Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
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.
Circ Heart Fail ; 5(1): 79-86, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22135403

ABSTRACT

BACKGROUND: Recognizing that inhibitors of phosphodiesterase type 5 (PDE5) are increasingly employed in patients with pulmonary hypertension and right ventricular (RV) failure, we examined PDE5 expression in the human RV and its impact on myocardial contractility. METHODS AND RESULTS: Tissue extracts from the RV of 20 patients were assayed for PDE5 expression using immunoblot and immunohistochemical staining. Tissues were selected from groups of nonfailing organ donors and transplant recipients with endstage ischemic cardiomyopathy or idiopathic dilated cardiomyopathy. Among dilated cardiomyopathy patients, subgroups with mild or severe RV dysfunction and prior left ventricular assist devices were analyzed separately. Our results showed that PDE5 abundance increased more than 4-fold in the RVs of the ischemic cardiomyopathy compared with the nonfailing group. In dilated cardiomyopathy, PDE5 upregulation was more moderate and varied with the severity of RV dysfunction. Immunohistochemical staining confirmed that cardiac myocytes contributed to the upregulation in the failing hearts. In functional studies, PDE5 inhibition produced little change in developed force in RV trabeculae from nonfailing hearts but produced a moderate increase in RV trabeculae from failing hearts. CONCLUSIONS: Our results showed the etiology- and severity-dependent upregulation of myocyte PDE5 expression in the RV and the impact of this upregulation on myocardial contractility. These findings suggest that RV PDE5 expression could contribute to the pathogenesis of RV failure, and direct myocardial responses to PDE5 inhibition may modulate the indirect responses mediated by RV afterload reduction.


Subject(s)
Cardiomyopathies/enzymology , Cardiomyopathy, Dilated/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Heart Ventricles/enzymology , Myocardium/enzymology , Adult , Biomarkers/metabolism , Cardiomyopathies/surgery , Cardiomyopathy, Dilated/surgery , Female , Heart Transplantation , Heart Ventricles/physiopathology , Humans , Male , Middle Aged , Myocardial Contraction/physiology , Myocytes, Cardiac/enzymology , Severity of Illness Index , Tissue Donors , Transplantation
3.
Toxicol Sci ; 120(2): 262-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21242511

ABSTRACT

In the clinical setting, natriuretic peptides (NPs) have proven to be reliable noninvasive markers for diagnostic, prognostic, and therapeutic monitoring of heart failure. Given their proven utility in humans, NPs are potential candidates for translational biomarkers during drug development to detect drug-induced hemodynamic stress resulting in cardiac hypertrophy in preclinical species. We evaluated the intra- and interassay precision and the stability of serum N-terminal-proatrial natriuretic peptide (NT-proANP) using a commercially available enzyme-linked immunoassay (EIA). We then measured NT-proANP concentrations in 532 serum samples from 337 male Crl:CD(SD) rats with or without pressure-induced cardiac hypertrophy. Additionally, we established a reference range using samples from control animals across multiple studies. The data demonstrate that the NT-proANP EIA is a robust and reproducible assay for the measurement of NT-proANP. The noninvasive translational utility, minimal sample volume requirement, and the lack of existing hypertrophic biomarkers in the male rat make NT-proANP an excellent candidate for further interrogation as a biomarker of cardiac hypertrophy in preclinical toxicology investigations.


Subject(s)
Atrial Natriuretic Factor/blood , Cardiomyopathy, Hypertrophic/blood , Protein Precursors/blood , Animals , Biomarkers/blood , Disease Models, Animal , Drug Evaluation, Preclinical , Male , Protein Stability , Rats , Rats, Sprague-Dawley , Time Factors
4.
Toxicol Appl Pharmacol ; 243(3): 340-7, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20004680

ABSTRACT

Metformin is a first-line drug for the treatment of type 2 diabetes (T2D) and is often prescribed in combination with other drugs to control a patient's blood glucose level and achieve their HbA1c goal. New treatment options for T2D will likely include fixed dose combinations with metformin, which may require preclinical combination toxicology studies. To date, there are few published reports evaluating the toxicity of metformin alone to aid in the design of these studies. Therefore, to understand the toxicity of metformin alone, Crl:CD(SD) rats were administered metformin at 0, 200, 600, 900 or 1200 mg/kg/day by oral gavage for 13 weeks. Administration of > or =900 mg/kg/day resulted in moribundity/mortality and clinical signs of toxicity. Other adverse findings included increased incidence of minimal necrosis with minimal to slight inflammation of the parotid salivary gland for males given 1200 mg/kg/day, body weight loss and clinical signs in rats given > or =600 mg/kg/day. Metformin was also associated with evidence of minimal metabolic acidosis (increased serum lactate and beta-hydroxybutyric acid and decreased serum bicarbonate and urine pH) at doses > or =600 mg/kg/day. There were no significant sex differences in mean AUC(0-24) or C(max) nor were there significant differences in mean AUC(0-24) or C(max) following repeated dosing compared to a single dose. The no observable adverse effect level (NOAEL) was 200 mg/kg/day (mean AUC(0-24)=41.1 microg h/mL; mean C(max)=10.3 microg/mL based on gender average week 13 values). These effects should be taken into consideration when assessing potential toxicities of metformin in fixed dose combinations.


Subject(s)
Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/toxicity , Metformin/pharmacokinetics , Metformin/toxicity , Animals , Area Under Curve , Blood Cell Count , Blood Chemical Analysis , Body Weight/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Eye/drug effects , Female , Liver Function Tests , Male , Mass Spectrometry , Ophthalmoscopy , Rats , Sex Characteristics , Survival Analysis , Urinalysis
5.
Circ Heart Fail ; 2(1): 39-46, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19808314

ABSTRACT

BACKGROUND: Urotensin-II (U-II) is an endogenous peptide upregulated in failing hearts. To date, insights into the myocardial actions of U-II have been obscured by its potent vasoconstrictor effects and interspecies differences in physiological responses to U-II. METHODS AND RESULTS: We examined the direct effects of exogenous U-II on in vitro contractility in nonfailing and failing human myocardial trabeculae (n=47). Rapid cooling contractures (RCC) were used to examine sarcoplasmic reticulum Ca(2+) load. In nonfailing myocardium, exogenous U-II increased developed force (DF), rates of force generation and decline and RCC amplitude suggesting increased sarcoplasmic reticulum Ca(2+) load. In isolated myocyte suspensions from nonfailing hearts, U-II increased phospholamban phosphorylation. In failing myocardium, exogenous U-II reduced DF and rates of force generation and decline without a significant change in RCC amplitude in trabeculae or a change in phospholamban phosphorylation in myocytes. To examine the effects of endogenous U-II, we administered the peptidic U-II receptor antagonist (UT-A) GSK248451A to isolated trabeculae. UT-A induced a decrease in DF in nonfailing myocardium and an increase in DF in failing myocardium. UT-A increased RCC amplitude slightly in both nonfailing and failing myocardium. During ongoing UT-A, exogenous U-II had little effect on DF and RCC amplitude, confirming effective receptor blockade. CONCLUSIONS: U-II modulates contractility independent of vasoconstriction with opposite effects in failing and nonfailing hearts. Positive inotropic responses to UT-A alone suggests that increased endogenous U-II constrains contractility in failing hearts via an autocrine or paracrine mechanism. These findings support a potential therapeutic role for UT-A in heart failure.


Subject(s)
Heart Failure/metabolism , Myocardial Contraction/drug effects , Myocardium/metabolism , Urotensins/metabolism , Urotensins/pharmacology , Adult , Blotting, Western , Enzyme Inhibitors/pharmacology , Heart Failure/pathology , Humans , Middle Aged , Myocardial Contraction/physiology , Myocardium/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Signal Transduction , Sodium/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism
6.
J Mol Cell Cardiol ; 45(1): 81-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18513742

ABSTRACT

Claudin-5 is a transmembrane cell junction protein that is a component of tight junctions in endothelial cell layers. We have previously shown that claudin-5 also localizes to lateral membranes of murine cardiomyocytes at their junction with the extracellular matrix. Claudin-5 levels are specifically reduced in myocytes from a mouse model of muscular dystrophy with cardiomyopathy. To establish whether claudin-5 is similarly specifically reduced in human cardiomyopathy, we compared the levels of claudin-5 with other cell junction proteins in 62 cardiomyopathic end-stage explant samples. We show that claudin-5 levels are reduced in at least 60% of patient samples compared with non-failing controls. Importantly, claudin-5 reductions can be independent of connexin-43, a gap junction protein previously reported to be reduced in failing heart samples. Other cell junction proteins including alpha-catenin, beta-catenin, gamma-catenin, desmoplakin, and N-cadherin are reduced in only a small number of failing samples and only in combination with reduced claudin-5 or connexin-43 levels. We also show that reduced claudin-5 levels can be present independently from dystrophin alterations, which are known to be capable of causing and resulting from cardiomyopathy. These data are the first to show alterations of a tight junction protein in human cardiomyopathy samples and suggest that claudin-5 may participate in novel mechanisms in the pathway to end-stage heart failure.


Subject(s)
Cardiomyopathies/metabolism , Gap Junctions/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Tight Junctions/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cardiomyopathies/pathology , Catenins/metabolism , Claudin-5 , Connexin 43 , Desmoplakins/metabolism , Female , Gap Junctions/pathology , Humans , Male , Mice , Myocytes, Cardiac/pathology , Tight Junctions/pathology
7.
J Mol Cell Cardiol ; 43(5): 552-63, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17931654

ABSTRACT

Myocardial contractile reserve is significantly attenuated in patients with advanced heart failure. The aim of this study was to identify mechanisms of impaired contractile reserve in a large animal model that closely mimics human myocardial failure. Progressive right ventricular hypertrophy and failure were induced by banding the pulmonary artery in kittens. Isometric contractile force was measured in right ventricular trabeculae (n=115) from age-matched Control and Banded feline hearts. Rapid cooling contractures (RCC) were used to determine sarcoplasmic reticulum (SR) Ca(2+) load while assessing the ability of changes in rate, adrenergic stimulation and bath Ca(2+) to augment contractility. The positive force-frequency relationship and robust pre- and post-receptor adrenergic responses observed in Control trabeculae were closely paralleled by increases in RCC amplitude and the RCC2/RCC1 ratio. Conversely, the severely blunted force-frequency and adrenergic responses in Banded trabeculae were paralleled by an unchanged RCC amplitude and RCC2/RCC1 ratio. Likewise, supraphysiologic levels of bath Ca(2+) were associated with severely reduced contractility and RCC amplitude in Banded trabeculae compared to Controls. There were no differences in myofilament Ca(2+) sensitivity or length-dependent increases in contractility between Control and Banded trabeculae. There was a significant decrease in SR Ca(2+)-ATPase pump abundance and phosphorylation of phospholamban and ryanodine receptor in Banded trabeculae compared with Controls. A reduced ability to increase SR Ca(2+) load is the primary mechanism of reduced contractile reserve in failing feline myocardium. The similarity of impaired contractile reserve phenomenology in this feline model and transplanted hearts suggests mechanistic relevance to human myocardial failure.


Subject(s)
Calcium/physiology , Myocardial Contraction/physiology , Sarcoplasmic Reticulum/physiology , Ventricular Function, Right/physiology , Acepromazine/pharmacology , Animals , Cats , Colforsin/pharmacology , Echocardiography , Ketamine/pharmacology , Models, Animal , Myocardial Contraction/drug effects , Sarcomeres/drug effects , Sarcomeres/physiology , Sarcomeres/ultrastructure , Ventricular Dysfunction, Right/physiopathology
8.
Am J Physiol Regul Integr Comp Physiol ; 292(2): R810-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17008460

ABSTRACT

Recent studies have identified sex differences in heart function that may affect the risk of developing heart failure. We hypothesized that there are fundamental differences in calcium (Ca) regulation in cardiac myocytes of males and premenopausal females. Isometric force transients (n = 45) were measured at various stimulation frequencies to define the force frequency responses (FFR) (0.5, 1.0, 1.5, and 2.0 Hz) during either changes in bath Ca ([Ca]o) (1.0, 1.75, 3.5, and 7.0 mM) or length-tension (20, 40, 60, 80, and 100% L(max)) in right ventricle trabeculae from normal male (MT) and premenopausal female (FT) cats. Force-Ca measurements were also obtained in chemically skinned trabeculae. Under basal conditions (0.5 Hz, 1.75 mM Ca, 80% L(max)) both MT and FT achieved similar developed forces (DF) (MT 11 +/- 1, FT = 10 +/- 1 mN/mm2). At low rates and lengths, there is no sex difference. At higher preloads and rates, there is a separation in DF in MT and FT. At basal [Ca]o both MT and FT exhibited positive FFR (2.0 Hz, 1.75 mM Ca: MT 38 +/- 3, FT 21 +/- 4 mN/mm2); however, at higher [Ca]o, MT achieved greater DF (2.0 Hz, 7.0 mM Ca: MT 40 +/- 3 and FT = 24 +/- 4 mN/mm2). We detected no sex difference in myofilament Ca sensitivity at a sarcomere length of 2.1 mum. However, rapid cooling contractures indicated greater sarcoplasmic reticulum (SR) Ca load in MT at higher frequencies. Despite virtually identical contractile performance under basal conditions, significant sex differences emerge under conditions of increased physiological stress. Given the lack of sex differences in myofilament Ca sensitivity, these studies suggest fundamental sex differences in cellular Ca regulation to achieve contractile reserve, with myocardium from males exhibiting higher SR Ca load.


Subject(s)
Myocardial Contraction/physiology , Actin Cytoskeleton/physiology , Animals , Calcium/physiology , Cats , Extracellular Space/metabolism , Extracellular Space/physiology , Female , Heart Rate/physiology , In Vitro Techniques , Male , Myocardium/ultrastructure , Sarcomeres/physiology , Sarcoplasmic Reticulum/physiology , Sex Characteristics
9.
Amyloid ; 12(4): 246-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16399650

ABSTRACT

OBJECTIVE: The goal of this study was to examine in vitro tissue stiffness and contractile performance in myocardial amyloidosis. BACKGROUND: Primary systemic amyloidosis involves the deposition of amyloid protein in mesodermal tissues including the heart. Functional assessment of cardiac amyloidosis is usually performed using echocardiography. However, this technique does not involve assessment of preload-dependent contractile reserve (the Frank-Starling mechanism). METHODS: At the time of heart transplantation, isolated myocardial trabeculae were dissected from the right ventricle of a patient with primary systemic amyloidosis. In vitro length-tension experiments were performed and trabeculae were subsequently fixed, sectioned and stained with crystal violet to determine amyloid deposition. RESULTS: Among the nine trabeculae capable of generating force transients, various combinations of myocardial stiffness and contractile performance were observed including normal stiffness and contractility, severely increased stiffness with impaired contractility and hybrid patterns. Histological analysis demonstrated varying degrees of amyloid deposition among sampled trabeculae. CONCLUSIONS: Our findings extend previous reports of functional heterogeneity among patients by demonstrating functional heterogeneity within a single patient's heart. Our findings also highlight the functional interdependence of passive stiffness and systolic performance in the diseased myocardium and demonstrate the value of dynamic assessments of myocardial performance.


Subject(s)
Amyloidosis/physiopathology , Heart/physiopathology , Isometric Contraction , Myocardial Contraction , Myocardial Ischemia/physiopathology , Amyloidosis/pathology , Amyloidosis/surgery , Female , Heart Transplantation , Humans , Kidney Transplantation , Middle Aged , Myocardial Ischemia/pathology , Myocardial Ischemia/surgery , Myocardium/pathology , Tensile Strength
10.
J Pharmacol Exp Ther ; 312(2): 517-24, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15550574

ABSTRACT

Historically, inhibitors of type III phosphodiesterases (PDE-III) have been effective inotropes in mammalian myocardium, but their clinical utility has been limited by adverse events, including arrhythmias that are considered to be due to Ca(2+) overload. ATI22-107 [2-(2-{2-[2-chloro-4-(6-oxo-1,4,5,6-tetrahydro-pyridazin-3-yl)-phenoxy]-acetylamino}-ethoxymethyl)-4-(2-chlorophenyl)-6-methyl-1,4-dihydro-pyridine-3,5-dicarboxylic acid dimethyl ester)], a novel, dual pharmacophore compound, was designed to simultaneously inhibit the cardiac phosphodiesterase (PDE-III) and produce inotropic effects, whereas inhibiting the L-type calcium channel (LTCC) was designed to minimize increases in diastolic Ca(2+). We compared the effects of ATI22-107 and enoximone, a pure PDE-III inhibitor, on the Fluo-3 calcium transient in normal feline ventricular myocytes and trabeculae. Enoximone-induced dose-dependent increases in peak [Ca(2+)](i), diastolic [Ca(2+)](i), T50, and T75. ATI22-107 demonstrated similar dose-dependent increases in peak [Ca(2+)](i) at 300 nM and 1.0 microM doses, with no further increases at higher doses. Throughout the dosing range, ATI22-107 induced much smaller, if any, increases in diastolic [Ca(2+)](i), T(25), and T(75). Current measurement of LTCC via patch-clamp techniques revealed dose-dependent decreases in LTCC current with an increasing dose of ATI22-107, thereby validating the dual functionality of the drug that has been proposed in this study. Studies in isolated trabeculae demonstrated that enoximone-induced a dose-dependent augmentation of the entire force-frequency relation in normal myocardium, whereas augmentation of contractility was only observed at lower stimulation frequencies with ATI22-107. These results demonstrate the effects of the LTCC-antagonizing moiety of ATI22-107 and suggest that the novel simultaneous combination of PDE-III and LTCC inhibition by one molecule may produce a favorable profile of limited inotropy without detrimental effects of increased diastolic [Ca(2+)](i).


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Cardiotonic Agents/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Nifedipine/analogs & derivatives , Nifedipine/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Aniline Compounds , Animals , Calcium Channels, L-Type/drug effects , Cats , Cell Separation , Cyclic Nucleotide Phosphodiesterases, Type 3 , Dose-Response Relationship, Drug , Enoximone/pharmacology , Fluorescent Dyes , In Vitro Techniques , Myocytes, Cardiac/drug effects , Phosphodiesterase Inhibitors/pharmacology , Xanthenes
SELECTION OF CITATIONS
SEARCH DETAIL
...