Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 231, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38418926

ABSTRACT

Current differentiation protocols for human induced pluripotent stem cells (hiPSCs) produce heterogeneous cardiomyocytes (CMs). Although chamber-specific CM selection using cell surface antigens enhances biomedical applications, a cell surface marker that accurately distinguishes between hiPSC-derived atrial CMs (ACMs) and ventricular CMs (VCMs) has not yet been identified. We have developed an approach for obtaining functional hiPSC-ACMs and -VCMs based on CD151 expression. For ACM differentiation, we found that ACMs are enriched in the CD151low population and that CD151 expression is correlated with the expression of Notch4 and its ligands. Furthermore, Notch signaling inhibition followed by selecting the CD151low population during atrial differentiation leads to the highly efficient generation of ACMs as evidenced by gene expression and electrophysiology. In contrast, for VCM differentiation, VCMs exhibiting a ventricular-related gene signature and uniform action potentials are enriched in the CD151high population. Our findings enable the production of high-quality ACMs and VCMs appropriate for hiPSC-derived chamber-specific disease models and other applications.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Cell Differentiation/physiology , Heart Ventricles , Myocytes, Cardiac/metabolism , Tetraspanin 24/genetics , Tetraspanin 24/metabolism
2.
Stem Cell Reports ; 18(11): 2108-2122, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37802074

ABSTRACT

Engineered cardiac tissue (ECT) using human induced pluripotent stem cell-derived cardiomyocytes is a promising tool for modeling heart disease. However, tissue immaturity makes robust disease modeling difficult. Here, we established a method for modeling hypertrophic cardiomyopathy (HCM) malignant (MYH7 R719Q) and nonmalignant (MYBPC3 G115∗) pathogenic sarcomere gene mutations by accelerating ECT maturation using an ERRγ agonist, T112, and mechanical stretching. ECTs treated with T112 under 10% elongation stimulation exhibited more organized and mature characteristics. Whereas matured ECTs with the MYH7 R719Q mutation showed broad HCM phenotypes, including hypertrophy, hypercontraction, diastolic dysfunction, myofibril misalignment, fibrotic change, and glycolytic activation, matured MYBPC3 G115∗ ECTs displayed limited phenotypes, which were primarily observed only under our new maturation protocol (i.e., hypertrophy). Altogether, ERRγ activation combined with mechanical stimulation enhanced ECT maturation, leading to a more accurate manifestation of HCM phenotypes, including non-cardiomyocyte activation, consistent with clinical observations.


Subject(s)
Cardiomyopathy, Hypertrophic , Induced Pluripotent Stem Cells , Humans , Tissue Engineering , Carrier Proteins/genetics , Induced Pluripotent Stem Cells/pathology , Cardiomyopathy, Hypertrophic/pathology , Phenotype , Myocytes, Cardiac/physiology , Mutation , Hypertrophy/pathology
3.
Nat Commun ; 12(1): 3596, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34155205

ABSTRACT

One of the earliest maturation steps in cardiomyocytes (CMs) is the sarcomere protein isoform switch between TNNI1 and TNNI3 (fetal and neonatal/adult troponin I). Here, we generate human induced pluripotent stem cells (hiPSCs) carrying a TNNI1EmGFP and TNNI3mCherry double reporter to monitor and isolate mature sub-populations during cardiac differentiation. Extensive drug screening identifies two compounds, an estrogen-related receptor gamma (ERRγ) agonist and an S-phase kinase-associated protein 2 inhibitor, that enhances cardiac maturation and a significant change to TNNI3 expression. Expression, morphological, functional, and molecular analyses indicate that hiPSC-CMs treated with the ERRγ agonist show a larger cell size, longer sarcomere length, the presence of transverse tubules, and enhanced metabolic function and contractile and electrical properties. Here, we show that ERRγ-treated hiPSC-CMs have a mature cellular property consistent with neonatal CMs and are useful for disease modeling and regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Receptors, Estrogen/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Myocytes, Cardiac/metabolism , Receptors, Estrogen/chemistry , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Sarcolemma/drug effects , Sarcolemma/metabolism , Sarcomeres/drug effects , Sarcomeres/metabolism , Transcriptome/drug effects , Troponin I/genetics , Troponin I/metabolism
4.
SLAS Discov ; 23(2): 154-163, 2018 02.
Article in English | MEDLINE | ID: mdl-28846466

ABSTRACT

Ghrelin O-acyl transferase (GOAT; MBOAT4) catalyzes O-acylation at serine-3 of des-acyl ghrelin. Acyl ghrelin is secreted by stomach X/A-like cells and plays a role in appetite and metabolism. Therefore, GOAT has been expected to be a novel antiobesity target because it is responsible for acyl ghrelin production. Here, we report homogeneous time-resolved fluorescence (HTRF) and enzyme-linked immunosorbent assay (ELISA) methods utilizing human GOAT-expressing microsomes as a novel high-throughput assay system for the discovery of hit compounds and optimization of lead compounds. Hit compounds exemplified by compound A (2-[(2,4-dichlorobenzyl)sulfanyl]-1,3-benzoxazole-5-carboxylic acid) were identified by high-throughput screening using the HTRF assay and confirmed to have GOAT inhibitory activity using the ELISA. Based on the hit compound information, the novel lead compound (compound B, (4-chloro-6-{[2-methyl-6-(trifluoromethyl)pyridin-3-yl]methoxy}-1-benzothiophen-3-yl)acetic acid) was synthesized and exhibited potent GOAT inhibition with oral bioavailability. Both the hit compound and lead compound showed octanoyl-CoA competitive inhibitory activity. Moreover, these two compounds decreased acyl ghrelin production in the stomach of mice after their oral administration. These novel findings demonstrate that GOAT is a druggable target, and its inhibitors are promising antiobesity drugs.


Subject(s)
Acyltransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ghrelin/metabolism , Acyl Coenzyme A/metabolism , Acylation/drug effects , Administration, Oral , Animals , Biological Availability , Drug Discovery/methods , Enzyme Inhibitors/pharmacokinetics , High-Throughput Screening Assays/methods , Humans , Male , Mice , Mice, Inbred C57BL , Microsomes/drug effects , Microsomes/metabolism , Stomach/drug effects
5.
Biochem Biophys Res Commun ; 488(2): 362-367, 2017 06 24.
Article in English | MEDLINE | ID: mdl-28501616

ABSTRACT

ß1-adrenergic receptor (Adrb1) belongs to the superfamily of G-protein-coupled receptors (GPCRs) and plays a critical role in the regulation of heart rate and myocardial contraction force. GPCRs are phosphorylated at multiple sites to regulate distinct signal transduction pathways in different tissues. However, little is known about the location and function of distinct phosphorylation sites of Adrb1 in vivo. To clarify the mechanisms underlying functional regulation associated with Adrb1 phosphorylation in vivo, we aimed to identify Adrb1 phosphorylation sites in the mouse heart using phosphoproteomics techniques with nano-flow liquid chromatography/tandem mass spectrometry (LC-MS/MS). We revealed the phosphorylation residues of Adrb1 to be Ser274 and Ser280 in the third intracellular loop and Ser412, Ser417, Ser450, Ser451, and Ser462 at the C-terminus. We also found that phosphorylation at Ser274, Ser280, and Ser462 was enhanced in response to stimulation with an Adrb1 agonist. This is the first study to identify Adrb1 phosphorylation sites in vivo. These findings will provide novel insights into the regulatory mechanisms mediated by Adrb1 phosphorylation.


Subject(s)
Myocardium/chemistry , Myocardium/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/metabolism , Animals , Chromatography, Liquid , Heart , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Proteomics , Signal Transduction , Tandem Mass Spectrometry
6.
J Biomol Screen ; 17(9): 1192-203, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22706346

ABSTRACT

Human-induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) at different stages (approximate days 30, 60, and 90) were used to determine the appropriate stage for functional and morphological assessment of drug effects in vitro. The hiPS-CMs had spontaneous beating activity, and ß-adrenergic function was comparable in all stages of differentiation. Microelectrode array analyses using ion channel blockers indicated that the electrophysiological properties of these ion channels were comparable at all differentiation stages. Ultrastructural analysis using electron microscopy showed that myofibrillar structures at days 60 and 90 were similarly distributed and more mature than that at day 30. Analysis of motion vectors in contracting cells showed that the velocity of contraction was the highest at day 90 and was the most mature among the three stages. Gene expression analysis demonstrated that expression of some genes related to myofilament and sarcoplasmic reticulum increased with maturation of morphological and contractile properties. In conclusion, day 30 cardiomyocytes are useful for basic screening such as the assessment of electrophysiological properties, and days 60 and 90 are the appropriate differentiation stage for morphological assays. For the assay of contractile function associated with subcellular components such as sarcoplasmic reticulum, day 90 cardiomyocytes are the most suitable.


Subject(s)
Drug Evaluation, Preclinical , Induced Pluripotent Stem Cells/cytology , Ion Channels/metabolism , Myocytes, Cardiac/cytology , Action Potentials/drug effects , Action Potentials/physiology , Calcium Channel Blockers/pharmacology , Cell Differentiation/physiology , Gene Expression Profiling , Humans , Ion Channels/antagonists & inhibitors , Microelectrodes , Microscopy, Electron, Transmission , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Myofibrils/ultrastructure , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Time Factors
7.
J Biomol Screen ; 14(3): 239-45, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19211779

ABSTRACT

Although embryonic stem cell (ESC)-derived cardiomyocytes may be a powerful tool in drug discovery, their potential has not yet been fully explored. Nor has a detailed comparison with adult heart tissue been performed. We have developed a method for efficient production of cardiomyocyte-rich embryoid bodies (EBs) from murine ESCs. Analysis of global gene expression profiles showed that EBs on day 7 and/or 21 of differentiation (d7CMs and d21CMs, respectively) were similar to adult heart tissue for genes categorized as regulators of muscle contraction or voltage-gated ion channel activity, although d21CMs were more mature than d7CMs for contractile components related to morphological structures. Calcium and sodium channel blockers altered Ca2+ transients, and isoproterenol, a beta-adrenergic compound, increased the rate of beating in d7CMs and d21CMs. Our gene analytic system therefore enabled us to identify genes that are expressed in the physiological pathways associated with ion channels and structural components in d7CMs and d21CMs. We conclude that EBs might be of use for the basic screening of drugs that might affect contractile function through ion channels.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Profiling/methods , Heart/physiology , Myocytes, Cardiac/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Embryo, Mammalian , Embryonic Stem Cells/cytology , Embryonic Stem Cells/ultrastructure , Gene Expression Regulation , Heart Rate/drug effects , Immunohistochemistry , Ion Channels/metabolism , Ion Channels/physiology , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Microarray Analysis , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Myocytes, Cardiac/ultrastructure , Principal Component Analysis , Time Factors
8.
Circ Res ; 101(12): 1328-38, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17916779

ABSTRACT

Biomechanical overload induces cardiac hypertrophy and heart failure, and reactive oxygen species (ROS) play a role in both processes. Thioredoxin-Interacting Protein (Txnip) is encoded by a mechanically-regulated gene that controls cell growth and apoptosis in part through interaction with the endogenous dithiol antioxidant thioredoxin. Here we show that Txnip is a critical regulator of the cardiac response to pressure overload. We generated inducible cardiomyocyte-specific and systemic Txnip-null mice (Txnip-KO) using Flp/frt and Cre/loxP technologies. Compared with littermate controls, Txnip-KO hearts had attenuated cardiac hypertrophy and preserved left ventricular (LV) contractile reserve through 4 weeks of pressure overload; however, the beneficial effects were not sustained and Txnip deletion ultimately led to maladaptive LV remodeling at 8 weeks of pressure overload. Interestingly, these effects of Txnip deletion on cardiac performance were not accompanied by global changes in thioredoxin activity or ROS; instead, Txnip-KO hearts had a robust increase in myocardial glucose uptake. Thus, deletion of Txnip plays an unanticipated role in myocardial energy homeostasis rather than redox regulation. These results support the emerging concept that the function of Txnip is not as a simple thioredoxin inhibitor but as a metabolic control protein.


Subject(s)
Blood Pressure/genetics , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Carrier Proteins/genetics , Gene Deletion , Gene Targeting , Thioredoxins/genetics , Animals , Cardiomegaly/metabolism , Carrier Proteins/physiology , Female , Gene Targeting/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thioredoxins/physiology , Ventricular Remodeling/physiology
9.
Cell Metab ; 6(1): 25-37, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17618854

ABSTRACT

Downregulation and functional deactivation of the transcriptional coactivator PGC-1alpha has been implicated in heart failure pathogenesis. We hypothesized that the estrogen-related receptor alpha (ERRalpha), which recruits PGC-1alpha to metabolic target genes in heart, exerts protective effects in the context of stressors known to cause heart failure. ERRalpha(-/-) mice subjected to left ventricular (LV) pressure overload developed signatures of heart failure including chamber dilatation and reduced LV fractional shortening. (31)P-NMR studies revealed abnormal phosphocreatine depletion in ERRalpha(-/-) hearts subjected to hemodynamic stress, indicative of a defect in ATP reserve. Mitochondrial respiration studies demonstrated reduced maximal ATP synthesis rates in ERRalpha(-/-) hearts. Cardiac ERRalpha target genes involved in energy substrate oxidation, ATP synthesis, and phosphate transfer were downregulated in ERRalpha(-/-) mice at baseline or with pressure overload. These results demonstrate that the nuclear receptor ERRalpha is required for the adaptive bioenergetic response to hemodynamic stressors known to cause heart failure.


Subject(s)
Heart/physiopathology , Receptors, Estrogen/physiology , Ventricular Pressure/physiology , Ventricular Remodeling/physiology , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Blood Pressure , Cardiac Output, Low , Cardiomegaly/physiopathology , Energy Metabolism , Female , Gene Expression Profiling , Heart/embryology , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , ERRalpha Estrogen-Related Receptor
10.
Am J Physiol Heart Circ Physiol ; 292(5): H2237-47, 2007 May.
Article in English | MEDLINE | ID: mdl-17209001

ABSTRACT

In the myocardium, the Na(+)/H(+) exchanger isoform-1 (NHE1) activity is detrimental during ischemia-reperfusion (I/R) injury, causing increased intracellular Na(+) (Na(i)(+)) accumulation that results in subsequent Ca(2+) overload. We tested the hypothesis that increased expression of NHE1 would accentuate myocardial I/R injury. Transgenic mice were created that increased the Na(+)/H(+) exchanger activity specifically in the myocardium. Intact hearts from transgenic mice at 10-15 wk of age showed no change in heart performance, resting intracellular pH (pH(i)) or phosphocreatine/ATP levels. Transgenic and wild-type (WT) hearts were subjected to 20 min of ischemia followed by 40 min of reperfusion. Surprisingly, the percent recovery of rate-pressure product (%RPP) after I/R improved in NHE1-overexpressing hearts (64 +/- 5% vs. 41 +/- 5% in WT; P < 0.05). In addition, NMR spectroscopy revealed that NHE1 overexpressor hearts contained higher ATP during early reperfusion (levels P < 0.05), and there was no difference in Na(+) accumulation during I/R between transgenic and WT hearts. HOE642 (cariporide), an NHE1 inhibitor, equivalently protected both WT and NHE1-overexpressing hearts. When hearts were perfused with bicarbonate-free HEPES buffer to eliminate the contribution of HCO(3)(-) transporters to pH(i) regulation, there was no difference in contractile recovery after reperfusion between controls and transgenics, but NHE1-overexpressing hearts showed a greater decrease in ATP during ischemia. These results indicate that the basal activity of NHE1 is not rate limiting in causing damage during I/R, therefore, increasing the level of NHE1 does not enhance injury and can have some small protective effects.


Subject(s)
Myocardium/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium/metabolism , Water-Electrolyte Imbalance/metabolism , Animals , Hydrogen-Ion Concentration , Mice , Mice, Transgenic , Myocardial Reperfusion Injury , Sodium-Hydrogen Exchangers/genetics , Up-Regulation , Water-Electrolyte Imbalance/complications
11.
Circulation ; 112(22): 3451-61, 2005 Nov 29.
Article in English | MEDLINE | ID: mdl-16316967

ABSTRACT

BACKGROUND: Mitochondria and sarcomeres have a well-defined architectural relation that partially depends on the integrity of the cytoskeletal network. An R120G missense mutation in the small heat shock protein alpha-B-crystallin (CryAB) causes desmin-related cardiomyopathy. Desmin-related cardiomyopathy is characterized by the formation of intracellular aggregates containing CryAB and desmin that are amyloid positive, and disease can be recapitulated in transgenic mice by cardiac-specific expression of the mutant protein. METHODS AND RESULTS: To understand the resultant pathology, we explored the acute effects of R120G expression both in vitro and in vivo. In vitro, transfection of adult cardiomyocytes with R120G-expressing adenovirus resulted in altered contractile mechanics. In vivo, as the cytoskeletal network is disturbed but before deficits in organ function can be detected, alterations in mitochondrial organization and architecture occur, leading to a reduction in the maximal rate of oxygen consumption with substrates that utilize complex I activity, alterations in the permeability transition pore, and compromised inner membrane potential. Apoptotic pathways are subsequently activated, which eventually results in cardiomyocyte death, dilation, and heart failure. CONCLUSIONS: Cardiac chaperone dysfunction acutely leads to altered cardiomyocyte mechanics, perturbations in mitochondrial-sarcomere architecture, and deficits in mitochondrial function, which can result in activation of apoptosis and heart failure.


Subject(s)
Apoptosis , Cardiomyopathies/etiology , Desmin/physiology , Mitochondrial Diseases/complications , Mutation, Missense , alpha-Crystallin B Chain/genetics , Amyloid , Animals , Cardiomyopathies/pathology , Cells, Cultured , Dimerization , Humans , Ion Channels , Mice , Mice, Transgenic , Mitochondrial Diseases/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Myocardial Contraction , Myocytes, Cardiac/cytology , Rats , Transfection
12.
Trends Cardiovasc Med ; 15(8): 283-90, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16297765

ABSTRACT

Recent data suggest that in addition to regulating apoptosis, Bcl-2 (an anti-apoptotic protein overexpressed in B-cell lymphoma) and Bcl-2 family members also regulate mitochondrial and cell physiology. t-Bid, a Bcl-2 family member, has been shown to modulate reorganization of mitochondrial cristae. Bcl-2 appears to regulate voltage-dependent anion channel permeability, which has important consequences for mitochondrial transport of adenine nucleotides, Ca(2+), and other metabolites. BAD, a pro-apoptotic Bcl-2 family member, is required for the binding of glucokinase to a mitochondrial complex, and BAD null mice have altered glucose homeostasis. It has been suggested that Bcl-2 family members may regulate important mitochondrial/cell functions and serve as sentinels to detect abnormalities in these pathways and, when the abnormalities are severe enough, to initiate or facilitate cell death. Understanding the physiologic processes controlled by Bcl-2 will be important in understanding cell regulation, and it may also provide new insights into the regulation of apoptosis.


Subject(s)
Apoptosis/physiology , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Signal Transduction/physiology , Adenosine Triphosphate/biosynthesis , Animals , Cell Count , Cell Membrane/physiology , Electron Transport/physiology , Homeostasis/physiology , Humans , Membrane Potentials/physiology , Mitochondria/metabolism , bcl-2-Associated X Protein/physiology
13.
Circ Res ; 97(9): 916-21, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16179590

ABSTRACT

During ischemia and reperfusion, with an increase in intracellular Na+ and a depolarized membrane potential, Ca2+ may enter the myocyte in exchange for intracellular Na+ via reverse-mode Na+-Ca2+ exchange (NCX). To test the role of Ca2+ entry via NCX during ischemia and reperfusion, we studied mice with cardiac-specific ablation of NCX (NCX-KO) and demonstrated that reverse-mode Ca2+ influx is absent in the NCX-KO myocytes. Langendorff perfused hearts were subjected to 20 minutes of global ischemia followed by 2 hours of reperfusion, during which time we monitored high-energy phosphates using 31P-NMR and left-ventricular developed pressure. In another group of hearts, we monitored intracellular Na+ using 23Na-NMR. Consistent with Ca2+ entry via NCX during ischemia, we found that hearts lacking NCX exhibited less of a decline in ATP during ischemia, delayed ischemic contracture, and reduced maximum contracture. Furthermore, on reperfusion following ischemia, NCX-KO hearts had much less necrosis, better recovery of left-ventricular developed pressure, improved phosphocreatine recovery, and reduced Na+ overload. The improved recovery of function following ischemia in NCX-KO hearts was not attributable to the reduced preischemic contractility in NCX-KO hearts, because when the preischemic workload was matched by treatment with isoproterenol, NCX-KO hearts still exhibited improved postischemic function compared with wild-type hearts. Thus, NCX-KO hearts were significantly protected against ischemia-reperfusion injury, suggesting that Ca2+ entry via reverse-mode NCX is a major cause of ischemia/reperfusion injury.


Subject(s)
Myocardial Reperfusion Injury/etiology , Myocardium/metabolism , Sodium-Calcium Exchanger/physiology , Animals , Calcium/metabolism , Energy Metabolism , Isoproterenol/pharmacology , Mice , Mice, Knockout , Myocardial Contraction , Myocardial Reperfusion Injury/prevention & control , Phenotype , Sodium/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors
14.
Circ Res ; 95(7): 734-41, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15345651

ABSTRACT

The antiapoptotic protein Bcl-2 is targeted to the mitochondria, but it is uncertain whether Bcl-2 affects only myocyte survival after ischemia, or whether it also affects metabolic functions of mitochondria during ischemia. Hearts from mice overexpressing human Bcl-2 and from their wild-type littermates (WT) were subjected to 24 minutes of global ischemia followed by reperfusion. During ischemia, the decrease in pH(i) and the initial rate of decline in ATP were significantly reduced in Bcl-2 hearts compared with WT hearts (P<0.05). The reduced acidification during ischemia was dependent on the activity of mitochondrial F1F0-ATPase. In the presence of oligomycin (Oligo), an F1F0-ATPase inhibitor, the decrease in pH(i) was attenuated in WT hearts, but in Bcl-2 hearts, Oligo had no additional effect on pH(i) during ischemia. Likewise, addition of Oligo to WT hearts slowed the rate of decline in ATP during ischemia to a level similar to that observed in Bcl-2 hearts, but addition of Oligo had no significant effect on the rate of decline in ATP in Bcl-2 hearts during ischemia. These data are consistent with Bcl-2-mediated inhibition of consumption of glycolytic ATP. Furthermore, mitochondria from Bcl-2 hearts have a reduced rate of consumption of ATP on uncoupler addition. This could be accomplished by limiting ATP entry into the mitochondria through the voltage-dependent anion channel, and/or the adenine nucleotide transporter, or by direct inhibition of the F1F0-ATPase. Immunoprecipitation showed greater interaction between Bcl-2 and voltage-dependent anion channel during ischemia. These data indicate that Bcl-2 modulation of metabolism contributes to cardioprotection.


Subject(s)
Mitochondria, Heart/physiology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology , Adenosine Triphosphate/metabolism , Anaerobiosis , Animals , Apoptosis , Blotting, Western , Cytosol/chemistry , Energy Metabolism/drug effects , Female , Gene Expression Regulation , Genes, bcl-2 , Glycolysis , Humans , Hydrogen-Ion Concentration , Male , Mice , Mice, Transgenic , Mitochondria, Heart/drug effects , Myocardial Contraction , Myocardial Infarction/pathology , Nuclear Magnetic Resonance, Biomolecular , Oligomycins/pharmacology , Phosphocreatine/metabolism , Porins/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proton-Translocating ATPases/physiology , Recombinant Fusion Proteins/physiology , Voltage-Dependent Anion Channels
15.
J Mol Cell Cardiol ; 37(3): 747-53, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15350847

ABSTRACT

We previously showed that beta-adrenergic stimulation revealed male/female differences in susceptibility to ischemia/reperfusion (I/R) injury. To explore whether altered [Na(+)](i) regulation is involved in the mechanism of this sex difference, we measured [Na(+)](i) by (23)Na NMR spectroscopy in isolated perfused mouse hearts. [Na(+)](i) increased to 195 +/- 3% (mean +/- S.E.) of the pre-ischemic level at 20 min of ischemia in male hearts, whereas [Na(+)](i) accumulation was slightly less in female hearts (176 +/- 2%, P < 0.05). There was no significant difference in the recovery of contractile function after reperfusion (male: 30.6 +/- 3.8%; female: 35.0 +/- 1.9%; P > 0.05). If hearts were treated with isoproterenol (ISO, 10 nmol/l), males exhibited significantly poorer recovery of post-ischemic contractile function than females (male: 13.0 +/- 1.9%; female: 28.1 +/- 1.2%; P < 0.05), and a significantly higher [Na(+)](i) accumulation during ischemia (male: 218 +/- 8%; female: 171 +/- 2%; P < 0.05). This ISO-induced male/female difference in [Na(+)](i) accumulation or contractile function was blocked by the nitric oxide synthase inhibitor, N(omega)-nitro-l-arginine methyl ester (1 micromol/l). Furthermore, in ISO-treated hearts, the Na(+)/K(+)-ATPase inhibitor, ouabain (200 micromol/l) did not abolish the male/female difference in [Na(+)](i) accumulation during I/R or functional protection. Thus the data show that the sex difference in the [Na(+)](i) regulation is mediated through a NO-dependent mechanism, and the difference in susceptibility to I/R injury appears to result from a difference in Na(+) influx.


Subject(s)
Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Sex Characteristics , Sodium/metabolism , Animals , Cardiotonic Agents/administration & dosage , Female , Isoproterenol/administration & dosage , Male , Mice , Myocardial Contraction/drug effects , Myocardial Ischemia/complications , Myocardial Ischemia/drug therapy , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/pathology , Nitric Oxide/metabolism
16.
Circulation ; 108(19): 2304-7, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14597593

ABSTRACT

BACKGROUND: Current treatment for acute myocardial infarction (AMI) focuses on reestablishing blood flow (reperfusion). Paradoxically, reperfusion itself may cause additional injury to the heart. We previously found that delta-protein kinase C (deltaPKC) inhibition during simulated ischemia/reperfusion in isolated rat hearts is cardioprotective. We focus here on the role for deltaPKC during reperfusion only, using an in vivo porcine model of AMI. METHODS AND RESULTS: An intracoronary application of a selective deltaPKC inhibitor to the heart at the time of reperfusion reduced infarct size, improved cardiac function, inhibited troponin T release, and reduced apoptosis. Using 31P NMR in isolated perfused mouse hearts, we found a faster recovery of ATP levels in hearts treated with the deltaPKC inhibitor during reperfusion only. CONCLUSIONS: Reperfusion injury after cardiac ischemia is mediated, at least in part, by deltaPKC activation. This study suggests that including a deltaPKC inhibitor at reperfusion may improve the outcome for patients with AMI.


Subject(s)
Enzyme Inhibitors/therapeutic use , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/prevention & control , Oligopeptides/therapeutic use , Protein Kinase C/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biomarkers , Cardiac Catheterization , Caspase 3 , Caspases/analysis , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Female , Infusions, Intra-Arterial , Mice , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Ischemia/complications , Oligopeptides/administration & dosage , Phosphocreatine/analysis , Phosphorus/analysis , Protein Kinase C/physiology , Protein Kinase C-delta , Swine , Troponin T/analysis
17.
Circ Res ; 90(4): 377-9, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11884365

ABSTRACT

We previously reported that activation of phosphatidylinositol-3-kinase (PI3-kinase) is involved in ischemic preconditioning (PC). Our goal was to determine downstream targets of PI3-kinase. In perfused rat hearts, PC (4 cycles of 5 minutes of ischemia and 5 minutes of reflow) increased phosphorylation of glycogen synthase kinase-3beta (GSK-3beta), a downstream target of PI3-kinase and protein kinase B (PKB), an effect that was blocked by wortmannin. Because phosphorylation inactivates GSK-3beta, we examined whether PC-induced phosphorylation and inhibition of GSK-3beta is important in PC by using two inhibitors of GSK-3beta, lithium and SB 216763. Pretreatment of perfused rat hearts with lithium or SB 216763, before ischemia, mimicked the protective effects of PC; hearts treated with either lithium or SB 216763 had improved postischemic function and reduced infarct size. These findings indicate that inhibition of GSK-3beta is protective and that this PI3-kinase--dependent signaling pathway may play an important role in ischemic preconditioning.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Ischemic Preconditioning, Myocardial , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Androstadienes/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , In Vitro Techniques , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Lithium/pharmacology , Male , Maleimides/pharmacology , Myocardial Ischemia/prevention & control , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...