pathway under myocardial ischemia. During reperfusion, Na + /H + exchange protein (NHE) is activated; H + is transported out of the cell, and the intracellular Na + increases greatly. Due to insufficient ATP produced by glycolysis to meet the normal requirements of cardiomyocytes, the activity of Na + -K + -ATP is inhibited causing Na + overload (Na + cannot be expelled from cells) and activating the reverse Na + /Ca 2+ exchange. The abundant Ca 2+ inward flow eventually causes Ca 2+ overload [107] [108] [109] . Nicorandil treatment opened mito-K ATP channels allowing abundant K + to enter the mitochondria, hyperpolarized mitochondrial membranes, promoted K + -Ca 2+ exchange, inhibited Ca 2+ influx, and improved Ca 2+ overload in a rabbit ischemic cardiomyocyte model [98] (Table 1) . Oxidative Medicine and Cellular Longevity regulatory center of apoptosis. Mitochondria regulate apoptosis via the main executor, Caspase-3-a member of the Caspase cysteine protease family. Cytoplasmic Cyt C interacts with apoptosis protease activating factor-1 (Apaf1) in the presence of deoxyadenosine triphosphate (dATP) and ATP and polymerizes to form apoptotic bodies. Subsequently, it activates the upstream factor Caspase-9 and the downstream factors such as Caspase-3, Caspase-6, and Caspase-7 in a cascade event, eventually causing apoptosis [110] . In addition, either Bax or Bak induces the release of Cyt C into the cytoplasm [86, [111] [112] [113] . Decreased expression of Bcl-2 increases the expression of Bax and Bak to accelerate apoptosis, whereas increased expression of Bcl-2 produces the opposite effect on reperfused hearts. A surge in ROS causes oxidative stress and lipid peroxidation, whereas oxidative stress, in turn, increases ROS due to impaired mitochondrial electron transport chain (ETC) and reduced activity of antioxidant enzymes [79, 80, 114] . Ca 2+ overload, ROS surge, and pH changes work together to open the mPTP as well as prolong its opening time by activating cyclophilin D (Cyp D), an essential component of mPTP. mPTP is a nonspecific, highly conductive, multiprotein channel located in the mitochondrial membrane gap. It remains inactive under physiological conditions, maintains ion mobility both inside and outside the mitochondria along with a stable membrane potential difference, and regulates mitochondrial membrane permeability [111] . Its irreversible opening during IRI triggers cardiomyocyte necrosis rather than apoptosis [115] . mPTP remains closed during acute myocardial ischemia and opens within the first few minutes of reperfusion, allowing small molecules, which normally cannot cross the mitochondrial membranes, to pass through, thus, increasing the osmotic pressure of the mitochondrial matrix, inducing mitochondrial fragmentation, and releasing Cyt C [83, 112, [116] [117] [118] (Figure 2 ). Nicorandil-induced opening of mito-K ATP channels triggers the production of low levels of ROS that are incapable of causing oxidative damage [119] [120] [121] . In contrast, elevated ROS activate several signaling pathways, stimulate mitochondrial antioxidant effect, and inhibit mitochondrial nicotinamide adenine dinucleotide phosphate oxidase-a major source of ROS production in cardiomyocytes [122] . Furthermore, nicorandil inhibits the release of Cyt C. The simultaneous increase in mitochondrial Bcl2 levels and decrease in Bax levels (elevated Bcl2/Bax ratio) inhibit the apoptotic pathway [123] . Another study showed that Bcl2 overexpression prolonged the survival time of mice, represented by reduced mitochondrial abnormalities, restored cardiac function, and decreased apoptosis [124] (Table 1) . Nicorandil pretreatment reduces Ca 2+ overload, increases ROS at a low level, enhances ATP production, inhibits ATP consumption, decreases Cyt C release, reduces mPTP opening [125, 126] , shortens the ischemic PC period, and inhibits apoptotic signaling pathways, thereby ameliorating mitochondrial dysfunction and allowing better myocardial salvage during reperfusion therapy (Table 1) . To conclude, nicorandil improves coronary microvascular function and restores microvascular dynamics, making it one of the most promising drugs for treating CMD. How-ever, little literature is available on it, mostly including small-scale randomized controlled trials with a few reports on its anti-CMD long-term effects. Because multiple mechanisms are implicated in CMD, combination therapy of nicorandil can prove to be highly effective [127] . With the research on the role of nicorandil in maintaining mitochondrial homeostasis, the cardiovascular protective mechanism of nicorandil has become increasingly clear and will be more and more widely used in clinical. The authors declare that they have no conflicts of interest. Xiaosi Jiang, Dan Wu, Zichao Jiang, and Weiwei Ling contributed equally to this work and share first authorship. Thrombus aspiration during primary percutaneous coronary intervention Effect of no-reflow during primary percutaneous coronary intervention for acute myocardial infarction on six-month mortality Treating and preventing no reflow in the cardiac catheterization laboratory Role of mitochondrial quality surveillance in myocardial infarction: from bench to bedside Atherosclerotic plaque morphology indicates clinical symptoms of plaque progression Reappraisal of ischemic heart disease Mitochondrial quality surveillance as a therapeutic target in myocardial infarction Mitochondrial quality control mechanisms as molecular targets in cardiac ischemia-reperfusion injury Microvascular obstruction: underlying pathophysiology and clinical diagnosis New insights into the role of mitochondria in cardiac microvascular ischemia/reperfusion injury BI1 is associated with microvascular protection in cardiac ischemia reperfusion injury via repressing Syk-Nox2-Drp1-mitochondrial fission pathways Mitochondrial quality control in cardiac microvascular ischemia-reperfusion injury: new insights into the mechanisms and therapeutic potentials SERCA overexpression improves mitochondrial quality control and attenuates cardiac microvascular ischemia-reperfusion injury Coronary microvascular obstruction: the new frontier in cardioprotection Perspective: cardiovascular disease and the COVID-19 pandemic Impact of a single intravenous administration of nicorandil before reperfusion in patients with ST-segment-elevation myocardial infarction Nicorandil improves cardiac function and clinical outcome in patients with acute myocardial infarction undergoing primary percutaneous coronary intervention: role of inhibitory effect on reactive oxygen species formation The coronary circulation as a target of cardioprotection Overexpression of mitochondrial creatine kinase preserves cardiac energetics without ameliorating murine chronic heart failure Microvascular and lymphatic dysfunction in HFpEF and its associated comorbidities Impaired vascular endothelial growth factor expression and secretion during in vitro differentiation of human primary term cytotrophoblasts Role of VEGFs in metabolic disorders Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca2+]c/VDAC-[Ca2+]m axis by activation of MAPK/ERK signaling pathway NR4A1 aggravates the cardiac microvascular ischemia reperfusion injury through suppressing FUNDC1-mediated mitophagy and promoting Mff-required mitochondrial fission by CK2α Autophagy in animal development Understanding offtarget effects through hybridization kinetics and thermodynamics Pathological roles of mitochondrial oxidative stress and mitochondrial dynamics in cardiac microvascular ischemia/reperfusion injury Pum2-Mff axis fine-tunes mitochondrial quality control in acute ischemic kidney injury Liraglutide protects cardiac microvascular endothelial cells against hypoxia/reoxygenation injury through the suppression of the SR-Ca 2+ -XO-ROS axis via activation of the GLP-1R/PI3K/Akt/survivin pathways Higher mitochondrial potential and elevated mitochondrial respiration are associated with excessive activation of blood platelets in diabetic rats Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARγ/FUNDC1/mitophagy pathways FHL-1 is not involved in pressure overload-induced maladaptive right ventricular remodeling and dysfunction Universal classification system type of incident myocardial infarction in patients with stable atherosclerosis: observations from thrombin receptor antagonist in secondary prevention of atherothrombotic ischemic events (TRA 2°P)-TIMI 50 DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways Glucose withdrawal induces endothelin 1 release with significant angiogenic effect from first trimester (FTM), but not term human umbilical cord perivascular cells (HUCPVC) Stabilization of myeloid-derived HIFs promotes vascular regeneration in retinal ischemia DNA-PKcs promotes cardiac ischemia reperfusion injury through mitigating BI-1-governed mitochondrial homeostasis Translational large animal model of hibernating myocardium: characterization by serial multimodal imaging BI1 alleviates cardiac microvascular ischemia-reperfusion injury via modifying mitochondrial fission and inhibiting XO/R-OS/F-actin pathways Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTPmitophagy axis Bax inhibitor 1 preserves mitochondrial homeostasis in acute kidney injury through promoting mitochondrial retention of PHB2 The "noreflow" phenomenon after temporary coronary occlusion in the dog Endothelial cells, endoplasmic reticulum stress and oxysterols Murine sca1/flk1-positive cells are not endothelial progenitor cells, but B2 lymphocytes Angiopoietin-2 as a marker of endothelial activation is a good predictor factor for intensive care unit admission of COVID-19 patients Impact of high cholesterol and endoplasmic reticulum stress on metabolic diseases: an updated mini-review Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemiareperfusion injury Endothelial deletion of ADAM10, a key regulator of notch signaling, causes impaired decidualization and reduced fertility in female mice The inotropic agent digitoxin strengthens desmosomal adhesion in cardiac myocytes in an ERK1/2-dependent manner Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission TMEM100 is a key factor for specification of lymphatic endothelial progenitors Perturbations in myocardial perfusion and oxygen balance in swine with multiple risk factors: a novel model of ischemia and no obstructive coronary artery disease Stachydrine protects eNOS uncoupling and ameliorates endothelial dysfunction induced by homocysteine Selenium inhibits homocysteineinduced endothelial dysfunction and apoptosis via activation of AKT Heterogeneity and chimerism of endothelial cells revealed by singlecell transcriptome in orthotopic liver tumors Anaemia is associated with severe RBC dysfunction and a reduced circulating NO pool: vascular and cardiac eNOS are crucial for the adaptation to anaemia Secreted frizzled-related protein 2, a novel mechanism to induce myocardial ischemic protection through angiogenesis Monocytes: protagonists of infarct inflammation and repair after myocardial infarction ER-mitochondria microdomains in cardiac ischemia-reperfusion injury: a fresh perspective Microvascular dysfunction in ankylosing spondylitis is associated with disease activity and is improved by anti-TNF treatment Treatment with tumor necrosis factor inhibitors restores coronary microvascular function in young patients with severe psoriasis Pleiotrophin selectively binds to vascular endothelial growth factor receptor 2 and inhibits or stimulates cell migration depending on ανβ3 integrin expression Nitrite-mediated reduction of macrophage NADPH oxidase activity is dependent on xanthine oxidoreductase-derived nitric oxide but independent of Snitrosation Small extracellular vesicles secreted from human amniotic fluid mesenchymal stromal cells possess cardioprotective and promigratory potential TRPV3 enhances skin keratinocyte proliferation through EGFR-dependent signaling pathways ATF6 decreases myocardial ischemia/reperfusion damage and links ER stress and oxidative stress signaling pathways in the heart Ripk3 regulates cardiac microvascular reperfusion injury: the role of IP3R-dependent calcium overload, XO-mediated oxidative stress and F-action/filopodia-based cellular migration Mitochondrial oxidative stress and "mitoinflammation": actors in the diseases Recent progress in the use of mitochondrial membrane permeability transition pore in mitochondrial dysfunction-related disease therapies Mechanotranduction pathways in the regulation of mitochondrial homeostasis in cardiomyocytes The endocannabinoid anandamide has an anti-inflammatory effect on CCL2 expression in vascular smooth muscle cells Mitochondria and angiogenesis Mitochondria in endothelial cells: sensors and integrators of environmental cues Stromal cellderived factor 1 (SDF1) attenuates platelet-derived growth factor-B (PDGF-B)-induced vascular remodeling for adipose tissue expansion in obesity The involvement of autophagy in the maintenance of endothelial homeostasis: the role of mitochondria Role of calcium ions in regulation of mammalian intramitochondrial metabolism Sodium/calcium exchange: its physiological implications Mitochondrial calcium channels Leptin enhances NMDA-induced spinal excitation in rats: a functional link between adipocytokine and neuropathic pain Melatonin rescues cardiovascular dysfunction during hypoxic development in the chick embryo Leopold Auerbach's achievements in the field of vascular system Melatonin enhances mitophagy and mitochondrial biogenesis in rats with carbon tetrachloride-induced liver fibrosis SIRT1 inhibition causes oxidative stress and inflammation in patients with coronary artery disease A future for remote ischaemic conditioning in high-risk patients Cell death pathways: intricate connections and disease implications Apatinib as targeted therapy for advanced bone and soft tissue sarcoma: a dilemma of reversing multidrug resistance while suffering drug resistance itself Nicorandil: a drug with ongoing benefits and different mechanisms in various diseased conditions Use of nicorandil in cardiovascular disease and its optimization New insights into bioactivation of organic nitrates, nitrate tolerance and cross-tolerance Nicorandil and long-acting nitrates: vasodilator therapies for the management of chronic stable angina pectoris Prognostic value of CEC count in HER2-negative metastatic breast cancer patients treated with bevacizumab and chemotherapy: a prospective validation study (UCBG COMET) Cyclic nucleotide-dependent relaxation pathways in vascular smooth muscle Ischemic preconditioning DNA-PKcs promotes alcohol-related liver disease by activating Drp1-related mitochondrial fission and repressing FUNDC1-required mitophagy Nicorandil ameliorates mitochondrial dysfunction in doxorubicin-induced heart failure in rats: possible mechanism of cardioprotection Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium IQGAP1 causes choroidal neovascularization by sustaining VEGFR2-mediated Rac1 activation Nicorandil regulates Bcl-2 family proteins and protects cardiac myocytes against hypoxia-induced apoptosis Role of TLR4/MyD88/NF-κB signaling pathway in coronary microembolization-induced myocardial injury prevented and treated with nicorandil Nicorandil modulated macrophages activation and polarization via NF-κb signaling pathway Potassium channel openers protect cardiac mitochondria by attenuating oxidant stress at reoxygenation Nicorandil improves post-ischemic myocardial dysfunction in association with opening the mitochondrial K(ATP) channels and decreasing hydroxyl radicals in isolated rat hearts Nicorandil attenuates the mitochondrial Ca2+ overload with accompanying depolarization of the mitochondrial membrane in the heart Benefit of nicorandil using an immunologic murine model of experimental colitis Pharmacological preconditioning with nicorandil and pioglitazone attenuates myocardial ischemia/reperfusion injury in rats The late phase of preconditioning Nicorandil improves post-fatigue tension in slow skeletal muscle fibers by modulating glutathione redox state Mitochondrial membrane permeability transition and cell death Tryptophane-kynurenine pathway in the remote ischemic conditioning mechanism Mitochondria and cell death: outer membrane permeabilization and beyond The permeability transition pore in cell death Melatonin prevents the dynamin-related protein 1-dependent mitochondrial fission and oxidative insult in the cortical neurons after 1-methyl-4-phenylpyridinium treatment Angiogenic responses in a 3D micro-engineered environment of primary endothelial cells and pericytes Exacerbated inflammatory signaling underlies aberrant response to BMP9 in pulmonary arterial hypertension lung endothelial cells Myocardial ischemiareperfusion injury: a neglected therapeutic target Protein S-glutathionlyation links energy metabolism to redox signaling in mitochondria Neuraminidase-1 promotes heart failure after ischemia/reperfusion injury by affecting cardiomyocytes and invading monocytes/macrophages Non-coding RNAs: emerging players in cardiomyocyte proliferation and cardiac regeneration Nicorandil affects mitochondrial respiratory chain function by increasing complex III activity and ROS production in skeletal muscle mitochondria Reactive oxygen species, but not Ca2+ overloading, trigger pH-and mitochondrial permeability transition-dependent death of adult rat myocytes after ischemia-reperfusion Free fatty acid receptor 4 activation protects against choroidal neovascularization in mice Nicorandil, a K(atp) channel opener, alleviates chronic renal injury by targeting podocytes and macrophages Mitochondrial fission controls DNA fragmentation by regulating endonuclease G Manipulation of death pathways in desminrelated cardiomyopathy Mechanisms involved in postconditioning protection of cardiomyocytes against acute reperfusion injury Matrix deformations around angiogenic sprouts correlate to sprout dynamics and suggest pulling activity Effect of intracoronary agents on the no-reflow phenomenon during primary percutaneous coronary intervention in patients with ST-elevation myocardial infarction: a network meta-analysis