Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Vasc Med ; 24(5): 414-421, 2019 10.
Article in English | MEDLINE | ID: mdl-31277561

ABSTRACT

Extensive atherosclerotic plaque burden in the lower extremities often leads to symptomatic peripheral artery disease (PAD) including impaired walking performance and claudication. Interleukin-1ß (IL-1ß) may play an important pro-inflammatory role in the pathogenesis of this disease. Interruption of IL-1ß signaling was hypothesized to decrease plaque progression in the leg macrovasculature and improve the mobility of patients with PAD with intermittent claudication. Thirty-eight patients (mean age 65 years; 71% male) with symptomatic PAD (confirmed by ankle-brachial index) were randomized 1:1 to receive canakinumab (150 mg subcutaneously) or placebo monthly for up to 12 months. The mean vessel wall area (by 3.0 T black-blood magnetic resonance imaging (MRI)) of the superficial femoral artery (SFA) was used to measure plaque volume. Mobility was assessed using the 6-minute walk test. Canakinumab was safe and well tolerated. Markers of systemic inflammation (interleukin-6 and high-sensitivity C-reactive protein) fell as early as 1 month after treatment. MRI (32 patients at 3 months; 21 patients at 12 months) showed no evidence of plaque progression in the SFA in either placebo-treated or canakinumab-treated patients. Although an exploratory endpoint, placebo-adjusted maximum and pain-free walking distance (58 m) improved as early as 3 months after treatment with canakinumab when compared with placebo. Although canakinumab did not alter plaque progression in the SFA, there is an early signal that it may improve maximum and pain-free walking distance in patients with symptomatic PAD. Larger studies aimed at this endpoint will be required to definitively demonstrate this. ClinicalTrials.gov Identifier: NCT01731990.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Intermittent Claudication/drug therapy , Peripheral Arterial Disease/drug therapy , Aged , Anti-Inflammatory Agents/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Biomarkers/blood , Double-Blind Method , Exercise Tolerance/drug effects , Female , Germany , Humans , Inflammation Mediators/blood , Intermittent Claudication/blood , Intermittent Claudication/diagnosis , Intermittent Claudication/physiopathology , Jordan , Male , Middle Aged , Peripheral Arterial Disease/blood , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/physiopathology , Proof of Concept Study , Prospective Studies , Recovery of Function , Time Factors , Treatment Outcome , United States
3.
J Med Chem ; 58(23): 9382-94, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26540564

ABSTRACT

Human clinical studies conducted with LCI699 established aldosterone synthase (CYP11B2) inhibition as a promising novel mechanism to lower arterial blood pressure. However, LCI699's low CYP11B1/CYP11B2 selectivity resulted in blunting of adrenocorticotropic hormone-stimulated cortisol secretion. This property of LCI699 prompted its development in Cushing's disease, but limited more extensive clinical studies in hypertensive populations, and provided an impetus for the search for cortisol-sparing CYP11B2 inhibitors. This paper summarizes the discovery, pharmacokinetics, and pharmacodynamic data in preclinical species and human subjects of the selective CYP11B2 inhibitor 8.


Subject(s)
Cytochrome P-450 CYP11B2/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Indoles/chemistry , Mineralocorticoid Receptor Antagonists/chemistry , Pyridines/chemistry , Sulfonamides/chemistry , Aldosterone/metabolism , Animals , Cytochrome P-450 CYP11B2/metabolism , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Halogenation , Haplorhini , Humans , Hypertension/drug therapy , Indoles/pharmacokinetics , Indoles/pharmacology , Methylation , Mineralocorticoid Receptor Antagonists/pharmacokinetics , Mineralocorticoid Receptor Antagonists/pharmacology , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
4.
Cardiovasc Res ; 108(1): 197-208, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26378152

ABSTRACT

AIMS: Liver kinase B1 (LKB1) is a protein kinase that activates the metabolic regulator AMP-activated protein kinase (AMPK) and other related kinases. Deletion of LKB1 in mice leads to cardiomyopathy and atrial fibrillation (AF). However, the specific role of the LKB1 pathway in early atrial biology remains unknown. Thus, we investigated whether LKB1 deletion altered atrial channel expression and electrophysiological function in a cardiomyocyte-specific knockout mouse model. METHODS AND RESULTS: We performed a systematic comparison of αMHC-Cre LKB1(fl/fl) and littermate LKB1(fl/fl) male mice. This included analysis of gene expression, histology, and echocardiography, as well as cellular and tissue-level electrophysiology using patch-clamp recordings in vitro, optical mapping ex vivo, and ECG recordings in vivo. At postnatal day 1, atrial depolarization was prolonged, and Nav1.5 and Cx40 expression were markedly down-regulated in MHC-Cre LKB1(fl/fl) mice. Inward sodium current density was significantly decreased in MHC-Cre LKB1(fl/fl) neonatal atrial myocytes. Subsequently, additional alterations in atrial channel expression, atrial fibrosis, and spontaneous onset of AF developed by 2 weeks of age. In adult mice, abnormalities of interatrial conduction and bi-atrial electrical coupling were observed, likely promoting the perpetuation of AF. Mice with AMPK-inactivated hearts demonstrated modest overlap in channel expression with MHC-Cre LKB1(fl/fl) hearts, but retained normal structure, electrophysiological function and contractility. CONCLUSIONS: Deletion of LKB1 causes early defects in atrial channel expression, action potential generation and conduction, which precede widespread atrial remodelling, fibrosis and AF. LKB1 is critical for normal atrial growth and electrophysiological function.


Subject(s)
Atrial Fibrillation/etiology , Heart Atria/physiopathology , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinases/physiology , Animals , Atrial Fibrillation/physiopathology , Connexins/analysis , Male , Mice , Mice, Inbred C57BL , NAV1.5 Voltage-Gated Sodium Channel/analysis , Signal Transduction/physiology , Gap Junction alpha-5 Protein
5.
J Nucl Med ; 56(6): 933-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25908827

ABSTRACT

UNLABELLED: Calcific aortic valve disease (CAVD) is the most common cause of aortic stenosis. Matrix metalloproteinases (MMPs) are upregulated in CAVD and contribute to valvular remodeling and calcification. We investigated the feasibility and correlates of MMP-targeted molecular imaging for detection of valvular biology in CAVD. METHODS: Apolipoprotein E-deficient (apoE(-/-)) mice were fed a Western diet (WD) for 3, 6, and 9 mo (n = 108) to induce CAVD. Wild-type mice served as the control group (n = 24). The development of CAVD was tracked with CT, echocardiography, MMP-targeted small-animal SPECT imaging using (99m)Tc-RP805, and histologic analysis. RESULTS: Key features of CAVD­leaflet thickening and valvular calcification­were noted after 6 mo of WD and were more pronounced after 9 mo. These findings were associated with a significant reduction in aortic valve leaflet separation and a significant increase in transaortic valve flow velocity. On in vivo SPECT/CT images, MMP signal in the aortic valve area was significantly higher at 6 mo in WD mice than in control mice and decreased thereafter. The specificity of the signal was demonstrated by blocking, using an excess of nonlabeled precursor. Similar to MMP signal, MMP activity as determined by in situ zymography and valvular inflammation by CD68 staining were maximal at 6 mo. In vivo (99m)Tc-RP805 uptake correlated significantly with MMP activity (R(2) = 0.94, P < 0.05) and CD68 expression (R(2) = 0.98, P < 0.01) in CAVD. CONCLUSION: MMP-targeted imaging detected valvular inflammation and remodeling in a murine model of CAVD. If this ability is confirmed in humans, the technique may provide a tool for tracking the effect of emerging medical therapeutic interventions and for predicting outcome in CAVD.


Subject(s)
Aortic Valve Stenosis/diagnostic imaging , Aortic Valve Stenosis/diagnosis , Aortic Valve/diagnostic imaging , Aortic Valve/pathology , Calcinosis/diagnostic imaging , Calcinosis/diagnosis , Heart Defects, Congenital/diagnosis , Heart Valve Diseases/diagnosis , Matrix Metalloproteinases/metabolism , Molecular Imaging , Multimodal Imaging , Animals , Aortic Valve/enzymology , Aortic Valve Stenosis/pathology , Apolipoproteins E/genetics , Bicuspid Aortic Valve Disease , Calcinosis/pathology , Diet, High-Fat , Echocardiography , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/enzymology , Heart Valve Diseases/diagnostic imaging , Heart Valve Diseases/enzymology , Humans , Mice , Mice, Transgenic , Technetium/chemistry , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
6.
Arterioscler Thromb Vasc Biol ; 35(5): 1166-78, 2015 May.
Article in English | MEDLINE | ID: mdl-25814675

ABSTRACT

OBJECTIVES: Pulmonary hypertension (PH) is a process of lung vascular remodeling, which can lead to right heart dysfunction and significant morbidity. The underlying mechanisms leading to PH are not well understood, and therapies are limited. Using intermittent hypoxia (IH) as a model of oxidant-induced PH, we identified an important role for endothelial cell mitophagy via mitochondrial uncoupling protein 2 (Ucp2) in the development of IH-induced PH. APPROACH AND RESULTS: Ucp2 endothelial knockout (VE-KO) and Ucp2 Flox (Flox) mice were subjected to 5 weeks of IH. Ucp2 VE-KO mice exhibited higher right ventricular systolic pressure and worse right heart hypertrophy, as measured by increased right ventricle weight/left ventricle plus septal weight (RV/LV+S) ratio, at baseline and after IH. These changes were accompanied by increased mitophagy. Primary mouse lung endothelial cells transfected with Ucp2 siRNA and subjected to cyclic exposures to CoCl2 (chemical hypoxia) showed increased mitophagy, as measured by PTEN-induced putative kinase 1 and LC3BII/I ratios, decreased mitochondrial biogenesis, and increased apoptosis. Similar results were obtained in primary lung endothelial cells isolated from VE-KO mice. Moreover, silencing PTEN-induced putative kinase 1 in the endothelium of Ucp2 knockout mice, using endothelial-targeted lentiviral silencing RNA in vivo, prevented IH-induced PH. Human pulmonary artery endothelial cells from people with PH demonstrated changes similar to Ucp2-silenced mouse lung endothelial cells. CONCLUSIONS: The loss of endothelial Ucp2 leads to excessive PTEN-induced putative kinase 1-induced mitophagy, inadequate mitochondrial biosynthesis, and increased apoptosis in endothelium. An endothelial Ucp2-PTEN-induced putative kinase 1 axis may be effective therapeutic targets in PH.


Subject(s)
Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/metabolism , Hypoxia/complications , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Animals , Autophagy/drug effects , Autophagy/physiology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/physiopathology , Ion Channels/pharmacology , Mice , Mice, Knockout , Mitochondrial Proteins/pharmacology , Mitophagy/drug effects , Mitophagy/physiology , Protein Kinases/metabolism , Random Allocation , Reference Values , Uncoupling Protein 2
7.
Proc Natl Acad Sci U S A ; 111(46): 16604-9, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368166

ABSTRACT

Cardiac disorders are the main cause of mortality in autosomal-dominant polycystic kidney disease (ADPKD). However, how mutated polycystins predispose patients with ADPKD to cardiac pathologies before development of renal dysfunction is unknown. We investigate the effect of decreased levels of polycystin 2 (PC2), a calcium channel that interacts with the ryanodine receptor, on myocardial function. We hypothesize that heterozygous PC2 mice (Pkd2(+/-)) undergo cardiac remodeling as a result of changes in calcium handling, separate from renal complications. We found that Pkd2(+/-) cardiomyocytes have altered calcium handling, independent of desensitized calcium-contraction coupling. Paradoxically, in Pkd2(+/-) mice, protein kinase A (PKA) phosphorylation of phospholamban (PLB) was decreased, whereas PKA phosphorylation of troponin I was increased, explaining the decoupling between calcium signaling and contractility. In silico modeling supported this relationship. Echocardiography measurements showed that Pkd2(+/-) mice have increased left ventricular ejection fraction after stimulation with isoproterenol (ISO), a ß-adrenergic receptor (ßAR) agonist. Blockers of ßAR-1 and ßAR-2 inhibited the ISO response in Pkd2(+/-) mice, suggesting that the dephosphorylated state of PLB is primarily by ßAR-2 signaling. Importantly, the Pkd2(+/-) mice were normotensive and had no evidence of renal cysts. Our results showed that decreased PC2 levels shifted the ßAR pathway balance and changed expression of calcium handling proteins, which resulted in altered cardiac contractility. We propose that PC2 levels in the heart may directly contribute to cardiac remodeling in patients with ADPKD in the absence of renal dysfunction.


Subject(s)
Calcium Signaling/physiology , Excitation Contraction Coupling/physiology , Myocytes, Cardiac/physiology , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , TRPP Cation Channels/deficiency , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Blood Pressure/genetics , Caffeine/pharmacology , Calcium Signaling/drug effects , Calcium-Binding Proteins/metabolism , Cardiac Pacing, Artificial , Excitation Contraction Coupling/drug effects , Excitation Contraction Coupling/genetics , Heterozygote , Isoproterenol/pharmacology , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Messenger/biosynthesis , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/physiology , TRPP Cation Channels/genetics , TRPP Cation Channels/physiology , Troponin I/metabolism , Ventricular Remodeling/drug effects , Ventricular Remodeling/genetics , Ventricular Remodeling/physiology
8.
Proc Natl Acad Sci U S A ; 110(40): 16133-8, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24043794

ABSTRACT

Urocortin 2 (Ucn2), a peptide of the corticotropin-releasing factor (CRF) family, binds with high affinity to type 2 CRF receptors (CRFR2) on cardiomyocytes and confers protection against ischemia/reperfusion. The mechanisms by which the Ucn2-CRFR2 axis mitigates against ischemia/reperfusion injury remain incompletely delineated. Activation of AMP-activated protein kinase (AMPK) also limits cardiac damage during ischemia/reperfusion. AMPK is classically activated by alterations in cellular energetics; however, hormones, cytokines, and additional autocrine/paracrine factors also modulate its activity. We examined the effects of both the endogenous cardiac Ucn2 autocrine/paracrine pathway and Ucn2 treatment on AMPK regulation. Ucn2 treatment increased AMPK activation and downstream acetyl-CoA carboxylase phosphorylation and glucose uptake in isolated heart muscles. These actions were blocked by the CRFR2 antagonist anti-sauvagine-30 and by a PKCε translocation-inhibitor peptide (εV1-2). Hypoxia-induced AMPK activation was also blunted in heart muscles by preincubation with either anti-sauvagine-30, a neutralizing anti-Ucn2 antibody, or εV1-2. Treatment with Ucn2 in vivo augmented ischemic AMPK activation and reduced myocardial injury and cardiac contractile dysfunction after regional ischemia/reperfusion in mice. Ucn2 also directly activated AMPK in ex vivo-perfused mouse hearts and diminished injury and contractile dysfunction during ischemia/reperfusion. Thus, both Ucn2 treatment and the endogenous cardiac Ucn2 autocrine/paracrine pathway activate AMPK signaling pathway, via a PKCε-dependent mechanism, defining a Ucn2-CRFR2-PKCε-AMPK pathway that mitigates against ischemia/reperfusion injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Corticotropin-Releasing Hormone/pharmacology , Myocardium/enzymology , Receptors, Corticotropin-Releasing Hormone/metabolism , Reperfusion Injury/metabolism , Signal Transduction/physiology , Urocortins/pharmacology , Acetyl-CoA Carboxylase/metabolism , Analysis of Variance , Animals , Antibodies, Neutralizing/pharmacology , Corticotropin-Releasing Hormone/blood , Corticotropin-Releasing Hormone/metabolism , Enzyme Activation/drug effects , Immunoblotting , Immunohistochemistry , Mice , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects , Urocortins/blood , Urocortins/metabolism
9.
Endocrinology ; 154(10): 3937-46, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23861374

ABSTRACT

Altered glucose metabolism in the heart is an important characteristic of cardiovascular and metabolic disease. Because thyroid hormones have major effects on peripheral metabolism, we examined the metabolic effects of heart-selective increase in T3 using transgenic mice expressing human type 2 iodothyronine deiodinase (D2) under the control of the α-myosin heavy chain promoter (MHC-D2). Hyperinsulinemic-euglycemic clamps showed normal whole-body glucose disposal but increased hepatic insulin action in MHC-D2 mice as compared to wild-type (WT) littermates. Insulin-stimulated glucose uptake in heart was not altered, but basal myocardial glucose metabolism was increased by more than two-fold in MHC-D2 mice. Myocardial lipid levels were also elevated in MHC-D2 mice, suggesting an overall up-regulation of cardiac metabolism in these mice. The effects of doxorubicin (DOX) treatment on cardiac function and structure were examined using M-mode echocardiography. DOX treatment caused a significant reduction in ventricular fractional shortening and resulted in more than 50% death in WT mice. In contrast, MHC-D2 mice showed increased survival rate after DOX treatment, and this was associated with a six-fold increase in myocardial glucose metabolism and improved cardiac function. Myocardial activity and expression of AMPK, GLUT1, and Akt were also elevated in MHC-D2 and WT mice following DOX treatment. Thus, our findings indicate an important role of thyroid hormone in cardiac metabolism and further suggest a protective role of glucose utilization in DOX-mediated cardiac dysfunction.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Glucose/metabolism , Heart Ventricles/drug effects , Insulin Resistance , Iodide Peroxidase/biosynthesis , Ventricular Dysfunction/chemically induced , AMP-Activated Protein Kinases/biosynthesis , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Glucose Clamp Technique , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Heart Ventricles/diagnostic imaging , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Survival Analysis , Triiodothyronine/metabolism , Ultrasonography , Ventricular Dysfunction/diagnostic imaging , Ventricular Dysfunction/metabolism , Ventricular Dysfunction/physiopathology , Iodothyronine Deiodinase Type II
10.
Am J Physiol Heart Circ Physiol ; 304(9): H1192-200, 2013 May.
Article in English | MEDLINE | ID: mdl-23457013

ABSTRACT

Overexpression of mitochondrial uncoupling proteins (UCPs) attenuates ischemia-reperfusion (I/R) injury in cultured cardiomyocytes. However, it is not known whether UCPs play an essential role in cardioprotection in the intact heart. This study evaluated the cardioprotective efficacy of UCPs against I/R injury and characterized the mechanism of UCP-mediated protection in addition to the role of UCPs in ischemic preconditioning (IPC). Cardiac UCP3 knockout (UCP3(-/-)) and wild-type (WT) mice hearts were subjected to ex vivo and in vivo models of I/R injury and IPC. Isolated UCP3(-/-) mouse hearts were retrogradely perfused and found to have poorer recovery of left ventricular function compared with WT hearts under I/R conditions. In vivo occlusion of the left coronary artery resulted in twofold larger infarcts in UCP3(-/-) mice compared with WT mice. Moreover, the incidence of in vivo I/R arrhythmias was higher in UCP3(-/-) mice. Myocardial energetics were significantly impaired with I/R, as reflected by a decreased ATP content and an increase in the AMP-to-ATP ratio. UCP3(-/-) hearts generated more reactive oxygen species (ROS) than WT hearts during I/R. Pretreatment of UCP3(-/-) hearts with the pharmacological uncoupling agent carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone improved postischemic functional recovery. Also the protective efficacy of IPC was abolished in UCP3(-/-) mice. We conclude that UCP3 plays a critical role in cardioprotection against I/R injury and the IPC phenomenon. There is increased myocardial vulnerability to I/R injury in hearts lacking UCP3. The mechanisms of UCP3-mediated cardioprotection include regulation of myocardial energetics and ROS generation by UCP3 during I/R.


Subject(s)
Arrhythmias, Cardiac/genetics , Ion Channels/genetics , Ischemic Preconditioning, Myocardial , Mitochondrial Proteins/genetics , Myocardial Reperfusion Injury/genetics , Adenosine Triphosphate/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/therapeutic use , Coronary Occlusion/physiopathology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Uncoupling Protein 2 , Uncoupling Protein 3 , Ventricular Dysfunction/genetics , Ventricular Dysfunction/physiopathology
11.
Cardiovasc Res ; 93(3): 516-24, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22200588

ABSTRACT

AIMS: Neuregulins (NRG) are growth factors that are synthesized by endothelial cells (ECs) and bind to erbB receptors. We have shown previously that NRG is proangiogenic in vitro, and that NRG/erbB signalling is important for autocrine endothelial angiogenic signalling in vitro. However, the role of NRG in the angiogenic response to ischaemia is unknown. We hypothesized that endothelial NRG is required for ischaemia-induced angiogenesis in vivo and that exogenous administration of NRG will enhance angiogenic responses after ischaemic insult. METHODS AND RESULTS: An endothelial-selective inducible NRG knockout mouse was created and subjected to femoral artery ligation. Endothelial NRG deletion significantly decreased blood flow recovery (by 40%, P < 0.05), capillary density, α(v)ß(3) integrin activation, and arteriogenesis after ischaemic injury. Isolated ECs from knockout mice demonstrated significantly impaired cord formation in vitro, suggesting that NRG signalling performs an important cell autonomous function. Recombinant human NRG (rNRG) has not only reversed the angiogenic defect in knockout mice but also accelerated blood flow recovery in wild-type mice. CONCLUSION: Endothelial production of NRG is required for angiogenesis and arteriogenesis induced by ischaemic injury. Furthermore, exogenous administration of rNRG can enhance this process, suggesting a potential role for NRG in vascular disease.


Subject(s)
Endothelium, Vascular/metabolism , Femoral Artery/physiology , Ischemia/metabolism , Neovascularization, Physiologic/physiology , Neuregulin-1/metabolism , Animals , Aorta, Thoracic/physiology , Biocompatible Materials , Cell Division/physiology , Collagen , Disease Models, Animal , Drug Combinations , Endothelium, Vascular/cytology , Gene Expression/physiology , Humans , Integrin alphaVbeta3/metabolism , Laminin , Mice , Mice, Knockout , Neuregulin-1/genetics , Proteoglycans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regional Blood Flow/physiology
12.
Circulation ; 123(20): 2254-62, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555713

ABSTRACT

BACKGROUND: Removal of cardiac endothelial cells (EC) has been shown to produce significant detrimental effects on the function of adjacent cardiac myocytes, suggesting that EC play a critical role in autocrine/paracrine regulation of the heart. Despite this important observation, the mediators of the protective function of EC remain obscure. Neuregulin (NRG, a member of the epidermal growth factor family) is produced by EC and cardiac myocytes contain receptors (erbB) for this ligand. We hypothesized that NRG is an essential factor produced by EC, which promotes cardioprotection against ischemic injury. METHODS AND RESULTS: We demonstrate that human cardiac EC express and release NRG in response to hypoxia-reoxygenation. Under conditions where hypoxia--reoxygenation causes significant cardiac myocyte cell death, NRG can significantly decrease apoptosis of isolated adult ventricular myocytes. Coculturing adult murine myocytes with human umbilical vein, murine lung microvascular, or human coronary artery EC can also protect myocytes against hypoxia--reoxygenation--induced apoptosis. These protective effects are abolished by NRG gene deletion or silencing of NRG expression in EC. Finally, endothelium-selective deletion of NRG in vivo leads to significantly decreased tolerance to ischemic insult, as demonstrated by impaired postischemic contractile recovery in a perfused whole-organ preparation and larger infarct sizes after coronary artery ligation. CONCLUSION: Together, these data demonstrate that EC-derived NRG plays an important role in cardiac myocyte protection against ischemic injury in the heart and supports the idea that manipulation of this signaling pathway may be an important clinical target in this setting.


Subject(s)
Endothelial Cells/metabolism , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Neuregulin-1/metabolism , Adult , Animals , Apoptosis/physiology , Cells, Cultured , Coculture Techniques , Coronary Vessels/cytology , Endothelial Cells/cytology , Humans , Hypoxia/metabolism , Hypoxia/pathology , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Neuregulin-1/genetics , Signal Transduction/physiology , Umbilical Veins/cytology
13.
Cardiovasc Res ; 90(2): 305-14, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21233252

ABSTRACT

AIMS: Doxorubicin (DOX) is a highly effective chemotherapeutic agent; however, cumulative dose-dependent cardiotoxicity is a significant side effect of this therapy. Because DOX is a polyaromatic hydrocarbon, we hypothesized that it will be metabolized by the activation of the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that is involved in the metabolism of numerous xenobiotic agents. These studies were performed to determine whether DOX activates AhR and whether this activation modulates the toxicity of DOX in cardiomyocytes. METHODS AND RESULTS: Treatment with DOX induced AhR migration to the nucleus, increased AhR binding with its co-factor, aryl hydrocarbon receptor nuclear translocator-1 (ARNT1), and increased the expression of AhR-regulated phase I (CYP1A1) and phase II (GSTA1) drug-metabolizing enzymes in both cardiomyocytes and in the intact heart. Knockdown of AhR in H9C2 cells abolished DOX-induced increases in CYP1A1 and GSTA1 expression. Similar results were obtained by treating adult rat ventricular myocytes with the AhR antagonist, CH-223191. Taken together, these findings indicate that DOX-induced upregulation of CYP1A1 and GSTA1 expression is AhR dependent. AhR null mice treated with 10 mg/kg DOX did not show any activation of CYP1A1 or GSTA1 expression. Moreover, lack of AhR in vivo resulted in a significant decrease in left ventricular function compared with wild-type animals, and increased p53 activation and apoptosis in the heart after treatment with DOX. CONCLUSIONS: These findings indicate that AhR plays an important role in DOX metabolism by the heart and further demonstrate that AhR is cardioprotective against DOX-induced cardiotoxicity.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Cardiotoxins/toxicity , Doxorubicin/toxicity , Heart/drug effects , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Apoptosis/physiology , Aryl Hydrocarbon Receptor Nuclear Translocator/agonists , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Cardiotoxins/pharmacokinetics , Doxorubicin/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
14.
Cancer Chemother Pharmacol ; 67(6): 1381-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20809120

ABSTRACT

PURPOSE: Doxorubicin-based chemotherapy is limited by the development of dose-dependent left ventricular dysfunction and congestive heart failure caused by reactive oxygen species (ROS). Uncoupling proteins (UCP) can inhibit mitochondrial ROS production as well as decrease myocyte damage from exogenous ROS. Prior studies have shown that cardiac UCP2 and UCP3 mRNA expression is decreased with acute doxorubicin treatment. However, the expression of UCP protein in hearts with doxorubicin cardiotoxicity and the resultant changes in mitochondrial function and oxidant stress have not been determined. METHODS: Heart failure was induced in Sprague-Dawley rats with intraperitoneal injections of doxorubicin (2 mg/kg t.i.w., total dose: 18 mg/kg). Mitochondria were isolated from mice receiving doxorubicin or saline injections for determination of UCP2 and UCP3 expression. In addition, mitochondrial respiration, ATP synthesis and ROS production were determined. RESULTS: Doxorubicin-induced heart failure was associated with significant decreases in UCP2 and UCP3 protein expression compared with nonfailing hearts (P < 0.05). While the rates of state 3 and state 4 respiration and ATP synthesis were lower in mitochondria isolated from failing hearts, the respiratory control ratio was 15% higher (P < 0.05), and the ratio of ATP production to oxygen consumption was 25% higher (P < 0.05) in mitochondria from failing hearts, indicating greater coupling between citric acid cycle flux and mitochondrial ATP synthesis. However, the decrease in UCP expression was associated with 50% greater mitochondrial ROS generation (P < 0.05). CONCLUSIONS: Downregulation of myocardial UCP2 and UCP3 in the setting of doxorubicin-induced heart failure is associated with improved efficiency of ATP synthesis, which might compensate for abnormal energy metabolism. However, this beneficial effect is counterbalanced by greater oxidant stress.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Heart Failure/metabolism , Ion Channels/biosynthesis , Mitochondria, Heart/drug effects , Mitochondrial Proteins/biosynthesis , Reactive Oxygen Species/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Down-Regulation , Heart Failure/chemically induced , Heart Failure/physiopathology , In Vitro Techniques , Male , Mice , Mitochondria, Heart/metabolism , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Uncoupling Protein 2 , Uncoupling Protein 3 , Ventricular Function, Left/drug effects
15.
Curr Cardiol Rev ; 7(4): 221-33, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22758623

ABSTRACT

Cardiac toxicity of molecularly targeted cancer agents is increasingly recognized as a significant side effect of chemotherapy. These new potent therapies may not only affect the survival of cancer cells, but have the potential to adversely impact normal cardiac and vascular function. Unraveling the mechanisms by which these therapies affect the heart and vasculature is crucial for improving drug design and finding alternative therapies to protect patients predisposed to cardiovascular disease. In this review, we summarize the classification and side effects of currently approved molecularly targeted chemotherapeutics.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxins/adverse effects , Cardiovascular Diseases/chemically induced , Molecular Targeted Therapy/adverse effects , Antineoplastic Agents/classification , Cardiotoxins/classification , Cardiovascular Diseases/physiopathology , Humans , Molecular Targeted Therapy/methods , Neoplasms/drug therapy
16.
FASEB J ; 24(7): 2567-75, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20215529

ABSTRACT

Inflammation is often accompanied by robust angiogenesis. Vascular endothelial cells (ECs) express erbB receptors and their ligand, neuregulin-1, and can respond to neuregulin by proliferation and angiogenesis. We hypothesized that some growth factor-like responses of ECs to inflammatory cytokines can be explained by cleavage of transmembrane neuregulin with subsequent release of its extracellular epidermal growth factor-like-containing domain and autocrine activation. Using a model of cultured human ECs, we found that interleukin-6 or interferon-gamma causes rapid cleavage and release of transmembrane neuregulin. Inhibitors of metalloproteinases abolish this effect. The addition of an inhibitor of tumor necrosis factor-alpha converting enzyme (TACE) blocks cytokine-induced neuregulin release. Silencing of TACE expression increases the amount of basal proneuregulin present in ECs but does not block neuregulin release in response to phorbol myristate acetate (PMA), suggesting that other proteinases are responsible for mediating protein kinase C-dependent cleavage. Cytokines capable of inducing neuregulin cleavage stimulated ERK activation and in vitro angiogenesis (Matrigel cord formation). This effect is blocked by inhibitors that block neuregulin cleavage, erbB protein tyrosine kinase inhibitors, or antineuregulin-neutralizing antibodies. Cytokine-activated metalloproteinase cleavage of neuregulin may play an important role in autocrine activation of EC signaling pathways, contributing to key biological effects, perhaps including inflammation-associated angiogenesis.


Subject(s)
Autocrine Communication , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Metalloproteases/metabolism , Neuregulins/metabolism , Cells, Cultured , Cytokines , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Hydrolysis , Inflammation , Interferon-gamma/pharmacology , Interleukin-6/pharmacology , Neovascularization, Physiologic
17.
Nanomedicine ; 5(4): 410-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19341815

ABSTRACT

We hypothesize that the efficacy of doxorubicin (DOX) can be maximized and dose-limiting cardiotoxicity minimized by controlled release from PEGylated nanoparticles. To test this hypothesis, a unique surface modification technique was used to create PEGylated poly(lactic-co-glycolic acid) (PLGA) nanoparticles encapsulating DOX. An avidin-biotin coupling system was used to control poly(ethylene glycol) conjugation to the surface of PLGA nanoparticles, of diameter approximately 130 nm, loaded with DOX to 5% (wt/wt). Encapsulation in nanoparticles did not compromise the efficacy of DOX; drug-loaded nanoparticles were found to be at least as potent as free DOX against A20 murine B-cell lymphoma cells in culture and of comparable efficacy against subcutaneously implanted tumors. Cardiotoxicity in mice as measured by echocardiography, serum creatine phosphokinase (CPK), and histopathology was reduced for DOX-loaded nanoparticles as compared with free DOX. Administration of 18 mg/kg of free DOX induced a sevenfold increase in CPK levels and significant decreases in left ventricular fractional shortening over control animals, whereas nanoparticle-encapsulated DOX produced none of these pathological changes. FROM THE CLINICAL EDITOR: The efficacy of doxorubicin (DOX) may be maximized and dose-limiting cardiotoxicity minimized by controlled release from PEGylated nanoparticles. Administration of 18 mg/kg of free DOX induced a sevenfold increase in CPK levels and significant decreases in left ventricular fractional shortening in mice, whereas nanoparticle-encapsulated DOX produced none of these pathological changes.


Subject(s)
Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Drug Delivery Systems/standards , Glycolates/chemistry , Nanoparticles/administration & dosage , Polyethylene Glycols/chemistry , Adsorption/drug effects , Animals , Cattle , Cell Death/drug effects , Doxorubicin/blood , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Glycolates/pharmacokinetics , Heart/drug effects , Heart/physiopathology , Lactic Acid , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/physiopathology , Mice , Mice, Inbred BALB C , Nanoparticles/toxicity , Nanoparticles/ultrastructure , Particle Size , Polyethylene Glycols/pharmacokinetics , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Serum Albumin, Bovine/metabolism , Surface Properties/drug effects , Tissue Distribution/drug effects , Ventricular Function, Left/drug effects
19.
FASEB J ; 23(4): 1272-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19088180

ABSTRACT

The purpose of this study was to determine both the short-term effects on cardiac development and embryo growth and the long-term effects on cardiac function and body composition of in utero caffeine exposure. Pregnant mice (C57BL/6) were exposed to hypoxia (10% O(2)) or room air from embryonic days (E) 8.5-10.5, and treated with caffeine (20 mg/kg, i.p.) or vehicle (normal saline, 0.9% NaCl). This caffeine dose results in a circulating level that is equivalent to 2 cups of coffee in humans. Hypoxic exposure acutely reduced embryonic growth by 30%. Exposure to a single dose of caffeine inhibited cardiac ventricular development by 53% in hypoxia and 37% in room air. Caffeine exposure resulted in inhibition of hypoxia-induced HIF1alpha protein expression in embryos by 40%. When offspring from dams treated with a single dose of caffeine were studied in adulthood, we observed that caffeine treatment alone resulted in a decrease in cardiac function of 38%, as assessed by echocardiography. We also observed a 20% increase in body fat with male mice exposed to caffeine. Caffeine was dissolved in normal saline, so it was used as a control. Room air controls were used to compare to the hypoxic mice. Exposure to a single dose of caffeine during embryogenesis results in both short-term effects on cardiac development and long-term effects on cardiac function.


Subject(s)
Caffeine/toxicity , Growth/drug effects , Maternal Exposure , Prenatal Exposure Delayed Effects/metabolism , Time , Animals , Caffeine/metabolism , Embryo, Mammalian/metabolism , Female , Hypoxia/metabolism , Maternal-Fetal Exchange/drug effects , Mice , Mice, Inbred C57BL , Pregnancy
20.
Diabetes ; 54(12): 3530-40, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16306372

ABSTRACT

Type 2 diabetes is a heterogeneous disease characterized by insulin resistance and altered glucose and lipid metabolism in multiple organs. To understand the complex series of events that occur during the development of obesity-associated diabetes, we examined the temporal pattern of changes in insulin action and glucose metabolism in individual organs during chronic high-fat feeding in C57BL/6 mice. Insulin-stimulated cardiac glucose metabolism was significantly reduced after 1.5 weeks of high-fat feeding, and cardiac insulin resistance was associated with blunted Akt-mediated insulin signaling and GLUT4 levels. Insulin resistance in skeletal muscle, adipose tissue, and liver developed in parallel after 3 weeks of high-fat feeding. Diet-induced whole-body insulin resistance was associated with increased circulating levels of resistin and leptin but unaltered adiponectin levels. High-fat feeding caused insulin resistance in skeletal muscle that was associated with significantly elevated intramuscular fat content. In contrast, diet-induced hepatic insulin resistance developed before a marked increase in intrahepatic triglyceride levels. Cardiac function gradually declined over the course of high-fat feeding, and after 20 weeks of high-fat diet, cardiac dysfunction was associated with mild hyperglycemia, hyperleptinemia, and reduced circulating adiponectin levels. Our findings demonstrate that cardiac insulin resistance is an early adaptive event in response to obesity and develops before changes in whole-body glucose homeostasis. This suggests that obesity-associated defects in cardiac function may not be due to insulin resistance per se but may be attributable to chronic alteration in cardiac glucose and lipid metabolism and circulating adipokines.


Subject(s)
Animal Feed , Dietary Fats , Heart Diseases/physiopathology , Insulin Resistance/physiology , Insulin/physiology , Adipose Tissue/anatomy & histology , Animals , Blood Glucose/metabolism , Glucose/metabolism , Glucose Clamp Technique , Heart/drug effects , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Organ Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...