Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cardiovasc Drugs Ther ; 27(4): 279-87, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23722418

ABSTRACT

PURPOSE: In patients undergoing primary percutaneous coronary intervention (PCI) for the treatment of ST-segment elevation myocardial infarction (STEMI), coronary microvascular dysfunction is associated with poor prognosis. Coronary microvascular resistance is predominantly regulated by ATP-sensitive potassium (KATP) channels. The aim of this study was to clarify whether nicorandil, a hybrid KATP channel opener and nitric oxide donor, may be a good candidate for improving microvascular dysfunction even when administered after primary PCI. METHODS: We compared the beneficial effects of nicorandil and nitroglycerin on microvascular function in 60 consecutive patients with STEMI. After primary PCI, all patients received single intracoronary administrations of nitroglycerin (250 µg) and nicorandil (2 mg) in a randomized order; 30 received nicorandil first, while the other 30 received nitroglycerin first. Microvascular dysfunction was evaluated with the index of microcirculatory resistance (IMR), defined as the distal coronary pressure multiplied by the hyperemic mean transit time. RESULTS: As a first administration, nicorandil decreased IMR significantly more than did nitroglycerin (median [interquartile ranges]: 10.8[5.2-20.7] U vs. 2.1[1.0-6.0] U, p=0.0002).As a second administration, nicorandil further decreased IMR, while nitroglycerin did not (median [interquartile ranges]: 6.0[1.3-12.7] U vs. -1.4[-2.6 to 1.3] U, p<0.0001). The IMR after the second administration was significantly associated with myocardial blush grade, angiographic TIMI frame count after the procedure, and peak creatine kinase level. CONCLUSION: Intracoronary nicorandil reduced microvascular dysfunction after primary PCI more effectively than did nitroglycerin in patients with STEMI, probably via its KATP channel-opening effect.


Subject(s)
Myocardial Infarction/therapy , Nicorandil/administration & dosage , Nitric Oxide Donors/administration & dosage , Nitroglycerin/administration & dosage , Percutaneous Coronary Intervention , Aged , Cross-Over Studies , Drug Administration Routes , Female , Humans , Male , Microvessels/drug effects , Microvessels/physiopathology , Middle Aged , Myocardial Infarction/physiopathology , Vascular Resistance/drug effects
2.
Circ J ; 75(1): 94-8, 2011.
Article in English | MEDLINE | ID: mdl-21116072

ABSTRACT

BACKGROUND: The objective of this study was to investigate whether a distal protection (DP) device can preserve the index of microcirculatory resistance (IMR) after primary percutaneous coronary intervention (PCI) in patients with anterior ST-segment elevation myocardial infarction (STEMI). METHODS AND RESULTS: The study group of 36 consecutive patients with anterior STEMI were randomized into 2 groups of primary PCI with or without DP: stenting without DP (non-DP group, n = 17) and with DP (DP group, n = 19). The DP in all cases was Filtrap (Nipro, Japan). Following final coronary angiography after successful PCI, IMR was measured using PressureWire™ Certus (St Jude Medical, USA) at maximal hyperemia. The averaged IMR of the 36 patients with STEMI after primary PCI was 31.6U. The IMR in the DP group was significantly lower than that in the non-DP group (26.6 ± 25.8U vs. 37.2 ± 23.2U, P = 0.03242). CONCLUSIONS: DP as an adjunctive therapy of PCI for acute anterior STEMI may have beneficial effects on myocardial microcirculation because of preservation of IMR.


Subject(s)
Angioplasty, Balloon, Coronary/instrumentation , Anterior Wall Myocardial Infarction/therapy , Coronary Circulation , Embolic Protection Devices , Microcirculation , Stents , Vascular Resistance , Angioplasty, Balloon, Coronary/adverse effects , Angioplasty, Balloon, Coronary/mortality , Anterior Wall Myocardial Infarction/diagnostic imaging , Anterior Wall Myocardial Infarction/mortality , Anterior Wall Myocardial Infarction/physiopathology , Coronary Angiography , Humans , Japan , Prospective Studies , Time Factors , Treatment Outcome
3.
Circulation ; 122(4): 361-9, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20625112

ABSTRACT

BACKGROUND: Apoptosis may contribute to the development of heart failure, but the role of apoptotic signaling initiated by the endoplasmic reticulum in this condition has not been well clarified. METHODS AND RESULTS: In myocardial samples from patients with heart failure, quantitative real-time polymerase chain reaction revealed an increase in messenger RNA for C/EBP homologous protein (CHOP), a transcriptional factor that mediates endoplasmic reticulum-initiated apoptotic cell death. We performed transverse aortic constriction or sham operation on wild-type (WT) and CHOP-deficient mice. The CHOP-deficient mice showed less cardiac hypertrophy, fibrosis, and cardiac dysfunction compared with WT mice at 4 weeks after transverse aortic constriction, although the contractility of isolated cardiomyocytes from CHOP-deficient mice was not significantly different from that in the WT mice. In the hearts of CHOP-deficient mice, phosphorylation of eukaryotic translation initiation factor 2alpha, which may reduce protein translation, was enhanced compared with WT mice. In the hearts of WT mice, CHOP-increased apoptotic cell death with activation of caspase-3 was observed at 4 weeks after transverse aortic constriction. In contrast, CHOP-deficient mice had less apoptotic cell death and lower caspase-3 activation at 4 weeks after transverse aortic constriction. Furthermore, the Bcl2/Bax ratio was decreased in WT mice, whereas this change was significantly blunted in CHOP-deficient mice. Real-time polymerase chain reaction microarray analysis revealed that CHOP could regulate several Bcl2 family members in failing hearts. CONCLUSIONS: We propose the novel concept that CHOP, which may modify protein translation and mediate endoplasmic reticulum-initiated apoptotic cell death, contributes to development of cardiac hypertrophy and failure induced by pressure overload.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Heart Failure/genetics , Animals , Apoptosis/physiology , CCAAT-Enhancer-Binding Proteins/deficiency , Cardiomyopathies/pathology , Cardiomyopathies/surgery , DNA Primers , Humans , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stroke Volume
4.
Circ J ; 74(5): 909-15, 2010 May.
Article in English | MEDLINE | ID: mdl-20234097

ABSTRACT

BACKGROUND: Although microvascular dysfunction following percutaneous coronary intervention (PCI) can be evaluated with the index of microcirculatory resistance (IMR), no method of treatment has been established. We hypothesized that intracoronary administration of nicorandil can improve IMR after successful primary PCI in patients with ST-segment elevation myocardial infarction (STEMI). METHODS AND RESULTS: In 40 patients with first STEMI after successful primary PCI, IMR was measured using PressureWire(TM) Certus (St. Jude Medical, MN, USA). In 20 of the patients (Group N), IMR was measured at baseline and after intracoronary nicorandil (2 mg/10 ml). In the other 20 patients (Control), IMR was measured at baseline, after intracoronary saline (10 ml) and after intracoronary nicorandil (2 mg/10 ml). In Group N, IMR significantly decreased after intracoronary nicorandil (median IMR, 27.7-18.7 U, P<0.0001). In the Control group, IMR did not change after saline administration (median IMR, 24.3-23.8 U, P=0.8193), but was significantly decreased after intracoronary nicorandil (median IMR, 23.8-14.9 U, P<0.0001). Next, all 40 patients were divided into subgroups by tertile of baseline IMR. In those with intermediate to high IMR (baseline IMR > or =21), intracoronary nicorandil significantly decreased IMR, although it did not change IMR in those with low IMR (baseline IMR <21). CONCLUSIONS: High IMR levels in patients with STEMI after successful primary PCI can be improved by intracoronary administration of nicorandil.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Catheter Ablation , Microcirculation/drug effects , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Nicorandil/administration & dosage , Vascular Resistance/drug effects , Aged , Female , Humans , Male , Middle Aged , Prospective Studies
5.
J Mol Cell Cardiol ; 48(6): 1280-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20170659

ABSTRACT

The unfolded protein response (UPR) is triggered to assist protein folding when endoplasmic reticulum (ER) function is impaired. Recent studies demonstrated that ER stress can also induce cell-specific genes. In this study, we examined whether X-box binding protein 1 (XBP1), a major UPR-linked transcriptional factor, regulates the expression of brain natriuretic peptide (BNP) in cardiomyocytes. In samples from failing human hearts, extensive splicing of XBP1 was observed along with increased expression of glucose-regulated protein of 78 kDa (GRP78), a target of spliced XBP1 (sXBP1), suggesting that the UPR was induced in heart failure in humans. Interestingly, quantitative real-time PCR revealed a positive correlation between cardiac expression of GRP78 and BNP, leading us to test the hypothesis that sXBP1 regulates BNP as well as GRP78 in cardiomyocytes. A pharmacological ER stressor caused a dose-dependent increase in the expression of sXBP1 and BNP by cultured cardiomyocytes. Short interfering RNA targeting XBP1 suppressed the induction of BNP expression by a pharmacological ER stressor or norepinephrine, which was rescued by the adenovirus-mediated overexpression of sXBP1. The promoter assay with overexpression of sXBP1 or norepinephrine showed that the proximal AP1/CRE-like element in the promoter region of BNP was critical for transcriptional regulation of BNP by sXBP1. Direct binding of sXBP1 to this element was confirmed by the chromatin immunoprecipitation assay. These findings suggest that ER stress observed in failing hearts regulates cardiac BNP expression through a novel promoter region of the AP1/CRE-like element.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Myocytes, Cardiac/cytology , Natriuretic Peptide, Brain/biosynthesis , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Animals , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Microscopy, Confocal/methods , Myocardium/metabolism , Promoter Regions, Genetic , RNA Interference , Rats , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
6.
Circulation ; 116(11): 1226-33, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17709641

ABSTRACT

BACKGROUND: The endoplasmic reticulum (ER) responds to various stresses by upregulation of ER chaperones, but prolonged ER stress eventually causes apoptosis. Although apoptosis is considered to be essential for the progression and rupture of atherosclerotic plaques, the influence of ER stress and apoptosis on rupture of unstable coronary plaques remains unclear. METHODS AND RESULTS: Coronary artery segments were obtained at autopsy from 71 patients, and atherectomy specimens were obtained from 40 patients. Smooth muscle cells and macrophages in the fibrous caps of thin-cap atheroma and ruptured plaques, but not in the fibrous caps of thick-cap atheroma and fibrous plaques, showed a marked increase of ER chaperone expression and apoptotic cells. ER chaperones also showed higher expression in atherectomy specimens from patients with unstable angina pectoris than in specimens from those with stable angina. Expression of 7-ketocholesterol was increased in the fibrous caps of thin-cap atheroma compared with thick-cap atheroma. Treatment of cultured coronary artery smooth muscle cells or THP-1 cells with 7-ketocholesterol induced upregulation of ER chaperones and apoptosis, whereas these changes were prevented by antioxidants. We also investigated possible signaling pathways for ER-initiated apoptosis and found that the CHOP (a transcription factor induced by ER stress)-dependent pathway was activated in unstable plaques. In addition, knockdown of CHOP expression by small interfering RNA decreased ER stress-dependent death of cultured coronary artery smooth muscle cells and THP-1 cells. CONCLUSIONS: Increased ER stress occurs in unstable plaques. Our findings suggest that ER stress-induced apoptosis of smooth muscle cells and macrophages may contribute to plaque vulnerability.


Subject(s)
Coronary Artery Disease/metabolism , Endoplasmic Reticulum/metabolism , Myocardial Ischemia/metabolism , Apoptosis/genetics , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Vessels/metabolism , Endoplasmic Reticulum/genetics , Humans , Molecular Chaperones/biosynthesis , Molecular Chaperones/genetics , Myocardial Ischemia/genetics , Up-Regulation/genetics
7.
Circulation ; 114(18): 1923-32, 2006 Oct 31.
Article in English | MEDLINE | ID: mdl-17043167

ABSTRACT

BACKGROUND: Adenosine inhibits proliferation of cardiac fibroblasts and hypertrophy of cardiomyocytes, both of which may play crucial roles in cardiac remodeling. In the present study, we investigated whether chronic stimulation of adenosine receptors begun after myocardial infarction (MI) prevents cardiac remodeling. METHODS AND RESULTS: MI was produced in Wistar rats by permanent ligation of the left anterior descending coronary artery. One week after the onset of MI, animals were randomized into 8 groups: vehicle, dipyridamole (DIP; the adenosine uptake inhibitor, 50 mg/kg), 2-chroloadenosine (CADO; the stable analogue of adenosine, 2 mg/kg), and CADO in the presence of the nonselective adenosine receptor antagonist 8-sulfophenyltheophylline (8-SPT) or the selective antagonist for adenosine A1, A2a, A2b, or A3 receptor. Three weeks after treatment, hemodynamic and echocardiographic parameters in the DIP and CADO groups were significantly improved compared with the vehicle group. These hemodynamic and echocardiographic improvements were blunted by either 8-SPT or the selective adenosine A2b antagonist MRS1754 but not by the selective antagonists for other subtypes of adenosine receptors. The collagen volume fraction was smaller, and gene expression of the molecules associated with cardiac remodeling such as matrix metalloproteinase in noninfarcted areas was reduced in the DIP and CADO groups compared with the vehicle group, both of which were attenuated by either 8-SPT or MRS1754. CONCLUSIONS: Long-term stimulation of adenosine A2b receptors begun after MI attenuates cardiac fibrosis in the noninfarcted myocardium and improves cardiac function. Drugs that stimulate adenosine A2b receptors or increase adenosine levels are new candidates for preventing cardiac remodeling after MI.


Subject(s)
Adenosine A2 Receptor Agonists , Myocardial Infarction/drug therapy , Ventricular Remodeling/drug effects , 2-Chloroadenosine/pharmacology , 2-Chloroadenosine/therapeutic use , Acetamides/pharmacology , Adenosine A2 Receptor Antagonists , Animals , Cardiomegaly/pathology , Collagen/analysis , Fibrosis , Heart Ventricles/diagnostic imaging , Heart Ventricles/pathology , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Purinergic P1 Receptor Antagonists , Purines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Theophylline/analogs & derivatives , Theophylline/pharmacology , Ultrasonography , Ventricular Function, Left/drug effects
8.
J Am Coll Cardiol ; 48(1): 176-84, 2006 Jul 04.
Article in English | MEDLINE | ID: mdl-16814664

ABSTRACT

OBJECTIVES: We investigated the effects of erythropoietin (EPO) on neovascularization and cardiac function after myocardial infarction (MI). BACKGROUND: Erythropoietin exerts antiapoptotic effects and mobilizes endothelial progenitor cells (EPCs). METHODS: We intravenously administered EPO (1,000 IU/kg) immediately [EPO(0) group], 6 h [EPO(6h) group], or 1 week [EPO(1wk) group] after the permanent ligation of the coronary artery in dogs. Control animals received saline immediately after the ligation. RESULTS: The infarct size 6 h after MI was significantly smaller in the EPO(0) group than in the control group (61.5 +/- 6.0% vs. 22.9 +/- 2.2%). One week after MI, the circulating CD34-positive mononuclear cell numbers in both the EPO(0) and the EPO(6h) groups were significantly higher than in the control group. In the ischemic region, the capillary density and myocardial blood flow 4 weeks after MI was significantly higher in both the EPO(0) and the EPO(6h) groups than in the control group. Four weeks after MI, left ventricular (LV) ejection fraction in the EPO(6h) (48.6 +/- 1.9%) group was significantly higher than that in either the control (41.9 +/- 0.9%) or the EPO(1wk) (42.6 +/- 1.2%) group but significantly lower than that in the EPO(0) group (56.1 +/- 2.3%). The LV end-diastolic pressure 4 weeks after MI in both the EPO(0) and the EPO(6h) groups was significantly lower than either the control or the EPO(1wk) group. Hematologic parameters did not differ among the groups. CONCLUSIONS: In addition to its acute infarct size-limiting effect, EPO enhances neovascularization, likely via EPC mobilization, and improves cardiac dysfunction in the chronic phase, although it has time-window limitations.


Subject(s)
Coronary Circulation/drug effects , Erythropoietin/therapeutic use , Myocardial Infarction/drug therapy , Neovascularization, Physiologic/drug effects , Ventricular Dysfunction, Left/physiopathology , Animals , Antigens, CD34/analysis , Capillaries/anatomy & histology , Dogs , Hemodynamics , Leukocytes, Mononuclear/immunology , Myocardial Infarction/blood , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Vascular Endothelial Growth Factor A/blood , Ventricular Dysfunction, Left/etiology
9.
Cardiovasc Drugs Ther ; 20(2): 93-102, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16761190

ABSTRACT

Aldosterone promotes cardiovascular inflammation and remodeling, both of which are characteristic changes in hypertensive and failing hearts. Since chronic inhibition of nitric oxide (NO) synthase with N(omega)-nitro-L-arginine methyl ester (L-NAME) induces systemic hypertension associated with cardiovascular inflammation and remodeling, we examined the potential role of aldosterone in this process using eplerenone, a selective aldosterone receptor antagonist. Ten-week-old male Wistar-Kyoto rats were randomly divided into 3 groups: the control group (no treatment), the L-NAME group (received L-NAME 1 g/L in drinking water), and the L-NAME+Eplerenone group (L-NAME plus eplerenone at 100 mg/kg/day). After 8 weeks of the treatment, the L-NAME group showed significantly higher systolic blood pressure than the control group (198 +/- 7 vs. 141 +/- 3 mmHg, P < 0.05). Eplerenone did not affect the increase in blood pressure caused by L-NAME (189 +/- 12 mmHg). Chronic inhibition of NO synthesis increased the plasma aldosterone concentration and CYP11B2 mRNA in adrenal glands. Cardiac inflammation and fibrosis were detected in the L-NAME group, while both changes were completely prevented by eplerenone. Cardiac hypertrophy was induced in L-NAME group, but was partially prevented by eplerenone. In the L-NAME group, left ventricular fractional shortening (LVFS: 27 +/- 2 vs. 38 +/- 1%) and E/A ratio (1.7 +/- 0.1 vs. 2.1 +/- 0.1) were significantly lower and LV end-diastolic pressure (LVEDP) was higher (4.9 +/- 0.6 vs. 13.9 +/- 0.5 mmHg) without LV enlargement, compared with those in the control group (P < 0.05). Eplerenone completely normalized LVFS (36 +/- 2%), E/A ratio (2.2 +/- 0.1), and LVEDP (6.2 +/- 0.7 mmHg). These results suggest that chronic inhibition of NO synthesis induces cardiac inflammation and dysfunction via an aldosterone receptor-dependent mechanism.


Subject(s)
Heart Failure/prevention & control , Hypertension/prevention & control , Mineralocorticoid Receptor Antagonists , Nitric Oxide/antagonists & inhibitors , Actins/drug effects , Actins/metabolism , Aldosterone/blood , Animals , Blotting, Western , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Drug Therapy, Combination , Eplerenone , Fibrosis/drug therapy , Fibrosis/pathology , Fibrosis/prevention & control , Heart Failure/complications , Heart Failure/metabolism , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hypertension/complications , Hypertension/metabolism , Immunohistochemistry , Male , NF-kappa B/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , NG-Nitroarginine Methyl Ester/therapeutic use , Nitric Oxide/biosynthesis , Peptide Fragments/metabolism , Proliferating Cell Nuclear Antigen/analysis , Protein Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Inbred WKY , Receptors, Mineralocorticoid/physiology , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , Spironolactone/therapeutic use , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
10.
Hypertens Res ; 29(4): 277-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16778335

ABSTRACT

The prevalence of atrial fibrillation (AF) increases in patients with hypertension. Angiotensin II is involved in structural atrial remodeling, which contributes to the onset and maintenance of AF in paced animal models. We investigated the role of angiotensin II in atrial structural remodeling in rats with hypertension. Ten-week-old male Wistar-Kyoto rats were randomly divided into 4 groups: a control group (no treatment), an Nomega-nitro-L-arginine methyl ester (L-NAME) group (administered L-NAME, an inhibitor of nitric oxide synthase, 1 g/l in drinking water), an L-NAME+candesartan group (L-NAME plus candesartan-an angiotensin II receptor blocker (ARB)-at 0.1 mg/kg/day), and an L-NAME + hydralazine group (L-NAME plus hydralazine at 120 mg/l in drinking water). Eight weeks after treatment, the L-NAME group showed significantly higher systolic blood pressure than the control group (197 +/- 12 vs.138 +/- 5 mmHg, p < 0.05). Candesartan or hydralazine with L-NAME reduced systolic blood pressure to baseline. Chronic inhibition of NO synthesis increased the extent of fibrosis and transforming growth factor-beta expression in atrial tissue, and both of these effects were prevented by candesartan, but not by hydralazine. Cardiac hypertrophy and dysfunction were induced in the L-NAME group, and these effects were also prevented by candesartan, but not by hydralazine. In contrast, the decrease in thrombomodulin expression in the atrial endocardium in hypertensive rats was restored by candesartan and hydralazine. The ARB prevented atrial structural remodeling, a possible contributing factor for the development of AF, in the hearts of rats with hypertension induced by long-term inhibition of NO synthesis.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Cardiomegaly/prevention & control , Hypertension/drug therapy , Nitric Oxide/antagonists & inhibitors , Tetrazoles/pharmacology , Animals , Antihypertensive Agents/pharmacology , Biphenyl Compounds , Blood Pressure , Cardiomegaly/etiology , Cardiomegaly/pathology , Enzyme Inhibitors/pharmacology , Heart Atria/drug effects , Heart Atria/metabolism , Heart Atria/pathology , Heart Rate , Hydralazine/pharmacology , Hypertension/chemically induced , Hypertension/complications , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Inbred WKY , Thrombomodulin/metabolism
11.
Cardiovasc Drugs Ther ; 20(3): 159-65, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16775664

ABSTRACT

PURPOSE: Recent studies suggest that G-CSF prevents cardiac remodeling following myocardial infarction (MI) likely through regeneration of the myocardium and coronary vessels. However, it remains unclear whether G-CSF administered at the onset of reperfusion prevents ischemia/reperfusion injury in the acute phase. We investigated acute effects of G-CSF on myocardial infarct size and the incidence of lethal arrhythmia and evaluated the involvement of the phosphatidylinositol-3 kinase (PI3K) in the in vivo canine models. METHODS: In open-chest dogs, left anterior descending coronary artery (LAD) was occluded for 90 minutes followed by 6 hours of reperfusion. We intravenously administered G-CSF (0.33 micro/kg/min) for 30 minutes from the onset of reperfusion. Wortmannin, a PI3K inhibitor, was selectively administered into the LAD after the onset of reperfusion. RESULTS: G-CSF significantly (p<0.05) reduced myocardial infarct size (38.7+/-4.3% to 15.7+/-5.3%) and the incidence of ventricular fibrillation during reperfusion periods (50% to 0%) compared with the control. G-CSF enhanced Akt phospholylation in ischemic canine myocardium. Wortmannin blunted both the infarct size-limiting and anti-arrhythmic effects of G-CSF. G-CSF did not change myeloperoxidase activity, a marker of neutrophil accumulation, in the infarcted myocardium. CONCLUSION: An intravenous administration of G-CSF at the onset of reperfusion attenuates ischemia/reperfusion injury through PI3K/Akt pathway in the in vivo model. G-CSF administration can be a promising candidate for the adjunctive therapy for patients with acute myocardial infarction.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Dogs , Myocardium/metabolism , Peroxidase/metabolism , Ventricular Fibrillation/drug therapy
12.
Biochem Biophys Res Commun ; 340(4): 1125-33, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16403436

ABSTRACT

The ubiquitin-proteasome system contributes to regulation of apoptosis degrading apoptosis-regulatory proteins. Marked accumulation of ubiquitinated proteins in cardiomyocytes of human failing hearts suggested impaired ubiquitin-proteasome system in heart failure. Since cardiomyocyte apoptosis contributes to the progression of cardiac dysfunction in pressure-overloaded hearts, we investigated the role of ubiquitin-proteasome system in such conditions. We found that proteasome activities already depressed before the onset of cardiac dysfunction in pressure-overloaded hearts of mice. Cardiomyocyte apoptosis was observed along with depression of proteasome activities and elevation of proapoptotic/antiapoptotic protein ratio in failing hearts. In cultured cardiomyocytes, pharmacological inhibition of proteasome accumulated proapoptotic proteins such as p53 and Bax. Gene silencing of these proapoptotic proteins by RNA interference prevented the accumulation of respective proteins and attenuated cardiomyocyte apoptosis induced by proteasome inhibition. We conclude that depression of proteasome activities contributes to cardiac dysfunction resulting from cardiomyocyte apoptosis through accumulation of proapoptotic proteins by impaired degradation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Adult , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred WKY
13.
Hypertension ; 46(1): 113-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15956114

ABSTRACT

Rapid nongenomic actions of aldosterone independent of mineralocorticoid receptors (MRs) on vascular tone are divergent. Until now, the rapid nongenomic actions of aldosterone on vascular tone of coronary artery and cardiac function in the in vivo ischemic hearts were not still fully estimated. Furthermore, although aldosterone can modulate protein kinase C (PKC) activity, there is no clear consensus whether PKC is involved in the nongenomic actions of aldosterone on the ischemic hearts. In open chest dogs, the selective infusion of aldosterone into the left anterior descending coronary artery (LAD) reduced coronary blood flow (CBF) in the nonischemic hearts in a dose-dependent manner. Also, in the ischemic state that CBF was decreased to 33% of the baseline, the intracoronary administration of aldosterone (0.1 nmol/L) rapidly decreased CBF (37.4+/-3.8 to 19.3+/-5.2 mL/100 g/min; P<0.05), along with decreases in fractional shortening (FS) (8.4+/-0.7 to 5.4+/-0.4%; P<0.05) and lactate extraction rate (LER) (-31.7+/-2.9 to -41.4+/-3.7%; P<0.05). The decrease in CBF was reproduced by the infusion of bovine serum albumin-conjugated aldosterone. Notably, these aldosterone-induced deteriorations of myocardial contractile and metabolic functions were blunted by the co-administration of GF109203X, an inhibitor of PKC, but not spironolactone. In addition, aldosterone activated vascular PKC. These results indicate that aldosterone nongenomically induces vasoconstriction via PKC-dependent pathways possibly through membrane receptors, which leads to the worsening of the cardiac contractile and metabolic functions in the ischemic hearts. Elevation of plasma or cardiac aldosterone levels may be deleterious to ischemic heart disease through its nongenomic effects.


Subject(s)
Aldosterone/pharmacology , Heart/physiopathology , Myocardial Ischemia/physiopathology , Protein Kinase C/metabolism , Aldosterone/administration & dosage , Animals , Coronary Circulation/drug effects , Coronary Vessels , Dogs , Enzyme Activation/drug effects , Heart/drug effects , Hemodynamics/drug effects , Injections, Intra-Arterial , Myocardial Contraction , Myocardium/enzymology , Vasoconstriction
14.
Cardiovasc Drugs Ther ; 19(1): 33-40, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15883754

ABSTRACT

Although recent studies suggest that erythropoietin (EPO) may reduce multiple features of the myocardial ischemia/reperfusion injury, the cellular mechanisms and the clinical implications of EPO-induced cardioprotection are still unclear. Thus, in this study, we clarified dose-dependent effects of EPO administered just before reperfusion on infarct size and the incidence of ventricular fibrillation and evaluated the involvement of the phosphatidylinositol-3 (PI3) kinase in the in vivo canine model. The canine left anterior descending coronary artery was occluded for 90 min followed by 6 h of reperfusion. A single intravenous administration of EPO just before reperfusion significantly reduced infarct size (high dose (1,000 IU/kg): 7.7 +/- 1.6%, low dose (100 IU/kg): 22.1 +/- 2.4%, control: 40.0 +/- 3.6%) in a dose-dependent manner. Furthermore, the high, but not low, dose of EPO administered as a single injection significantly reduced the incidence of ventricular fibrillation during reperfusion (high dose: 0%, low dose: 40.0%, control: 50.0%). An intracoronary administration of a PI3 kinase inhibitor, wortmannin, blunted the infarct size-limiting and anti-arrhythmic effects of EPO. Low and high doses of EPO equally induced Akt phosphorylation and decreased the equivalent number of TUNEL-positive cells in the ischemic myocardium of dogs. These effects of EPO were abolished by the treatment with wortmannin. In conclusion, EPO administered just before reperfusion reduced infarct size and the incidence of ventricular fibrillation via the PI3 kinase-dependent pathway in canine hearts. EPO administration can be a realistic strategy for the treatment of acute myocardial infarction.


Subject(s)
Cardiotonic Agents/therapeutic use , Erythropoietin/therapeutic use , Myocardial Infarction/prevention & control , Myocardial Reperfusion , Phosphatidylinositol 3-Kinases/metabolism , Ventricular Fibrillation/prevention & control , Animals , Apoptosis/drug effects , Blood Cell Count , Cardiotonic Agents/administration & dosage , Coronary Circulation/drug effects , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Erythropoietin/administration & dosage , Humans , In Situ Nick-End Labeling , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion/adverse effects , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Proteins , Ventricular Fibrillation/enzymology , Ventricular Fibrillation/pathology , Ventricular Fibrillation/physiopathology
15.
Circulation ; 111(13): 1672-8, 2005 Apr 05.
Article in English | MEDLINE | ID: mdl-15795331

ABSTRACT

BACKGROUND: Migration, proliferation, and matrix-degrading protease expression of smooth muscle cells (SMCs) are major features of intimal hyperplasia after vascular injury. Although MEK kinase 1 (MEKK1) has been shown to regulate cell migration and urokinase plasminogen activator (uPA) expression, the precise role of MEKK1 in this process remains unknown. METHODS AND RESULTS: We triggered a vascular remodeling model by complete ligation of the right common carotid artery in wild-type (WT) and MEKK1-null (MEKK1-/-) mice. The intimal areas 28 days after ligation were significantly decreased in the ligated MEKK1-/- arteries compared with WT arteries (28+/-8 versus 65+/-17 microm2, P<0.05). There were no differences in the ratios of proliferating cell nuclear antigen (PCNA)-positive cells to total cells within the arterial wall between WT and MEKK1-/- arteries. Proliferation capacity also did not differ between WT and MEKK1-/- cultured aortic smooth muscle cells (AoSMCs). In contrast, the number of intimal PCNA-positive cells 7 days after ligation was significantly smaller in MEKK1-/- arteries. Three different migration assays revealed that migration and invasion of MEKK1-/- AoSMCs were markedly impaired. Addition of full-length MEKK1 restored the migration capacity of MEKK1-/- AoSMCs. The number of MEKK1-/- AoSMCs showing lamellipodia formation by epithelial growth factor was significantly smaller compared with those of WT SMCs. Furthermore, uPA expression after ligation was markedly decreased in MEKK1-/- arteries. CONCLUSIONS: MEKK1 is implicated in vascular remodeling after blood-flow cessation by regulating the migration and uPA expression of SMCs. MEKK1 is a potential target for drug development to prevent vascular remodeling.


Subject(s)
Carotid Stenosis , Cell Movement , Hyperplasia/etiology , MAP Kinase Kinase Kinase 1/physiology , Muscle, Smooth, Vascular/cytology , Tunica Intima/pathology , Urokinase-Type Plasminogen Activator/genetics , Animals , Aorta/cytology , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , MAP Kinase Kinase Kinase 1/deficiency , Mice , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology
17.
J Mol Cell Cardiol ; 37(6): 1219-23, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15572052

ABSTRACT

Serotonin (5-hydroxytryptamine [5-HT]), which induces vasoconstriction via 5-HT2A receptors in smooth muscle cells and vasodilation through activating nitric oxide (NO) synthase (NOS) via 5-HT1B receptors in endothelial cells, possesses divergent effects on regulating vascular tone. These facts lead us to consider that sarpogrelate, a 5-HT2A receptor blocker, may increase coronary blood flow (CBF) via either attenuation of vasoconstriction through 5-HT2A receptor blockade or augmentation of vasodilation by relative stimulation of NOS through 5-HT1B receptor and we tested this hypothesis in ischemic canine hearts. In open chest dogs, coronary perfusion pressure was reduced so that CBF was decreased to 33% of the baseline and kept constant. Thereafter, sarpogrelate was infused selectively into the left anterior descending artery with and without either an inhibitor of NOS (NG-nitro-L-arginine methyl ester (L-NAME)) or a 5-HT1B receptor antagonist (GR55562). An intracoronary administration of sarpogrelate increased CBF (34.0 +/- 4.0 to 44.5 +/- 4.4 ml/100 g/min, P < 0.05), along with the cardiac NOx release (3.2 +/- 0.6 to 6.8 +/- 1.2 nmol/ml, P < 0.05). The increases in both CBF and NOx by sarpogrelate were completely blunted by the co-administration of either L-NAME or GR55562. Interestingly, sarpogrelate increased the cardiac serotonin release (-4.8 +/- 3.2 vs. 22.1 +/- 1.5 ng/ml, P < 0.05, respectively) in the hypoperfused heart. Immunohistochemical analysis showed that sarpogrelate induced serotonin production in ischemic cardiac myocytes. These results suggest that sarpogrelate increases CBF via augmented cardiac NO production through 5-HT1B receptor activation along with the blockade of 5-HT2A receptors. The increase in cardiac release of serotonin may increase NO production in the ischemic heart.


Subject(s)
Heart/drug effects , Myocardium/metabolism , Nitric Oxide/blood , Receptor, Serotonin, 5-HT1B/metabolism , Serotonin 5-HT2 Receptor Antagonists , Succinates/pharmacology , Animals , Dogs , Immunohistochemistry , Serotonin/biosynthesis , Serotonin/blood , Serotonin/genetics , Time Factors
18.
Circulation ; 110(6): 705-12, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15289376

ABSTRACT

BACKGROUND: The endoplasmic reticulum (ER) is recognized as an organelle that participates in folding secretory and membrane proteins. The ER responds to stress by upregulating ER chaperones, but prolonged and/or excess ER stress leads to apoptosis. However, the potential role of ER stress in pathophysiological hearts remains unclear. METHODS AND RESULTS: Mice were subjected to transverse aortic constriction (TAC) or sham operation. Echocardiographic analysis demonstrated that mice 1 and 4 weeks after TAC had cardiac hypertrophy and failure, respectively. Cardiac expression of ER chaperones was significantly increased 1 and 4 weeks after TAC, indicating that pressure overload by TAC induced prolonged ER stress. In addition, the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells increased, and caspase-3 was cleaved in failing hearts. The antagonism of angiotensin II type 1 receptor prevented upregulation of ER chaperones and apoptosis in failing hearts. On the other hand, angiotensin II upregulated ER chaperones and induced apoptosis in cultured adult rat cardiac myocytes. We also investigated possible signaling pathways for ER-initiated apoptosis. The CHOP- (a transcription factor induced by ER stress), but not JNK- or caspase-12-, dependent pathway was activated in failing hearts by TAC. Pharmacological ER stress inducers upregulated ER chaperones and induced apoptosis in cultured cardiac myocytes. Finally, mRNA levels of ER chaperones were markedly increased in failing hearts of patients with elevated brain natriuretic peptide levels. CONCLUSIONS: These findings suggest that pressure overload by TAC induces prolonged ER stress, which may contribute to cardiac myocyte apoptosis during progression from cardiac hypertrophy to failure.


Subject(s)
Aortic Valve Stenosis/complications , Apoptosis , Cardiomegaly/physiopathology , Endoplasmic Reticulum/physiology , Heart Failure/physiopathology , Myocytes, Cardiac/pathology , Stress, Physiological/physiopathology , Angiotensin II/pharmacology , Angiotensin II/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aortic Valve Stenosis/physiopathology , Cardiomegaly/complications , Cardiomegaly/genetics , Cells, Cultured/drug effects , Cells, Cultured/pathology , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum/drug effects , Gene Expression Regulation/drug effects , Heart Failure/etiology , Heart Failure/genetics , Humans , Imidazoles/pharmacology , Ligation , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones/biosynthesis , Molecular Chaperones/genetics , Myocytes, Cardiac/drug effects , Natriuretic Peptide, Brain/physiology , Olmesartan Medoxomil , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred WKY , Signal Transduction , Stress, Physiological/chemically induced , Tetrazoles/pharmacology , Thapsigargin/toxicity , Tunicamycin/toxicity
19.
Gene ; 302(1-2): 83-94, 2003 Jan 02.
Article in English | MEDLINE | ID: mdl-12527199

ABSTRACT

The mouse zinc-fingers and homeoboxes 1 (ZHX1) gene was cloned and its transcriptional regulatory mechanism analysed. The mouse ZHX1 gene spans approximately 29 kb and consists of five exons. Exons 1-3 contain the nucleotide sequence of the 5'-noncoding region of mouse ZHX1 cDNA, exon 4 contains a part of the 5'-noncoding region, an entire coding sequence, and a part of the 3'-noncoding sequence, and exon 5 contains the resulting 3'-noncoding sequence. The ZHX1 gene exists as one copy in the haploid mouse genome. Two species of ZHX1 mRNA with or without the nucleotide sequence of the third exon are produced by an alternative splicing. To investigate the regulatory elements involved in the transcription of the ZHX1 gene, transient DNA transfection experiments with ZHX1/firefly luciferase reporter genes were performed using a lipofection method. Functional analyses of a series of 5'- and 3'-deletion constructs of the reporter genes revealed that the nucleotide sequence between -59 and +50 is required for full promoter activity in mouse embryonal carcinoma F9 cells. Two positive regulatory cis-acting elements in the region were identified. These elements, designated as Box A and Box B, are located between nucleotides -47 and -42 and +22 and +27, respectively, and synergistically stimulate transcription of the mouse ZHX1 gene. Electrophoretic mobility shift assays with specific competitors and antibodies show that PEA3 and Yin and Yang 1 (YY1) bind to Box A and Box B, respectively. Thus, we conclude that PEA3 and YY1 synergistically stimulate the transcription of the ZHX1 gene.


Subject(s)
Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Zinc Fingers/genetics , Alternative Splicing , Animals , Base Sequence , Binding Sites/genetics , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Erythroid-Specific DNA-Binding Factors , Exons , Gene Expression Regulation , Genes/genetics , Introns , Male , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Sequence Analysis, DNA , Transcription Factors/metabolism , Transcription Initiation Site , Tumor Cells, Cultured , YY1 Transcription Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...