Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Am Heart Assoc ; 6(5)2017 May 09.
Article in English | MEDLINE | ID: mdl-28487390

ABSTRACT

BACKGROUND: The amino acid response (AAR) is an evolutionarily conserved protective mechanism activated by amino acid deficiency through a key kinase, general control nonderepressible 2. In addition to mobilizing amino acids, the AAR broadly affects gene and protein expression in a variety of pathways and elicits antifibrotic, autophagic, and anti-inflammatory activities. However, little is known regarding its role in cardiac stress. Our aim was to investigate the effects of halofuginone, a prolyl-tRNA synthetase inhibitor, on the AAR pathway in cardiac fibroblasts, cardiomyocytes, and in mouse models of cardiac stress and failure. METHODS AND RESULTS: Consistent with its ability to inhibit prolyl-tRNA synthetase, halofuginone elicited a general control nonderepressible 2-dependent activation of the AAR pathway in cardiac fibroblasts as evidenced by activation of known AAR target genes, broad regulation of the transcriptome and proteome, and reversal by l-proline supplementation. Halofuginone was examined in 3 mouse models of cardiac stress: angiotensin II/phenylephrine, transverse aortic constriction, and acute ischemia reperfusion injury. It activated the AAR pathway in the heart, improved survival, pulmonary congestion, left ventricle remodeling/fibrosis, and left ventricular function, and rescued ischemic myocardium. In human cardiac fibroblasts, halofuginone profoundly reduced collagen deposition in a general control nonderepressible 2-dependent manner and suppressed the extracellular matrix proteome. In human induced pluripotent stem cell-derived cardiomyocytes, halofuginone blocked gene expression associated with endothelin-1-mediated activation of pathologic hypertrophy and restored autophagy in a general control nonderepressible 2/eIF2α-dependent manner. CONCLUSIONS: Halofuginone activated the AAR pathway in the heart and attenuated the structural and functional effects of cardiac stress.


Subject(s)
Amino Acids/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Heart Failure/prevention & control , Myocytes, Cardiac/drug effects , Piperidines/pharmacology , Protein Synthesis Inhibitors/pharmacology , Quinazolinones/pharmacology , Stress, Physiological , Amino Acids/deficiency , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Amino Acyl-tRNA Synthetases/metabolism , Animals , Autophagy/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Serine-Threonine Kinases/metabolism , Time Factors , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
2.
ACS Med Chem Lett ; 8(5): 549-554, 2017 May 11.
Article in English | MEDLINE | ID: mdl-28523109

ABSTRACT

Transient Receptor Potential Vanilloid 4 (TRPV4) is a member of the Transient Receptor Potential (TRP) superfamily of cation channels. TRPV4 is expressed in the vascular endothelium in the lung and regulates the integrity of the alveolar septal barrier. Increased pulmonary vascular pressure evokes TRPV4-dependent pulmonary edema, and therefore, inhibition of TRPV4 represents a novel approach for the treatment of pulmonary edema associated with conditions such as congestive heart failure. Herein we report the discovery of an orally active, potent, and selective TRPV4 blocker, 3-(1,4'-bipiperidin-1'-ylmethyl)-7-bromo-N-(1-phenylcyclopropyl)-2-[3-(trifluoromethyl)phenyl]-4-quinolinecarboxamide (GSK2193874, 28) after addressing an unexpected off-target cardiovascular liability observed from in vivo studies. GSK2193874 is a selective tool for elucidating TRPV4 biology both in vitro and in vivo.

3.
PLoS One ; 10(6): e0130894, 2015.
Article in English | MEDLINE | ID: mdl-26098939

ABSTRACT

Previous studies have shown that glucagon-like peptide-1 (GLP-1) provides cardiovascular benefits independent of its role on peripheral glycemic control. However, the precise mechanism(s) by which GLP-1 treatment renders cardioprotection during myocardial ischemia remain unresolved. Here we examined the role for GLP-1 treatment on glucose and fatty acid metabolism in normal and ischemic rat hearts following a 30 min ischemia and 24 h reperfusion injury, and in isolated cardiomyocytes (CM). Relative carbohydrate and fat oxidation levels were measured in both normal and ischemic hearts using a 1-13C glucose clamp coupled with NMR-based isotopomer analysis, as well as in adult rat CMs by monitoring pH and O2 consumption in the presence of glucose or palmitate. In normal heart, GLP-1 increased glucose uptake (↑64%, p<0.05) without affecting glycogen levels. In ischemic hearts, GLP-1 induced metabolic substrate switching by increasing the ratio of carbohydrate versus fat oxidation (↑14%, p<0.01) in the LV area not at risk, without affecting cAMP levels. Interestingly, no substrate switching occurred in the LV area at risk, despite an increase in cAMP (↑106%, p<0.05) and lactate (↑121%, p<0.01) levels. Furthermore, in isolated CMs GLP-1 treatment increased glucose utilization (↑14%, p<0.05) and decreased fatty acid oxidation (↓15%, p<0.05) consistent with in vivo finding. Our results show that this benefit may derive from distinct and complementary roles of GLP-1 treatment on metabolism in myocardial sub-regions in response to this injury. In particular, a switch to anaerobic glycolysis in the ischemic area provides a compensatory substrate switch to overcome the energetic deficit in this region in the face of reduced tissue oxygenation, whereas a switch to more energetically favorable carbohydrate oxidation in more highly oxygenated remote regions supports maintaining cardiac contractility in a complementary manner.


Subject(s)
Cardiotonic Agents/pharmacology , Energy Metabolism/drug effects , Glucagon-Like Peptide 1/pharmacology , Myocardium/metabolism , Reperfusion Injury/metabolism , Animals , Carbon Isotopes/metabolism , Cardiotonic Agents/administration & dosage , Cyclic AMP/metabolism , Energy Metabolism/physiology , Fatty Acids/metabolism , Glucagon-Like Peptide 1/administration & dosage , Glucose/metabolism , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Myocytes, Cardiac/metabolism , Oxygen Consumption/physiology , Rats
4.
Front Pharmacol ; 4: 149, 2013.
Article in English | MEDLINE | ID: mdl-24348421

ABSTRACT

Thymosin beta 4 (Tß4) was previously shown to reduce infarct size and improve contractile performance in chronic myocardial ischemic injury via two phases of action: an acute phase, just after injury, when Tß4 preserves ischemic myocardium via antiapoptotic or anti-inflammatory mechanisms; and a chronic phase, when Tß4 activates the growth of vascular or cardiac progenitor cells. In order to differentiate between the effects of Tß4 during the acute and during the chronic phases, and also in order to obtain detailed hemodynamic and biomarker data on the effects of Tß4 treatment suitable for use in clinical studies, we tested Tß4 in a rat model of chronic myocardial ischemia using two dosing regimens: short term dosing (Tß4 administered only during the first 3 days following injury), and long term dosing (Tß4 administered during the first 3 days following injury and also every third day until the end of the study). Tß4 administered throughout the study reduced infarct size and resulted in significant improvements in hemodynamic performance; however, chamber volumes and ejection fractions were not significantly improved. Tß4 administered only during the first 3 days following injury tended to reduce infarct size, chamber volumes and improve hemodynamic performance. Plasma biomarkers of myocyte injury were significantly reduced by Tß4 treatment during the acute injury period, and plasma ANP levels were significantly reduced in both dosing groups. Surprisingly, neither acute nor chronic Tß4 treatment significantly increased blood vessel density in peri-infarct regions. These results suggest the following: repeated dosing may be required to achieve clinically measureable improvements in cardiac function post-myocardial infarction (MI); improvement in cardiac function may be observed in the absence of a high degree of angiogenesis; and that plasma biomarkers of cardiac function and myocardial injury are sensitive pharmacodynamic biomarkers of the effects of Tß4.

5.
Cardiovasc Diabetol ; 12: 148, 2013 Oct 14.
Article in English | MEDLINE | ID: mdl-24125539

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 (GLP-1) and its mimetics reduce infarct size in the setting of acute myocardial ischemia/reperfusion (I/R) injury. However, the short serum half-life of GLP-1 and its mimetics may limit their therapeutic use in acute myocardial ischemia. Domain antibodies to serum albumin (AlbudAbs) have been developed to extend the serum half-life of short lived therapeutic proteins, peptides and small molecules. In this study, we compared the effect of a long acting GLP-1 agonist, DPP-IV resistant GLP-1 (7-36, A8G) fused to an AlbudAb (GAlbudAb), with the effect of the GLP-1 mimetic, exendin-4 (short half-life GLP-1 agonist) on infarct size following acute myocardial I/R injury. METHODS: Male Sprague-Dawley rats (8-week-old) were treated with vehicle, GAlbudAb or exendin-4. Myocardial ischemia was induced 2 h following the final dose for GAlbudAb and 30 min post the final dose for exendin-4. In a subgroup of animals, the final dose of exendin-4 was administered (1 µg/kg, SC, bid for 2 days) 6 h prior to myocardial ischemia when plasma exendin-4 was at its minimum concentration (C(min)). Myocardial infarct size, area at risk and cardiac function were determined 24 h after myocardial I/R injury. RESULTS: GAlbudAb and exendin-4 significantly reduced myocardial infarct size by 28% and 23% respectively, compared to vehicle (both p < 0.01 vs. vehicle) after I/R injury. Moreover, both GAlbudAb and exendin-4 markedly improved post-ischemic cardiac contractile function. Body weight loss and reduced food intake consistent with the activation of GLP-1 receptors was observed in all treatment groups. However, exendin-4 failed to reduce infarct size when administered 6 h prior to myocardial ischemia, suggesting continuous activation of the GLP-1 receptors is needed for cardioprotection. CONCLUSIONS: Cardioprotection provided by GAlbudAb, a long acting GLP-1 mimetic, following myocardial I/R injury was comparable in magnitude, but more sustained in duration than that produced by short-acting exendin-4. Very low plasma concentrations of exendin-4 failed to protect the heart from myocardial I/R injury, suggesting that sustained GLP-1 receptor activation plays an important role in providing cardioprotection in the setting of acute myocardial I/R injury. Long-acting GLP-1 agonists such as GAlbudAb may warrant additional evaluation as novel therapeutic agents to reduce myocardial I/R injury during acute coronary syndrome.


Subject(s)
Cardiotonic Agents/pharmacology , Glucagon-Like Peptide 1/pharmacology , Immunoconjugates/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/pharmacology , Serum Albumin/immunology , Single-Domain Antibodies/pharmacology , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/blood , Cardiotonic Agents/pharmacokinetics , Disease Models, Animal , Exenatide , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/pharmacokinetics , Glucagon-Like Peptide-1 Receptor , Immunoconjugates/administration & dosage , Immunoconjugates/blood , Immunoconjugates/pharmacokinetics , Injections, Subcutaneous , Male , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Peptide Fragments/pharmacokinetics , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/blood , Venoms/pharmacology , Ventricular Function, Left/drug effects
6.
J Appl Physiol (1985) ; 114(9): 1202-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23449942

ABSTRACT

The loss of cardiac reserve is, in part, responsible for exercise intolerance in late-stage heart failure (HF). Exercise tolerance testing (ETT) has been performed in mouse models of HF; however, treadmill performance and at-rest cardiac indexes determined by magnetic resonance imaging (MRI) rarely correlate. The present study adopted a stress-MRI technique for comparison with ETT in HF models, using isoproterenol (ISO) to evoke cardiac reserve responses. Male C57BL/6J mice were randomly subjected to myocardial infarction (MI), transverse aortic constriction (TAC), or sham surgery under general anesthesia. Mice underwent serial ETT on a graded treadmill with follow-up ISO stress-MRI. TAC mice showed consistent exercise intolerance, with a 16.2% reduction in peak oxygen consumption vs. sham at 15-wk postsurgery (WPS). MI and sham mice had similar peak oxygen consumption from 7 WPS onward. Time to a respiratory exchange ratio of 1.0 correlated with ETT distance (r = 0.64; P < 0.001). The change in ejection fraction under ISO stress was reduced in HF mice at 4 WPS [10.1 ± 3.9% change (Δ) and 8.9 ± 3.5%Δ in MI and TAC, respectively, compared with 32.0 ± 3.5%Δ in sham; P < 0.001]. However, cardiac reserve differences between surgery groups were not observed at 16 WPS in terms of ejection fraction or cardiac output. In addition, ETT did not correlate with cardiac indexes under ISO stress. In conclusion, ISO stress was unable to reflect consistent differences in ETT between HF and healthy mice, suggesting cardiac-specific indexes are not the sole factors in defining exercise intolerance in mouse HF models.


Subject(s)
Heart Failure/physiopathology , Physical Exertion/physiology , Adrenergic beta-Agonists , Animals , Cardiotonic Agents , Disease Models, Animal , Exercise Test , Exercise Tolerance/physiology , Heart Failure/etiology , Humans , Isoproterenol , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/complications , Oxygen Consumption , Ventricular Function, Left
7.
Sci Transl Med ; 4(159): 159ra148, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23136043

ABSTRACT

Pulmonary edema resulting from high pulmonary venous pressure (PVP) is a major cause of morbidity and mortality in heart failure (HF) patients, but current treatment options demonstrate substantial limitations. Recent evidence from rodent lungs suggests that PVP-induced edema is driven by activation of pulmonary capillary endothelial transient receptor potential vanilloid 4 (TRPV4) channels. To examine the therapeutic potential of this mechanism, we evaluated TRPV4 expression in human congestive HF lungs and developed small-molecule TRPV4 channel blockers for testing in animal models of HF. TRPV4 immunolabeling of human lung sections demonstrated expression of TRPV4 in the pulmonary vasculature that was enhanced in sections from HF patients compared to controls. GSK2193874 was identified as a selective, orally active TRPV4 blocker that inhibits Ca(2+) influx through recombinant TRPV4 channels and native endothelial TRPV4 currents. In isolated rodent and canine lungs, TRPV4 blockade prevented the increased vascular permeability and resultant pulmonary edema associated with elevated PVP. Furthermore, in both acute and chronic HF models, GSK2193874 pretreatment inhibited the formation of pulmonary edema and enhanced arterial oxygenation. Finally, GSK2193874 treatment resolved pulmonary edema already established by myocardial infarction in mice. These findings identify a crucial role for TRPV4 in the formation of HF-induced pulmonary edema and suggest that TRPV4 blockade is a potential therapeutic strategy for HF patients.


Subject(s)
Heart Failure/complications , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/therapeutic use , Pulmonary Edema/drug therapy , Pulmonary Edema/prevention & control , TRPV Cation Channels/antagonists & inhibitors , Administration, Oral , Animals , Blood Pressure/drug effects , Calcium/metabolism , Disease Models, Animal , Diuretics/pharmacology , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Heart Failure/pathology , Heart Failure/physiopathology , Heart Rate/drug effects , Humans , In Vitro Techniques , Ion Channel Gating/drug effects , Lung/drug effects , Lung/metabolism , Lung/pathology , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/pharmacology , Mice , Mice, Knockout , Permeability/drug effects , Protein Transport/drug effects , Pulmonary Edema/etiology , Pulmonary Edema/pathology , Rats , TRPV Cation Channels/metabolism , Water-Electrolyte Balance/drug effects
8.
Magn Reson Med ; 67(1): 191-200, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21671268

ABSTRACT

The temporal evolution of heart failure and associated pulmonary congestion in rodent heart failure models has not yet been characterized simultaneously and noninvasively. In this study, MRI was used to assess the serial progression of left-ventricular dysfunction and lung congestion in mice following myocardial infarction (MI). Cardiac and lung (1) H MRI was performed at baseline and every 3 days up to 13 days postsurgery in sham and MI mice. Respiratory parameters and terminal lung mechanics were assessed followed by histological analysis. MRI revealed that the MI induced significant pulmonary congestion/edema as detected by increased MRI signal intensity and was associated with increased lung volume and reduced cardiac contractility. Pulmonary function was also depressed in MI-mice, reflected by a reduced tidal volume and a low minute ventilation rate. Additionally, MI significantly increased lung resistance, markedly reduced lung compliance and total lung capacity and significantly increased lung weights by 57%. Significant correlations were observed between the MRI measured lung congestion, lung volume, ejection fraction, and lung wet-weight parameters. This study demonstrates that MRI may be of significant value in evaluating therapies aimed at primary intervention for lung congestion and secondary prevention of unfavorable cardiac remodeling.


Subject(s)
Magnetic Resonance Imaging/methods , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling/physiology , Animals , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Edema/complications , Ventricular Dysfunction, Left/complications
9.
PLoS One ; 6(8): e23570, 2011.
Article in English | MEDLINE | ID: mdl-21887274

ABSTRACT

BACKGROUND: The cardioprotective effects of glucagon-like peptide-1 (GLP-1) and analogs have been previously reported. We tested the hypothesis that albiglutide, a novel long half-life analog of GLP-1, may protect the heart against I/R injury by increasing carbohydrate utilization and improving cardiac energetic efficiency. METHODS/PRINCIPAL FINDINGS: Sprague-Dawley rats were treated with albiglutide and subjected to 30 min myocardial ischemia followed by 24 h reperfusion. Left ventricle infarct size, hemodynamics, function and energetics were determined. In addition, cardiac glucose disposal, carbohydrate metabolism and metabolic gene expression were assessed. Albiglutide significantly reduced infarct size and concomitantly improved post-ischemic hemodynamics, cardiac function and energetic parameters. Albiglutide markedly increased both in vivo and ex vivo cardiac glucose uptake while reducing lactate efflux. Analysis of metabolic substrate utilization directly in the heart showed that albiglutide increased the relative carbohydrate versus fat oxidation which in part was due to an increase in both glucose and lactate oxidation. Metabolic gene expression analysis indicated upregulation of key glucose metabolism genes in the non-ischemic myocardium by albiglutide. CONCLUSION/SIGNIFICANCE: Albiglutide reduced myocardial infarct size and improved cardiac function and energetics following myocardial I/R injury. The observed benefits were associated with enhanced myocardial glucose uptake and a shift toward a more energetically favorable substrate metabolism by increasing both glucose and lactate oxidation. These findings suggest that albiglutide may have direct therapeutic potential for improving cardiac energetics and function.


Subject(s)
Cardiotonic Agents/pharmacology , Glucagon-Like Peptide 1/analogs & derivatives , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cyclic AMP/metabolism , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/pharmacology , Heart , Heart Function Tests , Hemodynamics/drug effects , In Vitro Techniques , Insulin/blood , Lactic Acid/blood , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/physiopathology , Principal Component Analysis , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
10.
J Cardiovasc Pharmacol ; 56(2): 147-55, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20714241

ABSTRACT

BACKGROUND: Hypoxia inducible factors (HIFs) are transcription factors that are regulated by HIF-prolyl 4-hydroxylases (PHDs) in response to changes in oxygen tension. Once activated, HIFs play an important role in angiogenesis, erythropoiesis, proliferation, cell survival, inflammation, and energy metabolism. We hypothesized that GSK360A, a novel orally active HIF-PHD inhibitor, could facilitate local and systemic HIF-1 alpha signaling and protect the failing heart after myocardial infarction (MI). METHODS AND RESULTS: GSK360A is a potent (nanomolar) inhibitor of HIF-PHDs (PHD1>PHD2 = PHD3) capable of activating the HIF-1 alpha pathway in a variety of cell types including neonatal rat ventricular myocytes and H9C2 cells. Male rats treated orally with GSK360A (30 mg x kg x d) had a sustained elevation in circulating levels of erythropoietin and hemoglobin and increased hemoxygenase-1 expression in the heart and skeletal muscle. In a rat model of established heart failure with systolic dysfunction induced by ligation of left anterior descending coronary artery, chronic treatment with GSK360A for 28 days prevented the progressive reduction in ejection fraction, ventricular dilation, and increased lung weight, which were observed in the vehicle-treated animals, for up to 3 months. In addition, the microvascular density in the periinfarct region was increased (>2-fold) in GSK360A-treated animals. Treatment was well tolerated (survival was 89% in the GSK360A group vs. 82% in the placebo group). CONCLUSIONS: Chronic post-myocardial infarction treatment with a selective HIF PHD inhibitor (GSK360A) exerts systemic and local effects by stabilizing HIF-1 alpha signaling and improves long-term ventricular function, remodeling, and vascularity in a model of established ventricular dysfunction. These results suggest that HIF-PHD inhibitors may be suitable for the treatment of post-MI remodeling and heart failure.


Subject(s)
Coronary Vessels/drug effects , Glycine/analogs & derivatives , Hypoxia-Inducible Factor 1/metabolism , Myocardial Infarction/drug therapy , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Quinolones/pharmacology , Ventricular Remodeling/drug effects , Animals , Cell Line , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Glycine/pharmacology , Hemodynamics/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley
11.
J Pharmacol Exp Ther ; 326(2): 443-52, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18499744

ABSTRACT

The transient receptor potential (TRP) vanilloid subtype 4 (V4) is a nonselective cation channel that exhibits polymodal activation and is expressed in the endothelium, where it contributes to intracellular Ca2+ homeostasis and regulation of cell volume. The purpose of the present study was to evaluate the systemic cardiovascular effects of GSK1016790A, a novel TRPV4 activator, and to examine its mechanism of action. In three species (mouse, rat, and dog), the i.v. administration of GSK1016790A induced a dose-dependent reduction in blood pressure, followed by profound circulatory collapse. In contrast, GSK1016790A had no acute cardiovascular effects in the TRPV4-/- null mouse. Hemodynamic analyses in the dog and rat demonstrate a profound reduction in cardiac output. However, GSK1016790A had no effect on rate or contractility in the isolated, buffer-perfused rat heart, and it produced potent endothelial-dependent relaxation of rodent-isolated vascular ring segments that were abolished by nitric-oxide synthase (NOS) inhibition (N-nitro-L-arginine methyl ester; L-NAME), ruthenium red, and endothelial NOS (eNOS) gene deletion. However, the in vivo circulatory collapse was not altered by NOS inhibition (L-NAME) or eNOS gene deletion but was associated with (concentration and time appropriate) profound vascular leakage and tissue hemorrhage in the lung, intestine, and kidney. TRPV4 immunoreactivity was localized in the endothelium and epithelium in the affected organs. GSK1016790A potently induced rapid electrophysiological and morphological changes (retraction/condensation) in cultured endothelial cells. In summary, inappropriate activation of TRPV4 produces acute circulatory collapse associated with endothelial activation/injury and failure of the pulmonary microvascular permeability barrier. It will be important to determine the role of TRPV4 in disorders associated with edema and microvascular congestion.


Subject(s)
Aorta, Thoracic/drug effects , Endothelium, Vascular/drug effects , Hemodynamics/drug effects , Leucine/analogs & derivatives , Sulfonamides/adverse effects , TRPV Cation Channels/agonists , Ventricular Function, Left/drug effects , Animals , Aorta, Thoracic/metabolism , Capillary Permeability/drug effects , Cell Adhesion/drug effects , Cell Line , Dogs , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Female , Humans , Immunohistochemistry , Leucine/adverse effects , Leucine/pharmacokinetics , Male , Mice , Mice, Knockout , Molecular Structure , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacokinetics , TRPV Cation Channels/genetics , Vasoconstriction/drug effects
12.
J Pharmacol Exp Ther ; 325(2): 466-74, 2008 May.
Article in English | MEDLINE | ID: mdl-18287212

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR)-delta is a transcription factor that belongs to the PPAR family. PPAR-delta is abundantly expressed in the heart, and its role in the heart is largely unknown. We tested whether pharmacological activation of PPAR-delta protects the heart from ischemia/reperfusion (I/R) injury in male Zucker fatty rats, a rodent model of obesity and dyslipidemia. A highly selective PPAR-delta agonist, [4-[[[2-[3-fluoro-4-(trifluoromethyl)phenyl]-4-methyl-5-thiazolyl]methyl] thio]-2-methylphenoxy]acetic acid (GW0742), was administered for 7 days at 10 mg/kg/day (p.o., once a day). Ischemic injury was produced by occlusion of the left anterior descending artery for 30 min followed by reperfusion for up to 24 h. Treatment with GW0742 reduced serum levels of cardiac troponin-I and infarct size by 63% (p < 0.01) and 32% (p < 0.01), respectively, and improved left ventricular function. Treatment with GW0742 up-regulated gene expression involved in cardiac fatty acid oxidation, increased fat use in the heart, and reduced serum levels of free fatty acids. The enhanced cardiac expression of interleukin (IL)-6, IL-8, intercellular adhesion molecule-1, and monocyte chemoattractant protein-1 induced by I/R were significantly attenuated by GW0742. Treatment with GW0742 also reduced apoptotic cardiomyocytes by 34% and cardiac caspase-3 activity by 61% (both p < 0.01 versus vehicle). GW0742 differentially regulated Bcl family members, favoring cell survival, and attenuated I/R-induced cardiac mitochondrial damage. In addition, GW0742 treatment augmented the cardiac Akt signaling pathway, as reflected by enhanced phospho-3-phosphoinositide-dependent kinase-1 and p-Akt. The results indicate that activation of PPAR-delta protected the heart from I/R injury in Zucker fatty rats, and multiple mechanisms including amelioration of lipotoxicity, anti-inflammation, and up-regulation of prosurvival signaling contribute together to the cardioprotection.


Subject(s)
Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/prevention & control , PPAR delta/agonists , Thiazoles/therapeutic use , Animals , Blood Pressure/drug effects , Cytokines/genetics , Disease Models, Animal , Fatty Acids/blood , Fatty Acids/metabolism , Heart/physiopathology , Ketones/metabolism , Male , Metabolic Syndrome/blood , Metabolic Syndrome/physiopathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/physiopathology , PPAR delta/physiology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Rats, Zucker , Troponin I/blood
13.
J Cardiovasc Pharmacol ; 49(6): 362-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17577100

ABSTRACT

Angiotensin II (Ang II) activates p38 mitogen-activated protein kinase (p38 MAPK) and increases reactive oxygen species (ROS), but the nature of the relationship in vivo is not fully understood. We assess the effect of SB239063AN, a highly selective, orally active, p38 MAPK inhibitor, on Ang II-dependent hypertension, target-organ damage and ROS production. Sprague-Dawley rats and MAPKAP kinase-2 knockout mice were infused with Ang II. Ang II infusion increased the levels of phosphorylated p38 MAPK in the heart and aorta. Production of superoxide anion and expression of NAD(P)H oxidase subunit gp91 in the aorta were increased 4- and 5-fold, respectively. In addition, Ang II infusion led to endothelial dysfunction, progressive and sustained hypertension, and cardiac hypertrophy. Treatment with SB239063AN (800 ppm in the diet) significantly attenuated the levels of phosphorylated p38 MAPK in the heart and aorta, reduced superoxide anion generation by 57% (P < 0.01), markedly suppressed gp91 mRNA expression, prevented endothelial dysfunction, and blunted both the hypertension and cardiac hypertrophy. Ang II-dependent hypertension was also significantly attenuated in MAPKAP kinase-2 knockout mice. The results suggest that Ang II induced hypertension, organ damage, and ROS production are possibly mediated by p38 MAPK and inhibition of p38 MAPK may offer a therapeutic approach for cardiovascular disease.


Subject(s)
Angiotensin II/adverse effects , Enzyme Inhibitors , Hypertension/drug therapy , Imidazoles , Pyrimidines , Superoxides/metabolism , Ventricular Remodeling/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/enzymology , Aorta, Abdominal/metabolism , Blood Pressure/drug effects , Carotid Arteries/drug effects , Carotid Arteries/enzymology , Carotid Arteries/metabolism , Echocardiography , Endothelium, Vascular/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Hypertension/chemically induced , Hypertension/enzymology , Hypertension/metabolism , Imidazoles/administration & dosage , Imidazoles/pharmacology , Imidazoles/therapeutic use , Intracellular Signaling Peptides and Proteins , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Myocardium/enzymology , Myocardium/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects , p38 Mitogen-Activated Protein Kinases/biosynthesis
14.
Diabetes ; 54(2): 554-62, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15677515

ABSTRACT

The mechanism responsible for the enhanced myocardial susceptibility to ischemic insult in patients with type 2 diabetes is not clear. The present study examines the effect of rosiglitazone treatment on cardiac insulin sensitization and its association with cardioprotection from ischemia/reperfusion injury in an animal model of diabetes. Male Zucker diabetic fatty (ZDF) rats were treated with rosiglitazone (3 mg . kg(-1) . day(-1) orally) or vehicle for 8 days before undergoing 30 min of coronary artery ligation, followed by reperfusion for 4 h (apoptosis) or 24 h (infarction). Rosiglitazone reduced the blood levels of glucose, triglycerides, and free fatty acids; enhanced cardiac glucose oxidation; and increased Akt phosphorylation (Akt-pS473) 2.1-fold and Akt kinase activity 1.8-fold in the ischemic myocardium. The phosphorylation of two downstream targets of Akt, glycogen synthase kinase-3beta and FKHR (forkhead transcription factor), was also enhanced by 2- and 2.9-fold, respectively. In rosiglitazone-treated rats, the number of apoptotic cardiomyocytes and the myocardial infarct size were decreased by 58 and 46%, respectively, and the myocardial contractile dysfunction was improved. Blockade of the insulin-Akt signaling pathway by wortmannin in the 8-day rosiglitazone-treated ZDF rats resulted in a markedly diminished cardioprotective effect of rosiglitazone. In addition, 8-day rosiglitazone treatment in Zucker lean rats or 2-day rosiglitazone treatment in ZDF rats, both of which showed no change in whole-body insulin sensitivity, resulted in a significant reduction in cardiac infarct size, but to a lesser degree when compared with that observed in 8-day rosiglitazone-treated ZDF rats. These results suggest that chronic treatment with rosiglitazone protects the heart against ischemia/reperfusion injury in ZDF rats, and that the enhanced cardiac protection observed after rosiglitazone treatment might be attributable in part to an improvement in cardiac insulin sensitivity.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Heart/physiopathology , Insulin Resistance/physiology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Thiazolidinediones/therapeutic use , Administration, Oral , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Coronary Vessels/physiopathology , Fatty Acids, Nonesterified/blood , Heart/drug effects , Hypoglycemic Agents/therapeutic use , Male , Myocardial Infarction/pathology , Myocardium/metabolism , Rats , Rats, Zucker , Rosiglitazone , Triglycerides/blood
15.
Cardiovasc Res ; 61(3): 548-58, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14962485

ABSTRACT

OBJECTIVE: To investigate the role of Rho A and Rho-kinase in acute myocardial ischemia/reperfusion injury and the protective effect of Rho-kinase inhibitor, Y-27632 [(R)-(+)-trans-N-(4-pyridyl)-4-(1-aminoethyl)cyclohexanecarboxamide]. METHODS AND RESULTS: Male CD1 mice were subjected to 30 min of coronary occlusion and 24 h reperfusion. Ischemia/reperfusion upregulated expression of Rho A in ischemic myocardium, and subsequently activated Rho-kinase. Y-27632 significantly inhibited the activation of Rho-kinase following ischemia/reperfusion. Treatment with Y-27632 at 10 and 30 mg/kg oral administration, reduced infarct size by 30.2% and 41.1%, respectively (P<0.01 vs. vehicle). Y-27632 also enhanced post-ischemia cardiac function. Left ventricular systolic pressure, +dP/dt and -dP/dt were significantly improved by 23.5%, 52.3%, and 59.4%, respectively (P<0.01 vs. vehicle). Moreover, Y-27632 reduced ischemia/reperfusion-induced myocardial apoptosis. The apoptotic myocytes in ischemic myocardium after 4 h reperfusion were reduced from 13.1% in vehicle group to 6.4% in Y-27632-treated group (P<0.01). Meanwhile, ischemia/reperfusion-induced downregulation of Bcl-2 in myocardium was remarkably attenuated in the treated animals. Ischemia/reperfusion resulted in remarkable elevation in serum levels of proinflammatory cytokines, interleukin-6 (IL-6), keratinocyte chemoattractant (KC) and granulocyte colony-stimulating factor (G-CSF), which was significantly suppressed by Y-27632. In addition, Y-27632 decreased ischemia/reperfusion-induced accumulation of neutrophils in the heart by 45% (P<0.01). CONCLUSIONS: These results suggest that Rho-kinase plays a pivotal role in myocardial ischemia/reperfusion injury. The cardiac protection provided by treatment with a selective Rho-kinase inhibitor is likely via anti-apoptotic effect and attenuation of ischemia/reperfusion-induced inflammatory responses. The finding of this study suggest a novel therapeutic approach to the treatment of acute myocardial ischemia/reperfusion injury.


Subject(s)
Amides/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/therapeutic use , Animals , Apoptosis , Blotting, Western/methods , Immunohistochemistry/methods , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred Strains , Myocardial Contraction/drug effects , Myocardial Ischemia/drug therapy , Myocardial Ischemia/enzymology , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Neutrophils/immunology , Protein Serine-Threonine Kinases/analysis , Proto-Oncogene Proteins c-bcl-2/analysis , rho-Associated Kinases
16.
NMR Biomed ; 17(8): 620-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15761951

ABSTRACT

Owing to its signal-enhancing characteristics in viable well-perfused tissue, divalent manganese (Mn2+) has been used as a myocardial imaging contrast agent. Because Mn2+ can enter excitable cells through the voltage-gated L-type calcium channels, manganese-enhanced MRI (MEMRI) has been used to determine the viability and the inotropic state of the heart. In this study, we examined the correlation between left ventricular infarction zone as assessed by cardiac MEMRI and function in mice with permanent coronary artery occlusion. At an Mn2+ infusion dose of 1.72+/-0.47 nmol/min/g body weight, the steady-state signal intensity (SI) enhancement 20-26 min post-Mn2+ infusion of the normal septum and left-ventricular wall during diastole was 128.2+/-14.4 and 127.9+/-26.5%, respectively, whereas the infarction zone was 56.0+/-7.1%. During systole, the SI enhancement was 144.6+/-33.0, 116.0+/-18.7 and 48.3+/-20.0% for the normal septum, left-ventricular wall and infarction zone, respectively. A good correlation was obtained between the MEMRI determined infarction volume and conventional histological TTC staining (r = 0.9582, p<0.01). There was also a strong negative correlation between MEMRI determined infarction percentage (compared with whole left ventricle) and ejection fraction (r = -0.94, p<0.05). These data suggest that the Mn2+ concentration at steady state in the heart may reflect altered tissue viability in the infarcted tissue as well as surrounding region following myocardial infarction. In conclusion, in vivo cardiac MEMRI offers a manner in which functional, pathologic and viability data may be obtained simultaneously in myocardial tissue.


Subject(s)
Contrast Media , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Manganese , Myocardial Infarction/diagnosis , Ventricular Dysfunction, Left/diagnosis , Animals , Mice , Mice, Inbred ICR , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Severity of Illness Index , Tissue Survival , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism
17.
Circulation ; 108(19): 2393-9, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14557369

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor-alpha (PPAR-alpha) is expressed in the heart and regulates genes involved in myocardial fatty acid oxidation (FAO). The role of PPAR-alpha in acute ischemia/reperfusion myocardial injury remains unclear. METHODS AND RESULTS: The coronary arteries of male mice were ligated for 30 minutes. After reperfusion for 24 hours, ischemic and infarct sizes were determined. A highly selective and potent PPAR-alpha agonist, GW7647, was administered by mouth for 2 days, and the third dose was given 1 hour before ischemia. GW7647 at 1 and 3 mg x kg(-1) x d(-1) reduced infarct size by 28% and 35%, respectively (P<0.01), and myocardial contractile dysfunction was also improved. Cardioprotection by GW7647 was completely abolished in PPAR-alpha-null mice. Ischemia/reperfusion downregulated mRNA expression of cardiac PPAR-alpha and FAO enzyme genes, decreased myocardial FAO enzyme activity and in vivo cardiac fat oxidation, and increased serum levels of free fatty acids. All of these changes were reversed by GW7647. Moreover, GW7647 attenuated ischemia/reperfusion-induced release of multiple proinflammatory cytokines and inhibited neutrophil accumulation and myocardial expression of matrix metalloproteinases-9 and -2. Furthermore, GW7647 inhibited nuclear factor-kappaB activation in the heart, accompanied by enhanced levels of inhibitor-kappaBalpha. CONCLUSIONS: Activation of PPAR-alpha protected the heart from reperfusion injury. This cardioprotection might be mediated through metabolic and antiinflammatory mechanisms. This novel effect of the PPAR-alpha agonist could provide an added benefit to patients treated with PPAR-alpha activators for dyslipidemia.


Subject(s)
Butyrates/therapeutic use , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/prevention & control , Phenylurea Compounds/therapeutic use , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Animals , Butyrates/administration & dosage , Chemotaxis, Leukocyte/drug effects , Cytokines/metabolism , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Fatty Acids/blood , I-kappa B Proteins/biosynthesis , Ligation , Male , Matrix Metalloproteinases/biosynthesis , Mice , Mice, Knockout , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/enzymology , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Oxidation-Reduction , Phenylurea Compounds/administration & dosage , Premedication , RNA, Messenger/biosynthesis , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology
18.
Cardiovasc Res ; 54(3): 549-58, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12031700

ABSTRACT

OBJECTIVE: Matrix metalloproteinase-9 (MMP-9) activity is up regulated in the heart subjected to ischemic insult. Whether increased MMP-9 activity contributes to acute myocardial injury after ischemia-reperfusion remains unknown. To investigate the role of MMP-9 in myocardial infarction, we utilized a MMP-9 knockout mouse. METHODS AND RESULTS: Standard homologous recombination in embryonic stem cells was used to generate a mouse lacking MMP-9. The left anterior descending coronary artery was occluded for 30 min followed by 24 h reperfusion, and the ischemic and infarct sizes were determined. Targeted deletion of MMP-9 protected the heart from no-flow ischemia-reperfusion-induced myocardial injury. The myocardial infarct size was reduced by 17.5% in MMP-9 heterozygotes (+/-) (P<0.01) and 35.4% in MMP-9 knockout (-/-) mice (P<0.01) versus the wild-type (+/+) mice, respectively. Analysis of MMP activity in myocardial extracts by zymography demonstrated that ischemia-reperfusion-induced expression of proMMP-9 and active MMP-9 was reduced by 77.8% (P<0.01) and 69.1% (P<0.001), respectively, in (+/-) mice compared to (+/+) mice, and was absent in (-/-) animals. The expression of TIMP-1, an endogenous inhibitor of MMP-9, was elevated 4.7-fold (P<0.05) and 21.4-fold (P<0.05) in the (+/-) and (-/-) mice, respectively, compared to (+/+) mice. Immunohistochemical analysis revealed that neutrophils were the primary cellular source of MMP-9, and less neutrophils were detected in the ischemic region of the heart following ischemia-reperfusion in (-/-) mice compared to (+/+) mice. Measurement of myeloperoxidase activity, a marker enzyme of neutrophils, demonstrated a 44% reduction in neutrophils infiltrated into the ischemic myocardium in the (-/-) mice compared to the (+/+) mice (P<0.05). CONCLUSION: These results suggest that MMP-9 plays an important role in ischemia-reperfusion-induced myocardial infarction and MMP-9 could be a target for prevention or treatment of acute ischemic myocardial injury.


Subject(s)
Gene Deletion , Matrix Metalloproteinase 9/genetics , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Animals , Immunohistochemistry/methods , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/analysis , Mice , Mice, Knockout , Models, Animal , Myocardial Reperfusion Injury/genetics , Myocardium/immunology , Neutrophil Infiltration , Neutrophils/enzymology , Tissue Inhibitor of Metalloproteinase-1/analysis , Tissue Inhibitor of Metalloproteinase-1/metabolism , Ventricular Remodeling
19.
J Mol Cell Cardiol ; 34(1): 5-15, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11812160

ABSTRACT

Recent evidence has shown that the cardioprotection afforded by the late phase of ischemic preconditioning (PC) is mediated by upregulation of inducible nitric oxide synthase (iNOS). However, the specific cardiac cell type(s) that express(es) iNOS in response to ischemic PC remains unknown. Thus, mice underwent a sequence of six cycles of 4-min coronary occlusion/4-min reperfusion, which induces late PC, and tissue samples were collected at serial times for measurement of mRNA (Northern) and protein levels (Western). In addition, whole heart samples were cryosectioned for in situ hybridization and immunohistochemistry. The steady-state levels of iNOS mRNA in the ischemic regions started to increase at 1 h after ischemic PC, peaked at 3 h (201+/-31% of sham, n=5 P<0.01) and remained elevated at 24 h (177+/-22% of sham, n=5 P<0.01). In accordance with these data, iNOS protein expression was increased at 24 h (219+/-41% of sham, n=5 P<0.01). In contrast, neither endothelial nitric oxide synthase (eNOS) mRNA levels nor its protein expression changed at any time-point. The magnitude of iNOS upregulation after ischemic PC was mild compared with that noted 66 h after permanent coronary occlusion (360+/-53% of sham) or 8 h after endotoxin (3117+/-61% of control). After ischemic PC, diffuse iNOS signals were detected with in situ hybridization and immunohistochemistry in the cytoplasmic space of cardiac myocytes and, to a lesser degree, in the wall of large vessels, but were absent in smooth muscle and endothelium of small vessels and in fibroblasts. This pattern contrasted with that observed in mouse hearts subjected to permanent coronary occlusion where strong iNOS signals were concentrated in inflammatory cells but absent in cardiac myocytes. Thus, not only the degree of iNOS expression but also its cellular distribution were profoundly different in reversibly injured (preconditioned) v infarcted myocardium. We conclude that iNOS is rapidly upregulated after ischemic PC and that cardiac myocytes are the main source of ischemic PC-induced iNOS expression. This study demonstrates, for the first time, a differential pattern of iNOS expression in sublethal (PC) v lethal ischemia, which may have important implication for the role of iNOS in these two settings.


Subject(s)
Ischemic Preconditioning , Myocardium/cytology , Nitric Oxide Synthase/metabolism , Up-Regulation , Animals , Blotting, Northern , Blotting, Western , Immunohistochemistry , In Situ Hybridization , Ischemia , Mice , Mice, Inbred ICR , Myocardium/enzymology , Nitric Oxide Synthase Type II , RNA, Messenger/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...