Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biochem Mol Toxicol ; 32(11): e22213, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30152906

ABSTRACT

We investigated the effect of apigenin, a dietary flavonoid, on isoproterenol hydrochloride (ISO)-induced apoptotic signaling in cardiomyoblast H9C2 cells. The results showed that apigenin treatment (10 µM) prevented ISO (31.25 µM)-induced lipid peroxidative levels and antioxidants status in H9C2 cells. Furthermore, apigenin inhibited expression of inflammatory markers in ISO-treated cells. In addition, apigenin prevented ISO-induced DNA damage and apoptotic signaling through modulating the expression of Bax, caspase-3, -8 and -9, cytochrome c, and Fas proteins in H9C2 cells. It is concluded that apigenin prevents ISO-induced antioxidants depletion, oxidative DNA damage, inflammatory, and apoptotic signaling in H9C2 cells. Thus, the present results demonstrated that apigenin has a cardioprotective effect on cardiomyoblasts cells.


Subject(s)
Antioxidants/pharmacology , Apigenin/pharmacology , Apoptosis/drug effects , Cardiotonic Agents/adverse effects , Isoproterenol/adverse effects , Myoblasts, Cardiac/drug effects , Oxidative Stress/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biomarkers/metabolism , Cardiotonic Agents/antagonists & inhibitors , Cell Line , DNA Damage/drug effects , Inflammation Mediators/agonists , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Isoproterenol/antagonists & inhibitors , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial , Mitochondria, Heart/drug effects , Mitochondria, Heart/immunology , Mitochondria, Heart/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/immunology , Mitochondrial Membranes/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/immunology , Myoblasts, Cardiac/metabolism , Rats , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/metabolism , fas Receptor/agonists , fas Receptor/antagonists & inhibitors , fas Receptor/metabolism
2.
Biochem Biophys Res Commun ; 486(3): 774-780, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28343995

ABSTRACT

Ischemia/reperfusion (I/R) induces additional damage to the restoration of blood flow to ischemic myocardium. This study examined the effects of urolithin A (UA) on myocardial injury of ischemia/reperfusion in vivo and vitro and explored its underlying mechanisms. Mice were subjected to myocardial ischemia followed by reperfusion. Cells were subjected to hypoxia followed by reoxygenation. UA alleviated hypoxia/reoxygenation (H/R) injury in myocardial cells, reduced myocardial infarct size and cell death in mice after ischemia/reperfusion. Meanwhile, UA enhanced antioxidant capacity in cardiomyocytes following hypoxia/reoxygenation. UA reduced myocardial apoptosis following ischemia/reperfusion. The protection of UA was abolished by LY294002, a PI3K/Akt-inhibitor. These results demonstrated that UA alleviates myocardial ischemia/reperfusion injury probably through PI3K/Akt pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Coumarins/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/genetics , Animals , Animals, Newborn , Apoptosis/drug effects , Cardiotonic Agents/antagonists & inhibitors , Cell Survival/drug effects , Chromones/pharmacology , Coumarins/antagonists & inhibitors , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction
3.
Can J Physiol Pharmacol ; 94(5): 542-53, 2016 May.
Article in English | MEDLINE | ID: mdl-27070866

ABSTRACT

Astragaloside IV (ASI), a traditional Chinese medicine, is a main active ingredient of Astragalus membranaceus. Many clinical studies have found that ASI protects cardiomyocytes in cardiovascular diseases, but the underlying mechanisms remain obscure. The aim of this study was to investigate the molecular mechanisms responsible for the protective effects of ASI in cardiomyocytes from anoxia/reoxygenation (A/R) injury. According to the previous studies, we hypothesized that the cardioprotective effects of ASI against A/R injury might be associated with Notch1/Hes1 signaling pathway. In this study, neonatal rat primary cardiomyocytes were preconditioned with ASI prior to A/R injury. Our results showed that ASI effectively increased the cell viability, decreased the content of MDA, decreased the activities of CPK and LDH, increased the activities of GSH-Px and SOD, and reduced the reactive oxygen species (ROS) generation and the loss of mitochondrial membrane potential (Δψm). ASI inhibited the mitochondrial permeability transition pore (mPTP) opening and activation of caspase-3, and finally decreased the cell apoptosis in cardiomyocytes. Furthermore, ASI upregulated Hes1 protein expression. However, pretreatment with DAPT, a Notch1 inhibitor, effectively attenuated the cardioprotective effects of ASI against A/R injury, except MDA, SOD, GSH-Px, and the ROS generation. Taken together, we demonstrated that ASI could protect against A/R injury via the Notch1/Hes1 signaling pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Saponins/pharmacology , Transcription Factor HES-1/agonists , Triterpenes/pharmacology , Up-Regulation/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Astragalus propinquus/chemistry , Cardiotonic Agents/antagonists & inhibitors , Caspase 3/chemistry , Caspase 3/metabolism , Cells, Cultured , Diamines/pharmacology , Drugs, Chinese Herbal/chemistry , Enzyme Activation/drug effects , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Saponins/antagonists & inhibitors , Signal Transduction/drug effects , Thiazoles/pharmacology , Transcription Factor HES-1/metabolism , Triterpenes/antagonists & inhibitors
4.
Pharmazie ; 70(4): 244-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26012254

ABSTRACT

Triptolide (TPL) is a diterpene triepoxide with potent immunosuppressive and anti-inflammatory properties. It is the main effective component of the traditional Chinese herb Tripterygium wilfordii Hook F and has been used in China for centuries to treat immune-related disorders. The present study was conducted to investigate the effects of TPL on cardiac remodeling in rats. Age matched male Wistar rats were used in this study. Cardiac remodeling rat model was established by hypodermic injection of isoprenaline for ten days. The rats were treated with TPL (20 or 100 µg/kg/d) for six consecutive weeks. At the end of the study, the cardiac function, collagen volume fraction, perivascular collagen area and hydroxyproline concentration were studied. Echocardiography, Masson staining, immunohistochemistry, western blot and real-time polymerase chain reaction were performed. The protein and mRNA expression of transforming growth factor-ß1 (TGF-ß1), drosophila mothers against decapentaplegic protein 3 (Smad3) and p38 mitogen activated protein kinase (p38 MAPK) were analyzed. The results indicated that TPL treatment significantly reduced the collagen volume fraction, perivascular collagen area, ventricular weight/body weight ratio and hydroxyproline concentration in myocardial tissue compared with the model group. In addition, it also improved the cardiac function. TPL attenuated cardiac remodeling in rats by down-regulating the p38 MAPK and TGF-ß1/Smad3 signaling pathways. TPL treatment significantly attenuated cardiac fibrosis and improved cardiac function through suppressing the p38 MAPK and TGF-ß1/Smad3 signaling pathway in isoprenaline-induced cardiac remodeling rats. Our findings suggested that TPL might be a novel complementary medicine in the treatment of chronic heart failure.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cardiotonic Agents/antagonists & inhibitors , Diterpenes/pharmacology , Isoproterenol/antagonists & inhibitors , Phenanthrenes/pharmacology , Smad3 Protein/physiology , Transforming Growth Factor beta1/physiology , Ventricular Remodeling/drug effects , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Blood Pressure/drug effects , Diterpenes/adverse effects , Epoxy Compounds/adverse effects , Epoxy Compounds/pharmacology , Fibrosis/prevention & control , Hydroxyproline/metabolism , Male , Myocardium/pathology , Organ Size/drug effects , Phenanthrenes/adverse effects , Rats , Rats, Wistar , Signal Transduction/drug effects
5.
J Mol Cell Cardiol ; 66: 12-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24184997

ABSTRACT

This study investigated if zinc plays a role in postconditioning-induced cardioprotection in rat hearts. Isolated rat hearts were subjected to 30 min regional ischemia followed by 2h of reperfusion. Postconditioning was elicited by 6 cycles of 10s reperfusion and 10s ischemia. Cytosolic zinc concentrations were measured with inductively coupled plasma optical emission spectroscopy (ICPOES). Infarct size was assessed by triphenyltetrazolium chloride staining. Cytosolic zinc concentrations were decreased dramatically upon reperfusion in the control hearts. In contrast, postconditioning increased cytosolic zinc levels at reperfusion. The anti-infarct effect of postconditioning was inhibited by the selective zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethyl) ethylenediamine (TPEN). Postconditioning significantly increased phosphorylation levels of the reperfusion injury salvage kinases (RISK) including Akt (Ser(473)), extracellular signal-regulated kinase1/2 (ERK1/2) (Thr(202)/Tyr(204)), and glycogen synthase kinase-3ß (GSK-3ß) (Ser(9)) at reperfusion, which were nullified by TPEN. Postconditioning decreased the activity of protein phosphatase 2A (PP2A) in a zinc-dependent manner. Knockdown of the zinc transporter Zip2 inhibited the protective effect of postconditioning on hypoxia/reoxygenation injury in H9c2 cells. These results suggest that zinc plays an important role in the cardioprotective effect of postconditioning presumably by enhancing the activation of the RISK pathway. Zip2 and inactivation of PP2A by zinc may, at least in part, account for the activation of the RISK pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Ischemic Postconditioning , Myocardial Reperfusion Injury/prevention & control , Zinc/pharmacology , Animals , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/metabolism , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cations, Divalent , Chelating Agents/pharmacology , Ethylenediamines/pharmacology , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction , Zinc/metabolism
6.
Toxicol Appl Pharmacol ; 276(1): 55-62, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24480152

ABSTRACT

Doxorubicin (Dox) is an indispensable chemotherapeutic agent for the treatment of various forms of neoplasia such as lung, breast, ovarian, and bladder cancers. Cardiotoxicity is a major concern for patients receiving Dox therapy. Previous work from our laboratory indicated that glucocorticoids (GCs) alleviate Dox-induced apoptosis in cardiomyocytes. Here we have found glucocorticoid-induced leucine zipper (GILZ) to be a mediator of GC-induced cytoprotection. GILZ was found to be induced in cardiomyocytes by GC treatment. Knocking down of GILZ using siRNA resulted in cancelation of GC-induced cytoprotection against apoptosis by Dox treatment. Overexpressing GILZ by transfection was able to protect cells from apoptosis induced by Dox as measured by caspase activation, Annexin V binding and morphologic changes. Western blot analyses indicate that GILZ overexpression prevented cytochrome c release from mitochondria and cleavage of caspase-3. When bcl-2 family proteins were examined, we found that GILZ overexpression causes induction of the pro-survival protein Bcl-xL. Since siRNA against Bcl-xL reverses GC induced cytoprotection, Bcl-xL induction represents an important event in GILZ-induced cytoprotection. Our data suggest that GILZ functions as a cytoprotective gene in cardiomyocytes.


Subject(s)
Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Cardiotoxins/antagonists & inhibitors , Doxorubicin/antagonists & inhibitors , Glucocorticoids/pharmacology , Myocytes, Cardiac/drug effects , Transcription Factors/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Cardiotonic Agents/antagonists & inhibitors , Cardiotoxins/adverse effects , Cell Line , Cell Survival/drug effects , Corticosterone/antagonists & inhibitors , Corticosterone/pharmacology , Doxorubicin/adverse effects , Glucocorticoids/antagonists & inhibitors , Mice , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Up-Regulation/drug effects , bcl-X Protein/agonists , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
7.
Nat Med ; 13(10): 1185-92, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17873880

ABSTRACT

The cardioprotective effects of estrogen are mediated by receptors expressed in vascular cells. Here we show that 27-hydroxycholesterol (27HC), an abundant cholesterol metabolite that is elevated with hypercholesterolemia and found in atherosclerotic lesions, is a competitive antagonist of estrogen receptor action in the vasculature. 27HC inhibited both the transcription-mediated and the non-transcription-mediated estrogen-dependent production of nitric oxide by vascular cells, resulting in reduced estrogen-induced vasorelaxation of rat aorta. Furthermore, increasing 27HC levels in mice by diet-induced hypercholesterolemia, pharmacologic administration or genetic manipulation (by knocking out the gene encoding the catabolic enzyme CYP7B1) decreased estrogen-dependent expression of vascular nitric oxide synthase and repressed carotid artery reendothelialization. As well as antiestrogenic effects, there were proestrogenic actions of 27HC that were cell-type specific, indicating that 27HC functions as an endogenous selective estrogen receptor modulator (SERM). Taken together, these studies point to 27HC as a contributing factor in the loss of estrogen protection from vascular disease.


Subject(s)
Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Estrogens/pharmacology , Hydroxycholesterols/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Animals , Aorta, Thoracic/drug effects , Binding, Competitive/drug effects , Cardiotonic Agents/metabolism , Cell Culture Techniques , Cell Line , Cells, Cultured , Cholesterol, Dietary/administration & dosage , DNA, Complementary , Dose-Response Relationship, Drug , Drug Administration Schedule , Estrogens/metabolism , Female , Glutathione Transferase/metabolism , Humans , Hydroxycholesterols/administration & dosage , Hydroxycholesterols/blood , Inhibitory Concentration 50 , Injections, Subcutaneous , Kidney/cytology , Kinetics , Male , Mice , Mice, Knockout , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type III , RNA, Messenger/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , Selective Estrogen Receptor Modulators/administration & dosage , Selective Estrogen Receptor Modulators/blood , Vasodilation/drug effects
8.
Can J Physiol Pharmacol ; 90(8): 1117-25, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22808991

ABSTRACT

Genistein, an isoflavone and a rich constituent of soy, possesses important regulatory effects on nitric oxide (NO) synthesis and oxidative stress. Transient and low release of NO by endothelial nitric oxide synthase (eNOS) has been shown to be beneficial, while high and sustained release by inducible nitric oxide synthase (iNOS) may be detrimental in pathological cardiac hypertrophy. The present study was designed to evaluate whether genistein could prevent isoproterenol-induced cardiac hypertrophy in male Wistar rats (150-200 g, 10-12 weeks old) rats. Isoproterenol (5 mg·(kg body weight)(-1)) was injected subcutaneously once daily for 14 days to induced cardiac hypertrophy. Genistein (0.1 and 0.2 mg·kg(-1), subcutaneous injection once daily) was administered along with isoproterenol. Heart tissue was studied for myocyte size and fibrosis. Myocardial thiobarbituric acid reactive substances (TBARS), glutathione (GSH), superoxide dismutase (SOD), catalase levels, and 1-OH proline (collagen content) were also estimated. Genistein significantly prevented any isoproterenol-induced increase in heart weight to body weight ratio, left ventricular mass (echocardiographic), myocardial 1-OH proline, fibrosis, myocyte size and myocardial oxidative stress. These beneficial effects of genistein were blocked by a nonselective NOS inhibitor (L-NAME), but not by a selective iNOS inhibitor (aminoguanidine). Thus, the present study suggests that the salutary effects of genistein on isoproterenol-induced cardiac hypertrophy may be mediated through inhibition of iNOS and potentiation of eNOS activities.


Subject(s)
Cardiomegaly/drug therapy , Cardiomegaly/prevention & control , Cardiotonic Agents/therapeutic use , Genistein/therapeutic use , Myocardium/metabolism , Myocardium/pathology , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Catalase/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fibrosis , Genistein/antagonists & inhibitors , Genistein/pharmacology , Glutathione/metabolism , Guanidines/pharmacology , Hydroxyproline/metabolism , Isoproterenol/antagonists & inhibitors , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
9.
Can J Physiol Pharmacol ; 90(10): 1386-93, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22966876

ABSTRACT

Natriuretic peptides and digitalis-like compounds serve as regulators of homeostasis, including control of volume expansion and blood pressure. The aim of the present study was to explore possible interactions between atrial natriuretic peptide (ANP) and ouabain in the heart. ANP (1 nmol/L) had no effect in papillary muscle preparations from guinea pigs. Ouabain (1 µmol/L) induced positive inotropic effect. The addition of ANP prior to ouabain resulted in a significant decrease in the ouabain-induced positive inotropic effect, manifested as an attenuated increase in twitch maximal upward force slope and resting muscular tension. In addition, ANP caused an increase in Na⁺-K⁺-ATPase activity in heart microsomal preparations. The effect of ouabain on Na⁺-K⁺-ATPase activity was shown in a biphasic manner. Ouabain (0.01-1 nmol/L) had a small but significant increase on pump activity, but higher doses of ouabain inhibited activity. ANP attenuated ouabain-induced Na⁺-K⁺-ATPase activity. Furthermore, ouabain (50 nmol/L) or ANP (10 nmol/L) alone induced Akt activation in cardiomyocytes. However, ANP blocked ouabain-induced Akt activation. These results point to the existence of interactions between ANP and ouabain on Na⁺-K⁺-ATPase signaling and function in the heart, which may be mediated by regulation of Na⁺-K⁺-ATPase activity and (or) signal transduction mechanisms.


Subject(s)
Atrial Natriuretic Factor/metabolism , Cardiotonic Agents/pharmacology , Myocardial Contraction/drug effects , Ouabain/pharmacology , Papillary Muscles/drug effects , Animals , Cardiotonic Agents/antagonists & inhibitors , Cells, Cultured , Guinea Pigs , In Vitro Techniques , Male , Mice , Mice, Transgenic , Microsomes/drug effects , Microsomes/enzymology , Microsomes/metabolism , Muscle Tonus/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Ouabain/antagonists & inhibitors , Papillary Muscles/enzymology , Papillary Muscles/metabolism , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Up-Regulation/drug effects
10.
J Pharmacol Exp Ther ; 339(3): 825-31, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21865439

ABSTRACT

Isoflurane (ISO) is known to depress cardiac contraction. Here, we hypothesized that decreasing myofilament Ca(2+) responsiveness is central to ISO-induced reduction in cardiac force development. Moreover, we also tested whether the nitroxyl (HNO) donor 1-nitrosocyclohexyl acetate (NCA), acting as a myofilament Ca(2+) sensitizer, restores force in the presence of ISO. Trabeculae from the right ventricles of LBN/F1 rats were superfused with Krebs-Henseleit solution at room temperature, and force and intracellular Ca(2+) ([Ca(2+)](i)) were measured. Steady-state activations were achieved by stimulating the muscles at 10 Hz in the presence of ryanodine. The same muscles were chemically skinned with 1% Triton X-100, and the force-Ca(2+) relation measurements were repeated. ISO depressed force in a dose-dependent manner without significantly altering [Ca(2+)](i). At 1.5%, force was reduced over 50%, whereas [Ca(2+)](i) remained unaffected. At 3%, contraction was decreased by ∼75% with [Ca(2+)](i) reduced by only 15%. During steady-state activation, 1.5% ISO depressed maximal Ca(2+)-activated force (F(max)) and increased the [Ca(2+)](i) required for 50% activation (Ca(50)) without affecting the Hill coefficient. After skinning, the same muscles showed similar decreases in F(max) and increases in Ca(50) in the presence of ISO. NCA restored force in the presence of ISO without affecting [Ca(2+)](i). These results show that 1) ISO depresses cardiac force development by decreasing myofilament Ca(2+) responsiveness, and 2) myofilament Ca(2+) sensitization by NCA can effectively restore force development without further increases in [Ca(2+)](i). The present findings have potential translational value because of the efficiency and efficacy of HNO on ISO-induced myocardial contractile dysfunction.


Subject(s)
Anesthetics, Inhalation/toxicity , Calcium/physiology , Heart Ventricles/drug effects , Isoflurane/toxicity , Myocardial Contraction/drug effects , Myofibrils/drug effects , Nitrogen Oxides/metabolism , Acetates/pharmacology , Anesthetics, Inhalation/pharmacology , Animals , Cardiotonic Agents/antagonists & inhibitors , Drug Evaluation, Preclinical , Free Radicals/metabolism , Glucose , Isoflurane/pharmacology , Myocardial Contraction/physiology , Myofibrils/physiology , Nitroso Compounds/pharmacology , Rats , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/physiology , Tromethamine , Ventricular Function/drug effects
11.
Drug Metab Dispos ; 39(4): 675-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21205911

ABSTRACT

Cinobufagin (CB), a major bioactive component of the traditional Chinese medicine Chansu, has been reported to have potent antitumor activity. In this study, in vitro metabolism of CB among species was compared with respect to metabolic profiles, enzymes involved, and catalytic efficiency by using liver microsomes from human (HLM), mouse (MLM), rat (RLM), dog (DLM), minipig (PLM), and monkey (CyLM). Significant species differences in CB metabolism were revealed. In particular, species-specific deacetylation and epimerization combined with hydroxylation existed in RLM, whereas hydroxylation was a major pathway in HLM, MLM, DLM, PLM, and CyLM. Two monohydroxylated metabolites of CB in human and animal species were identified as 1α-hydroxylcinobufagin and 5ß-hydroxylcinobufagin by using liquid chromatography-mass spectrometry and two-dimensional NMR techniques. CYP3A4 was identified as the main isoform involved in CB hydroxylation in HLM on the basis of the chemical inhibition studies and screen assays with recombinant human cytochrome P450s. Furthermore, ketoconazole, a specific inhibitor of CYP3A, strongly inhibited CB hydroxylation in MLM, DLM, PLM, and CyLM, indicating that CYP3A was responsible for CB hydroxylation in these animal species. The apparent substrate affinity and catalytic efficiency for 1α- and 5ß-hydroxylation of CB in liver microsomes from various species were also determined. PLM appears to have K(m) and total intrinsic clearance value (V(max)/K(m)) similar to those for HLM, and the total microsomal intrinsic clearance values for CB obeyed the following order: mouse > dog > monkey > human > minipig. These findings provide vital information to better understand the metabolic behaviors of CB among various species.


Subject(s)
Bufanolides/metabolism , Cardiotonic Agents/metabolism , Drugs, Chinese Herbal/metabolism , Microsomes, Liver/metabolism , Animals , Biotransformation , Bufanolides/antagonists & inhibitors , Bufanolides/pharmacokinetics , Bufanolides/toxicity , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/pharmacokinetics , Cardiotonic Agents/toxicity , Cell Proliferation/drug effects , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Dogs , Drugs, Chinese Herbal/pharmacokinetics , Drugs, Chinese Herbal/toxicity , Haplorhini , Humans , Hydroxylation , Male , Mice , Mixed Function Oxygenases/metabolism , Rats , Species Specificity , Swine , Swine, Miniature
12.
Toxicol Appl Pharmacol ; 256(2): 168-73, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21854795

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) α and PPARγ ligands can attenuate myocardial fibrosis. Osthole, an active constituent isolated from the fruit of Cnidium monnieri (L.) Cusson, may be a dual PPARα/γ agonist, but there has been no report on its effect on myocardial fibrosis. In the present study, we investigated the inhibitory effect of osthole on myocardial fibrotic formation in mice and its possible mechanisms. A mouse model with myocardial fibrosis was induced by hypodermic injection of isoprenaline while the mice were simultaneously treated with 40 and 80 mg/kg osthole for 40 days. After the addition of osthole, the cardiac weight index and hydroxyproline content in the myocardial tissues were decreased, the degree of collagen accumulation in the heart was improved, and the downregulation of myocardial PPARα/γ mRNA expression induced by isoprenaline was reversed. Moreover, the mRNA expression of transforming growth factor (TGF)-ß1 and the protein levels of nuclear factor (NF)-κB and TGF-ß1 in the myocardial tissues were decreased. These findings suggest that osthole can prevent isoprenaline-induced myocardial fibrosis in mice, and its mechanisms may be related to the reduction of TGF-ß1 expression via the activation of PPARα/γ and subsequent inhibition of NF-κB in myocardial tissues.


Subject(s)
Cardiotonic Agents/pharmacology , Coumarins/pharmacology , Heart/drug effects , Isoproterenol/pharmacology , Myocardium/metabolism , Transforming Growth Factor beta1/biosynthesis , Animals , Cardiotonic Agents/antagonists & inhibitors , Collagen/analysis , Fibrosis , Gene Expression Regulation/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Isoproterenol/antagonists & inhibitors , Male , Mice , Myocardium/chemistry , Myocardium/pathology , PPAR alpha/analysis , PPAR alpha/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/analysis
13.
Diabetes Metab Res Rev ; 27(4): 331-40, 2011 May.
Article in English | MEDLINE | ID: mdl-21309057

ABSTRACT

BACKGROUND: Diabetes increases cardiac damage after myocardial ischaemia. Cannabinoids can protect against myocardial ischaemia/reperfusion injury. The aim of this study was to examine the cardioprotective effect of the cannabinoid agonist WIN 55,212-2 (WIN) against ischaemia/reperfusion injury in an experimental model of type 2 diabetes. We performed these experiments in the Zucker diabetic fatty rat, and focused on the role of cannabinoid receptors in modulation of cardiac inducible nitric oxide synthase (iNOS)/endothelial-type nitric oxide synthase (eNOS) expression. METHODS: Male 20-week-old Zucker diabetic fatty rats were treated with vehicle, WIN, the selective CB1 or CB2 receptor antagonists AM251 and AM630, respectively, AM251 + WIN or AM630 + WIN. Hearts were isolated from these rats, and the cardiac functional response to ischaemia/reperfusion injury was evaluated. In addition, cardiac iNOS and eNOS expression were determined by western blot. RESULTS: WIN significantly improved cardiac recovery after ischaemia/ reperfusion in the hearts from Zucker diabetic fatty rats by restoring coronary perfusion pressure and heart rate to preischaemic levels. Additionally, WIN decreased cardiac iNOS expression and increased eNOS expression after ischaemia/reperfusion in diabetic hearts. WIN-induced cardiac functional recovery was completely blocked by the CB2 antagonist AM630. However, changes in NOS isoenzyme expression were not affected by the CB antagonists. CONCLUSIONS: This study shows a cardioprotective effect of a cannabinoid agonist on ischaemia/reperfusion injury in an experimental model of a metabolic disorder. The activation mainly of CB2 receptors and the restoration of iNOS/eNOS cardiac equilibrium are mechanisms involved in this protective effect. These initial studies have provided the basis for future research in this field.


Subject(s)
Benzoxazines/therapeutic use , Cannabinoids/therapeutic use , Cardiotonic Agents/therapeutic use , Morpholines/therapeutic use , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/prevention & control , Naphthalenes/therapeutic use , Receptor, Cannabinoid, CB2/agonists , Animals , Benzoxazines/antagonists & inhibitors , Cannabinoids/antagonists & inhibitors , Cardiotonic Agents/antagonists & inhibitors , Coronary Vessels/drug effects , Diabetes Mellitus, Type 2/complications , Heart/drug effects , Heart/physiopathology , Heart Rate/drug effects , Indoles/pharmacology , Male , Morpholines/antagonists & inhibitors , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Naphthalenes/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Zucker , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism
14.
Steroids ; 146: 50-56, 2019 06.
Article in English | MEDLINE | ID: mdl-30904504

ABSTRACT

Bisphenol A (BPA) is an environmental oestrogen or xenoestrogen (XEs). XEs represent a health risk due to their potential for endocrine disruption and ability to mimic estrogenic activity. The effects of BPA on isolated hearts under normal and ischemia/reperfusion (I/R) conditions were investigated for the first time, with a focus on the effects of BPA and 17ß-oestradiol (E2) co-administration on I/R injury. Our results indicated that BPA at 10-7 M and 10-5 M did not significantly affect heart rate (HR), coronary flow (CF), lactate dehydrogenase (LDH) or creatine kinase (CK) release in normal or I/R isolated hearts within the 90 min. However, E2 exerted a protective effect against I/R injury, whereas, BPA inhibited the cardio-protective effects of E2 on HR, CF, and LDH and CK release. Furthermore, BPA in combination with E2 aggravated I/R injury by increasing infarct size and causing a more severe ultrastructural disruption as compared to treatment with E2 alone. Based on our results, we conclude that BPA inhibits the cardio-protective effects of E2 on I/R-injured hearts, despite not significantly affecting normal or I/R isolated hearts.


Subject(s)
Benzhydryl Compounds/pharmacology , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Estradiol/pharmacology , Heart/drug effects , Heart/physiopathology , Myocardial Reperfusion Injury/physiopathology , Phenols/pharmacology , Animals , Coronary Circulation/drug effects , Creatine Kinase/metabolism , Drug Interactions , Female , Guinea Pigs , Heart Rate/drug effects , L-Lactate Dehydrogenase/metabolism , Male , Myocardial Reperfusion Injury/metabolism
15.
Free Radic Biol Med ; 44(1): 82-90, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18045550

ABSTRACT

In this study, we determined the changes in the intracellular redox environment of the heart during ischemia and reperfusion and the effects of resveratrol on such changes. Because redox regulation by thioredoxin (Trx) plays a crucial role in signal transduction and cytoprotection against ROS, the effects of resveratrol on the changes in the amounts of thioredoxin were monitored in an attempt to determine the role of intracellular thioredoxin in resveratrol-mediated changes in intracellular redox environment and its role in resveratrol-mediated cardioprotection. Rats were randomly divided into four groups: group I, control (rats were gavaged with vehicle only); group II, rats were gavaged with 2.5 mg/kg body wt resveratrol per day for 10 days; group III, rats were given resveratrol for 10 days, but on the 7th day, they were treated with shRNA against Trx-1; group IV, rats were given resveratrol for 10 days, but were injected (iv) with cisplatin (1 mg/kg body wt) on days 1, 3, 5, 7, and 9. In concert, two groups of mice (Dn-Trx-1) and a corresponding wild-type group were also gavaged with 2.5 mg/kg body wt resveratrol for 10 days. After 10 days, isolated rat and mouse hearts perfused via working mode were made globally ischemic for 30 min followed by 2 h of reperfusion. Ischemia/reperfusion developed an infarct size of about 40% and resulted in about 25% apoptotic cardiomyocytes, which were reduced by resveratrol. Cisplatin, but not shRNA-Trx-1, abolished the cardioprotective abilities of resveratrol. In the experiments with mouse hearts, similar to rat hearts, resveratrol significantly reduced the ischemia/reperfusion-mediated increase in infarct size and apoptosis in both groups. MDA formation, a presumptive marker for lipid peroxidation, was increased in the I/R group and reduced in the resveratrol group, and resveratrol-mediated reduction in MDA formation was abolished with cisplatin, but not with shRNA-Trx-1. I/R-induced reduction in GSH/GSSH ratio was prevented by resveratrol, and resveratrol-mediated preservation of GSH/GSSG ratio was reduced by cisplatin, but not by sh-RNA-Trx-1. RT-PCR revealed an increase in both Trx-1 and Trx-2 transcripts; but only Trx-2 protein, not Trx-1 protein, was enhanced with resveratrol by Western blot analysis. Electron paramagnetic resonance spectroscopic study revealed that resveratrol treatment significantly increased the decay rates of nitroxide radicals compared to control hearts, suggesting that resveratrol can switch into the reduction state more compared to control heart. Finally, resveratrol generated a survival signal by phosphorylation of Akt and increase in induction of Bcl-2 expression, which was inhibited by cisplatin, but not by shRNA-Trx-1. Taken together, the results of this study indicate that resveratrol provides cardioprotection by maintaining intracellular redox environments, and Trx-2 is likely to play a role in switching I/R-induced death signal into survival signal.


Subject(s)
Cell Death/physiology , Cell Survival/physiology , Signal Transduction , Stilbenes/metabolism , Thioredoxins/metabolism , Animals , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/metabolism , Cardiotonic Agents/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Antagonism , Genes, bcl-2 , Male , Mice , Mice, Transgenic , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Organ Specificity , Oxidation-Reduction , RNA, Antisense/genetics , RNA, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Resveratrol , Signal Transduction/drug effects , Signal Transduction/physiology , Stilbenes/antagonists & inhibitors , Stilbenes/pharmacology , Thioredoxins/antagonists & inhibitors , Thioredoxins/genetics , Thioredoxins/pharmacology
16.
Life Sci ; 83(7-8): 264-71, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18625248

ABSTRACT

We have previously demonstrated the participation of reactive oxygen species (ROS) in the positive inotropic effect of a physiological concentration of Angiotensin II (Ang II, 1 nM). The objective of the present work was to evaluate the role and source of ROS generation in the positive inotropic effect produced by an equipotent concentration of endothelin-1 (ET-1, 0.4 nM). Isolated cat ventricular myocytes were used to measure sarcomere shortening with a video-camera, superoxide anion (()O(2)(-)) with chemiluminescence, and ROS production and intracellular pH (pH(i)) with epifluorescence. The ET-1-induced positive inotropic effect (40.4+/-3.1%, n=10, p<0.05) was associated to an increase in ROS production (105+/-29 fluorescence units above control, n=6, p<0.05). ET-1 also induced an increase in ()O(2)(-) production that was inhibited by the NADPH oxidase blocker, apocynin, and by the blockers of mitochondrial ATP-sensitive K(+) channels (mK(ATP)), glibenclamide and 5 hydroxydecanoic acid. The ET-1-induced positive inotropic effect was inhibited by apocynin (0.3 mM; 6.3+/-6.6%, n=13), glibenclamide (50 microM; 8.8+/-3.5%, n=6), 5 hydroxydecanoic acid (500 microM; 14.1+/-8.1, n=9), and by scavenging ROS with MPG (2 mM; 0.92+/-5.6%, n=8). ET-1 enhanced proton efflux (J(H)) carried by the Na(+)/H(+) exchanger (NHE) after an acid load, effect that was blocked by MPG. Consistently, the ET-induced positive inotropic effect was also inhibited by the NHE selective blocker HOE642 (5 microM; 9.37+/-6.07%, n=7). The data show that the effect of a concentration of ET-1 that induces an increase in contractility of about 40% is totally mediated by an intracellular pathway triggered by mitochondrial ROS formation and stimulation of the NHE.


Subject(s)
Cardiotonic Agents/pharmacology , Endothelin-1/pharmacology , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Superoxides/metabolism , Angiotensin II/antagonists & inhibitors , Angiotensin II/pharmacology , Animals , Cardiotonic Agents/antagonists & inhibitors , Cats , Cells, Cultured , Dose-Response Relationship, Drug , Endothelin-1/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Heart Ventricles/cytology , Heart Ventricles/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Sarcomeres/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Sulfhydryl Compounds/pharmacology , Superoxides/antagonists & inhibitors , Vasoconstrictor Agents/antagonists & inhibitors , Vasoconstrictor Agents/pharmacology
17.
Acta Anaesthesiol Scand ; 52(1): 111-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17996001

ABSTRACT

BACKGROUND: Post-ischemic administration of volatile anesthetics activates ischemia-reperfusion injury salvage process and decreases myocardial damage. However, the mechanisms underlying anesthetic post-conditioning and effects of lidocaine on it remain unclear. Here we report the cardioprotection of sevoflurane-induced post-conditioning and the effects of lidocaine on it. METHODS: Isolated perfused rat hearts were exposed to 40 min of ischemia followed by 1 h of reperfusion. Volatile anesthetic post-conditioning was induced by 15 min of 3 vol% sevourane (1.5 minimum alveolar concentration) administered at the onset of reperfusion. In some experiments, lidocaine was coadministered with sevoflurane in different concentrations (2, 10 and 20 microg/ml). Infarct size was determined by dividing the total necrotic area of the left ventricle by the total left ventricular slice area (percent necrotic area). RESULTS: Sevoflurane-induced post-conditioning signicantly improved post-ischemia functional recovery and decreased infarct size (47.3+/-5.6% in unprotected hearts vs. 18.6+/-3.1% in anesthetic post-conditioning, P<0.05). Sevourane-mediated cardioprotection was abolished by 20 microg/ml lidocaine. CONCLUSIONS: Sevourane-induced post-conditioning effectively protected myocardium against reperfusion damage and its cytoprotection was reversed by 20 microg/ml lidocaine.


Subject(s)
Cardiotonic Agents/antagonists & inhibitors , Heart/drug effects , Lidocaine/toxicity , Methyl Ethers/antagonists & inhibitors , Myocardial Reperfusion Injury/prevention & control , Animals , Biomarkers , Cardiotonic Agents/pharmacology , Heart Ventricles/pathology , In Vitro Techniques , KATP Channels/drug effects , KATP Channels/physiology , L-Lactate Dehydrogenase/analysis , Lidocaine/pharmacology , Male , Methyl Ethers/pharmacology , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Necrosis , Rats , Rats, Sprague-Dawley , Sevoflurane
18.
Yakugaku Zasshi ; 128(3): 469-77, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18311068

ABSTRACT

The present study was designed to investigate the possible role of free radicals in cardioprotective effects of ischemic, pharmacological and remote preconditioning. Isolated rat heart was perfused on Langendorff apparatus with Kreb's Henseleit solution and subjected to 30 min global ischemia followed by 120 min reperfusion. To assess myocardial injury, coronary effluent was analyzed for lactate dehydrogenase and creatine kinase activity. Myocardial infarct size was estimated using triphenyl tetrazolium chloride staining. Ischemic preconditioning, pharmacological preconditioning (angiotensin II; H2O2), remote aortic preconditioning markedly attenuated I/R induced increase in lactate dehydrogenase and creatine kinase release and myocardial infarct size. Administration of N-Acetyl Cysteine (NAC), in vitro, during ischemic and pharmacological, and in vivo during remote preconditioning attenuated the cardioprotective effects of preconditioning. On the basis of these results, it may be concluded that sub threshold generation of Reactive Oxygen Species (ROS) may activate redox signaling which may be responsible for preconditioning induced cardioprotection.


Subject(s)
Acetylcysteine/pharmacology , Cardiotonic Agents/pharmacology , Free Radical Scavengers/pharmacology , Ischemic Preconditioning , Reactive Oxygen Species/pharmacology , Angiotensin II , Animals , Cardiotonic Agents/antagonists & inhibitors , Female , Hydrogen Peroxide , In Vitro Techniques , Male , Myocardial Infarction/prevention & control , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/antagonists & inhibitors
19.
Pharmacol Rep ; 70(4): 720-729, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29935398

ABSTRACT

BACKGROUND: The current study is focusing on the role of brain natriuretic peptide (BNP), a substrate of dipeptidyl peptidase-4 (DPP-4) enzyme, and its signaling survival pathway in the cardioprotective mechanism of sitagliptin, a DPP-4 inhibitor. METHODS: Male Wistar rats were randomized into 7 groups, sham, I/R, KT-5823 (selective protein kinase (PK) G inhibitor), 5-HD (selective mito-KATP channel blocker), sitagliptin (300mg/kg, po), sitagliptin+KT-5823, and sitagliptin+5-HD. Sitagliptin was administered for 3 days prior to induction of coronary I/R, while either KT-5823 or 5-HD was administered intravenously 5min before coronary ligation. RESULTS: Pretreatment with sitagliptin provided significant protection against I/R injury as manifested by decreasing, percentage of infarct size, suppressing the elevated ST segment, reducing the increased cardiac enzymes, as well as DPP-4 activity and elevating both heart rate (HR) and left ventricular developed pressure (LVDP). However, the addition of either blocker to sitagliptin regimen reversed partly its cardioprotective effects. Although I/R increased BNP content, it unexpectedly decreased that of cGMP; nevertheless, sitagliptin elevated both parameters, an effect that was not affected by the use of the two blockers. On the molecular level, sitagliptin decreased caspase-3 activity and downregulated the mRNA levels of BNP, Bax, and Cyp D, while upregulated that of Bcl2. The use of either KT-5823 or 5-HD with sitagliptin hindered its effect on the molecular markers tested. CONCLUSIONS: The results of the present study suggest that the cardioprotective effect of sitagliptin is mediated partly, but not solely, through the BNP/cGMP/PKG survival signaling pathway.


Subject(s)
Natriuretic Peptide, Brain/metabolism , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Sitagliptin Phosphate/pharmacology , Animals , Carbazoles/pharmacology , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Caspase 3/biosynthesis , Cyclic GMP/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/biosynthesis , Decanoic Acids/pharmacology , Dipeptidyl Peptidase 4/metabolism , Hemodynamics/drug effects , Hydroxy Acids/pharmacology , Male , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Sitagliptin Phosphate/antagonists & inhibitors , bcl-2-Associated X Protein/biosynthesis
20.
Circulation ; 114(13): 1395-402, 2006 Sep 26.
Article in English | MEDLINE | ID: mdl-16966583

ABSTRACT

BACKGROUND: Intracellular proteins involved in oxidative stress and apoptosis are nitrated in diseased tissues but not in normal tissues; definitive evidence to support a causative link between a specific protein that is nitratively modified with tissue injury in a specific disease is limited, however. The aims of the present study were to determine whether thioredoxin (Trx), a novel antioxidant and antiapoptotic molecule, is susceptible to nitrative inactivation and to establish a causative link between Trx nitration and postischemic myocardial apoptosis. METHODS AND RESULTS: In vitro exposure of human Trx-1 to 3-morpholinosydnonimine resulted in significant Trx-1 nitration and almost abolished Trx-1 activity. 3-morpholinosydnonimine-induced nitrative Trx-1 inactivation was completely blocked by MnTE-2-PyP(5+) (a superoxide dismutase mimetic) and markedly attenuated by PTIO (a nitric oxide scavenger). Administration of either reduced or oxidized Trx-1 in vivo attenuated myocardial ischemia/reperfusion injury (>50% reduction in apoptosis and infarct size, P<0.01). However, administration of nitrated Trx-1 failed to exert a cardioprotective effect. In cardiac tissues obtained from ischemic/reperfused heart, significant Trx-1 nitration was detected, Trx activity was markedly inhibited, Trx-1/ASK1 (apoptosis signal-regulating kinase-1) complex formation was abolished, and apoptosis signal-regulating kinase-1 activity was increased. Treatment with either FP15 (a peroxynitrite decomposition catalyst) or MnTE-2-PyP(5+) 10 minutes before reperfusion blocked nitrative Trx inactivation, attenuated apoptosis signal-regulating kinase-1 activation, and reduced postischemic myocardial apoptosis. CONCLUSIONS: These results strongly suggest that nitrative inactivation of Trx plays a proapoptotic role under those pathological conditions in which production of reactive nitrogen species is increased and that antinitrating treatment may have therapeutic value in those diseases, such as myocardial ischemia/reperfusion, in which pathological apoptosis is increased.


Subject(s)
Apoptosis/physiology , Molsidomine/analogs & derivatives , Myocardial Ischemia/metabolism , Myocardium/pathology , Thioredoxins/antagonists & inhibitors , Amino Acid Substitution , Animals , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cyclic N-Oxides/pharmacology , Free Radical Scavengers/pharmacology , Humans , Imidazoles/pharmacology , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System/drug effects , Male , Metalloporphyrins/pharmacology , Metalloporphyrins/therapeutic use , Mice , Molsidomine/antagonists & inhibitors , Molsidomine/pharmacology , Mutagenesis, Site-Directed , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/drug therapy , NADP/metabolism , Oxidation-Reduction , Oxidative Stress , Peroxynitrous Acid/pharmacology , Thioredoxins/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL