Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Cardiovasc Diabetol ; 19(1): 7, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924211

ABSTRACT

BACKGROUND: Diabetes mellitus type 2 (DM2) is a risk factor for developing heart failure but there is no specific therapy for diabetic heart disease. Sodium glucose transporter 2 inhibitors (SGLT2I) are recently developed diabetic drugs that primarily work on the kidney. Clinical data describing the cardiovascular benefits of SGLT2Is highlight the potential therapeutic benefit of these drugs in the prevention of cardiovascular events and heart failure. However, the underlying mechanism of protection remains unclear. We investigated the effect of Dapagliflozin-SGLT2I, on diabetic cardiomyopathy in a mouse model of DM2. METHODS: Cardiomyopathy was induced in diabetic mice (db/db) by subcutaneous infusion of angiotensin II (ATII) for 30 days using an osmotic pump. Dapagliflozin (1.5 mg/kg/day) was administered concomitantly in drinking water. Male homozygous, 12-14 weeks old WT or db/db mice (n = 4-8/group), were used for the experiments. Isolated cardiomyocytes were exposed to glucose (17.5-33 mM) and treated with Dapagliflozin in vitro. Intracellular calcium transients were measured using a fluorescent indicator indo-1. RESULTS: Angiotensin II infusion induced cardiomyopathy in db/db mice, manifested by cardiac hypertrophy, myocardial fibrosis and inflammation (TNFα, TLR4). Dapagliflozin decreased blood glucose (874 ± 111 to 556 ± 57 mg/dl, p < 0.05). In addition it attenuated fibrosis and inflammation and increased the left ventricular fractional shortening in ATII treated db/db mice. In isolated cardiomyocytes Dapagliflozin decreased intracellular calcium transients, inflammation and ROS production. Finally, voltage-dependent L-type calcium channel (CACNA1C), the sodium-calcium exchanger (NCX) and the sodium-hydrogen exchanger 1 (NHE) membrane transporters expression was reduced following Dapagliflozin treatment. CONCLUSION: Dapagliflozin was cardioprotective in ATII-stressed diabetic mice. It reduced oxygen radicals, as well the activity of membrane channels related to calcium transport. The cardioprotective effect manifested by decreased fibrosis, reduced inflammation and improved systolic function. The clinical implication of our results suggest a novel pharmacologic approach for the treatment of diabetic cardiomyopathy through modulation of ion homeostasis.


Subject(s)
Benzhydryl Compounds/pharmacology , Blood Glucose/drug effects , Diabetes Mellitus/drug therapy , Diabetic Cardiomyopathies/prevention & control , Glucosides/pharmacology , Myocytes, Cardiac/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Ventricular Function, Left/drug effects , Angiotensin II , Animals , Biomarkers/blood , Blood Glucose/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Diabetes Mellitus/blood , Diabetic Cardiomyopathies/chemically induced , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Fibrosis , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/metabolism , Sodium-Hydrogen Exchanger 1/metabolism
2.
Article in English | MEDLINE | ID: mdl-29707604

ABSTRACT

We have previously shown that an Epoxyeicosatrienoic Acid (EET) -agonist has pleiotropic effects and reverses cardiomyopathy by decreasing inflammatory molecules and increasing antioxidant signaling. We hypothesized that administration of an EET agonist would increase Peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α), which controls mitochondrial function and induction of HO-1 and negatively regulates the expression of the proinflammatory adipokines CCN3/NOV in cardiac and pericardial tissues. This pathway would be expected to further improve left ventricular (LV) systolic function as well as increase insulin receptor phosphorylation. Measurement of the effect of an EET agonist on oxygen consumption, fractional shortening, blood glucose levels, thermogenic and mitochondrial signaling proteins was performed. Control obese mice developed signs of metabolic syndrome including insulin resistance, hypertension, inflammation, LV dysfunction, and increased NOV expression in pericardial adipose tissue. EET agonist intervention decreased pericardial adipose tissue expression of NOV, while normalized FS, increased PGC-1α, HO-1 levels, insulin receptor phosphorylation and improved mitochondrial function, theses beneficial effect were reversed by deletion of PGC-1α. These studies demonstrate that an EET agonist increases insulin receptor phosphorylation, mitochondrial and thermogenic gene expression, decreased cardiac and pericardial tissue NOV levels, and ameliorates cardiomyopathy in an obese mouse model of the metabolic syndrome.

3.
Biochem Pharmacol ; 85(11): 1626-33, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23537701

ABSTRACT

UNLABELLED: Tetrahydrocannabinol (THC), the major psychoactive component of marijuana, is a cannabinoid agonist that exerts its effects by activating at least two specific receptors (CB1 and CB2) that belong to the seven transmembrane G-protein coupled receptor (GPCR) family. Both CB1 and CB2 mRNA and proteins are present in the heart. THC treatment was beneficial against hypoxia in neonatal cardiomyocytes in vitro. We also observed a neuroprotective effect of an ultra low dose of THC when applied to mice before brain insults. The present study was aimed to test and characterize the cardioprotective effects of a very low dose (0.002mg/kg) of THC which is 3-4 orders of magnitude lower than the conventional doses, administered before myocardial infarction in mice in vivo. Three regimens of THC administration were tested: single THC application 2h or 48h before the induction of infarct, or 3 weeks continuous treatment before MI. All protocols of THC administration were found to be beneficial. In the case of THC treatment 2h before MI, fractional shortening was elevated (37±4% vs. 42±1%, p<0.04), troponin T leakage to the blood was reduced (14±3ng/ml vs. 10±4ng/ml, p<0.008), infarct size decreased (29±4% vs. 23±4%, p<0.02), and the accumulation of neutrophils to the infarct area declined (36±10cells/field vs. 19±4cells/field, p<0.007) in THC- compared to vehicle-pretreated mice, 24h after MI. ERK1/2 phosphorylation following infarct was also inhibited by pre-treatment with THC (p<0.01). CONCLUSION: A single ultra low dose of THC before ischemia is a safe and effective treatment that reduces myocardial ischemic damage.


Subject(s)
Cardiotonic Agents/pharmacology , Dronabinol/pharmacology , Animals , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/prevention & control , Phosphorylation , Tumor Necrosis Factor-alpha/blood
4.
Apoptosis ; 18(5): 547-55, 2013 May.
Article in English | MEDLINE | ID: mdl-23435964

ABSTRACT

Ischemia/reperfusion (I/R) injury is the main cause of primary graft dysfunction of liver allografts. Cobalt-protoporphyrin (CoPP)-dependent induction of heme oxygenase (HO)-1 has been shown to protect the liver from I/R injury. This study analyzes the apoptotic mechanisms of HO-1-mediated cytoprotection in mouse liver exposed to I/R injury. HO-1 induction was achieved by the administration of CoPP (1.5 mg/kg body weight i.p.). Mice were studied in in vivo model of hepatic segmental (70 %) ischemia for 60 min and reperfusion injury. Mice were randomly allocated to four main experimental groups (n = 10 each): (1) A control group undergoing sham operation. (2) Similar to group 1 but with the administration of CoPP 72 h before the operation. (3) Mice undergoing in vivo hepatic I/R. (4) Similar to group 3 but with the administration of CoPP 72 h before ischemia induction. When compared with the I/R mice group, in the I/R+CoPP mice group, the increased hepatic expression of HO-1 was associated with a significant reduction in liver enzyme levels, fewer apoptotic hepatocytes cells were identified by morphological criteria and by immunohistochemistry for caspase-3, there was a decreased mean number of proliferating cells (positively stained for Ki67), and a reduced hepatic expression of: C/EBP homologous protein (an index of endoplasmic reticulum stress), the NF-κB's regulated genes (CIAP2, MCP-1 and IL-6), and increased hepatic expression of IκBa (the inhibitory protein of NF-κB). HO-1 over-expression plays a pivotal role in reducing the hepatic apoptotic IR injury. HO-1 may serve as a potential target for therapeutic intervention in hepatic I/R injury during liver transplantation.


Subject(s)
Heme Oxygenase-1/biosynthesis , Hepatocytes/enzymology , Liver/enzymology , Membrane Proteins/biosynthesis , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Biomarkers/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cytoprotection/drug effects , Enzyme Induction/drug effects , Gene Expression , Heme Oxygenase-1/genetics , Hepatocytes/drug effects , Hepatocytes/pathology , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Injections, Intraperitoneal , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Liver/drug effects , Liver/injuries , Male , Membrane Proteins/genetics , Mice , Protoporphyrins/pharmacology
5.
Eur J Pharm Sci ; 45(5): 592-9, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22234377

ABSTRACT

Butyroyloxymethyl diethylphosphate (AN-7) is a prodrug of butyric acid effective in reducing cardiotoxicity caused by chemotherapy. In this study, we tested whether AN-7 protects the heart and cardiomyocytes against ischemia injury. A single oral dose of AN-7 was given to mice or rats. Animals were sacrificed 1.5 or 24 h later and the hearts were subjected to ischemia and reperfusion ex-vivo (Langendorff). The mechanical performance was recorded throughout and the infarct size was measured at the end of reperfusion. Neonatal rat cardiomyocytes were subjected to 24-48 h hypoxia (1% O(2)) in the absence or presence of AN-7 and mitochondria damage and cell death were assessed. Proteins were analyzed by Western immunoblotting. In the two rodents, a single dose of AN-7 given in vivo preconditioned the hearts for improved functional recovery from ischemia and reperfusion performed ex-vivo. Both 1.5 h and 24 h treatments improved the pressure-related parameters whereas the coronary flow was ameliorated in the 24 h treatment only. Infarct size was smaller in the AN-7 treated hearts. In cardiomyocytes, AN-7 diminished the hypoxia induced dissipation of mitochondria membrane potential and cell death. Compared with untreated controls, AN-7-treated hearts recovering from global ischemia and cardiomyocytes undergoing hypoxia, displayed significantly higher levels of the cytoprotective heme oxygenase-1. Our findings indicate that AN-7 imparts cardioprotection against ischemia both in vivo and in vitro and emerges as a potential treatment modality for cardiac injury.


Subject(s)
Heart/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Organophosphates/pharmacology , Prodrugs/pharmacology , Animals , Butyrates/pharmacology , Cell Death/drug effects , Heart/physiopathology , Heme Oxygenase-1/metabolism , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Organophosphorus Compounds/pharmacology , Rats , Rats, Wistar , Regional Blood Flow , Reperfusion
6.
Mol Cell Biochem ; 363(1-2): 167-78, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22160856

ABSTRACT

The hypothesis of the present study is that cardiomyocytes subjected to prolonged ischemia, may release survival factors that will protect new cardiac cells from ischemic stress. We exposed neonatal rat cardiomyocyte primary cultures to hypoxia, collected the supernatant, treated intact cardiac cells by this posthypoxic supernatant, and exposed them to hypoxia. The results show cardioprotection of the treated cells compared with the untreated ones. We named the collected posthypoxic supernatant "conditioned medium" (CM), which acts in a dose-dependent manner to protect new cardiac cells from hypoxia: 100 or 75% of CM diluted in phosphate-buffered saline (PBS) protected cells as if they were not exposed to hypoxia (P < 0.001). When CM was removed from the cells before hypoxia, protection was not observed. CM also protected skeletal muscle cultures from hypoxia, but not cardiac cells against H(2)O(2)-induced cell damage. Finally, CM treatment protected the isolated heart in Langendorff set-up against ischemia. Smaller infarct size (9.9 ± 4.4% vs. 28.3 ± 8.5%, P < 0.05), better Rate Pressure Product (67 ± 11% vs. 48.6 ± 13.4%, P < 0.05) and better rate of contraction and relaxation were observed following ischemia and reperfusion (1341 ± 399 mmHg/s vs. 951 ± 349 mmHg/s, P < 0.05 and 1053 ± 347 mmHg/s vs. 736 ± 314 mmHg/s, P < 0.05). To conclude, there are factors that are released from the heart cells subjected to ischemia/hypoxia that protects cardiomyocytes from ischemic stress.


Subject(s)
Autocrine Communication , Myocardial Infarction/prevention & control , Myocardial Ischemia/prevention & control , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cell Hypoxia , Cells, Cultured , Culture Media, Conditioned/metabolism , Hydrogen Peroxide/toxicity , Male , Muscle Fibers, Skeletal/metabolism , Myocardial Contraction , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Perfusion , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Ventricular Function , Ventricular Pressure
7.
Biochem Pharmacol ; 82(9): 1126-33, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21839729

ABSTRACT

Pyrimidine nucleotides are signaling molecules, which activate G protein-coupled membrane receptors of the P2Y family. P2Y(2) and P2Y(4) receptors are part of the P2Y family, which is composed of 8 subtypes that have been cloned and functionally defined. We have previously found that uridine-5'-triphosphate (UTP) reduces infarct size and improves cardiac function following myocardial infarct (MI). The aim of the present study was to determine the role of P2Y(2) receptor in cardiac protection following MI using knockout (KO) mice, in vivo and wild type (WT) for controls. In both experimental groups used (WT and P2Y(2)(-/-) receptor KO mice) there were 3 subgroups: sham, MI, and MI+UTP. 24h post MI we performed echocardiography and measured infarct size using triphenyl tetrazolium chloride (TTC) staining on all mice. Fractional shortening (FS) was higher in WT UTP-treated mice than the MI group (44.7±4.08% vs. 33.5±2.7% respectively, p<0.001). However, the FS of P2Y(2)(-/-) receptor KO mice were not affected by UTP treatment (34.7±5.3% vs. 35.9±2.9%). Similar results were obtained with TTC and hematoxylin and eosin stainings. Moreover, troponin T measurements demonstrated reduced myocardial damage in WT mice pretreated with UTP vs. untreated mice (8.8±4.6 vs. 12±3.1 p<0.05). In contrast, P2Y(2)(-/-) receptor KO mice pretreated with UTP did not demonstrate reduced myocardial damage. These results indicate that the P2Y(2) receptor mediates UTP cardioprotection, in vivo.


Subject(s)
Myocardial Infarction/drug therapy , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/metabolism , Uridine Triphosphate/therapeutic use , Animals , Diphosphates/metabolism , Genotype , Inflammation/metabolism , L-Lactate Dehydrogenase/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardium/cytology , Myocardium/metabolism , Troponin T/blood , Ventricular Remodeling/drug effects
8.
Mol Cell Biochem ; 345(1-2): 153-60, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20730620

ABSTRACT

Activation of either the A(1) adenosine receptor (A(1)R) or the A(3) adenosine receptor (A(3)R), by their specific agonists CCPA and Cl-IB-MECA, respectively, protects cardiac cells in culture against ischemic injury. Yet the full protective mechanism remains unclear. In this study, we therefore examined the involvement of p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinases (ERK) phosphorylation in this protective intracellular signaling mechanism. Furthermore, we investigated whether p38 MAPK phosphorylation occurs upstream or downstream from the opening of mitochondrial ATP-sensitive potassium (K(ATP)) channels. The role of p38 MAPK activation in the intracellular signaling process was studied in cultured cardiomyocytes subjected to hypoxia, that were pretreated with CCPA or Cl-IB-MECA or diazoxide (a mitochondrial K(ATP) channel opener) with and without SB203580 (a specific inhibitor of phosphorylated p38 MAPK). Cardiomyocytes were also pretreated with anisomycin (p38 MAPK activator) with and without 5-hydroxy decanoic acid (5HD) (a mitochondrial K(ATP) channel blocker). SB203580 together with the CCPA, Cl-IB-MECA or diazoxide abrogated the protection against hypoxia as shown by the level of ATP, lactate dehydrogenase (LDH) release, and propidium iodide (PI) staining. Anisomycin protected the cardiomyocytes against ischemic injury and this protection was abrogated by SB203580 but not by 5HD. Conclusions Activation of A(1)R or A(3)R by CCPA or Cl-IB-MECA, respectively, protects cardiomyocytes from hypoxia via phosphorylation of p38 MAPK, which is located downstream from the mitochondrial K(ATP) channel opening. Elucidating the signaling pathway by which adenosine receptor agonists protect cardiomyocytes from hypoxic damage, will facilitate the development of anti ischemic drugs.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Hypoxia/prevention & control , Myocytes, Cardiac/pathology , p38 Mitogen-Activated Protein Kinases/physiology , Adenosine/analogs & derivatives , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart , Hypoxia/diet therapy , Myocytes, Cardiac/drug effects , Phosphorylation , Potassium Channels , Protective Agents/pharmacology , Rats
9.
Cell Biochem Biophys ; 47(1): 11-20, 2007.
Article in English | MEDLINE | ID: mdl-17406056

ABSTRACT

We have previously found that, following myocardial ischemia/reperfusion injury, isolated hearts from bax gene knockout mice [Bax(-/-)] exhibited higher cardioprotection than the wild-type. We here explore the effect of Bax(-/-), following myocardial infarction (MI) in vivo. Homozygotic Bax(-/-) and matched wild-type were studied. Mice underwent surgical ligation of the left anterior descending coronary artery (LAD). The progressive increase in left-ventricular end diastolic diameter, end systolic diameter, in Bax(-/-) was significantly smaller than in Bax(+/+) at 28 d following MI (p < 0.03) as seen by echocardiography. Concomitantly, fractional shortening was higher (35 +/- 4.1% and 27 +/- 2.5%, p < 0.001) and infarct size was smaller in Bax(-/-) compared to the wild-type at 28 days following MI (24 +/- 3.7 % and 37 +/- 3.3%, p < 0.001). Creatine kinase and lactate dehydrogenase release in serum were lower in Bax(-/-) than in Bax(+/+) 24 h following MI. Caspase 3 activity was elevated at 2 h after MI only in the wild-type, but reduced to baseline values at 1 and 28 d post-MI. Bax knockout mice hearts demonstrated reduced infarct size and improved myocardial function following permanent coronary artery occlusion. The Bax gene appears to play a significant role in the post-MI response that should be further investigated.


Subject(s)
Myocardial Infarction/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis , Body Weight , Caspases/metabolism , Echocardiography/methods , Female , Heterozygote , Homozygote , Male , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Time Factors
10.
Apoptosis ; 10(6): 1261-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16215674

ABSTRACT

BACKGROUND: A major mechanism underlying warm ischemia/reperfusion (I/R) injury during liver transplantation is the activation of the caspase chain, which leads to apoptosis. Recently, it was demonstrated that the release of cathepsin B, a cysteine protease, from the cytosol in liver injury induces mitochondrial release of cytochrome c and the activation of caspase-3 and -9, thereby leading to apoptosis. The aim of this study was to ascertain if cathepsin B inactivation attenuates the apoptotic injury due to I/R in mouse liver. METHODS: A model of segmental (70%) hepatic ischemia was used. Eighteen mice were anesthetized and randomly divided into three groups: (1) CONTROL GROUP: sham operation (laparotomy); (2) Ischemic group: midline laparotomy followed by occlusion of all structures in the portal triad to the left and median lobes for 60 min (ischemic period); (3) STUDY GROUP: like group 2, but with intraperitoneal administration of a pharmacological inhibitor of cathepsin B (4 mg/100 g) 30 min before induction of ischemia. Serum liver enzyme levels were measured by biochemical analysis, and intrahepatic caspase-3 activity was measured by fluorometric assay; apoptotic cells were identified by morphological criteria, the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) fluorometric assay, and immunohistochemistry for caspase-3. RESULTS: Showed that at 6 h of reperfusion, there was a statistically significant reduction in liver enzyme levels in the animals pretreated with cathepsin B inhibitor (p<0.05). On fluorometric assay, caspase-3 activity was significantly decreased in group 3 compared to group 2 (p<0.0001). The reduction in postischemic apoptotic hepatic injury in the cathepsin B inhibitor -treated group was confirmed morphologically, by the significantly fewer apoptotic hepatocyte cells detected (p<0.05); immunohistochemically, by the significantly weaker activation of caspase-3 compared to the ischemic group (p<0.05); and by the TUNEL assay (p<0.05). CONCLUSION: The administration of cathepsin B inhibitor before induction of ischemia can attenuate postischemic hepatocyte apoptosis and thereby minimize liver damage. Apoptotic hepatic injury seems to be mediated through caspase-3 activity. These findings have important implications for the potential use of cathepsin B inhibitors in I/R injury during liver transplantation.


Subject(s)
Apoptosis , Cathepsin B/metabolism , Liver/enzymology , Liver/pathology , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Animals , Caspase 3/metabolism , Enzyme Activation , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL
11.
Apoptosis ; 10(5): 955-62, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16151631

ABSTRACT

Ischemia/reperfusion injury during liver transplantation is a major cause of primary nonfunctioning graft for which there is no effective treatment other than retransplantation. Adenosine prevents ischemia-reperfusion-induced hepatic injury via its A2A receptors. The aim of this study was to investigate the role of A2A receptor agonist on apoptotic ischemia/reperfusion-induced hepatic injury in rats. Isolated rat livers within University of Wisconsin solution were randomly divided into four groups: (1) continuous perfusion of Krebs-Henseleit solution through the portal vein for 165 minutes (control); (2) 30-minute perfusion followed by 120 minutes of ischemia and 15 minutes of reperfusion; (3) like group 2, but with the administration of CGS 21680, an A2A receptor agonist, 30 microg/100 ml, for 1 minute before ischemia; (4) like group 3, but with administration of SCH 58261, an A2A receptor antagonist. Serum liver enzyme levels were measured by biochemical analysis, and intrahepatic caspase-3 activity was measured by fluorometric assay; apoptotic cells were identified by morphological criteria, the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) fluorometric assay, and immunohistochemistry for caspase-3. Results showed that at 1 minute of reperfusion, there was a statistically significant reduction in liver enzyme levels in the animals pretreated with CGS (p < 0.05). On fluorometric assay, caspase-3 activity was significantly decreased in group 3 compared to group 2 (p < 0.0002). The reduction in postischemic apoptotic hepatic injury in the CGS-treated group was confirmed morphologically, by the significantly fewer apoptotic hepatocyte cells detected (p < 0.05); immunohistochemically, by the significantly weaker activation of caspase-3 compared to the ischemic group (p < 0.05); and by the TUNEL assay (p < 0.05). In conclusion, the administration of A2A receptor agonist before induction of ischemia can attenuate postischemic apoptotic hepatic injury and thereby minimize liver injury. Apoptotic hepatic injury seems to be mediated through caspase-3 activity.


Subject(s)
Adenosine A2 Receptor Agonists , Adenosine/analogs & derivatives , Liver/blood supply , Phenethylamines/therapeutic use , Reperfusion Injury/drug therapy , Adenosine/therapeutic use , Animals , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , In Situ Nick-End Labeling , In Vitro Techniques , Liver/enzymology , Male , Rats , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control
12.
Apoptosis ; 10(1): 53-62, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15711922

ABSTRACT

The release of cardioactive substances during hepatic ischemia/reperfusion injury generates toxic free radicals that inflict hepatic and remote cardiac damage. The aim of the study was to determine whether TPEN, a potent iron chelator, ameliorates the apoptotic hepatic and cardiac function injuries. Three groups of isolated rat livers were studied: (1) continuously perfused with Krebs-Henseleit solution; (2) subjected to 120 min of ischemia and 15 min of reperfusion; (3) as in group 2, with TPEN administered prior to ischemia. Isolated hearts were perfused for 65 min with the effluent of the reperfused livers. Results showed that TPEN administration reduced the release of norepinephrine, epinephrine, dopamine, prostaglandin E2 and angiotensin II, decreased intrahepatic caspase-3 activity, and decreased the mean hepatocyte apoptotic index (TUNEL assay) (p = 0.001). Perfusion with post-ischemic hepatic effluent caused a transient 15-min increase in left ventricular contraction and coronary flow (p < 0.05), followed by a decrease in cardiac function at one hour. TPEN reduced the transient elevation in left ventricular contraction p < 0.05), but did not prevent the subsequent decrease in cardiac function. In conclusion, TPEN attenuates post-ischemic apoptotic hepatic injury by modulating caspase-3-like activity and reduces the cardioactive substances released from the liver.


Subject(s)
Apoptosis/drug effects , Chelating Agents/pharmacology , Ethylenediamines/pharmacology , Ischemia/therapy , Liver/blood supply , Reperfusion Injury/prevention & control , Animals , Caspases/physiology , Liver/pathology , Male , Rats , Rats, Wistar , Time Factors
13.
Med Sci Monit ; 7(6): 1137-44, 2001.
Article in English | MEDLINE | ID: mdl-11687721

ABSTRACT

BACKGROUND: N-acetyl-L-cysteine (NAC) both replenishes reduced glutathione (GSH) and mitigates reperfusion injury. We hypothesized that liver content of GSH could affect remote myocardial reperfusion injury following liver ischemia-reperfusion. MATERIAL AND METHODS: Following stabilization (30 min), isolated rat livers (6/group) were perfused with Krebs-Henseleit solution (two control groups) or made globally ischemic (two ischemia groups) for 120 min. Paired livers + paced hearts (Langendorff preparation) were then reperfused for 15 min after which the hearts were recirculated alone for 50 min. NAC was added to Krebs (2 mM) that perfused livers during stabilization and reperfusion phases in one control and one ischemia group. RESULTS: GSH levels in the two control liver groups were identical (30.1 +/- 5.7 [SD] nmol/mg protein), and similar to that of the ischemia + NAC livers (28.6 +/- 2.8) but 2-fold that of the ischemia + 0 livers (15.8 +/- 2.4 nmol/mg protein, p<0.05). While hearts paired with control livers maintained unchanged their myocardial velocity of contraction, the contraction in the ischemia + NAC-paired hearts reduced, but was better than in the ischemia + 0-paired hearts (71 +/- 8% vs. 41 +/- 6% off baseline, p<0.05). Coronary flow also decreased dissimilarly in the two ischemia-associated groups of heart: 72 +/- 9% (ischemia + NAC) vs. 46 +/- 7% (ischemia + 0, p<0.05). Xanthine oxidase in the ischemia + 0 livers was 7.5-folds higher than in the ischemia-treated livers. CONCLUSIONS: NAC treatment of ischemia-reperfused livers, associated with GSH replenishment, prevents remote myocardial reperfusion injury. The role of NAC and GSH in reducing liver-associated oxidative burst propagation is discussed.


Subject(s)
Glutathione/metabolism , Ischemia/metabolism , Liver/blood supply , Liver/metabolism , Myocardial Reperfusion Injury/prevention & control , Acetylcysteine/administration & dosage , Acetylcysteine/pharmacology , Animals , In Vitro Techniques , Male , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Wistar
14.
Med Sci Monit ; 7(6): 1145-52, 2001.
Article in English | MEDLINE | ID: mdl-11687722

ABSTRACT

BACKGROUND: We recently demonstrated that isolated paced hearts perfused with modified Krebs-Henseleit solution containing high dose allopurinol (1 mM) were protected from liver ischemia-reperfusion (IR)-induced reperfusion injury. The objective was to study the effects of low dose allopurinol together with external pacing in attenuating myocardial reperfusion dysfunction following liver IR in the same double organ model. MATERIAL AND METHODS: Isolated rat livers were perfused with modified Krebs-Henseleit solution (groups 1 and 2, n=8/all groups) or underwent global ischemia (groups 3-6) for 120 minutes. Following a 15-minute conjoint reperfusion of earlier separately isolated liver+heart, the hearts were recirculated alone for additional 45 minutes. The organs of three-group donating animals (groups 2, 4, and 6) were treated with allopurinol 18 hours and 1 hour before the experiment (50 mg x kg(-1) intraperitoneally) and it was also added during perfusion (0.1 mM in Krebs). The hearts in groups 5 and 6 were paced (300 x min(-1)). RESULTS: The hearts perfused with IR Krebs (group 3) experienced decreased myocardial left ventricular-developed pressure (LVP), heart rate (in the unpaced hearts) and later coronary flow (by 68%, 21% and 32%, respectively); LVP and coronary flow also decreased correspondingly in the IR-paced hearts (group 4). Xanthine oxidase (XO) was high in groups 3 and 4 compared to group 1. IR allopurinol-treated hearts, both unpaced and paced (groups 5 and 6) had normal, similar myocardial performance, while their circulating XO was as low as in group 2 (allopurinol-treated controls). CONCLUSIONS: External pacing in the double organ, isolated-perfused liver-heart did not ameliorate XO-mediated dysfunction compared to low dose allopurinol. The unexpected delayed coronary insufficiency in myocardial reperfusion injury is discussed.


Subject(s)
Allopurinol/pharmacology , Ischemia/physiopathology , Liver/blood supply , Myocardial Reperfusion Injury/prevention & control , Pacemaker, Artificial , Animals , In Vitro Techniques , Models, Animal , Rats
15.
Exp Clin Cardiol ; 6(1): 29-34, 2001.
Article in English | MEDLINE | ID: mdl-20428441

ABSTRACT

BACKGROUND: Cardiovascular dysfunction frequently occurs after major vascular surgery or liver transplantation. OBJECTIVE: To evaluate the effects on myocardial activity of vasoactive agents released from ischemic-reperfused liver. ANIMALS AND METHODS: Isolated rat livers were perfused with Krebs-Henseleit solution (KH), propranolol 10(-5) M, losartan 2x10(-5) M and indomethacin 10(-5) M, then made globally ischemic for 120 min (37 degrees C) and reperfused. Isolated hearts from other rats were stabilized with KH and reperfused for 15 min with the perfusate exiting the livers. Livers were disconnected, and the hearts continued to be recirculated with the accumulated liver and heart effluent for an additional 50 min. Enzyme leakage, different vasoactive substances, left ventricular developed pressure (LVP) and coronary flow were measured during the experimental protocol. RESULTS: Hepatic release of adrenaline, noradrenaline, angiotensin II, prostaglandin E(2) and thromboxane B(2) was significantly increased in the liver effluent following ischemia. When this effluent was directed to the heart, LVP was significantly raised in the first 10 min of reperfusion (137+/-5%) followed by marked decreased (46+/-6%) during the following 65 min of myocardial reperfusion. In the ischemic-reperfused drug-treated groups, the initial positive effect on LVP was milder than in controls (propranolol 112+/-12%, losartan 111+/-11%, indomethacin 113+/-9%) and the final LVP was lower (propranolol 29+/-6%, losartan 27+/-7% [P<0.05 versus ischemic control], indomethacin 46 +/-12%). CONCLUSION: During the initial phase of reperfusion, vasoactive substances released in the hepatic effluent potentiated LVP of the hearts exposed to this effluent. When the three inhibitory drugs were added to KH, this initial augmentation was not sustained. Propranolol and losartan, but not indomethacin, further depressed LVP. Vasoactive substances released from ischemic reperfused livers directly influenced heart function.

16.
Isr Med Assoc J ; 2(6): 450-4, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10897237

ABSTRACT

BACKGROUND: The beneficial effect of aprotinin, a naturally occurring protease inhibitor, on preservation of organs such as the liver, kidney and lung has been documented. OBJECTIVE: To explore the effects of hepatic ischemia and reperfusion on both liver and myocardial function, using a dual isolated perfused organ model with and without aprotinin. METHODS: Isolated rat livers were stabilized for 30 minutes with oxygenated modified Krebs-Henseleit solution at 37 degrees C. Livers were then perfused continuously with KH or KH + aprotinin 10(6) KIU/L for an additional 135 min. Livers of two other groups were made globally ischemic for 120 min, then perfused for 15 min with KH or with KH + aprotinin. Isolated hearts (Langendorff preparation) were stabilized for 30 min and then reperfused with KH or KH + aprotinin exiting the liver for 15 min. The liver's circuit was disconnected, and hearts were re-circulated with the accumulated liver + heart effluent for an additional 50 min. RESULTS: In the ischemia and ischemia + aprotinin groups, portal vein pressure (1 and 15 min reperfusion) was 331 +/- 99% and 339 +/- 61% vs. 308 +/- 81% and 193 +/- 35% of baseline, respectively (P < 0.03 vs. ischemia). There were no other differences in the enzyme leakage between aprotinin-treated or untreated ischemic livers. Left ventricular pressure was stable in the controls. However, LV pressure in groups perfused with ischemic liver effluent declined within 65 min reperfusion, whether aprotinin treated or not (84 +/- 8% and 73 +/- 5% of baseline, respectively, P < 0.004 only for ischemia vs. control). CONCLUSION: When aprotinin was used, LV pressure was inclined to be higher while liver portal vein pressure was lower, thus providing protection against liver and heart reperfusion injury.


Subject(s)
Aprotinin/pharmacology , Heart/physiopathology , Liver/blood supply , Reperfusion Injury/physiopathology , Serine Proteinase Inhibitors/pharmacology , Animals , Heart/drug effects , Hemodynamics/drug effects , In Vitro Techniques , Male , Rats , Rats, Wistar
17.
J Trauma ; 47(4): 691-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10528603

ABSTRACT

OBJECTIVES: Cardiovascular, respiratory, and vascular dysfunction can follow trauma-induced no-flow-reflow states: hemorrhage, blunt trauma, or neurogenic shock. Liver ischemia-reperfusion (IR) induces remote lung damage by means of xanthine oxidase (XO) pro-oxidant activity. This damage was not proven in the heart, neither was the independent role of radical oxygen species (ROS) established in such cases. We investigated whether multiple organ dysfunction after a trauma-like IR is XO and ROS related and whether clinically used ROS scavengers could be beneficial. METHODS: A controlled, randomized trial in which isolated rat livers, hearts, lungs, and aortic rings were perfused with Krebs-Henseleit solutions. After stabilization, livers were either perfused or made ischemic (2 hours). Then, pairs of liver plus heart, lung, or ring were reperfused in series (15 minutes), and then the second organ circulated alone for 45 minutes. Remote organ protection against the pro-oxidant hepatic-induced toxicity was evaluated by using allopurinol (1 mmol/L, heart), mannitol (0.25 g/kg, lung), or methylene blue (40 mg/kg, ring). RESULTS: IR liver effluents typically contained high lactate dehydrogenase, XO, and uric acid concentrations compared with control organs. IR was associated with doubled lung peak inspiratory pressure and reduced static compliance. Myocardial velocity of contraction and relaxation decreased by one third of baseline, and rings contracted abnormally and responded inadequately to phenylephrine. Wet-weight to dry-weight ratios in the remote organs increased as well. Most remote reperfusion injuries were attenuated by the drugs. CONCLUSION: Liver no-flow-reflow directly induces myocardial, pulmonary, and vascular dysfunction. These are likely mediated by XO and ROS. The tested drugs protected against these pro-oxidants, even in the presence of circulating XO.


Subject(s)
Aorta/metabolism , Heart Arrest/complications , Liver/metabolism , Lung/metabolism , Multiple Organ Failure/etiology , Multiple Organ Failure/metabolism , Multiple Trauma/complications , Myocardium/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Xanthine Oxidase/metabolism , Allopurinol/therapeutic use , Animals , Coronary Vessels , Disease Models, Animal , Drug Evaluation, Preclinical , Free Radical Scavengers/therapeutic use , In Vitro Techniques , Liver/blood supply , Lung/blood supply , Male , Mannitol/therapeutic use , Methylene Blue/therapeutic use , Multiple Organ Failure/drug therapy , Random Allocation , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Xanthine Oxidase/antagonists & inhibitors
18.
Anesth Analg ; 88(4): 710-6, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10195509

ABSTRACT

UNLABELLED: The administration of protamine sulfate (protamine) to reverse the action of heparin is associated with adverse reactions. We studied the effects of protamine and isoflurane on isolated, perfused rat hearts previously subjected to cardioplegic ischemia. Hearts were perfused with oxygenated Krebs-Henseleit (KH) solution for 30 min, then subjected to cardioplegic ischemia for 30 min (KCl 16 mEq/L at 31 degrees C) and 5 min reperfusion. Drug exposure lasted 15 min, and the recovery period was 60 min. Test groups were control, protamine (10 microg/mL), isoflurane (1.5%), protamine +/- isoflurane, sodium nitroprusside (SNP) (2.5 ng/mL), and SNP +/- protamine. Left ventricular developed pressure (LVP), coronary flow, and myocardial oxygen consumption were depressed by protamine to 30% +/- 4%, 47% +/- 4%, and 39% +/- 4% of baseline (P < 0.001 versus control), respectively. Isoflurane and SNP afforded partial protection from the effects of protamine: LVP was 57% +/- 5% and 51% +/- 3% of baseline, respectively (P < 0.05 versus protamine alone and control); coronary flow was 70% +/- 6% and 97% +/- 12% of baseline, respectively (P < 0.05 versus protamine alone; P < 0.05 for isoflurane versus control); and O2 consumption was 69% +/- 6% and 88% +/- 15% of baseline, respectively (P < 0.05 versus protamine; P < 0.05 for isoflurane versus control). In this model, protamine-induced myocardial depression and coronary vasoconstriction were less pronounced in the presence of either isoflurane or SNP. IMPLICATIONS: We examined the interactions of isoflurane, sodium nitroprusside, and protamine in a rat heart model and found that both isoflurane and sodium nitroprusside partially protect the heart from the depressant effects of protamine. This finding is significant, as these drugs are often used in heart surgery.


Subject(s)
Anesthetics, Inhalation/pharmacology , Heparin Antagonists/pharmacology , Isoflurane/pharmacology , Myocardial Ischemia/physiopathology , Nitroprusside/pharmacology , Protamines/pharmacology , Vasodilator Agents/pharmacology , Animals , Drug Interactions , Heart Arrest, Induced , Hemodynamics , Male , Myocardial Reperfusion , Oxygen Consumption , Rats , Rats, Wistar
19.
Phys Med Biol ; 43(10): 3129-37, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9814539

ABSTRACT

The current induced in the outer circuit of a fast response ionization chamber exposed to pulsed radiation consists of two components, a fast one induced by free electrons and a slow one induced by ions. The fast electron component may be used for the representation of the shape of the ionizing pulse. In order to avoid interference with the slow ion current, the latter has to be removed from the signal. This is achieved by deriving a voltage course from the chamber signal which fits the shape of the ion component and subtracting this from the entire signal. The function of the electronic circuit used for this purpose is described. Some considerations about the time resolution of the chamber gas are to be found in the appendix.


Subject(s)
Radiation, Ionizing , Electronics , Electrons , Ions , Particle Accelerators
20.
J Trauma ; 43(4): 627-33; discussion 633-5, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9356059

ABSTRACT

OBJECTIVES: To investigate whether liver ischemia and reperfusion (IR) directly affect functions of remote organs. BACKGROUND: Cardiovascular and respiratory dysfunction follows hemorrhage, spinal shock, or trauma as a result of no-flow-reflow phenomena. Hepatic IR induces remote organ damage probably by xanthine oxidase and oxygen species. MATERIALS AND METHODS: Isolated rat livers, lungs, and hearts were perfused with Krebs-Henseleit solutions. After stabilization, livers were either perfused or made ischemic. Then, livers and hearts or livers and lungs were reperfused in series, and the liver was disconnected and the second organ continued to perfuse with the accumulated effluents. MEASUREMENTS AND MAIN RESULTS: Ischemic and reperfused liver effluent contained high lactate dehydrogenase and uric acid concentrations compared with controls; xanthine oxidase increased 60 to 100 times. Ischemic and reperfused lung peak inspiratory pressure almost doubled; airway static compliance halved; myocardial contractility decreased to 70% of baseline; wet weight-to-dry weight ratios of lungs and livers increased. CONCLUSION: Ischemic and reperfused liver can directly induce myocardial and pulmonary dysfunction, presumably by oxidant-induced injury.


Subject(s)
Heart Diseases/etiology , Liver/blood supply , Lung Diseases/etiology , Reperfusion Injury/complications , Acute Disease , Animals , Heart Diseases/physiopathology , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Liver/metabolism , Lung Diseases/physiopathology , Male , Myocardial Contraction , Rats , Rats, Wistar , Reperfusion Injury/physiopathology , Uric Acid/metabolism , Xanthine Oxidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...