cardiovascular-agents and Reperfusion-Injury

cardiovascular-agents has been researched along with Reperfusion-Injury* in 52 studies

Reviews

13 review(s) available for cardiovascular-agents and Reperfusion-Injury

ArticleYear
Histone Deacetylase Inhibitors: A Novel Strategy for Neuroprotection and Cardioprotection Following Ischemia/Reperfusion Injury.
    Journal of the American Heart Association, 2020, 06-02, Volume: 9, Issue:11

    Ischemia/reperfusion injury is a complex molecular cascade that causes deleterious cellular damage and organ dysfunction. Stroke, sudden cardiac arrest, and acute myocardial infarction are the most common causes of ischemia/reperfusion injury without effective pharmacologic therapies. Existing preclinical evidence suggests that histone deacetylase inhibitors may be an efficacious, affordable, and clinically feasible therapy that can improve neurologic and cardiac outcomes following ischemia/reperfusion injury. In this review, we discuss the pathophysiology and epigenetic modulations of ischemia/reperfusion injury and focus on the neuroprotective and cardioprotective effects of histone deacetylase inhibitors. We also summarize the protective effects of histone deacetylase inhibitors for other vital organs and highlight the key research priorities for their successful translation to the bedside.

    Topics: Animals; Brain; Cardiovascular Agents; Cerebrovascular Circulation; Coronary Circulation; Epigenesis, Genetic; Histone Deacetylase Inhibitors; Humans; Myocardial Reperfusion Injury; Myocardium; Neuroprotective Agents; Reperfusion Injury

2020
Preconditioning is hormesis part I: Documentation, dose-response features and mechanistic foundations.
    Pharmacological research, 2016, Volume: 110

    This article provides the first extensive documentation of the dose response features of pre- and postconditioning. Pre- and postconditioning studies with rigorous study designs, using multiple doses/concentrations along with refined dose/concentration spacing strategies, often display hormetic dose/concentration response relationships with considerable generality across biological model, inducing (i.e., conditioning) agent, challenging dose treatment, endpoint, and mechanism. Pre- and postconditioning hormesis dose/concentration-response relationships are reported for 154 diverse conditioning agents, affecting more than 550 dose/concentration responses, across a broad range of biological models and endpoints. The quantitative features of the pre- and postconditioning-induced protective responses are modest, typically being 30-60% greater than control values at maximum, findings that are consistent with a large body (>10,000) of hormetic dose/concentration responses not related to pre- and postconditioning. Regardless of the biological model, inducing agent, endpoint or mechanism, the quantitative features of hormetic dose/concentration responses are similar, suggesting that the magnitude of response is a measure of biological plasticity. This paper also provides the first documentation that hormetic effects account for preconditioning induced early (1-3h) and delayed (12-72h) windows of protection. These findings indicate that pre- and postconditioning are specific types of hormesis.

    Topics: Animals; Brain; Brain Ischemia; Cardiovascular Agents; Cytoprotection; Disease Models, Animal; Heart; Hormesis; Humans; Myocardial Reperfusion Injury; Myocardium; Neuroprotective Agents; Reperfusion Injury; Time Factors; Treatment Outcome

2016
Medical Functions of Hydrogen Sulfide.
    Advances in clinical chemistry, 2016, Volume: 74

    Hydrogen sulfide (H(2)S) is a gasomediator synthesized from L- and D-cysteine in various tissues. It is involved in a number of physiological and pathological processes. H(2)S exhibits antiatherosclerotic, vasodilator, and proangiogenic properties, and protects the kidney and heart from damage following ischemia/reperfusion injury. H(2)S donors may be natural or synthetic, and may be used for the safe treatment of a wide range of diseases. This review article summarizes the current state of knowledge of the therapeutic function of H(2)S.

    Topics: Angiogenesis Inducing Agents; Cardiovascular Agents; Cysteine; Diclofenac; Disulfides; Heart; Humans; Hydrogen Sulfide; Kidney; Levodopa; Prostaglandins F, Synthetic; Reperfusion Injury; Sulfides; Thioctic Acid; Thiones; Vasodilator Agents

2016
Ischemia-reperfusion injury: influencing the microcirculatory and cellular environment.
    Annals of plastic surgery, 2014, Volume: 72, Issue:2

    Ischemia-reperfusion injury forms the basis of tissue damage and cellular apoptosis in many pathologic and traumatic processes. The tissue damage follows a natural progression of cellular and metabolic events initiated by an ischemic episode. Ischemia causes intracellular/extracellular changes principally resulting in increased intracellular calcium, pH changes, and adenosine triphosphate depletion that end in cell death if the process is not interrupted. This interruption takes the form of reperfusion, characterized by a "flushing" of tissues with toxic metabolites, principally reactive oxygen species. The immediate effect is mitochondrial pore permeability, complement activation, cytochrome release, cytokine activation, inflammation, edema, neutrophil platelet adhesion, capillary plugging, and thrombosis. This sets the stage for the long recognized "no-reflow" phenomenon and progressive tissue death. Current recognition of cellular "cross-talk" and molecular events have introduced new logical strategies to sequentially combat the events occurring in relation to ischemia-reperfusion injury. These include mechanical preconditioning and pharmacological preconditioning and postconditioning strategies. It is likely that success in reversing or limiting tissue damage will be found in a sequential multitargeted approach using a combination of these strategies-clinical trials in this regard are sorely needed.

    Topics: Cardiovascular Agents; Cell Adhesion Molecules; Endothelium-Dependent Relaxing Factors; Free Radical Scavengers; Humans; Hydrogen Sulfide; Hypothermia, Induced; Ischemia; Ischemic Postconditioning; Ischemic Preconditioning; Microcirculation; Nitric Oxide; Reperfusion Injury

2014
Targeting adenosine receptors in the development of cardiovascular therapeutics.
    Expert review of clinical pharmacology, 2012, Volume: 5, Issue:2

    Adenosine receptor stimulation has negative inotropic and dromotropic actions, reduces cardiac ischemia-reperfusion injury and remodeling, and prevents cardiac arrhythmias. In the vasculature, adenosine modulates vascular tone, reduces infiltration of inflammatory cells and generation of foam cells, and may prevent the development of atherosclerosis as a result. Modulation of insulin sensitivity may further add to the anti-atherosclerotic properties of adenosine signaling. In the kidney, adenosine plays an important role in tubuloglomerular feedback and modulates tubular sodium reabsorption. The challenge is to take advantage of the beneficial actions of adenosine signaling while preventing its potential adverse effects, such as salt retention and sympathoexcitation. Drugs that interfere with adenosine formation and elimination or drugs that allosterically enhance specific adenosine receptors seem to be most promising to meet this challenge.

    Topics: Adenosine; Animals; Arrhythmias, Cardiac; Atherosclerosis; Cardiovascular Agents; Cardiovascular Diseases; Humans; Insulin; Kidney; Receptors, Purinergic P1; Reperfusion Injury; Signal Transduction

2012
Soluble epoxide hydrolase: a promising therapeutic target for cardiovascular diseases.
    Die Pharmazie, 2011, Volume: 66, Issue:3

    Epoxyeicosatrienoic acids (EETs) are cytochrome P450 (CYP450) products of arachidonic acid and EETs are endogenous lipid mediators synthesized by the vascular endothelium which perform important biological functions, including vasodilation, anti-inflammation, antimigratory, and cellular signaling regulations. However, EETs are rapidly degraded by soluble epoxide hydrolase (sEH) to the corresponding diols: dihydroxyeicosatrienoic acids (DHETs), which have little active in causing vasorelaxation. A number of studies have supported that the inhibition of sEH (sEHIs) had cardiovascular protective effects in hypertension, cardiac hypertrophy, atherosclerosis, ischemia-reperfusion injury, and ischemic stroke. Moreover, sEHIs could slow the progression of inflammation, protect end-organ damage and prevent ischemic events, also, attenuate endothelial dysfunction, suggesting that the pharmacological blockade of sEH might provide a broad and novel avenue for the treatment of many cardiovascular diseases.

    Topics: Animals; Brain Ischemia; Cardiomegaly; Cardiovascular Agents; Cardiovascular Diseases; Enzyme Inhibitors; Epoxide Hydrolases; Humans; Hypertension; Polymorphism, Genetic; Reperfusion Injury; Risk; Stroke

2011
Ex vivo and in vivo approaches to study mechanisms of cardioprotection targeting ischemia/reperfusion (i/r) injury: useful techniques for cardiovascular drug discovery.
    Current drug discovery technologies, 2008, Volume: 5, Issue:4

    The last few decades have seen significant advancement in the therapy of Ischemic Heart Diseases (IHD). This is a direct outcome of the increasing knowledge of the molecular mechanisms involved during an ischemic insult of the myocardium. Even then there is still a major unmet need for better strategies or drug therapies to reduce ventricular remodeling and improve post-ischemic myocardial function. The ex-vivo isolated working heart model and the in vivo myocardial infarction model are the best known techniques to elucidate the contribution of a drug therapy to confer cardioprotection in the event of an ischemic insult/reperfusion. Our review aims to provide an insight into the state of the art techniques that lay the foundations for cardiovascular drug discovery and present the prospects for further development from a preclinical perspective. The first section of the review provides an overview of the rat/mouse ex-vivo and in vivo models of myocardial ischemia. The following section will then present various applications of these clinically relevant models in characterizing cardiac functions, screening for drugs and identifying the drug induced changes in cardiac functions. Finally the role of these models in drug development is discussed with respect to functional relevance of drug treatment on heart rate, aortic flow, coronary flow, infarct size and the mechanisms by which these drugs promote myocardial protection. This review may serve as a basic knowledge for researchers who intend to study the efficacy of a drug in the treatment of ischemic heart diseases.

    Topics: Animals; Cardiotonic Agents; Cardiovascular Agents; Disease Models, Animal; Drug Discovery; In Vitro Techniques; Mice; Myocardial Ischemia; Perfusion; Rats; Reperfusion Injury; Technology, Pharmaceutical; Ventricular Remodeling

2008
Ameliorating effects of compounds derived from Salvia miltiorrhiza root extract on microcirculatory disturbance and target organ injury by ischemia and reperfusion.
    Pharmacology & therapeutics, 2008, Volume: 117, Issue:2

    Ischemia and reperfusion (I/R) exerts multiple insults in microcirculation, frequently accompanied by endothelial cell injury, enhanced adhesion of leukocytes, macromolecular efflux, production of oxygen free radicals, and mast cell degranulation. Since the microcirculatory disturbance results in injury of organ involved, protection of organ after I/R is of great importance in clinic. Salvia miltiorrhiza root has long been used in Asian countries for clinical treatment of various microcirculatory disturbance-related diseases. This herbal drug contains many active water-soluble compounds, including protocatechuic aldehyde (PAl), 3,4-dihydroxyphenyl lactic acid (DLA) and salvianolic acid B (SalB). These compounds, as well as water-soluble fraction of S. miltiorrhiza root extract (SMRE), have an ability to scavenge peroxides and are able to inhibit the expression of adhesion molecules in vascular endothelium and leukocytes. Moreover, lipophilic compounds of SMRE also prevent the development of vascular damage; NADPH oxidase and platelet aggregation are inhibited by tanshinone IIA and tanshinone IIB, respectively, and the mast cell degranulation is blunted by cryptotanshinone and 15,16-dihydrotanshinone I. Thus, the water-soluble and lipophilic compounds of SMRE appear to improve the I/R-induced vascular damage multifactorially and synergically. This review will summarize the ameliorating effect of compounds derived from SMRE on microcirculatory disturbance and target organ injury after I/R and will provide a new perspective on remedy with multiple drugs.

    Topics: Animals; Blood Platelets; Brain; Cardiovascular Agents; Cell Degranulation; Cerebrovascular Circulation; Coronary Circulation; Cytokines; Drugs, Chinese Herbal; Endothelial Cells; Humans; Kidney; Leukocytes; Liver; Liver Circulation; Lung; Mast Cells; Microcirculation; Molecular Structure; Myocardial Reperfusion Injury; Plant Extracts; Plant Roots; Renal Circulation; Reperfusion Injury; Salvia miltiorrhiza

2008
Potential role and mechanisms of subcellular remodeling in cardiac dysfunction due to ischemic heart disease.
    Journal of cardiovascular medicine (Hagerstown, Md.), 2007, Volume: 8, Issue:4

    Several studies have revealed varying degrees of changes in sarcoplasmic reticular and myofibrillar activities, protein content, gene expression and intracellular Ca-handling during cardiac dysfunction due to ischemia-reperfusion (I/R); however, relatively little is known about the sarcolemmal and mitochondrial alterations, as well as their mechanisms in the I/R hearts. Because I/R is associated with oxidative stress and intracellular Ca-overload, it has been indicated that changes in subcellular activities, protein content and gene expression due to I/R are related to both oxidative stress and Ca-overload. Intracellular Ca-overload appears to induce changes in subcellular activities, protein contents and gene expression (subcellular remodeling) by activation of proteases and phospholipases, as well as by affecting the genetic apparatus, whereas oxidative stress is considered to cause oxidation of functional groups of different subcellular proteins in addition to modifying the genetic machinery. Ischemic preconditioning, which is known to depress the development of both intracellular Ca-overload and oxidative stress due to I/R, was observed to attenuate the I/R-induced subcellular remodeling and improve cardiac performance. It is suggested that a combination therapy with antioxidants and interventions, which reduce the development of intracellular Ca-overload, may improve cardiac function by preventing or attenuating the occurrence of subcellular remodeling due to ischemic heart disease. It is proposed that defects in the activities of subcellular organelles may serve as underlying mechanisms for I/R-induced cardiac dysfunction under acute conditions, whereas subcellular remodeling due to alterations in gene expression may explain the impaired cardiac performance under chronic conditions of I/R.

    Topics: Animals; Antioxidants; Calcium; Cardiovascular Agents; Humans; Ischemic Preconditioning, Myocardial; Mitochondria, Heart; Myocardial Ischemia; Myocardium; Oxidative Stress; Reperfusion Injury; Sarcolemma; Ventricular Function; Ventricular Remodeling

2007
Antiplatelet and antileukocyte effects of cardiovascular, immunomodulatory and chemotherapeutic drugs.
    Cardiovascular & hematological agents in medicinal chemistry, 2006, Volume: 4, Issue:3

    In vitro and ex vivo interactions of betaadrenoceptor blocking drugs, antihistamines and chloroquine with blood platelets and polymorphonuclear leukocytes resulted in different alterations of regulatory functions of these blood cells. Inhibition of platelet aggregation, arachidonate regulatory pathway, 5-hydroxytryptamine transportation, removal of platelet membrane receptors, inhibition of second messenger pathways at subcellular level and suppression of phagocytosis are indicative of nonreceptor rather than specific receptor interactions. Binding of drugs with biomembranes is reversible depending on the ionic charge of the molecule and hydrophobicity of the bilayer, partition coefficient, pH and pKa of the amphiphilic molecules and other physico-chemical properties of amphiphilic drugs. Alterations in the drug molecule structure alters the drug-phospholipid binding profile. Any change in the metabolism of membrane phospholipids directly or indirectly influences one or more of the important components of the phospholipid-signalling pathway. In addition to changes in phospholipase A, C and D activities, protein kinase C, calmodulin-phosphodiesterase, Ca2+,Mg2+-ATPase, Na+,K+-ATPase and other messengers were found to be changed in cells and tissue after cationic amphiphilic drug (CAD) administration. Although not much has been understood of the mechanism by which some CAD affect immune functions, there are good reasons to suggest that these effects might occur. CADs share sufficient similarities in their structure even though they come from diverse pharmacological classes. CADs affect ion transport, immune functions, tumour growth, serotonin metabolism and several other functions in the body. Extensive therapeutic use and associated side effects have generated a great deal of interest in understanding the nonreceptor interactions with CADs.

    Topics: Adjuvants, Immunologic; Animals; Antineoplastic Agents; Blood Platelets; Cardiovascular Agents; Chloroquine; Histamine H1 Antagonists; Humans; Inflammation; Leukocytes; Reperfusion Injury; Thromboembolism

2006
Vascular and anti-oxidant actions of flavonols and flavones.
    Clinical and experimental pharmacology & physiology, 2004, Volume: 31, Issue:11

    1. Flavonols and flavones are plant-derived polyphenolic compounds that are commonly consumed in the diet. Epidemiological studies indicating that high dietary intake of flavonols reduces the risk of mortality due to coronary heart disease have provoked interest in the mechanism of this cardioprotective effect. 2. We have investigated the structure-activity relationships of a range of flavonols and flavones with regard to their vascular relaxant and anti-oxidant activity. In rat isolated thoracic aorta, the synthetic flavonol 3',4'-dihydroxyflavonol (DiOHF) was found to be a significantly more potent vasorelaxant than the naturally occurring compounds chrysin, apigenin, luteolin, quercetin and fisetin. Similarly, DiOHF was significantly more potent than those compounds in the inhibition of calcium-induced contraction of the rat aorta. 3. 3',4'-Dihydroxyflavonol was also found to significantly inhibit superoxide radical generation in a cell-free system in the presence of xanthine/xanthine oxidase or by rat isolated aorta in the presence of NADPH. In the presence of oxidant stress generated by pyrogallol or xanthine/xanthine oxidase, endothelium-dependent relaxation of rat aortic rings was impaired. 3',4'-Dihydroxyflavonol was able to significantly improve endothelium-dependent relaxation in the presence of those oxygen radical generators. 4. In addition, DiOHF was found to significantly improve dilatation in the rat hindquarters vasculature after exposure to ischaemia and reperfusion. 3',4'-Dihydroxyflavonol was found to be equally effective whether applied before ischaemia or during ischaemia just before reperfusion. 5. In conclusion, DiOHF is an effective vasodilator and anti-oxidant that is able to prevent vascular reperfusion injury. We suggest that DiOHF may be useful as an adjunct to thrombolytic therapy in the management of reperfusion injury.

    Topics: Animals; Antioxidants; Cardiovascular Agents; Flavonols; Free Radical Scavengers; Humans; Muscle Relaxation; Muscle, Smooth, Vascular; Plants, Medicinal; Reperfusion Injury

2004
Cardiac arrest.
    Emergency medicine clinics of North America, 1996, Volume: 14, Issue:1

    This article reviews the critical resuscitations necessary during prehospital and emergency department treatment of cardiac arrest. Standard therapy for cardiac arrest rhythms is presented. Novel pharmacologic agents, types of cardiopulmonary resuscitation, and circulatory-assist devices are discussed.

    Topics: Adult; Algorithms; Arrhythmias, Cardiac; Cardiovascular Agents; Child; Emergency Medical Services; Heart Arrest; Hemodynamics; Humans; Intubation, Intratracheal; Monitoring, Physiologic; Physical Examination; Reperfusion Injury; Respiration; Respiration, Artificial; Resuscitation

1996
Reactive oxygen species in developmental toxicity: review and hypothesis.
    Teratology, 1996, Volume: 53, Issue:3

    Topics: Abnormalities, Drug-Induced; Animals; Antioxidants; Cardiovascular Agents; Constriction; Embryo, Mammalian; Female; Humans; Hypoxia; Organ Culture Techniques; Pregnancy; Pregnancy Outcome; Reactive Oxygen Species; Reperfusion Injury

1996

Trials

1 trial(s) available for cardiovascular-agents and Reperfusion-Injury

ArticleYear
L-arginine and tetrahydrobiopterin protects against ischemia/reperfusion-induced endothelial dysfunction in patients with type 2 diabetes mellitus and coronary artery disease.
    Atherosclerosis, 2009, Volume: 204, Issue:1

    Diminished levels of L-arginine and endothelial nitric oxide synthase (eNOS) uncoupling through deficiency of tetrahydrobiopterin (BH(4)) may contribute to endothelial dysfunction. We investigated the effect of L-arginine and BH(4) administration on ischemia-reperfusion (I/R)-induced endothelial dysfunction in patients with type 2 diabetes and coronary artery disease (CAD). Forearm blood flow was measured by venous occlusion plethysmography in 12 patients with type 2 diabetes or impaired glucose tolerance and CAD. Forearm ischemia was induced for 20 min, followed by 60 min of reperfusion. The patients received a 15 min intra-brachial infusion of L-arginine (20 mg/min) and BH(4) (500 microg/min) or 0.9% saline starting at 15 min of ischemia on two separate study occasions. Compared with pre-ischemia the endothelium-dependent vasodilatation (EDV) induced by acetylcholine was significantly reduced at 15 and 30 min of reperfusion when saline was infused (P<0.001), but not following L-arginine and BH(4) infusion. EDV was also significantly less reduced at 15 and 30 min of reperfusion following L-arginine and BH(4) infusion, compared to saline infusion (P<0.02). Endothelium-independent vasodilatation (EIDV) induced by nitroprusside was unaffected by I/R. Venous total biopterin levels in the infused arm increased from 37+/-7 at baseline to 6644+/-1240 nmol/l during infusion of L-arginine and BH(4) (P<0.0001), whereas there was no difference in biopterin levels during saline infusion. In conclusion L-arginine and BH(4) supplementation reduces I/R-induced endothelial dysfunction, a finding which may represent a novel treatment strategy to limit I/R injury in patients with type 2 diabetes and CAD.

    Topics: Aged; Arginine; Biopterins; Blood Flow Velocity; Cardiovascular Agents; Coronary Artery Disease; Cross-Over Studies; Diabetes Mellitus, Type 2; Dose-Response Relationship, Drug; Drug Therapy, Combination; Endothelium, Vascular; Female; Forearm; Humans; Infusions, Intra-Arterial; Male; Regional Blood Flow; Reperfusion Injury; Treatment Outcome; Vasodilation; Vasodilator Agents

2009

Other Studies

38 other study(ies) available for cardiovascular-agents and Reperfusion-Injury

ArticleYear
Combined levosimendan and Sacubitril/Valsartan markedly protected the heart and kidney against cardiorenal syndrome in rat.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2022, Volume: 148

    Cardiorenal syndrome (CRS) remains the leading cause of death in hospitalized patients for all disease entities. Sacubitril/Valsartan (Sac/Val) therapy has been proved to improve prognostic outcome in patients with heart failure or chronic kidney disease. This study tested the hypothesis that combined levosimendan and Sac/Val was superior to just one therapy on protecting the heart and kidney against simultaneous heart and kidney ischemia (I) (for 50-min)-reperfusion (R) (for 7-days) (i.e., double IR) injury (defined as CRS).. Adult-male Spraque-Dawley rats (n = 40) were equally categorized into group 1 (sham-operated control), group 2 (double IR), group 3 [double IR+levosimendan (10 mg/kg by intra-peritoneum administration at 30 min/followed by days 1-5 once daily after IR procedure)], group 4 [double IR+Sac/Val (10 mg/kg, orally at 30 min/followed by days 1-5 twice daily after IR procedure)], and group 5 (double IR+Sac/Val+levosimendan). By day 7 after double-IR, the left-ventricular-ejection fraction (LVEF)/left-ventricular-fraction-shortening (LVFS) were highest in group 1, lowest in group 2 and significantly higher in group 5 than in groups 3/4, but they showed no difference between groups 3/4, whereas the circulatory heart-failure (brain-natriuretic peptide)/proinflammatory (suppression of tumorigenicity-2) biomarkers, blood-urea-nitrogen/creatinine and ratio of urine protein to creatinine (all p < 0.0001) exhibited an opposite pattern of LVEF among the groups. The protein expressions of inflammatory (tumor necrosis factor-α/interleukin-1ß/matrix metalloproteinase-9)/oxidative-stress (NOX-1/NOX-2/NOX-4)/apoptotic (mitochondrial-Bax/caspase-3/poly-(ADP-ribose)-polymerase)/fibrotic (Smad3/transforming growth factor-ß)/mitochondrial-damaged (cytosolic-cytochrome-C)/myocardial-hypertrophic (ß-MHC) biomarkers in LV myocardium exhibited an opposite pattern of LVEF among the groups (all p < 0.0001). The cellular expressions of inflammatory (CD68)/DNA-damaged (γ-H2AX) biomarkers and infarct/fibrotic areas in LV myocardium and kidney displayed an opposite pattern of LVEF among the groups (all p < 0.0001).. Combined levosimendan and Sac/Val was superior to merely one therapy on protecting the heart and kidney as well as preserving their functions against double IR injury.

    Topics: Aminobutyrates; Animals; Apoptosis; Biphenyl Compounds; Cardio-Renal Syndrome; Cardiovascular Agents; Drug Combinations; Fibrosis; Humans; Inflammation; Kidney; Male; Myocardium; Oxidative Stress; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Simendan; Stroke Volume; Valsartan; Ventricular Function, Left

2022
Bretschneider Solution and Two Antianginal Drugs Protect Peripheral Tissue in an Animal Model of Hemorrhagic Shock.
    Journal of cardiovascular pharmacology, 2022, 06-01, Volume: 79, Issue:6

    Shock and subsequent resuscitation provoke ischemia-reperfusion injury. Trimetazidine (TMZ), allopurinol (ALO), and histidine-tryptophan-ketoglutarate (HTK) solution, can protect from ischemia-reperfusion injury in chronic coronary syndromes and in transplantation. The objective of the current study is to compare, in a hemorrhagic shock and standard resuscitation animal model, organ damage parameters between placebo and treatment with TMZ, ALO, or HTK. Shock was induced in Wistar rats by controlled arterial bleeding, maintaining mean arterial pressure between 38 and 42 mm Hg for 60 minutes; then, drawn blood was reinfused. Animals were divided into: Sham (n = 4), Control (n = 6), TMZ (n = 7), ALO (n = 9), and HTK (n = 7). At the end of the experiment, animals were sacrificed and tissue harvested. TMZ, ALO and HTK decreased histopathologic damage in heart [Control: 1.72 (1.7-1.77); TMZ: 1.75 (1.72-1.79); ALO: 1.75 (1.74-1.8); HTK: 1.82 (1.78-1.85); all P < 0.05], kidney [Control: 3 (2-3); TMZ: 1 (1-2); ALO: 1 (1-1); HTK: 1(1-1); all P < 0.05] and intestine [Control: 3 (2-3); TMZ: 1 (1-2); ALO: 1 (1-1); HTK: 1 (0-2); all P < 0.05]. Also, treatment with TMZ, ALO, and HTK increased immunohistochemical expression of thioredoxin-1 in heart [Control: 6.6 (5.6-7.4); TMZ: 9.5 (8.1-9.7); ALO: 9.1 (8.4-10.2); HTK: 14.2 (12.6-15); all P < 0.05]; and kidney [Control: 4.6 (4-5.1); TMZ: 9.7 (9.3-9.9); ALO: 9.6 (9-9.9); HTK: 16.7 (16.1-17); all P < 0.05]. In an experimental model of hemorrhagic shock, TMZ, ALO, and HTK solution attenuated cell damage in multiple parenchyma and increased antioxidant defenses.

    Topics: Allopurinol; Animals; Cardiovascular Agents; Disease Models, Animal; Glucose; Glutathione; Insulin; Mannitol; Organ Preservation Solutions; Potassium Chloride; Procaine; Rats; Rats, Wistar; Reperfusion Injury; Shock, Hemorrhagic

2022
Transcriptomic Hallmarks of Ischemia-Reperfusion Injury.
    Cells, 2021, 07-20, Volume: 10, Issue:7

    Ischemia reperfusion injury (IRI) is associated with a broad array of life-threatening medical conditions including myocardial infarct, cerebral stroke, and organ transplant. Although the pathobiology and clinical manifestations of IRI are well reviewed by previous publications, IRI-related transcriptomic alterations are less studied. This study aimed to reveal a transcriptomic hallmark for IRI by using the RNA-sequencing data provided by several studies on non-human preclinical experimental models. In this regard, we focused on the transcriptional responses of IRI in an acute time-point up to 48 h. We compiled a list of highly reported genes in the current literature that are affected in the context of IRI. We conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses and found many of the up-regulated genes to be involved in cell survival, cell surface signaling, response to oxidative stress, and inflammatory response, while down-regulated genes were predominantly involved in ion transport. Furthermore, by GO analysis, we found that multiple inflammatory and stress response processes were affected after IRI. Tumor necrosis factor alpha (TNF) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways were also highlighted in the Kyoto Encyclopedia of Genes and Genomes enrichment analysis. In the last section, we discuss the treatment approaches and their efficacy for IRI by comparing RNA sequencing data from therapeutic interventions with the results of our cross-comparison of differentially expressed genes and pathways across IRI.

    Topics: Animals; Cardiovascular Agents; Cell- and Tissue-Based Therapy; Cerebral Cortex; Gene Expression Profiling; Gene Expression Regulation; Gene Ontology; Gene Regulatory Networks; Humans; Ischemic Postconditioning; Ischemic Preconditioning; Kidney; Liver; Metabolic Networks and Pathways; Mice; Molecular Sequence Annotation; Myocardium; NF-kappa B; Rats; Reperfusion Injury; Sequence Analysis, RNA; Signal Transduction; Spinal Cord; Transcriptome; Tumor Necrosis Factor-alpha

2021
Pediatric Post-Cardiac Arrest Care: A Scientific Statement From the American Heart Association.
    Circulation, 2019, 08-06, Volume: 140, Issue:6

    Successful resuscitation from cardiac arrest results in a post-cardiac arrest syndrome, which can evolve in the days to weeks after return of sustained circulation. The components of post-cardiac arrest syndrome are brain injury, myocardial dysfunction, systemic ischemia/reperfusion response, and persistent precipitating pathophysiology. Pediatric post-cardiac arrest care focuses on anticipating, identifying, and treating this complex physiology to improve survival and neurological outcomes. This scientific statement on post-cardiac arrest care is the result of a consensus process that included pediatric and adult emergency medicine, critical care, cardiac critical care, cardiology, neurology, and nursing specialists who analyzed the past 20 years of pediatric cardiac arrest, adult cardiac arrest, and pediatric critical illness peer-reviewed published literature. The statement summarizes the epidemiology, pathophysiology, management, and prognostication after return of sustained circulation after cardiac arrest, and it provides consensus on the current evidence supporting elements of pediatric post-cardiac arrest care.

    Topics: Acute Kidney Injury; Adrenal Insufficiency; Anticonvulsants; Brain Damage, Chronic; Cardiomyopathies; Cardiopulmonary Resuscitation; Cardiovascular Agents; Child; Combined Modality Therapy; Fluid Therapy; Glucose Metabolism Disorders; Heart Arrest; Humans; Hypnotics and Sedatives; Hypothermia, Induced; Hypoxia-Ischemia, Brain; Infections; Inflammation; Monitoring, Physiologic; Multiple Organ Failure; Neuromuscular Blocking Agents; Oxygen Inhalation Therapy; Prognosis; Reperfusion Injury; Respiratory Therapy; Time Factors

2019
Protective effects of pentoxifylline in small intestine after ischemia-reperfusion.
    The Journal of international medical research, 2018, Volume: 46, Issue:10

    Objective This study was performed to determine the healing effects of pentoxifylline on molecular responses and protection against severe ischemic damage in the small intestine. Methods Thirty-six Wistar albino rats were divided into six groups. The superior mesenteric artery was clamped for 120 minutes, and reperfusion was performed for 60 minutes. Saline (0.4 mL), pentoxifylline (1 mg/kg), and pentoxifylline (10 mg/kg) were intraperitoneally administered to the rats in the C

    Topics: Animals; Cardiovascular Agents; Disease Models, Animal; Hematologic Agents; Infusions, Parenteral; Intestine, Small; Ischemia; Male; Pentoxifylline; Protective Agents; Rats; Rats, Wistar; Reperfusion Injury; Wound Healing

2018
Protective effect of mangiferin on myocardial ischemia-reperfusion injury in streptozotocin-induced diabetic rats: role of AGE-RAGE/MAPK pathways.
    Scientific reports, 2017, 02-09, Volume: 7

    Topics: Animals; Cardiovascular Agents; Diabetes Mellitus, Experimental; Disease Models, Animal; Glycation End Products, Advanced; Male; MAP Kinase Signaling System; Myocardial Ischemia; Rats, Wistar; Receptor for Advanced Glycation End Products; Reperfusion Injury; Signal Transduction; Treatment Outcome; Xanthones

2017
Use of dextran sulfate in tourniquet-induced skeletal muscle reperfusion injury.
    The Journal of surgical research, 2014, Volume: 187, Issue:1

    Lower extremity ischemia-reperfusion injury (IRI)-prolonged ischemia and the subsequent restoration of circulation-may result from thrombotic occlusion, embolism, trauma, or tourniquet application in surgery. The aim of this study was to assess the effect of low-molecular-weight dextran sulfate (DXS) on skeletal muscle IRI.. Rats were subjected to 3 h of ischemia and 2 or 24 h of reperfusion. To induce ischemia the femoral artery was clamped and a tourniquet placed under the maintenance of the venous return. DXS was injected systemically 10 min before reperfusion. Muscle and lung tissue samples were analyzed for deposition of immunoglobulin M (IgM), IgG, C1q, C3b/c, fibrin, and expression of vascular endothelial-cadherin and bradykinin receptors b1 and b2.. Antibody deposition in reperfused legs was reduced by DXS after 2 h (P < 0.001, IgM and IgG) and 24 h (P < 0.001, IgM), C3b/c deposition was reduced in muscle and lung tissue (P < 0.001), whereas C1q deposition was reduced only in muscle (P < 0.05). DXS reduced fibrin deposits in contralateral legs after 24 h of reperfusion but did not reduce edema in muscle and lung tissue or improve muscle viability. Bradykinin receptor b1 and vascular endothelial-cadherin expression were increased in lung tissue after 24 h of reperfusion in DXS-treated and non-treated rats but bradykinin receptor b2 was not affected by IRI.. In contrast to studies in myocardial infarction, DXS did not reduce IRI in this model. Neither edema formation nor viability was improved, whereas deposition of complement and coagulation components was significantly reduced. Our data suggest that skeletal muscle IRI may not be caused by the complement or coagulation alone, but the kinin system may play an important role.

    Topics: Animals; Antigens, CD; Cadherins; Cardiovascular Agents; Complement C1q; Complement C3b; Dextran Sulfate; Disease Models, Animal; Edema; Femoral Artery; Fibrin; Hindlimb; Immunoglobulin G; Immunoglobulin M; Male; Muscle, Skeletal; Peptide Fragments; Rats; Rats, Wistar; Receptors, Bradykinin; Reperfusion Injury; Tourniquets

2014
The fibrin-derived peptide bβ15-42 attenuates liver damage in a rat model of liver ischemia/reperfusion injury.
    Shock (Augusta, Ga.), 2013, Volume: 39, Issue:4

    The inflammatory response after liver ischemia/reperfusion (I/R) contributes to increased risk of liver failure after liver surgery. Strategies aimed to preventing inflammation could be beneficial in reducing liver I/R injury. Recent studies have demonstrated that peptide Bβ15-42 is able to decrease the injury of I/R in heart and kidney by inhibition of leukocyte migration and preserving endothelial barrier function. Prompted by these results, we hypothesized that Bβ15-42 could also possess anti-inflammatory abilities to protect from or reduce hepatic I/R injury. Therefore, in this study, we aimed to evaluate the effects of Bβ15-42 in a model of liver I/R injury in rats. Rats were treated with Bβ15-42 at initiation of reperfusion and 2 h thereafter. Rats were killed at 0.5, 6, 24, and 48 h after reperfusion. Hepatic mRNA levels of fibrinogen-α (Fgα), Fgβ, Fgγ were significantly increased after I/R. Treatment with Fg-derived Bβ15-42 ameliorated liver I/R injury, as indicated by lower serum aminotransferase levels and fewer I/R-associated histopathologic changes. Bβ15-42 treatment decreased leukocyte infiltration and expression of hepatic inflammatory cytokines. Moreover, Bβ15-42 significantly reduced high-mobility group box 1 release and altered mitogen-activated protein kinase activation. In conclusion, Bβ15-42 treatment protected against liver warm I/R injury. The mechanism of protective action of Bβ15-42 seemed to involve its ability to reduce hepatic inflammatory response through preventing high-mobility group box 1 release and altering mitogen-activated protein kinase activation.

    Topics: Animals; Biomarkers; Cardiovascular Agents; Cytokines; Fibrin Fibrinogen Degradation Products; HMGB1 Protein; Liver; Male; MAP Kinase Signaling System; Neutrophil Infiltration; Peptide Fragments; Rats; Rats, Inbred Lew; Reperfusion Injury; Transaminases

2013
The effects of adenocaine (adenosine and lidocaine) on early post-resuscitation cardiac and neurological dysfunction in a porcine model of cardiac arrest.
    Resuscitation, 2013, Volume: 84, Issue:11

    Return of spontaneous circulation (ROSC) elicits ischaemia/reperfusion injury and myocardial dysfunction. The combination of adenosine and lidocaine (AL, adenocaine) has been shown to (1) inhibit neutrophil inflammatory activation and (2) improve left ventricular function after ischaemia. We hypothesized that resuscitation with adenocaine during early moments of cardiopulmonary resuscitation (CPR) attenuates leucocyte oxidant generation and myocardial dysfunction.. Pigs were randomized to: (1) sham (n=7), (2) cardiac arrest (CA; n=16), or 3) cardiac arrest+adenocaine (CA+AL; n=12). After 7 min of electrically induced ventricular fibrillation, start of CPR was followed by infusion of saline (CA) or adenocaine (CA+AL) for 6 min. Haemodynamics, cardiodynamics (pressure-volume loops) and leucocyte superoxide anion generation were assessed. Neurological function was evaluated after 24h by histology and neurological deficit score (0=normal; 500=brain dead).. Rate of ROSC was comparable between groups: CA group 11/16 and CA+AL group 7/12 p=0.57). Cardiac index transiently increased after ROSC in both groups. Left ventricular dysfunction demonstrated by a rightward shift of the intercept of end-systolic pressure-volume relations in CA was avoided in the CA+AL group. Leucocyte superoxide anion generation 2h after ROSC was significantly attenuated in the CA+AL group compared to the CA group. Neurological deficit scores [CA: median: 17.5(IQR:0-75) and CA+AL: 35(IQR:15-150)] and histopathological damage were comparable in both groups (p=0.37).. Infusion of adenocaine during early resuscitation from CA significantly improved early post-resuscitation cardiac function and attenuated leucocyte superoxide anion generation, without a change in post-ROSC neurological function. (IACUC protocol number 023-2009).

    Topics: Adenosine; Animals; Brain Death; Cardiopulmonary Resuscitation; Cardiovascular Agents; Coronary Angiography; Disease Models, Animal; Drug Combinations; Heart Arrest; Hemodynamics; Lidocaine; Oxygen; Random Allocation; Reperfusion Injury; Survival Rate; Swine

2013
An experimental model of aortic surgery and the effect of Iloprost on lung injury.
    Scandinavian cardiovascular journal : SCJ, 2013, Volume: 47, Issue:4

    Ischemia/reperfusion (I/R) damage of the lung is a frequently encountered complication following aortic surgery. The aim of the present study is to investigate the histopathological effects of Iloprost on pulmonary damage developed after I/R.. Twenty-four Sprague-Dawley rats were randomly divided into 3 groups. In the control group, aortas were not clamped. In the I/R group, aortas were occluded, and after 1 h of ischemia, clamps were removed. After 2 h of reperfusion period, lungs of the rats were extracted. In the I/R + Iloprost group after 1 h of ischemia, Iloprost infusion was initiated, and maintained for the duration of 2 h reperfusion period. For histopathological scoring, density of polymorphonuclear leucocytes, congestion, interstitial edema, and bleeding were semiquantitatively evaluated, and histopathological changes were scored.. In the I/R group, multifocal-marked histopathological changes in 5 (62.5%), and multifocal-moderate histopathological changes in 3 (37.5%) rats were detected. In the I/R + Iloprost group, multifocal-moderate histopathological changes in 4 (50%), and multifocal-mild changes in 4 (50%) rats were detected.. In the experimental rat model, administration of Iloprost has been shown to have preventive effects for pulmonary damage occurring after I/R generated by infrarenal aortic occlusion.

    Topics: Animals; Aorta; Cardiovascular Agents; Cytoprotection; Disease Models, Animal; Drug Administration Schedule; Female; Iloprost; Infusions, Intravenous; Lung Injury; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Time Factors

2013
Synergistic protective effect of astragaloside IV-tetramethylpyrazine against cerebral ischemic-reperfusion injury induced by transient focal ischemia.
    Journal of ethnopharmacology, 2012, Mar-06, Volume: 140, Issue:1

    Astragaloside IV and tetramethylpyrazine have been extensively used in the cardio-cerbrovascular diseases of medicine as a chief ingredient of glycoside or alkaloid formulations for the treatment of stroke and myocardial ischemia diseases.. To investigate the effects of astragaloside IV (ASG IV) and tetramethylpyrazine (TMPZ) on cerebral ischemia-reperfusion (IR) injury model in rat model.. Rats were randomly divided into the following five groups: sham group, IR group and treatment group including ASG IV, ASG IV-TMPZ and nimodipine treatment. The therapeutic effect was evaluated by micro-positron emission tomography (Micro-PET) using (18)F-fluoro-2-deoxy-d-glucose. The neurological examination, infarct volume and the levels of oxidative stress- and cell apoptosis-related molecules were assessed.. Micro-PET imaging showed that glucose metabolism in the right hippocampus was significantly decreased in the IR group compared to the sham group (P<0.01). ASG IV and ASG IV-TMPZ treatments reversed the decreased glucose metabolism in the model group (P<0.05 and P<0.01, respectively). IR induced the increase of Caspase-3 mRNA levels, MDA content and iNOS activity, but it caused the decrease of SOD activity and Bcl-2 expression compared the sham group (P<0.01). ASG IV-TMPZ and ASG IV reversed the IR-induced changes of these parameters, i.e. the down regulation of Caspase-3 mRNA, MDA content and iNOS activity, and the up regulation of SOD activity and Bcl-2 expression (P<0.05).. This study showed that ASG IV-TMPZ played a pivotal synergistic protective role against focal cerebral ischemic reperfusion damage in a rat experimental model.

    Topics: Animals; Astragalus Plant; Cardiovascular Agents; Caspase 3; Disease Models, Animal; Drug Synergism; Glucose; Hippocampus; Ischemic Attack, Transient; Male; Malondialdehyde; Neuroprotective Agents; Nimodipine; Nitric Oxide Synthase Type II; Phytotherapy; Plant Extracts; Positron-Emission Tomography; Proto-Oncogene Proteins c-bcl-2; Pyrazines; Random Allocation; Rats; Rats, Sprague-Dawley; Reperfusion Injury; RNA, Messenger; Saponins; Superoxide Dismutase; Triterpenes

2012
Effect of methylene blue on the hemodynamic instability resulting from liver ischemia and reperfusion in rabbits.
    Transplantation proceedings, 2011, Volume: 43, Issue:10

    The experimental investigation was performed to study the effects of methylene blue (MB) on hemodynamic, biochemical, and tissue changes among rabbits undergoing liver ischemia and reperfusion (IR). Twenty-four rabbits were randomized into 5 groups: 1, SHAM, control; 2, MB infusion bolus (3 mg/kg); 3, IR, hepatic ischemia for 60 minutes followed by 120 minutes of reperfusion; 4, MB-R, undergoing ischemia that had received an MB bolus infusion (3 mg/kg) prior to reperfusion; 5, R-MB, undergoing ischemia and MB bolus infusion after hemodynamic instability caused by reperfusion. The analysis included continuous recording of vital signs. Blood samples were collected at 0, 60, and 180 minutes of IR to determine blood gases as well as biochemical markers of liver function, nitric oxide, lipid peroxidation, and neutrophil activity. At the end of each experiment, liver tissue samples were collected for histological evaluation of parenchymae markers. Statistical analysis used two-way analysis of variance (ANOVA) tests with significance set at P<.05. Vital signs significantly improved with MB infusion, irrespective of whether it was applied before or after reperfusion. Blood gas data revealed different patterns among the SHAM, MB, IR, MB-R, and R-MB groups, without statistical significance, except for favorable lactate results in the R-MB group (P<.01), which displayed greater survival. Biochemical tests did not show significant differences among the groups, whereas histological analysis revealed favorable appearances for the MB-R and R-MB groups. The MB effect lasted long after reperfusion, suggesting that improvement in the hemodynamic parameters was not based on liver integrity, but rather was possibly related to endothelial function.

    Topics: Animals; Biomarkers; Blood Gas Analysis; Cardiovascular Agents; Disease Models, Animal; Hemodynamics; Liver; Liver Circulation; Male; Methylene Blue; Primary Graft Dysfunction; Rabbits; Reperfusion Injury; Time Factors

2011
Stachydrine, a major constituent of the Chinese herb leonurus heterophyllus sweet, ameliorates human umbilical vein endothelial cells injury induced by anoxia-reoxygenation.
    The American journal of Chinese medicine, 2010, Volume: 38, Issue:1

    Stachydrine is a major constituent of Chinese herb leonurus heterophyllus sweet, which is used in clinics to promote blood circulation and dispel blood stasis. Our study aimed to investigate the role of stachydrine in human umbilical vein endothelial cells (HUVECs) injury induced by anoxia-reoxygenation. Cultured HUVECs were divided randomly into control group, anoxia-reoxygenation (A/R) group and 4 A/R+stachydrine groups. HUVECs in the control group were exposed to normoxia for 5 hours, while in all A/R groups, HUVECs underwent 3 hours anoxia followed by 2 hours reoxygenation, and HUVECs in the 4 A/R+stachydrine groups were treated with 10(-8) M, 10(-7) M, 10(-6) M and 10(-5) M (final concentration) of stachydrine respectively. After anoxia-reoxygenation, tissue factor (TF) was over-expressed, cell viability and the concentrations of SOD, GSH-PX and NO were declined, while LDH, MDA and ET-1 were over-produced (p < 0.05 to 0.001 vs. the control group). However, in stachydrine treated groups, TF expression was inhibited at both mRNA and protein levels, while the declined cell viability and SOD, GSH-PX, NO as well as the enhanced LDH, MDA and ET-1 levels occurred during anoxia-reoxygenation were ameliorated and reversed effectively (p < 0.05 to 0.01 versus A/R group). Consequently, our findings indicate that TF plays an important role in the development of anoxia-reoxygenation injury of HUVECs, stachydrine ameliorates HUVECs injury induced by anoxia-reoxygenation and its putative mechanisms are related to inhibition of TF expression.

    Topics: Cardiovascular Agents; Cell Culture Techniques; Cell Survival; Drugs, Chinese Herbal; Endothelial Cells; Endothelin-1; Glutathione Peroxidase; Humans; L-Lactate Dehydrogenase; Leonurus; Malondialdehyde; Nitric Oxide; Phytotherapy; Proline; Reperfusion Injury; RNA, Messenger; Superoxide Dismutase; Thromboplastin; Umbilical Veins

2010
Indomethacin corrects alterations associated with ischemia/reperfusion in an in vitro proximal tubular model.
    American journal of nephrology, 2010, Volume: 32, Issue:1

    Recent in vivo data indicate that indomethacin improves renal outcome after ischemia via improvement of renal cell survival and function. To examine direct effects of indomethacin on isolated proximal tubular cells, we investigated the influence of indomethacin on markers of ischemia/reperfusion (I/R) damage in an established in vitro model of ischemia and reperfusion.. Ischemia was applied for 2 h followed by reperfusion for up to 48 h. Indomethacin was added at the beginning of reperfusion. Parameters were investigated after 6, 24 or 48 h of reperfusion.. Indomethacin diminished cell death by necrosis and apoptosis, release of prostaglandin E2, induction of I/R-induced protein, dedifferentiation or induction of inducible nitric oxide synthase. Moreover, indomethacin totally prevented the ischemia-induced inhibition of basolateral organic anion transport. Indomethacin did not affect ischemia-mediated induction of nuclear factor-kappaB or monocyte chemoattractant protein 1. Ischemia did not induce matrix protein synthesis.. We have shown that: (a) indomethacin applied after ischemia has a beneficial effect on proximal tubule cell survival after model ischemia and impairs changes of parameters characteristically induced by ischemia via direct action on proximal tubule cells; (b) the inflammatory response of proximal tubule cells was not affected by indomethacin, and (c) fibrosis does not take place after model ischemia in isolated proximal tubule cells.

    Topics: Animals; Apoptosis; Biomarkers; Cardiovascular Agents; Cells, Cultured; Chemokine CCL2; Dinoprostone; Epithelial Cells; Extracellular Matrix; In Vitro Techniques; Indomethacin; Kidney Tubules, Proximal; Necrosis; NF-kappa B; Nitric Oxide Synthase Type II; Opossums; Organic Anion Transporters; Rats; Reperfusion Injury; RNA, Messenger

2010
Cerebralcare Granule, a Chinese herb compound preparation, improves cerebral microcirculatory disorder and hippocampal CA1 neuron injury in gerbils after ischemia-reperfusion.
    Journal of ethnopharmacology, 2010, Jul-20, Volume: 130, Issue:2

    Cerebralcare Granule (CG) is a Chinese herb compound preparation that has been used for treatment of cerebrovascular related diseases. However, the effect of post-treatment with CG on ischemia and reperfusion (I/R) induced cerebral injury is so far unclear.. In present study, cerebral global I/R was induced in Mongolian gerbils by clamping bilateral carotid arteries for 30 min followed by reperfusion for 5 days, and CG (0.4 g/kg or 0.8 g/kg) was administrated 3h after the initiation of reperfusion.. Post-treatment with CG for 5 days attenuated the I/R-induced production of hydrogen peroxide in, leukocyte adhesion to, and albumin leakage from cerebral microvessels, and, meanwhile, protected neuron from death, reduced the number of caspase-3- and Bax-positive cells, and increased Bcl-2-positive cells in hippocampal CA1 region.. The results suggest that CG given after initiation of reperfusion is able to ameliorate cerebral microvascular dysfunction and hippocampal CA1 neuron damage caused by I/R.

    Topics: Animals; Apoptosis; bcl-2-Associated X Protein; Brain Ischemia; CA1 Region, Hippocampal; Capillary Permeability; Cardiovascular Agents; Caspase 3; Cerebral Veins; Cerebrovascular Circulation; Disease Models, Animal; Drugs, Chinese Herbal; Gerbillinae; Hydrogen Peroxide; Leukocyte Rolling; Male; Microcirculation; Neurons; Neuroprotective Agents; Proto-Oncogene Proteins c-bcl-2; Reperfusion Injury; Serum Albumin; Venules

2010
Cardiac protection takes off.
    Cardiovascular research, 2009, Jul-15, Volume: 83, Issue:2

    Topics: Angioplasty, Balloon, Coronary; Animals; Cardiovascular Agents; Cytoprotection; Humans; Ischemic Preconditioning, Myocardial; Myocardial Infarction; Myocardial Revascularization; Myocardium; Reperfusion Injury; Treatment Outcome

2009
Mitochondrial permeability transition pore opening as a promising therapeutic target in cardiac diseases.
    The Journal of pharmacology and experimental therapeutics, 2009, Volume: 330, Issue:3

    In addition to their central role in ATP synthesis, mitochondria play a critical role in cell death. Oxidative stress accompanied by calcium overload, ATP depletion, and elevated phosphate levels induces mitochondrial permeability transition (MPT) with formation of nonspecific MPT pores (MPTP) in the inner mitochondrial membrane. Pore opening results in mitochondrial dysfunction with uncoupled oxidative phosphorylation and ATP hydrolysis, ultimately leading to cell death. For the past 20 years, three proteins have been accepted as key structural components of the MPTP: adenine nucleotide translocase (ANT) in the inner membrane, cyclophilin D (CyP-D) in the matrix, and the voltage-dependent anion channel (VDAC) in the outer membrane. However, most recent studies have questioned the molecular identity of the pores. Genetic studies have eliminated the VDAC as an essential component of MPTP and attributed a regulatory (rather than structural) role to ANT. Currently, the phosphate carrier appears to play a crucial role in MPTP formation. MPTP opening has been examined extensively in cardiac pathological conditions, including ischemia/reperfusion as well as heart failure. Accordingly, MPTP is accepted as a therapeutic target for both pharmacological and conditional strategies to block pore formation by direct interaction with MPTP components or indirectly by decreasing MPTP inducers. Inhibition of MPTP opening by reduction of CyP-D activity by nonimmunosuppressive analogs of cyclosporine A or sanglifehrin A, as well as attenuation of reactive oxygen species accumulation through mitochondria-targeted antioxidants, is the most promising. This review outlines our current knowledge of the structure and function of the MPTP and describes possible approaches for cardioprotection.

    Topics: Animals; Cardiovascular Agents; Heart Diseases; Heart Failure; Humans; Mitochondria; Mitochondrial Membrane Transport Proteins; Mitochondrial Permeability Transition Pore; Reperfusion Injury

2009
Remote ischemic preconditioning in vascular surgery patients: the additional value to medical treatment.
    Journal of endovascular therapy : an official journal of the International Society of Endovascular Specialists, 2009, Volume: 16, Issue:6

    Topics: Albuminuria; Aortic Aneurysm, Abdominal; Biomarkers; Cardiovascular Agents; Elective Surgical Procedures; Glomerular Filtration Rate; Humans; Ischemic Preconditioning; Kidney Diseases; Lower Extremity; Minimally Invasive Surgical Procedures; Myocardial Reperfusion Injury; Reperfusion Injury; Time Factors; Tourniquets; Treatment Outcome; Vascular Surgical Procedures

2009
[Hemodynamic effects of N-acetylcysteine and ischemic preconditioning in a liver ischemia-reperfusion model].
    Orvosi hetilap, 2008, Nov-23, Volume: 149, Issue:47

    The aim of the study was to investigate whether repeated ischemic preconditioning or N-acetylcysteine (NAC) prevents ischemic-reperfusion injury as determined by having favourable hemodynamic effects during reperfusion in canine livers.. The control group ( n = 10) underwent 60 minutes of hepatic ischemia followed by 180 minutes reperfusion. In the NAC group ( n = 5) 150 mg kg -1 of NAC was administered intravenously before inducing ischemia. In the preconditioned group ( n = 5) animals received ischemic preconditioning (10 minutes of ischemia followed by 10 minutes of reperfusion repeated three times) before clamping the portal triad.. 18 dogs survived the study period. One dog in the NAC group died due to circulatory failure unresponsive to inotropic drugs. The cardiac index and the intrathoracic blood volume index were significantly higher in the preconditioning group compared to the controls throughout the study period.. Repeated ischemic preconditioning might improve hemodynamic parameters, whereas we were unable to find any significant differences between the groups regarding N-acetylcysteine.

    Topics: Acetylcysteine; Animals; Cardiovascular Agents; Disease Models, Animal; Dogs; Hemodynamics; Ischemic Preconditioning; Liver; Reperfusion Injury

2008
Postischemic infusion of 17-beta-estradiol protects myocardial function and viability.
    The Journal of surgical research, 2008, May-15, Volume: 146, Issue:2

    Females demonstrate improved cardiac recovery after ischemia/reperfusion injury compared with males. Attenuation of myocardial dysfunction with preischemic estradiol suggests that estrogen may be an important mediator of this cardioprotection. However, it remains unclear whether post-injury estradiol may have clinical potential in the treatment of acute myocardial infarction. We hypothesize that postischemic administration of 17beta-estradiol will decrease myocardial ischemia/reperfusion injury and improve left ventricular cardiac function.. Adult male Sprague Dawley rat hearts (n = 20) (Harlan, Indianapolis, IN) were isolated, perfused with Krebs-Henseleit solution via Langendorff model, and subjected to 15 min of equilibration, 25 min of warm ischemia, and 40 min reperfusion. Experimental hearts received postischemic 17beta-estradiol infusion, 1 nm (n = 4), 10 nm (n = 4), 25 nm (n = 4), or 50 nm (n = 4), throughout reperfusion. Control hearts (n = 4) were infused with perfusate vehicle.. Postischemic recovery of left ventricular developed pressure was significantly greater with 1 nm (51.6% +/- 7.4%) and 10 nm estradiol (47.7% +/- 8.6%) than with vehicle (37.8% +/- 9.7%) at end reperfusion. There was also greater recovery of the end diastolic pressure with 1 nm (47.8 +/- 4.0 mmHg) and 10 nm estradiol (54.0 +/- 4.0) compared with vehicle (75.3 +/- 7.5). Further, 1 nm and 10 nm estrogen preserved coronary flow after ischemia and decreased coronary effluent lactated dehydrogenase compared with controls. Estrogen at 25 nm and 50 nm did not provide additional benefit in terms of functional recovery. Estrogen at all concentrations increased extracellular signal-regulated protein kinase phosphorylation.. Postischemic infusion of 17beta-estradiol protects myocardial function and viability. The attractive potential for the clinical application of postischemic estrogen therapy warrants further study to elucidate the mechanistic pathways and differences between males and females.

    Topics: Animals; Cardiovascular Agents; Estradiol; Heart; Male; Myocardial Ischemia; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Tissue Survival; Ventricular Function, Left

2008
Preventive effects of Ginkgo biloba extract on ischemia-reperfusion injury in rat bladder.
    Urologia internationalis, 2007, Volume: 78, Issue:2

    The aim of this experimental study was to evaluate the effects of Ginkgo biloba (EGb 761) on ischemia-reperfusion (I-R) injury in the rat bladder.. A bladder I-R injury was induced by abdominal aorta occlusion by ischemia for 30 min, followed by 45 min reperfusion in rats. The rats were divided into four groups of 7 rats each; the control, I-R, and I-R groups were pretreated intraperitoneally with 50 or 100 mg/kg G. biloba 60 min before ischemia induction. Contractile responses to carbachol through isolated organ bath studies were recorded, histological sections were evaluated by light microscopy, and TUNEL staining was performed for the evaluation of apoptosis.. In the I-R group, the contractile responses of the bladder strips were lower than those of the control group (p < 0.01-0.001) and were restored by pretreatment with 100 mg/kg G. biloba (p < 0.05-0.001). Decreased polymorphonuclear leukocyte infiltration was detected in the G. biloba pretreatment groups when compared to the I-R group during histological evaluation. The ratio of TUNEL-positive nuclei was 1.84% in the I-R group, whereas it was decreased in both of the G. biloba pretreatment groups (p < 0.01, p < 0.01).. Our data indicated that G. biloba has a preventive effect on I-R injury in rat urinary bladder.

    Topics: Animals; Cardiovascular Agents; Ginkgo biloba; Male; Muscle Contraction; Plant Extracts; Rats; Rats, Wistar; Reperfusion Injury; Urinary Bladder

2007
Kinetic analysis of 99mTc-sestamibi evaluates the protective effects by ischaemic preconditioning on ischaemic myocardium in an isolated rabbit heart.
    Nuclear medicine communications, 2007, Volume: 28, Issue:11

    To analyse the kinetic changes of uptake, washout and retention of Tc-sestamibi in order to evaluate the protective effects and possible mechanism of ischaemic preconditioning and adenosine preconditioning on myocardium injured by ischaemia/reperfusion.. Isolated ischaemia/reperfusion rabbit heart models, as established by Langendorff, were used. Eighteen rabbit hearts perfused in Krebs-Henseleit (KH) buffer were randomly assigned to three groups: ischaemia/reperfusion (I/R, n=6), adenosine preconditioning (AD, n=6), and ischaemic preconditioning (IPC, n=6). Tc-sestamibi (55.5 MBq) in KH was perfused for 40 min and washed out for 40 min. The kinetic changes of Tc-sestamibi within myocardial tissue was monitored during the uptake and washout phases. Cardiac haemodynamic parameters, creatine kinase and lactate dehydrogenase leakage in coronary effluent, and myocardial infarct size were measured to assess myocardial injuries in rabbit hearts.. In the early phases of uptake, there were no significantly different uptake rates of Tc-sestamibi between AD (before 20 min), IPC (before 15 min) and I/R myocardium (all P>0.05). Uptake rates of Tc-sestamibi in myocardium of the three groups all tended to increase, with the uptake time increasing. In the late phases of uptake, AD and IPC were significantly higher than I/R (all P<0.05). In the washout phases, the retention fractions of Tc-sestamibi in myocardium of the three groups all showed a descending tendency with washout time increasing. The retention fractions in AD and IPC were all higher than I/R (all P<0.05). There were no statistical differences in uptake rates and retention fractions of Tc-sestamibi between AD and IPC (all P>0.05). Cardiac haemodynamic parameters, creatine kinase and lactate dehydrogenase leakage, and myocardial infarct size demonstrated there is lighter injury in AD and IPC myocardium than in I/R (all P<0.05). The retention of Tc-sestamibi and myocardial infarction weight were significantly negatively correlated (r=-0.8384, P<0.001).. Adenosine preconditioning has similar myocardial protective effects on ischaemia/reperfusion myocardium as does ischaemic preconditioning. Tc-sestamibi may be a sensitive and reliable measure for evaluating the importance and mechanism of ischaemic preconditioning and adenosine preconditioning.

    Topics: Adenosine; Animals; Blood Pressure; Cardiovascular Agents; Heart; Heart Rate; In Vitro Techniques; Ischemic Preconditioning, Myocardial; Male; Myocardial Infarction; Myocardial Ischemia; Myocardium; Positron-Emission Tomography; Rabbits; Radiopharmaceuticals; Reperfusion Injury; Technetium Tc 99m Sestamibi

2007
Preservation of mitochondrial function during ischemia as a possible mechanism for cardioprotection of diltiazem against ischemia/reperfusion injury.
    Biochemical pharmacology, 2004, Feb-01, Volume: 67, Issue:3

    A possible mechanism for D-cis-diltiazem (diltiazem)-mediated improvement of the contractile function of ischemic/reperfused hearts was examined. Thirty-five-min ischemia/60-min reperfusion recovered little the left ventricular developed pressure (LVDP) and decreased myocardial high-energy phosphates (HEPs). Ischemia induced an accumulation of tissue Na+ content, an increase in cytochrome c in the cytosolic fraction, and a decrease in the oxygen consumption rate (OCR) in perfused hearts. Treatment of the heart with 1 microM diltiazem for the last 3-min of pre-ischemia did not affect the decrease in HEPs during ischemia, whereas that with 3 microM partially attenuated the decrease in ATP, suggesting that 3 microM diltiazem exerted energy-sparing effect. Treatment with 1 microM diltiazem enhanced the post-ischemic recovery of LVDP associated with attenuation of the ischemia-induced accumulation of tissue Na+, increase in cytochrome c in the cytosolic fraction, and decrease in myocardial OCR, and restoration of the myocardial HEPs during reperfusion. Combined treatment with diltiazem and a Na+/H+ exchange inhibitor, but not a Na+ channel blocker, facilitated the attenuation of Na+ accumulation in the ischemic heart and the enhancement of the post-ischemic recovery of LVDP. Sodium lactate, a possible metabolite in ischemic hearts, and sodium chloride increased the Na+ concentration in mitochondria, released cytochrome c into incubation medium, and reduced the mitochondrial respiration. Treatment of isolated mitochondria with diltiazem failed to attenuate the sodium lactate- and sodium chloride-induced alterations. These results suggest that the cardioprotection of diltiazem may be exerted via attenuating cytosolic Na+ overload through Na+ channels in the ischemic heart, leading to preservation of mitochondrial functional ability during ischemia, followed by improvement of post-ischemic energy production and contractile recovery.

    Topics: Animals; Calcium; Cardiotonic Agents; Cardiovascular Agents; Cytochromes c; Diltiazem; Drug Interactions; Ischemia; Male; Mitochondria; Myocardial Contraction; Myocardial Reperfusion Injury; Myocardium; Oxygen Consumption; Phosphates; Rats; Rats, Wistar; Reperfusion Injury; Respiration; Sodium; Sodium Channel Blockers; Sodium-Hydrogen Exchangers

2004
Changes of vasoregulatory gene expression following hepatic ischemia/reperfusion and treatment with endothelin-A receptor blockade.
    Journal of cardiovascular pharmacology, 2004, Volume: 44 Suppl 1

    The objective of this study was to investigate the effect of a specific endothelin-A receptor antagonist on mRNA expression of genes encoding vasoactive mediators and proinflammatory cytokines following complete vascular exclusion of the porcine liver. Fourteen adult German Landrace pigs were subjected to 120 minutes of warm hepatic ischemia by total vascular exclusion. The animals were divided into two groups: the control group received saline solution and the therapy group was given the selective endothelin-A receptor antagonist BSF 208075. Liver tissue samples were collected 1 hour after reperfusion and mRNA expression for preproendothelin-1, prointerleukin-1beta, prointerleukin- 6, pro-tumor necrosis factor-alpha and endothelial nitric oxide synthase was analyzed quantitatively using the TaqMan system. Additionally, immunohistochemical analysis using a semiquantitative score for endothelin-1 and endothelin-A receptor was performed. One hour after reperfusion, quantitative reverse transcriptase-polymerase chain reaction revealed significantly lower expression of preproendothelin-1, pro-tumor necrosis factor-alpha, and prointerleukin-6 in the therapy group compared to controls. Immunohistochemical analysis demonstrated significantly reduced endothelin-1 immunostaining after therapy. Treatment with the selective endothelin-A receptor antagonist exerts a protective effect on the microcirculation after liver ischemia and reperfusion. We were able to show that the endothelin-A receptor antagonist not only has effects on the expression of vasoactive genes, it also decreases gene expression of proinflammatory cytokines such as tumor necrosis factor-alpha and interleukin-6.

    Topics: Angiogenic Proteins; Animals; Cardiovascular Agents; Cytokines; Disease Models, Animal; Down-Regulation; Endothelin A Receptor Antagonists; Endothelin-1; Female; Interleukin-1beta; Interleukin-6; Liver; Microcirculation; Nitric Oxide Synthase Type III; Phenylpropionates; Pyridazines; Receptor, Endothelin A; Reperfusion Injury; RNA, Messenger; Swine; Time Factors; Tumor Necrosis Factor-alpha; Warm Ischemia

2004
Endothelin-A receptor blockade improves postischemic hepatic microhemodynamics.
    Journal of cardiovascular pharmacology, 2004, Volume: 44 Suppl 1

    The aim of this study was to investigate a possible protective role of a selective endothelin-A receptor antagonist on hepatic microcirculation after ischemia/reperfusion. In a rat model, warm ischemia of the left liver lobe was induced for 90 minutes under intraperitoneal anesthesia with xylazine and ketamine. Shamoperated and untreated ischemic groups and a group treated with BSF 208075 were investigated. The effect of the endothelin-A receptor antagonist on ischemia/reperfusion was assessed by in-vivo microscopy and measurement of aspartate aminotransferase and alanine aminotransferase levels. In the untreated group, sinusoidal constriction to 70% of basal diameters was observed, leading to a significant decrease in perfusion rate. In addition, we found an increased percentage of stagnant leukocytes and platelets in sinusoids and in postsinusoidal venules (P < 0.05). A significant increase in liver enzymes was detected 6 hours after reperfusion (P < 0.05). In the treatment group, sinusoidal diameters were maintained at 108%, and perfusion rate was significantly increased (P < 0.05). Hepatocellular damage was decreased and leukocyte and platelet-endothelium interactions were reduced (P < 0.05). Our results provide evidence that the new therapeutic approach using an endothelin-A receptor antagonist is effective in reducing hepatic ischemia/reperfusion injury. It could be shown for the first time that endothelin receptor blockade also influences platelet-endothelium interactions.

    Topics: Alanine Transaminase; Animals; Aspartate Aminotransferases; Cardiovascular Agents; Disease Models, Animal; Endothelin A Receptor Antagonists; Endothelin-1; Endothelium, Vascular; Female; Hemodynamics; Leukocyte Rolling; Liver; Liver Circulation; Microcirculation; Microscopy, Fluorescence; Microscopy, Video; Phenylpropionates; Platelet Adhesiveness; Pyridazines; Rats; Rats, Wistar; Receptor, Endothelin A; Reperfusion Injury

2004
A new method of long-term preventive cardioprotection using Lactobacillus.
    American journal of physiology. Heart and circulatory physiology, 2000, Volume: 278, Issue:5

    Potential long-term cardioprotection was investigated in an extensive experimental study. Lactobacillus cultivation components (LCC) were administered intravenously in anesthetized rats 1, 7, and 21 days before global ischemia (GI). GI was produced by full stop flow in isolated Langendorff-perfused hearts for 20 min and was followed by reperfusion. Control animals were injected with saline. LCC reduced reperfusion tachyarrhythmia significantly and improved functional recovery of the ischemized rat heart. These beneficial effects were associated with reduction of release of norepinephrine (NE) and prostacyclin at the first minute of reperfusion, activation of myocardial catalase, and overexpression of 70-kDa heat stress protein (HSP-70) at ischemia and reperfusion (P < 0.05). This cardioprotection was documented up to 21 days after a single injection of LCC. Thus Lactobacillus cultivation components are new nontoxic materials that produce marked long-term cardioprotection against ischemia-reperfusion damage. This effect is attributed to an activation of the cellular defense system, manifested by activation of the antioxidant pathway and by expression of protective proteins. NE is involved in this process, and the data also suggest a role for prostacyclin in this model of cardioprotection. The potential of LCC and related compounds working through similar mechanisms in the prevention and therapy of various ischemic heart syndromes should be explored.

    Topics: Adenosine Triphosphate; Animals; Body Temperature; Cardiovascular Agents; Catalase; Coronary Circulation; Epoprostenol; Hemodynamics; HSP70 Heat-Shock Proteins; Lactic Acid; Lactobacillus; Male; Myocardial Ischemia; Norepinephrine; Pharmaceutical Preparations; Rats; Rats, Sprague-Dawley; Recovery of Function; Reperfusion Injury; Tachycardia

2000
Comparison of bisoprolol and carvedilol cardioprotection in a rabbit ischemia and reperfusion model.
    European journal of pharmacology, 2000, Oct-06, Volume: 406, Issue:1

    Carvedilol, a selective alpha(1) and non-selective beta-adrenoceptor antagonist and antioxidant, has been shown to provide significant cardiac protection in animal models of myocardial ischemia. To further explore the mechanisms contributing to the efficacy of carvedilol cardioprotection, the effects of carvedilol on hemodynamic variables, infarct size and myeloperoxidase activity (an index of neutrophil accumulation) were compared with a beta(1) selective adrenoceptor antagonist, bisoprolol. Carvedilol (1 mg/kg) or bisoprolol (1 mg/kg) was given intravenously 5 min before reperfusion. In vehicle-treated rabbits, ischemia (45 min) and reperfusion (240 min) resulted in significant increases in left ventricular end diastolic pressure, large myocardial infarction (64.7+/-2.6% of area-at-risk) and a marked increase in myeloperoxidase activity (64+/-14 U/g protein in area-at-risk). Carvedilol treatment resulted in sustained reduction of the pressure-rate-index and significantly smaller infarcts (30+/-2.9, P<0.01 vs. vehicle) as well as decreased myeloperoxidase activity (26+/-11 U/g protein in area-at-risk, P<0.01 vs. vehicle). Administration of bisoprolol at 1 mg/kg resulted in a pressure-rate-index comparable to that of carvedilol and also decreased infarct size (48.4+/-2.5%, P<0.001 vs. vehicle, P<0.05 vs. carvedilol), although to a significantly lesser extent than that observed with carvedilol. Treatment with bisoprolol failed to reduce myeloperoxidase activity in the ischemic myocardial tissue. In addition, carvedilol, but not bisoprolol, markedly decreased cardiac membrane lipid peroxidation measured by thiobarbituric acid formation. Taken together, this study suggests that the superior cardioprotection of carvedilol over bisoprolol is possibly the result of carvedilol's antioxidant and anti-neutrophil effects, not its hemodynamic properties.

    Topics: Adrenergic beta-Agonists; Adrenergic beta-Antagonists; Animals; Antioxidants; Bisoprolol; Carbazoles; Cardiovascular Agents; Carvedilol; Creatine Kinase; Disease Models, Animal; Dose-Response Relationship, Drug; Heart Rate; Isoproterenol; Lipid Peroxidation; Male; Membrane Lipids; Myocardial Ischemia; Myocardium; Peroxidase; Propanolamines; Rabbits; Reperfusion Injury; Ventricular Pressure

2000
Protective effect of preconditioning and adenosine pretreatment in experimental skeletal muscle reperfusion injury.
    The British journal of surgery, 1999, Volume: 86, Issue:7

    Prolonged ischaemia followed by reperfusion (I/R) of skeletal muscle results in significant tissue injury. Ischaemic preconditioning (IPC), achieved by repeated brief periods of I/R before prolonged ischaemia or adenosine pretreatment, can prevent I/R injury in cardiac muscle. The aim of this study was to ascertain in a rodent model if damage to skeletal muscle due to global hindlimb tourniquet-induced I/R could be similarly attenuated.. Anaesthetized rats were randomized (n = 6-10 per group) to five groups: sham-operated controls; I/R (4 h of ischaemia, 2 h of reperfusion); IPC (three cycles of 10 min of ischaemia/10 min of reperfusion) alone; IPC immediately preceding I/R; or adenosine 1000 microg/kg immediately before I/R. At the end of reperfusion, biopsies were taken from the left gastrocnemius muscle for measurement of myeloperoxidase (MPO) and reduced glutathione (GSH). Before ischaemia and at the end of reperfusion, blood samples were taken for measurement of nitric oxide metabolites, tumour necrosis factor (TNF) alpha and macrophage inflammatory protein (MIP) 2.. IPC before I/R resulted in lower levels of MPO (P < 0.001) and TNF-alpha (P = 0.004), and higher levels of GSH (P < 0.001) and nitric oxide metabolites (P = 0.002) than I/R alone. Adenosine had effects comparable to IPC pretreatment (P < 0.001 for MPO, P = 0.002 for GSH, P = 0.02 for nitric oxide metabolites and P = 0.001 for TNF-alpha). There was no difference in the blood pressure or the MIP-2 concentration among the groups.. IPC or pretreatment with adenosine ameliorates the I/R injury of skeletal muscle.

    Topics: Adenosine; Animals; Blood Pressure; Cardiovascular Agents; Chemokine CXCL2; Glutathione; Ischemic Preconditioning; Male; Monokines; Muscle, Skeletal; Nitric Oxide; Peroxidase; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Tumor Necrosis Factor-alpha

1999
Treatment with l-cis diltiazem before reperfusion reduces infarct size in the ischemic rabbit heart in vivo.
    Japanese journal of pharmacology, 1999, Volume: 80, Issue:4

    l-cis Diltiazem, an optical isomer of diltiazem, protects against myocardial dysfunction in vitro, whereas its Ca2+ channel blocking activity is about 100 times less potent than that of diltiazem. However, there is no evidence that l-cis diltiazem actually protects against ischemia/reperfusion injury in vivo. To assess this, we employed an anesthetized rabbit model, where the left circumflex artery was occluded for 15 min and reperfused for 360 min. Treatment with diltiazem before and during ischemia (bolus 200 microg/kg and 15 microg/kg per minute for 25 min, i.v.; 575 microg/kg total) showed slightly depressed hemodynamic parameters, while l-cis diltiazem (1150 microg/kg) had no effect. Treatment with l-cis diltiazem produced a high recovery of the thickening fraction and limited the infarct size in a dose-dependent manner. Furthermore, the treatment with l-cis diltiazem (1150 microg/kg) or diltiazem (575 microg/kg) 5 min before reperfusion also limited the infarct size, but not after reperfusion. These results suggest that l-cis diltiazem affects some events in the onset of reperfusion, independently of Ca2+-channel-blocking action. Our observations are the first to show that l-cis diltiazem demonstrated its cardioprotective action in the ischemic rabbit heart in vivo.

    Topics: Anesthesia; Animals; Cardiovascular Agents; Diltiazem; Dose-Response Relationship, Drug; Heart; Hemodynamics; Isomerism; Male; Myocardial Infarction; Myocardial Ischemia; Rabbits; Reperfusion Injury; Time Factors

1999
Cardioprotective effects of the aminopeptidase P inhibitor apstatin: studies on ischemia/reperfusion injury in the isolated rat heart.
    Journal of cardiovascular pharmacology, 1999, Volume: 34, Issue:4

    Aminopeptidase P and angiotensin-converting enzyme (ACE) are responsible for the metabolism of exogenously administered bradykinin in the coronary circulation of the rat. It has been shown that ACE inhibitors decrease cytosolic enzyme release from the ischemic rat heart and reduce reperfusion-induced ventricular arrhythmias by increasing endogenous levels of bradykinin. It was hypothesized that the aminopeptidase P inhibitor apstatin could do the same. In an isolated perfused rat heart preparation subjected to global ischemia and reperfusion, both apstatin and ramiprilat (an ACE inhibitor) significantly decreased creatine kinase (CK) and lactate dehydrogenase (LDH) release. The difference between the postischemia and preischemia levels of released CK was reduced 68% by apstatin and 68% by ramiprilat compared with control. The corresponding reductions in LDH release were 74% for apstatin and 81% for ramiprilat. A combination of the inhibitors was not significantly better than either one alone. Apstatin and ramiprilat also significantly reduced the duration of reperfusion-induced ventricular fibrillation by 69 and 61%, respectively. The antiarrhythmic effect of apstatin was reversed by HOE140, a bradykinin B2-receptor antagonist, suggesting that apstatin is acting by potentiating endogenously formed bradykinin. The results demonstrate that the aminopeptidase P inhibitor apstatin is cardioprotective in this model of cardiac ischemia/ reperfusion injury.

    Topics: Adrenergic beta-Antagonists; Angiotensin-Converting Enzyme Inhibitors; Animals; Arrhythmias, Cardiac; Bradykinin; Cardiovascular Agents; Creatine Kinase; Drug Interactions; In Vitro Techniques; L-Lactate Dehydrogenase; Male; Peptides; Perfusion; Protease Inhibitors; Ramipril; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Ventricular Fibrillation

1999
Effect of ursodeoxycholic acid on ischemia/reperfusion injury in isolated rat heart.
    Archives of pharmacal research, 1999, Volume: 22, Issue:5

    In this study, the effects of ursodeoxycholic acid (UDCA) on ischemia/reperfusion injury were investigated on isolated heart perfusion model. Hearts were perfused with oxygenated Krebs-Henseleit solution (pH 7.4, 37 degrees C) on a Langendorff apparatus. After equilibration, isolated hearts were treated with UDCA 20 to 160 microM or vehicle (0.04% DMSO) for 10 min before the onset of ischemia. After global ischemia (30 min), ischemic hearts were reperfused and allowed to recover for 30 min. The physiological (i.e. heart rate, left ventricular developed pressure, coronary flow, double product and time to contracture formation) and biochemical (lactate dehydrogenase; LDH) parameters were evaluated. In vehicle-treated group, time to contracture formation was 21.4 min during ischemia, LVDP was 18.5 mmHg at the endpoint of reperfusion and LDH activity in total reperfusion effluent was 54.0 U/L. Cardioprotective effects of UDCA against ischemia/reperfusion consisted of a reduced TTC (EC25=97.3 microM), reduced LDH release and enhanced recovery of cardiac contractile function during reperfusion. Especially, the treatments of UDCA 80 and 160 microM significantly increased LVDP and reduced LDH release. Our findings suggest that UDCA ameliorates ischemia/reperfusion-induced myocardial damage.

    Topics: Animals; Cardiovascular Agents; Coronary Circulation; Dose-Response Relationship, Drug; Hemodynamics; In Vitro Techniques; L-Lactate Dehydrogenase; Male; Myocardial Contraction; Rats; Reperfusion Injury; Time Factors; Ursodeoxycholic Acid

1999
Cardioprotective effects of N-hydroxyguanidine PR5 in myocardial ischaemia and reperfusion in rats.
    British journal of pharmacology, 1999, Volume: 128, Issue:5

    1. The potential for the N-hydroxyguanidine compound PR5 (N-(3, 4-dimethoxy-2-chlorobenzylideneamino)-N'-hydroxyguanidine) as a cardioprotective agent in heart ischaemia and reperfusion injury was investigated using rat models. 2. Administration of 1-10 mg kg-1 of PR5 5 min before 10 min of left coronary artery occlusion, followed by 20 min reperfusion, strongly inhibited reperfusion burst of arrhythmias and markedly improved the survival of the animals (e.g. ventricular fibrillation incidence 93 vs 43% (P<0.05); mortality 47 vs 0% (P<0.05), for controls and for 3 mg kg-1 of PR5, respectively). 3. Administration of 3 mg kg-1 of PR5 1 min before reperfusion to rats subjected to 10 min occlusion, 20 min reperfusion was most effective in reducing arrhythmias and decreasing mortality (43 vs 0%, P<0.05), but effects were also seen when PR5 was administered 0, 1 and 5 min after start of reperfusion. 4. Coronary occlusion/reperfusion (10 - 20 min) increased malondialdehyde (MDA) of rat hearts (0.88+/-0.13 for sham vs 1.45+/-0.12 nmol mg-1 protein for ischaemic; P<0.05). In rats where 3 mg kg-1 PR5 were administered 1 min before reperfusion the increase was attenuated (MDA being 1.04+/-0.12; P<0.05 vs ischaemic). 5. PR5 caused a substantial reduction of the infarction size in rats subjected to 180 min left coronary artery occlusion, followed by 120 min of reperfusion; the necrotic zone being 326+/-32 mg for controls vs 137+/-21 mg for animals treated with 3x3 mg kg-1 of PR5 (P<0.01). 6. PR5 reduced the elevation of the ST-segment of the ECGs, as well as caused pronounced attenuation of the rapid blood pressure drop seen at the start of reperfusion following coronary artery occlusion. 7 We conclude that the N-hydroxyguanidine PR5 provides remarkable protection against ischaemia and reperfusion induced myocardial necrosis and life-threatening arrhythmias. These effects of PR5 are discussed in relation to a recently discovered ability of N-hydroxyguanidines to function as electron acceptors at the xanthine oxidase enzyme.

    Topics: Animals; Antihypertensive Agents; Arrhythmias, Cardiac; Cardiovascular Agents; Dose-Response Relationship, Drug; Enzyme Inhibitors; Guanabenz; Guanidines; Hydroxylamines; Male; Malondialdehyde; Myocardial Ischemia; Myocardial Reperfusion Injury; Myocardium; Oxidation-Reduction; Rats; Rats, Wistar; Reperfusion Injury; Xanthine Oxidase

1999
Antiischemic and antiarrhythmic activities of some novel alinidine analogs in the rat heart.
    Journal of cardiovascular pharmacology, 1997, Volume: 29, Issue:4

    The antiischemic and antiarrhythmic effects of alinidine and a number of novel alinidine analogs were examined by using perfused rat-heart models. In the isolated working rat heart, the alinidine analog TH91:21 (10 microM; a butyl derivative) significantly increased the postischemic recovery of the heart in terms of both power and efficiency when compared with the control group. In the in situ perfused heart model, this same compound, along with TH91:22 (10 microM; a pentyl derivative) also significantly reduced the severity of both ischemia- and reperfusion-induced arrhythmias in both paced and unpaced hearts. Thus this study is the first to demonstrate the potent antiarrhythmic efficacy of two novel alinidine analogs TH91:21 and TH91:22, with TH91:21 also demonstrated to be a potent antiischemic agent in the isolated working rat heart. Although the mode of action of these compounds remains unclear, results from this study suggest that it is not simply a result of bradycardia or blockade of KATP channels, two actions these compounds possess. These compounds thus possess a novel and beneficial pharmacologic profile worthy of further study.

    Topics: Animals; Anti-Arrhythmia Agents; Arrhythmias, Cardiac; Bradycardia; Cardiac Pacing, Artificial; Cardiovascular Agents; Clonidine; Heart; Male; Potassium Channel Blockers; Rats; Rats, Inbred WKY; Reperfusion Injury; Structure-Activity Relationship

1997
Adenosine inhibition of neutrophil damage during reperfusion does not involve K(ATP)-channel activation.
    The American journal of physiology, 1997, Volume: 273, Issue:4

    This study tests the hypothesis that cardioprotection exerted by adenosine A2-receptor activation and neutrophil-related events involves stimulation of ATP-sensitive potassium (K(ATP)) channels on neutrophils during reperfusion. The adenosine A2 agonist CGS-21680 (CGS) inhibited superoxide radical generation from isolated rabbit polymorphonuclear neutrophils (PMNs) in a dose-dependent manner from 17.7 +/- 2.1 to 7.4 +/- 1.3 nmol/5 x 10(6) PMNs (P < 0.05). Pinacidil, a K(ATP)-channel opener, partially inhibited superoxide radical production, which was completely reversed by glibenclamide (Glib). Incremental doses of Glib in combination with CGS (1 microM) did not alter CGS-induced inhibition of superoxide radical generation. CGS significantly reduced PMN adherence to the endothelial surface of aortic segments in a dose-dependent manner from 189 +/- 8 to 50 +/- 6 PMNs/mm2 (P < 0.05), which was also not altered by incremental doses of Glib. Infusion of CGS (0.025 mg/kg) before reperfusion reduced infarct size from 29 +/- 2% in the Vehicle group to 15 +/- 1% in rabbits undergoing 30 min of ischemia and 120 min of reperfusion (P < 0.05). Glib (0.3 mg/kg) did not change the infarct size (28 +/- 2%) vs. the Vehicle group and did not attenuate infarct size reduction by CGS (16 +/- 1%). Glib did not change blood glucose levels. Cardiac myeloperoxidase activity was decreased in the ischemic tissue of the CGS group (0.15 +/- 0.03 U/100 mg tissue) compared with the Vehicle group (0.37 +/- 0.05 U/100 mg tissue; P < 0.05). We conclude that adenosine A2 activation before reperfusion partially reduces infarct size by inhibiting neutrophil activity and that this effect does not involve K(ATP)-channel stimulation.

    Topics: Adenosine; Adenosine Triphosphate; Animals; Cardiovascular Agents; Cell Adhesion; Creatine Kinase; Female; Glyburide; Hemodynamics; Male; Myocardial Infarction; Myocardium; Neutrophils; Peroxidase; Phenethylamines; Potassium Channels; Rabbits; Reperfusion Injury; Superoxides

1997
Liberation of vasoactive substances and its prevention with thromboxane A2 synthase inhibitor in pig liver transplantation.
    Transplant international : official journal of the European Society for Organ Transplantation, 1996, Volume: 9, Issue:1

    There are multiple causes of liver graft nonfunction in the early post-transplant period. Since a severe microcirculatory disturbance based on ischemia-reperfusion liver injury is considered to be the main underlying pathophysiology, it is suspected that various vasoactive substances are liberated after reperfusion of the graft. In order to investigate this matter, we conducted an experimental study with pig liver allotransplantation. Two groups of animals received donor grafts with or without thromboxane synthase inhibitor (sodium ozagrel), 1.25 mg/ kg body weight intravenously, given at the time of liver harvesting. All of the recipient animals in the treatment group (n = 10) survived longer than 7 days whereas three of ten animals in the control group died within 7 days. Serum lactate dehydrogenase (LDH) in the recipient serum at 1 h after reperfusion was significantly lower in the treatment group (915.1 +/- 167.3 U/l) than in the control group (1264.4 +/- 134.7 U/l). Serum thromboxane B2 (2261.7 +/- 1055.7 pg/ml) and endothelin-1 (6.3 +/- 2.2 pg/ml) after reperfusion in the treatment group were significantly lower than those in the control group (4220.0 +/- 1711.0 pg/ml and 11.2 +/- 3.1 pg/ml, respectively). Although serum angiotensin II after reperfusion tended to be lower in the treatment group than in the controls serum renin activity was less than 3 ng/ml in both groups of animals. There were no differences in the plasma endotoxin levels between the two groups. We conclude that the administration of sodium ozagrel to the donor animals provided better graft function in recipients than no such treatment. We speculate that the inhibition of thromboxane A2 production suppresses the liberation of other vasoconstrictive substances, preventing microcirculatory disturbance and, thereby, contributing to improved graft function after liver transplantation.

    Topics: Angiotensin II; Animals; Aspartate Aminotransferases; Cardiovascular Agents; Endothelins; Endotoxins; Enzyme Inhibitors; Female; L-Lactate Dehydrogenase; Liver Transplantation; Methacrylates; Prostaglandins F; Reperfusion Injury; Swine; Thromboxane B2; Thromboxane-A Synthase

1996
Enhancement of cardiac function by cyclocreatine in models of cardiopulmonary bypass.
    Journal of molecular and cellular cardiology, 1995, Volume: 27, Issue:4

    This study tests the hypothesis that the administration of cyclocreatine prior to global ischemia enhances recovery of cardiac function during reperfusion. Two models were used. First, in a Langendorff-working heart model of normothermic cardioplegic arrest, rats (n = 6 per group) were injected intravenously with saline or cyclocreatine (600, 300, or 150 mg/kg). After 30 min or 2 h, hearts were excised and perfused in the Langendorff mode for 5 min and then in the working heart mode for 20 min. Normothermic arrest was induced by infusing warm St. Thomas solution once; then hearts were kept at 37 degrees C for 40 min. Following arrest, hearts were reperfused in the Langendorff mode for 15 min and then in the working mode for 30 min. Cyclocreatine consistently produced significantly better recovery of aortic flow and cardiac output compared to that of saline hearts. Second, in an intact canine model of cold cardioplegic arrest, adult mongrel dogs (n = 3 to 6 per group) underwent aortic cross-clamping for 1 h, followed by reperfusion on bypass for 45 min and off bypass for 4 h. Dogs were injected intravenously with saline or cyclocreatine (500 mg/kg) for 1 h before experiment. Post-bypass segmental contractility and cardiac output were significantly better in cyclocreatine hearts compared to that of controls. In a limited study, after a 3 h aortic cross-clamp time, cyclocreatine hearts achieved 91% baseline function while control hearts failed after 2 h. Results of this study suggest that cyclocreatine, without inotropic or chronotropic effect, protects the heart from global ischemic injury.

    Topics: Animals; Cardiac Output; Cardiopulmonary Bypass; Cardiovascular Agents; Coronary Circulation; Creatinine; Dogs; In Vitro Techniques; Male; Models, Cardiovascular; Myocardial Contraction; Myocardial Ischemia; Myocardial Reperfusion; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Sodium Chloride; Time Factors

1995
Bradycardic agent UL-FS 49 attenuates ischemic regional myocardial dysfunction and reduces infarct size in swine: comparison with the beta-blocker atenolol.
    Journal of cardiovascular pharmacology, 1995, Volume: 25, Issue:2

    Heart rate (HR) is a major factor determining the severity of myocardial ischemia, and HR reduction is an effective therapy for myocardial ischemia. We tested the effects of HR reduction induced by either UL-FS 49 or atenolol on regional myocardial blood flow, function, and infarct size (IS) in a porcine model of 90-min low-flow ischemia and 2-h reperfusion. In 24 Göttinger miniswine, the left anterior descending coronary artery (LAD) was cannulated and hypoperfused at constant inflow to reduce anterior systolic wall thickening (AWT, sonomicrometry) by approximately 85%. Eight swine served as a placebo group, and 8 other swine received UL-FS 49 (0.60 mg/kg intravenously, i.v.) after 10-min ischemia. In the remaining 8 swine, atenolol was infused after 10-min ischemia at a dosage [mean 1.75 +/- 1.20 (SD) mg/kg i.v.] to mimic the HR reduction observed with UL-FS 49. Systemic hemodynamics, subendocardial blood flow (ENDO, microspheres) and AWT were measured under control conditions, at 10 and 90 min of ischemia. In the swine receiving UL-FS 49 or atenolol, additional measurements were made 5 min after administration of the respective drug. After 2-h reperfusion, IS (percentage of area at risk) was determined with TTC-staining. Five minutes after administration of UL-FS 49, HR was decreased from 113 +/- 9 to 83 +/- 13 beats/min (p < 0.05) and remained unchanged when ischemia was prolonged to 90 min. In the swine receiving atenolol, HR was reduced from 117 +/- 14 to 93 +/- 7 beats/min (p < 0.05) 5 min after drug administration and decreased further to 87 +/- 10 beats/min when ischemia was prolonged to 90 min. After 10 min of ischemia, AWT in the placebo, UL-FS 49, and atenolol group was decreased to 7.0 +/- 5.5, 6.4 +/- 3.5, and 6.2 +/- 3.3% (all p < 0.05 vs. control), respectively. The reduction in ENDO was also comparable among the three groups. In the placebo group, AWT remained unchanged when ischemia was prolonged to 90 min (4.4 +/- 2.6%). In swine receiving atenolol, AWT tended to increase (13.6 +/- 10.5%), whereas in swine receiving UL-FS 49, AWT was significantly increased to 21.4 +/- 7.1% (p < 0.05 vs. 10-min ischemia and vs. the placebo and atenolol groups). IS was significantly reduced in swine receiving atenolol (3.9 +/- 3.5%) or UL-FS 49 (5.8 +/- 4.6%) as compared with the placebo-group (10.4 +/- 8.9%).(ABSTRACT TRUNCATED AT 400 WORDS)

    Topics: Adenosine Triphosphate; Animals; Atenolol; Benzazepines; Blood Pressure; Bradycardia; Cardiovascular Agents; Coronary Circulation; Disease Models, Animal; Female; Heart; Heart Rate; Injections, Intravenous; Lactates; Male; Myocardial Infarction; Myocardial Ischemia; Myocardium; Oxygen Consumption; Phosphocreatine; Reperfusion Injury; Swine; Swine, Miniature

1995
Iloprost infusion decreases skeletal muscle ischemia-reperfusion injury.
    Journal of vascular surgery, 1990, Volume: 11, Issue:1

    Iloprost (a long-acting prostacyclin analog) has been demonstrated to decrease cardiac muscle infarct size after ischemia-reperfusion injury. We investigated the ability of iloprost to decrease skeletal muscle injury and platelet sequestration after ischemia-reperfusion injury in a canine bilateral isolated gracilis muscle model. Anesthesized animals (n = 13) were subjected to 6 hours of gracilis muscle ischemia and 1 hour of reperfusion. Fifteen minutes before muscle reperfusion, the animals were infused with radium 111-labeled autogenous platelets. Experimental animals (n = 7) received a continuous preischemic intravenous infusion of iloprost (0.45 microgram/kg/hr) and two 0.45 microgram/kg intravenous injections of iloprost (10 minutes before the ischemic interval and 10 minutes before reperfusion). Muscle injury was measured with triphenyltetrazolium chloride histochemical staining. Platelet sequestration within ischemic muscle specimens was determined by measuring indium 111 activity in a gamma counter. Iloprost infusion decreased muscle infarct size from 57.0% +/- 12.6% in control animals to 15.8% +/- 4.4% in experimental animals (p less than 0.05). Platelet uptake in experimental and control muscle was 1.2 +/- 0.21 x 10(7) and 2.17 +/- 0.48 x 10(7) platelets/gm ischemic muscle, respectively (p = 0.1). Although platelet sequestration was not altered significantly in this experiment, a reduction in skeletal muscle injury was confirmed. Further investigation on the mechanisms of action of iloprost in chronic and acute skeletal muscle ischemia is warranted.

    Topics: Animals; Blood Platelets; Blood Pressure; Body Temperature; Cardiovascular Agents; Dogs; Epoprostenol; Extremities; Female; Iloprost; Male; Muscles; Muscular Diseases; Reperfusion Injury

1990