Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 45
1.
Redox Biol ; 67: 102884, 2023 11.
Article En | MEDLINE | ID: mdl-37725888

Adenosine kinase (ADK) plays the major role in cardiac adenosine metabolism, so that inhibition of ADK increases myocardial adenosine levels. While the cardioprotective actions of extracellular adenosine against ischemia/reperfusion (I/R) are well-established, the role of cellular adenosine in protection against I/R remains unknown. Here we investigated the role of cellular adenosine in epigenetic regulation on cardiomyocyte gene expression, glucose metabolism and tolerance to I/R. Evans blue/TTC staining and echocardiography were used to assess the extent of I/R injury in mice. Glucose metabolism was evaluated by positron emission tomography and computed tomography (PET/CT). Methylated DNA immunoprecipitation (MeDIP) and bisulfite sequencing PCR (BSP) were used to evaluate DNA methylation. Lentiviral/adenovirus transduction was used to overexpress DNMT1, and the OSI-906 was administered to inhibit IGF-1. Cardiomyocyte-specific ADK/IGF-1-knockout mice were used for mechanistic experiments.Cardiomyocyte-specific ADK knockout enhanced glucose metabolism and ameliorated myocardial I/R injury in vivo. Mechanistically, ADK deletion caused cellular adenosine accumulation, decreased DNA methyltransferase 1 (DNMT1) expression and caused hypomethylation of multiple metabolic genes, including insulin growth factor 1 (IGF-1). DNMT1 overexpression abrogated these beneficial effects by enhancing apoptosis and decreasing IGF-1 expression. Inhibition of IGF-1 signaling with OSI-906 or genetic knocking down of IGF-1 also abrogated the cardioprotective effects of ADK knockout, revealing the therapeutic potential of increasing IGF-1 expression in attenuating myocardial I/R injury. In conclusion, the present study demonstrated that cardiomyocyte ADK deletion ameliorates myocardial I/R injury via epigenetic upregulation of IGF-1 expression via the cardiomyocyte adenosine/DNMT1/IGF-1 axis.


Myocardial Reperfusion Injury , Myocytes, Cardiac , Mice , Animals , Myocytes, Cardiac/metabolism , Epigenesis, Genetic , Adenosine/metabolism , Insulin-Like Growth Factor I/metabolism , Positron Emission Tomography Computed Tomography , Ischemia/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Mice, Knockout , Apoptosis , Reperfusion , DNA/metabolism , Glucose/metabolism
2.
Eur J Pharmacol ; 927: 175050, 2022 Jul 15.
Article En | MEDLINE | ID: mdl-35618039

Pharmacological inhibition of adenosine kinase (ADK), the major route of myocardial adenosine metabolism, can elicit acute cardioprotection against ischemia-reperfusion (IR) by increasing adenosine signaling. Here, we identified a novel, extended effect of the ADK inhibitor, ABT-702, on cardiac ADK protein longevity and investigated its impact on sustained adenosinergic cardioprotection. We found that ABT-702 treatment significantly reduced cardiac ADK protein content in mice 24-72 h after administration (IP or oral). ABT-702 did not alter ADK mRNA levels, but strongly diminished (ADK-L) isoform protein content through a proteasome-dependent mechanism. Langendorff perfusion experiments revealed that hearts from ABT-702-treated mice maintain higher adenosine release long after ABT-702 tissue elimination, accompanied by increased basal coronary flow (CF) and robust tolerance to IR. Sustained cardioprotection by ABT-702 did not involve increased nitric oxide synthase expression, but was completely dependent upon increased adenosine release in the delayed phase (24 h), as indicated by the loss of cardioprotection and CF increase upon perfusion of adenosine deaminase or adenosine receptor antagonist, 8-phenyltheophylline. Importantly, blocking adenosine receptor activity with theophylline during ABT-702 administration prevented ADK degradation, preserved late cardiac ADK activity, diminished CF increase and abolished delayed cardioprotection, indicating that early adenosine receptor signaling induces late ADK degradation to elicit sustained adenosine release. Together, these results indicate that ABT-702 induces a distinct form of delayed cardioprotection mediated by adenosine receptor-dependent, proteasomal degradation of cardiac ADK and enhanced adenosine signaling in the late phase. These findings suggest ADK protein stability may be pharmacologically targeted to achieve sustained adenosinergic cardioprotection.


Adenosine Kinase , Morpholines , Pyrimidines , Adenosine Kinase/antagonists & inhibitors , Adenosine Kinase/metabolism , Animals , Cardiotonic Agents/pharmacology , Heart/diagnostic imaging , Mice , Morpholines/pharmacology , Myocardium/enzymology , Proteolysis/drug effects , Pyrimidines/pharmacology , Receptors, Purinergic P1/metabolism
3.
Analyst ; 144(14): 4219-4232, 2019 Jul 21.
Article En | MEDLINE | ID: mdl-31199423

We present the ability to conduct single micrometer-sized uranium particle age-dating measurements on particles that are younger, smaller, and less enriched in 235U content than previously reported. Specifically, we use large geometry secondary ion mass spectrometry (LG-SIMS) to precisely measure the 230Th/234U radiochronometer, combined with a systematic treatment of relevant parameters such as particle size, enrichment, and age, to achieve this development. We describe the necessary requirements for instrument background, interference rejection, abundance sensitivity, and other instrumental conditions that allow for this advance in single-particle uranium age-dating. We introduce the use of statistics developed by Feldman and Cousins to generate 95% confidence intervals in particle age, even when 230Th daughter ions are not detected. For particles where counts are limited and are of identical isotopic signatures, we provide an option for aggregating individual measurements of single particles to reduce measurement uncertainty, as if the measurement had been performed on one larger particle. The methodology is validated on a range of certified reference materials and 'real-world' samples, ranging in age from 15 to 60 years, and on individual particles ranging in equivalent size from 0.6 to 6.8 micrometers. Additionally, we provide model age calculations for particles ranging in size from 1.0 to 3.0 micrometers across enrichments ranging from natural uranium to highly-enriched uranium and on ages ranging from 0 to 60 years. Experimental results compare well with the predicted model ages, providing realistic guidance for expectations of single micrometer-sized uranium particle age-dating measurements. The age-dating capabilities described herein are directly relevant to the International Atomic Energy Agency (IAEA) and its mission to safeguard nuclear materials and monitor member state nuclear programs.

4.
J Mol Cell Cardiol ; 130: 49-58, 2019 05.
Article En | MEDLINE | ID: mdl-30910669

Adenosine exerts numerous protective actions in the heart, including attenuation of cardiac hypertrophy. Adenosine kinase (ADK) converts adenosine to adenosine monophosphate (AMP) and is the major route of myocardial adenosine metabolism, however, the impact of ADK activity on cardiac structure and function is unknown. To examine the role of ADK in cardiac homeostasis and adaptation to stress, conditional cardiomyocyte specific ADK knockout mice (cADK-/-) were produced using the MerCreMer-lox-P system. Within 4 weeks of ADK disruption, cADK-/- mice developed spontaneous hypertrophy and increased ß-Myosin Heavy Chain expression without observable LV dysfunction. In response to 6 weeks moderate left ventricular pressure overload (transverse aortic constriction;TAC), wild type mice (WT) exhibited ~60% increase in ventricular ADK expression and developed LV hypertrophy with preserved LV function. In contrast, cADK-/- mice exhibited significantly greater LV hypertrophy and cardiac stress marker expression (atrial natrurietic peptide and ß-Myosin Heavy Chain), LV dilation, reduced LV ejection fraction and increased pulmonary congestion. ADK disruption did not decrease protein methylation, inhibit AMPK, or worsen fibrosis, but was associated with persistently elevated mTORC1 and p44/42 ERK MAP kinase signaling and a striking increase in microtubule (MT) stabilization/detyrosination. In neonatal cardiomyocytes exposed to hypertrophic stress, 2-chloroadenosine (CADO) or adenosine treatment suppressed MT detyrosination, which was reversed by ADK inhibition with iodotubercidin or ABT-702. Conversely, adenoviral over-expression of ADK augmented CADO destabilization of MTs and potentiated CADO attenuation of cardiomyocyte hypertrophy. Together, these findings indicate a novel adenosine receptor-independent role for ADK-mediated adenosine metabolism in cardiomyocyte microtubule dynamics and protection against maladaptive hypertrophy.


Adenosine Kinase/metabolism , Cardiomegaly/metabolism , MAP Kinase Signaling System , Microtubules/metabolism , Myocytes, Cardiac/metabolism , Ventricular Dysfunction, Left/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Kinase/genetics , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Mice , Mice, Knockout , Microtubules/genetics , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Stroke Volume/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
5.
J Mol Cell Cardiol ; 128: 105-116, 2019 03.
Article En | MEDLINE | ID: mdl-30707992

While most mitochondrial proteins are encoded in the nucleus and translated on cytosolic/endoplasmic reticulum ribosomes, proteins encoded by mitochondrial DNA are translated on mitochondrial ribosomes. Mitochondrial GTPases 1 (MTG1) regulates mitochondrial ribosome assembly and translation, but its impact on cardiac adaptation to stress is unknown. Here, we found that MTG1 is dramatically elevated in hearts of dilated cardiomyopathy patients and in mice exposed to left ventricular pressure overload (AB). To examine the role of MTG1 in cardiac hypertrophy and heart failure, MTG1 loss/gain of function studies were performed in cultured cardiomyocytes and mice exposed to hypertrophic stress. MTG1 shRNA and adenoviral overexpression studies indicated that MTG1 expression attenuates angiotensin II-induced hypertrophy in cultured cardiomyocytes, while MTG1 KO mice exhibited no observable cardiac phenotype under basal conditions. MTG1 deficiency significantly exacerbated AB-induced cardiac hypertrophy, expression of hypertrophic stress markers, fibrosis, and LV dysfunction in comparison to WT mice. Conversely, transgenic cardiac MTG1 expression attenuated AB-induced hypertrophy and LV dysfunction. Mechanistically, MTG1 preserved mitochondrial respiratory chain complex activity during pressure overload, which further attenuated ROS generation. Moreover, we demonstrated that TAK1, P38 and JNK1/2 activity is downregulated in the MTG1 overexpression group. Importantly, dampening oxidative stress with N-acetylcysteine (NAC) lowered hypertrophy in MTG1 KO to WT levels. Collectively, our data indicate that MTG1 protects against pressure overload-induced cardiac hypertrophy and dysfunction by preserving mitochondrial function and reducing oxidative stress and downstream TAK1 stress signaling.


Cardiomyopathy, Dilated/genetics , GTP Phosphohydrolases/genetics , Heart Failure/genetics , MAP Kinase Kinase Kinases/genetics , Angiotensin II/genetics , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomyopathy, Dilated/pathology , Heart Failure/pathology , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/genetics
6.
Mol Pharmacol ; 93(4): 335-343, 2018 04.
Article En | MEDLINE | ID: mdl-29358221

According to current views, oxidation of aldehyde dehydrogenase-2 (ALDH2) during glyceryltrinitrate (GTN) biotransformation is essentially involved in vascular nitrate tolerance and explains the dependence of this reaction on added thiols. Using a novel fluorescent intracellular nitric oxide (NO) probe expressed in vascular smooth muscle cells (VSMCs), we observed ALDH2-catalyzed formation of NO from GTN in the presence of exogenously added dithiothreitol (DTT), whereas only a short burst of NO, corresponding to a single turnover of ALDH2, occurred in the absence of DTT. This short burst of NO associated with oxidation of the reactive C302 residue in the active site was followed by formation of low-nanomolar NO, even without added DTT, indicating slow recovery of ALDH2 activity by an endogenous reductant. In addition to the thiol-reversible oxidation of ALDH2, thiol-refractive inactivation was observed, particularly under high-turnover conditions. Organ bath experiments with rat aortas showed that relaxation by GTN lasted longer than that caused by the NO donor diethylamine/NONOate, in line with the long-lasting nanomolar NO generation from GTN observed in VSMCs. Our results suggest that an endogenous reductant with low efficiency allows sustained generation of GTN-derived NO in the low-nanomolar range that is sufficient for vascular relaxation. On a longer time scale, mechanism-based, thiol-refractive irreversible inactivation of ALDH2, and possibly depletion of the endogenous reductant, will render blood vessels tolerant to GTN. Accordingly, full reactivation of oxidized ALDH2 may not occur in vivo and may not be necessary to explain GTN-induced vasodilation.


Aldehyde Dehydrogenase, Mitochondrial/metabolism , Drug Tolerance/physiology , Muscle, Smooth, Vascular/metabolism , Nitrates/metabolism , Nitric Oxide/metabolism , Nitroglycerin/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Cell Line, Transformed , Cell Line, Tumor , Dithiothreitol/pharmacology , Female , Humans , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Nitrates/pharmacology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
8.
Basic Res Cardiol ; 112(5): 55, 2017 08 17.
Article En | MEDLINE | ID: mdl-28819685

Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthases that limits nitric oxide bioavailability. Dimethylarginine dimethylaminohydrolase-1 (DDAH1) exerts a critical role for ADMA degradation and plays an important role in NO signaling. In the heart, DDAH1 is observed in endothelial cells and in the sarcolemma of cardiomyocytes. While NO signaling is important for cardiac adaptation to stress, DDAH1 impact on cardiomyocyte homeostasis is not clear. Here we used the MerCreMer-LoxP model to specifically disrupt cardiomyocyte DDAH1 expression in adult mice to determine the physiological impact of cardiomyocyte DDAH1 under basal conditions and during hypertrophic stress imposed by transverse aortic constriction (TAC). Under control conditions, cardiomyocyte-specific DDAH1 knockout (cDDAH KO) had no detectable effect on plasma ADMA and left ventricular (LV) hypertrophy or function in adult or aging mice. In response to TAC, DDAH1 levels were elevated 2.5-fold in WT mice, which exhibited no change in LV or plasma ADMA content and moderate LV hypertrophy and LV dysfunction. In contrast, cDDAH1 KO mice exposed to TAC showed no increase in LV DDAH1 expression, slightly increased LV tissue ADMA levels, no increase in plasma ADMA, but significantly exacerbated LV hypertrophy, fibrosis, nitrotyrosine production, and LV dysfunction. These findings indicate cardiomyocyte DDAH1 activity is dispensable for cardiac function under basal conditions, but plays an important role in attenuating cardiac hypertrophy and ventricular remodeling under stress conditions, possibly through locally confined regulation of subcellular ADMA and NO signaling.


Amidohydrolases/metabolism , Hypertrophy, Left Ventricular/prevention & control , Myocytes, Cardiac/enzymology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left , Ventricular Remodeling , Amidohydrolases/deficiency , Amidohydrolases/genetics , Animals , Arginine/analogs & derivatives , Arginine/blood , Atrial Natriuretic Factor/metabolism , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Knockout , Myocytes, Cardiac/pathology , Nitric Oxide/metabolism , Phenotype , Signal Transduction , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
9.
J Anal At Spectrom ; 32(2): 393-401, 2017 Feb.
Article En | MEDLINE | ID: mdl-28626281

The determination of the relative isotopic abundance by secondary ion mass spectrometry of 236U in uranium-containing material is complicated by the presence of 235U1H+ ions at the same nominal mass as the uranium isotopic peak. The net intensity of the 236U signal is usually determined by a peak-stripping procedure, whereby the 235U1H+ contribution is obtained by applying the 238U1H+/238U+ ratio to the 235U+ signal. The subtraction of one signal from another has consequences for the uncertainty of the final 236U abundance determination that may be especially significant when the amount of sample is limited, as is the case with small uranium particles that are of great interest for nuclear safeguards. We have developed a model based on Poisson counting statistics to determine the effects of various parameters on the uncertainty of the 236U abundance, including uranium enrichment level, hydride-to-parent ratio, uranium mass consumed during analysis, single versus multichannel counting, and sample substrate composition. The model predictions have been successfully tested against experimental measurements of uranium oxide particles of both 3% and 90% enrichment in 235U.

10.
Basic Res Cardiol ; 112(3): 25, 2017 05.
Article En | MEDLINE | ID: mdl-28349258

Inflammatory responses play an important role in the development of left ventricular (LV) hypertrophy and dysfunction. Recent studies demonstrated that increased T-cell infiltration and T-cell activation contribute to LV hypertrophy and dysfunction. Dendritic cells (DCs) are professional antigen-presenting cells that orchestrate immune responses, especially by modulating T-cell function. In this study, we investigated the role of bone marrow-derived CD11c+ DCs in transverse aortic constriction (TAC)-induced LV fibrosis and hypertrophy in mice. We observed that TAC increased the number of CD11c+ cells and the percentage of CD11c+ MHCII+ (major histocompatibility complex class II molecule positive) DCs in the LV, spleen and peripheral blood in mice. Using bone marrow chimeras and an inducible CD11c+ DC ablation model, we found that depletion of bone marrow-derived CD11c+ DCs significantly attenuated LV fibrosis and hypertrophy in mice exposed to 24 weeks of moderate TAC. CD11c+ DC ablation significantly reduced TAC-induced myocardial inflammation as indicated by reduced myocardial CD45+ cells, CD11b+ cells, CD8+ T cells and activated effector CD8+CD44+ T cells in LV tissues. Moreover, pulsing of autologous DCs with LV homogenates from TAC mice promoted T-cell proliferation. These data indicate that bone marrow-derived CD11c+ DCs play a maladaptive role in hemodynamic overload-induced cardiac inflammation, hypertrophy and fibrosis through the presentation of cardiac self-antigens to T cells.


Dendritic Cells/immunology , Hypertrophy, Left Ventricular/immunology , Lymphocyte Activation/immunology , Ventricular Remodeling/immunology , Animals , Antigen Presentation/immunology , Bone Marrow Cells/immunology , CD11c Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Cardiomegaly/immunology , Disease Models, Animal , Flow Cytometry , Mice , Mice, Inbred C57BL , Myocarditis/immunology
11.
Vasc Med ; 22(3): 179-188, 2017 06.
Article En | MEDLINE | ID: mdl-28145161

Pharmacologic inhibition of nitric oxide production inhibits growth of coronary collateral vessels. Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is the major enzyme that degrades asymmetric dimethylarginine (ADMA), a potent inhibitor of nitric oxide synthase. Here we examined regulation of the ADMA-DDAH1 pathway in a canine model of recurrent myocardial ischemia during the time when coronary collateral growth is known to occur. Under basal conditions, DDAH1 expression was non-uniform across the left ventricular (LV) wall, with expression strongest in the subepicardium. In response to ischemia, DDAH1 expression was up-regulated in the midmyocardium of the ischemic zone, and this was associated with a significant reduction in myocardial interstitial fluid (MIF) ADMA. The decrease in MIF ADMA during ischemia was likely due to increased DDAH1 because myocardial protein arginine N-methyl transferase 1 (PRMT1) and the methylated arginine protein content (the source of ADMA) were unchanged or increased, respectively, at this time. The inflammatory mediators interleukin (IL-1ß) and tumor necrosis factor (TNF-α) were also elevated in the midmyocardium where DDAH1 expression was increased. Both of these factors significantly up-regulated DDAH1 expression in cultured human coronary artery endothelial cells. Taken together, these results suggest that inflammatory factors expressed in response to myocardial ischemia contributed to up-regulation of DDAH1, which was responsible for the decrease in MIF ADMA.


Amidohydrolases/metabolism , Coronary Vessels/enzymology , Myocardial Ischemia/enzymology , Myocardium/enzymology , Neovascularization, Physiologic , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Cell Hypoxia , Cells, Cultured , Collateral Circulation , Coronary Circulation , Coronary Vessels/physiopathology , Disease Models, Animal , Dogs , Endothelial Cells/enzymology , Humans , Interleukin-1beta/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/pathology , Nitric Oxide Synthase Type II/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
12.
Circulation ; 134(18): 1353-1368, 2016 Nov 01.
Article En | MEDLINE | ID: mdl-27678262

BACKGROUND: The recruitment of leukocytes to the vascular wall is a key step in hypertension development. Chemokine receptor CXCR2 mediates inflammatory cell chemotaxis in several diseases. However, the role of CXCR2 in hypertension development and the underlying mechanisms remain unknown. METHODS: Angiotensin II (490 ng·kg-1·min-1) or deoxycorticosterone acetate (DOCA) salt-induced mouse hypertensive models in genetic ablation, pharmacologic inhibition of CXCR2, and adoptive bone marrow transfer mice were used to determine the role of CXCR2 in hypertension (measured by radiotelemetry and tail-cuff system), inflammation (verified by flow cytometry and quantitative real-time polymerase chain reaction [PCR] analysis), vascular remodeling (studied by haematoxylin and eosin and Masson's trichrome staining), vascular dysfunction (assessed by aortic ring), and oxidative stress (indicated by nicotinamide adenine dinucleotide phosphate [NADPH] oxidase activity, dihydroethidium staining, and quantitative real-time PCR analysis). Moreover, the blood CXCR2+ cells in normotensive controls and hypertension patients were analyzed by flow cytometry. RESULTS: Angiotensin II significantly upregulated the expression of CXCR2 mRNA and protein and increased the number of CD45+ CXCR2+ cells in mouse aorta (n=8 per group). Selective CXCR2 knockout (CXCR2-/-) or pharmacological inhibition of CXCR2 markedly reduced angiotensin II- or DOCA-salt-induced blood pressure elevation, aortic thickness and collagen deposition, accumulation of proinflammatory cells into the vascular wall, and expression of cytokines (n=8 per group). CXCR2 inhibition also ameliorated angiotensin II-induced vascular dysfunction and reduced vascular superoxide formation, NADPH activity, and expression of NADPH oxidase subunits (n=6 per group). Bone marrow reconstitution of wild-type mice with CXCR2-/- bone marrow cells also significantly abolished angiotensin II-induced responses (n=6 per group). It is important to note that CXCR2 blockade reversed established hypertension induced by angiotensin II or DOCA-salt challenge (n=10 per group). Furthermore, we demonstrated that CXCR2+ proinflammatory cells were higher in hypertensive patients (n=30) compared with normotensive individuals (n=20). CONCLUSIONS: Infiltration of CXCR2+ cells plays a pathogenic role in arterial hypertension and vascular dysfunction. Inhibition of CXCR2 pathway may represent a novel therapeutic approach to treat hypertension.


Angiotensin II/pharmacology , Hypertension/prevention & control , Receptors, Interleukin-8B/biosynthesis , Up-Regulation/drug effects , Vascular Remodeling/drug effects , Animals , Disease Models, Animal , Hypertension/genetics , Hypertension/metabolism , Male , Mice , Mice, Knockout , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/genetics , Up-Regulation/genetics , Vascular Remodeling/genetics
13.
J Biol Chem ; 291(46): 24076-24084, 2016 Nov 11.
Article En | MEDLINE | ID: mdl-27679490

Aldehyde dehydrogenase-2 (ALDH2) catalyzes vascular bioactivation of the antianginal drug nitroglycerin (GTN), resulting in activation of soluble guanylate cyclase (sGC) and cGMP-mediated vasodilation. We have previously shown that a minor reaction of ALDH2-catalyzed GTN bioconversion, accounting for about 5% of the main clearance-based turnover yielding inorganic nitrite, results in direct NO formation and concluded that this minor pathway could provide the link between vascular GTN metabolism and activation of sGC. However, lack of detectable NO at therapeutically relevant GTN concentrations (≤1 µm) in vascular tissue called into question the biological significance of NO formation by purified ALDH2. We addressed this issue and used a novel, highly sensitive genetically encoded fluorescent NO probe (geNOp) to visualize intracellular NO formation at low GTN concentrations (≤1 µm) in cultured vascular smooth muscle cells (VSMC) expressing an ALDH2 mutant that reduces GTN to NO but lacks clearance-based GTN denitration activity. NO formation was compared with GTN-induced activation of sGC. The addition of 1 µm GTN to VSMC expressing either wild-type or C301S/C303S ALDH2 resulted in pronounced intracellular NO elevation, with maximal concentrations of 7 and 17 nm, respectively. Formation of GTN-derived NO correlated well with activation of purified sGC in VSMC lysates and cGMP accumulation in intact porcine aortic endothelial cells infected with wild-type or mutant ALDH2. Formation of NO and cGMP accumulation were inhibited by ALDH inhibitors chloral hydrate and daidzin. The present study demonstrates that ALDH2-catalyzed NO formation is necessary and sufficient for GTN bioactivation in VSMC.


Aldehyde Dehydrogenase, Mitochondrial/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Nitric Oxide/metabolism , Nitroglycerin/pharmacokinetics , Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial/genetics , Amino Acid Substitution , Animals , Cattle , Chloral Hydrate/pharmacology , Humans , Isoflavones/pharmacology , Mice , Mice, Knockout , Mutation, Missense , Nitroglycerin/pharmacology , Swine
14.
Hypertension ; 68(3): 688-96, 2016 09.
Article En | MEDLINE | ID: mdl-27432861

The inflammatory response regulates congestive heart failure (CHF) development. T cell activation plays an important role in tissue inflammation. We postulate that CD28 or B7 deficiency inhibits T cell activation and attenuates CHF development by reducing systemic, cardiac, and pulmonary inflammation. We demonstrated that chronic pressure overload-induced end-stage CHF in mice is characterized by profound accumulation of activated effector T cells (CD3(+)CD44(high) cells) in the lungs and a mild but significant increase of these cells in the heart. In knockout mice lacking either CD28 or B7, there was a dramatic reduction in the accumulation of activated effector T cells in both hearts and lungs of mice under control conditions and after transverse aortic constriction. CD28 or B7 knockout significantly attenuated transverse aortic constriction-induced CHF development, as indicated by less increase of heart and lung weight and less reduction of left ventricle contractility. CD28 or B7 knockout also significantly reduced transverse aortic constriction-induced CD45(+) leukocyte, T cell, and macrophage infiltration in hearts and lungs, lowered proinflammatory cytokine expression (such as tumor necrosis factor-α and interleukin-1ß) in lungs. Furthermore, CD28/B7 blockade by CTLA4-Ig treatment (250 µg/mouse every 3 days) attenuated transverse aortic constriction-induced T cell activation, left ventricle hypertrophy, and left ventricle dysfunction. Our data indicate that CD28/B7 deficiency inhibits activated effector T cell accumulation, reduces myocardial and pulmonary inflammation, and attenuates the development of CHF. Our findings suggest that strategies targeting T cell activation may be useful in treating CHF.


Abatacept/pharmacology , B7 Antigens/metabolism , CD28 Antigens/metabolism , Heart Failure/physiopathology , Pneumonia/physiopathology , Analysis of Variance , Animals , B7 Antigens/immunology , CD28 Antigens/immunology , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Heart Failure/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , Random Allocation , Statistics, Nonparametric , Systole/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
16.
Hypertension ; 68(1): 114-22, 2016 07.
Article En | MEDLINE | ID: mdl-27160197

Congestive heart failure (CHF) is associated with an increase of leukocyte infiltration, proinflammatory cytokines, and fibrosis in the heart and lung. Regulatory T cells (Tregs, CD4(+)CD25(+)FoxP3(+)) suppress inflammatory responses in various clinical conditions. We postulated that expansion of Tregs attenuates CHF progression by reducing cardiac and lung inflammation. We investigated the effects of interleukin-2 (IL-2) plus IL-2 monoclonal antibody clone JES6-1 complexes (IL2/JES6-1) on induction of Tregs, transverse aortic constriction-induced cardiac and lung inflammation, and CHF progression in mice. We demonstrated that end-stage CHF caused a massive increase of lung macrophages and T cells, as well as relatively mild left ventricular (LV) leukocyte infiltration. Administration of IL2/JES6-1 caused an ≈6-fold increase of Tregs within CD4(+) T cells in the spleen, lung, and heart of mice. IL2/JES6-1 treatment of mice with existing transverse aortic constriction-induced LV failure markedly reduced lung and right ventricular weight and improved LV ejection fraction and LV end-diastolic pressure. Mechanistically, IL2/JES6-1 treatment significantly increased Tregs; suppressed CD4(+) T-cell accumulation; dramatically attenuated leukocyte infiltration, including decreasing CD45(+) cells, macrophages, CD8(+) T cells, and effector memory CD8(+); and reduced proinflammatory cytokine expressions and fibrosis in the lung of mice. Furthermore, IL2/JES6-1 administered before transverse aortic constriction attenuated the development of LV hypertrophy and dysfunction in mice. Our data indicate that increasing Tregs through administration of IL2/JES6-1 effectively attenuates pulmonary inflammation, right ventricular hypertrophy, and further LV dysfunction in mice with existing LV failure, suggesting that strategies to properly expand Tregs may be useful in reducing CHF progression.


Antibodies, Monoclonal/pharmacology , Heart Failure/drug therapy , Interleukin-2/pharmacology , Pneumonia/drug therapy , Pneumonia/immunology , Analysis of Variance , Animals , Antigen-Antibody Complex/pharmacology , Cytokines/analysis , Disease Models, Animal , Disease Progression , Heart Failure/immunology , Heart Failure/physiopathology , Interleukin-2/immunology , Male , Mice , Mice, Inbred BALB C , Pneumonia/physiopathology , Random Allocation , Reference Values , Risk Assessment , Severity of Illness Index , Statistics, Nonparametric , T-Lymphocytes, Regulatory/immunology , Treatment Outcome , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/immunology , Ventricular Dysfunction, Left/physiopathology
17.
Nitric Oxide ; 54: 73-81, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26923818

Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthases that limits nitric oxide bioavailability and can increase production of NOS derived reactive oxidative species. Increased plasma ADMA is a one of the strongest predictors of mortality in patients who have had a myocardial infarction or suffer from chronic left heart failure, and is also an independent risk factor for several other conditions that contribute to heart failure development, including hypertension, coronary artery disease/atherosclerosis, diabetes, and renal dysfunction. The enzyme responsible for ADMA degradation is dimethylarginine dimethylaminohydrolase-1 (DDAH1). DDAH1 plays an important role in maintaining nitric oxide bioavailability and preserving cardiovascular function in the failing heart. Here, we examine mechanisms of abnormal NO production in heart failure, with particular focus on the role of ADMA and DDAH1.


Arginine/analogs & derivatives , Heart Failure/metabolism , Nitric Oxide/biosynthesis , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Arginine/metabolism , Heart Failure/etiology , Humans , Nitric Oxide Synthase/antagonists & inhibitors , Peroxynitrous Acid/metabolism , Signal Transduction , Superoxides/metabolism , omega-N-Methylarginine/metabolism
18.
Nitric Oxide ; 54: 8-14, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26805578

Scavenging of nitric oxide (NO) often interferes with studies on NO signaling in cell-free preparations. We observed that formation of cGMP by NO-stimulated purified soluble guanylate cyclase (sGC) was virtually abolished in the presence of cytosolic preparations of porcine coronary arteries, with the scavenging activity localized in the tunica media (smooth muscle layer). Electrochemical measurement of NO release from a donor compound and light absorbance spectroscopy showed that cytosolic preparations contained a reduced heme protein that scavenged NO. This protein, which reacted with anti-human hemoglobin antibodies, was efficiently removed from the preparations by haptoglobin affinity chromatography. The cleared cytosols showed only minor scavenging of NO according to electrochemical measurements and did not decrease cGMP formation by NO-stimulated sGC. In contrast, the column flow-through caused a nearly 2-fold increase of maximal sGC activity (from 33.1 ± 1.6 to 54.9 ± 2.2 µmol × min(-1) × mg(-1)). The proteins retained on the affinity column were identified as hemoglobin α and ß subunits. The results indicate that hemoglobin, presumably derived from vasa vasorum erythrocytes, is present and scavenges NO in preparations of porcine coronary artery smooth muscle. Selective removal of hemoglobin-mediated scavenging unmasked stimulation of maximal NO-stimulated sGC activity by a soluble factor expressed in vascular tissue.


Coronary Vessels/metabolism , Hemoglobins/metabolism , Nitric Oxide/metabolism , Tunica Media/metabolism , Animals , Cattle , Cyclic GMP/metabolism , Cytoglobin , Globins/metabolism , Haptoglobins/metabolism , Humans , In Vitro Techniques , Soluble Guanylyl Cyclase/metabolism , Swine
19.
Hypertension ; 64(4): 738-44, 2014 Oct.
Article En | MEDLINE | ID: mdl-24958502

Studies have reported that development of congestive heart failure is associated with increased endoplasmic reticulum stress. Double stranded RNA-activated protein kinase R-like endoplasmic reticulum kinase (PERK) is a major transducer of the endoplasmic reticulum stress response and directly phosphorylates eukaryotic initiation factor 2α, resulting in translational attenuation. However, the physiological effect of PERK on congestive heart failure development is unknown. To study the effect of PERK on ventricular structure and function, we generated inducible cardiac-specific PERK knockout mice. Under unstressed conditions, cardiac PERK knockout had no effect on left ventricular mass, or its ratio to body weight, cardiomyocyte size, fibrosis, or left ventricular function. However, in response to chronic transverse aortic constriction, PERK knockout mice exhibited decreased ejection fraction, increased left ventricular fibrosis, enhanced cardiomyocyte apoptosis, and exacerbated lung remodeling in comparison with wild-type mice. PERK knockout also dramatically attenuated cardiac sarcoplasmic reticulum Ca(2+)-ATPase expression in response to aortic constriction. Our findings suggest that PERK is required to protect the heart from pressure overload-induced congestive heart failure.


Endoplasmic Reticulum Stress , Heart Failure/physiopathology , Lung/physiopathology , eIF-2 Kinase/metabolism , Animals , Aorta/physiopathology , Apoptosis , Blotting, Western , Calcium-Transporting ATPases/metabolism , Cardiomegaly/physiopathology , Constriction , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Female , Fibrosis , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Lung/pathology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Phosphorylation , Pressure , Sarcoplasmic Reticulum/enzymology , Transcription Factor CHOP/metabolism , Ventricular Dysfunction, Left/physiopathology , Weight-Bearing , eIF-2 Kinase/genetics
20.
Circulation ; 129(13): 1397-406, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24463368

BACKGROUND: Double-stranded RNA-dependent protein kinase (PKR) is a eukaryotic initiation factor 2α kinase that inhibits mRNA translation under stress conditions. PKR also mediates inflammatory and apoptotic signaling independently of translational regulation. Congestive heart failure is associated with cardiomyocyte hypertrophy, inflammation, and apoptosis, but the role of PKR in left ventricular hypertrophy and the development of congestive heart failure has not been examined. METHODS AND RESULTS: We observed increased myocardial PKR expression and translocation of PKR into the nucleus in humans and mice with congestive heart failure. To determine the impact of PKR on the development of congestive heart failure, PKR knockout and wild-type mice were exposed to pressure overload produced by transverse aortic constriction. Although heart size increased similarly in wild-type and PKR knockout mice after transverse aortic constriction, PKR knockout mice exhibited very little pulmonary congestion, well-preserved left ventricular ejection fraction and contractility, and significantly less myocardial fibrosis compared with wild-type mice. Bone marrow-derived cells from wild-type mice did not abolish the cardiac protective effect observed in PKR knockout mice, whereas bone marrow-derived cells from PKR knockout mice had no cardiac protective effect in wild-type mice. Mechanistically, PKR knockout attenuated transverse aortic constriction-induced tumor necrosis factor-α expression and leukocyte infiltration and lowered cardiac expression of proapoptotic factors (Bax and caspase-3), so that PKR knockout hearts were more resistant to transverse aortic constriction-induced cardiomyocyte apoptosis. PKR depletion in isolated cardiomyocytes also conferred protection against tumor necrosis factor-α- or lipopolysaccharide-induced apoptosis. CONCLUSION: PKR is a maladaptive factor upregulated in hemodynamic overload that contributes to myocardial inflammation, cardiomyocyte apoptosis, and the development of congestive heart failure.


Blood Pressure/physiology , Heart Failure/prevention & control , Heart Failure/physiopathology , Hemodynamics/physiology , Ventricular Dysfunction, Left/prevention & control , eIF-2 Kinase/deficiency , Adult , Aged , Animals , Aorta/physiopathology , Apoptosis/physiology , Cytokines/metabolism , Disease Models, Animal , Female , Heart Failure/metabolism , Humans , Hypertrophy/physiopathology , Hypertrophy/prevention & control , Male , Mice , Mice, Knockout , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Up-Regulation/physiology , eIF-2 Kinase/genetics , eIF-2 Kinase/physiology
...