267 results on '"Bache RJ"'
Search Results
2. Bioenergetic and functional consequences of bone marrow-derived multipotent progenitor cell transplantation in hearts with postinfarction left ventricular remodeling.
- Author
-
Zeng L, Hu Q, Wang X, Mansoor A, Lee J, Feygin J, Zhang G, Suntharalingam P, Boozer S, Mhashilkar A, Panetta CJ, Swingen C, Deans R, From AH, Bache RJ, Verfaillie CM, and Zhang J
- Published
- 2007
3. Active compression-decompression CPR improves vital organ perfusion in a dog model of ventricular fibrillation: Chest 1994; 106/4: 1250–1259
- Author
-
Chang, MW, Coffeen, P, Lurie, KG, Shultz, J, Bache, RJ, and White, CW
- Published
- 1995
- Full Text
- View/download PDF
4. Adenosine kinase attenuates cardiomyocyte microtubule stabilization and protects against pressure overload-induced hypertrophy and LV dysfunction.
- Author
-
Fassett J, Xu X, Kwak D, Zhu G, Fassett EK, Zhang P, Wang H, Mayer B, Bache RJ, and Chen Y
- Subjects
- 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, Adenosine Kinase metabolism, Cardiomegaly metabolism, MAP Kinase Signaling System, Microtubules metabolism, Myocytes, Cardiac metabolism, Ventricular Dysfunction, Left metabolism
- Abstract
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 β-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 β-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., (Copyright © 2019. Published by Elsevier Ltd.)- Published
- 2019
- Full Text
- View/download PDF
5. Hints of Lyme: Disseminated Borreliosis Involving the Mitral and Tricuspid Valves.
- Author
-
Maheshwari A and Bache RJ
- Subjects
- Adult, Anti-Bacterial Agents therapeutic use, Atrioventricular Block etiology, Ceftriaxone therapeutic use, Cystic Fibrosis, Endocarditis, Bacterial diagnosis, Endocarditis, Bacterial pathology, Humans, Lung Transplantation, Lyme Disease drug therapy, Male, Endocarditis, Bacterial microbiology, Lyme Disease complications, Mitral Valve microbiology, Tricuspid Valve microbiology
- Published
- 2017
- Full Text
- View/download PDF
6. Repetitive ischemia increases myocardial dimethylarginine dimethylaminohydrolase 1 expression.
- Author
-
Zhang P, Fassett JT, Zhu G, Li J, Hu X, Xu X, Chen Y, and Bache RJ
- Subjects
- 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, Amidohydrolases metabolism, Coronary Vessels enzymology, Myocardial Ischemia enzymology, Myocardium enzymology, Neovascularization, Physiologic
- Abstract
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-1β) 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.
- Published
- 2017
- Full Text
- View/download PDF
7. Role of bone marrow-derived CD11c + dendritic cells in systolic overload-induced left ventricular inflammation, fibrosis and hypertrophy.
- Author
-
Wang H, Kwak D, Fassett J, Liu X, Yao W, Weng X, Xu X, Xu Y, Bache RJ, Mueller DL, and Chen Y
- Subjects
- 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, Dendritic Cells immunology, Hypertrophy, Left Ventricular immunology, Lymphocyte Activation immunology, Ventricular Remodeling immunology
- Abstract
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.- Published
- 2017
- Full Text
- View/download PDF
8. A Novel Method for Assessing Cardiac Output With the Use of Oxygen Circulation Time.
- Author
-
Kwon Y, Van't Hof J, Roy SS, Bache RJ, and Das G
- Subjects
- Aged, Blood Circulation Time methods, Blood Flow Velocity physiology, Cohort Studies, Female, Humans, Male, Middle Aged, Prospective Studies, Sensitivity and Specificity, Severity of Illness Index, Thermodilution methods, Cardiac Catheterization methods, Cardiac Output physiology, Heart Failure diagnosis, Oxygen Consumption physiology
- Abstract
Background: We investigated whether a simple breath hold would yield dynamic oxygen (O
2 ) saturation change and whether the derived circulation time would be useful in assessing cardiac function., Methods and Results: Patients undergoing right heart catheterization for clinical indications (n = 48), including heart failure (HF; n = 24), were prospectively recruited. Each subject was instructed to hold their breath for 20-40 seconds. Lung to finger circulation time (LFCT), defined as the time from the point of rebreathing to nadir O2 desaturation, was correlated with cardiac output. Among 48 subjects recruited, 37 manifested ≥3% O2 desaturation allowing for an LFCT measurement. Mean LFCT was 38.5 ± 17.5 seconds (range 18.9-94.7 s). LFCT in patients with a clinical diagnosis of HF was significantly longer than those without (45.9 ± 19.9 s vs 31.5 ± 11.5 s; P = .01). Overall, the LFCT was inversely correlated with cardiac output (Fick: r = -0.56; P < .001 [n = 37]; thermodilution: r = -0.6; P = .001 [n = 27])., Conclusions: LFCT is prolonged in low cardiac output. LFCT is a novel method that may be useful to noninvasively assess cardiac function in HF., (Copyright © 2015 Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
9. CD28/B7 Deficiency Attenuates Systolic Overload-Induced Congestive Heart Failure, Myocardial and Pulmonary Inflammation, and Activated T Cell Accumulation in the Heart and Lungs.
- Author
-
Wang H, Kwak D, Fassett J, Hou L, Xu X, Burbach BJ, Thenappan T, Xu Y, Ge JB, Shimizu Y, Bache RJ, and Chen Y
- Subjects
- 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, Abatacept pharmacology, B7 Antigens metabolism, CD28 Antigens metabolism, Heart Failure physiopathology, Pneumonia physiopathology
- Abstract
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-1β) 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., (© 2016 American Heart Association, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
10. Increasing Regulatory T Cells With Interleukin-2 and Interleukin-2 Antibody Complexes Attenuates Lung Inflammation and Heart Failure Progression.
- Author
-
Wang H, Hou L, Kwak D, Fassett J, Xu X, Chen A, Chen W, Blazar BR, Xu Y, Hall JL, Ge JB, Bache RJ, and Chen Y
- Subjects
- 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, Antibodies, Monoclonal pharmacology, Heart Failure drug therapy, Interleukin-2 pharmacology, Pneumonia drug therapy, Pneumonia immunology
- Abstract
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., (© 2016 American Heart Association, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
11. ATP sensitive K(+) channels are critical for maintaining myocardial perfusion and high energy phosphates in the failing heart.
- Author
-
Jameel MN, Xiong Q, Mansoor A, Bache RJ, and Zhang J
- Subjects
- Adenosine Triphosphate metabolism, Animals, Coronary Circulation drug effects, Disease Models, Animal, Dogs, Heart Failure metabolism, Heart Failure pathology, Humans, Mitochondria drug effects, Mitochondria pathology, Myocardial Ischemia drug therapy, Myocardial Ischemia metabolism, Myocardial Ischemia pathology, Myocardium pathology, Oxidative Phosphorylation drug effects, Oxygen Consumption drug effects, Potassium Channels drug effects, Receptors, Purinergic P1 drug effects, Receptors, Purinergic P1 metabolism, Vasodilation drug effects, Glyburide administration & dosage, Heart Failure drug therapy, Mitochondria metabolism, Myocardium metabolism, Potassium Channels metabolism
- Abstract
Congestive heart failure (CHF) is associated with intrinsic alterations of mitochondrial oxidative phosphorylation which lead to increased myocardial cytosolic free ADP. ATP sensitive K(+) channels (KATP) act as metabolic sensors that are important for maintaining coronary blood flow (MBF) and in mediating the response of the myocardium to stress. Coronary adenosine receptors (AdR) are not normally active but cause vasodilation during myocardial ischemia. This study examined the myocardial energetic response to inhibition of KATP and AdR in CHF. CHF (as evidenced by LVEDP>20mmHg) was produced in adult mongrel dogs (n=12) by rapid ventricular pacing for 4weeks. MBF was measured with radiolabeled microspheres during baseline (BL), AdR blockade with 8-phenyltheophylline (8-PT; 5mg/kg iv), and KATP blockade with glibenclamide (GLB; 20μg/kg/min ic). High energy phosphates were examined with (31)P magnetic resonance spectroscopy (MRS) while myocardial oxygenation was assessed from the deoxymyoglobin signal (Mb-δ) using (1)H MRS. During basal conditions the phosphocreatine (PCr)/ATP ratio (1.73±0.15) was significantly lower than in previously studied normal dogs (2.42±0.11) although Mb-δ was undetectable. 8-PT caused ≈21% increase in MBF with no change in PCr/ATP. GLB caused a 33±0.1% decrease in MBF with a decrease in PCr/ATP from 1.65±0.17 to 1.11±0.11 (p<0.0001). GLB did not change the pseudo-first-order rate constant of ATP production via CK (kf), but the ATP production rate via CK was reduced by 35±0.08%; this was accompanied by an increase in Pi/PCr and appearance of a Mb-δ signal indicating tissue hypoxia. Thus, in the failing heart the balance between myocardial ATP demands and oxygen delivery is critically dependent on functioning KATP channels., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
12. Endoplasmic reticulum stress sensor protein kinase R-like endoplasmic reticulum kinase (PERK) protects against pressure overload-induced heart failure and lung remodeling.
- Author
-
Liu X, Kwak D, Lu Z, Xu X, Fassett J, Wang H, Wei Y, Cavener DR, Hu X, Hall J, Bache RJ, and Chen Y
- Subjects
- 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, Endoplasmic Reticulum Stress, Heart Failure physiopathology, Lung physiopathology, eIF-2 Kinase metabolism
- Abstract
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., (© 2014 American Heart Association, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
13. NOX2-induced myocardial fibrosis and diastolic dysfunction: role of the endothelium.
- Author
-
Bache RJ and Chen Y
- Subjects
- Animals, Humans, Male, NADPH Oxidase 2, Cardiomegaly enzymology, Endothelium, Vascular enzymology, Inflammation Mediators physiology, Membrane Glycoproteins physiology, Mesenchymal Stem Cells enzymology, NADPH Oxidases physiology, Ventricular Dysfunction, Left enzymology
- Published
- 2014
- Full Text
- View/download PDF
14. Metformin protects against systolic overload-induced heart failure independent of AMP-activated protein kinase α2.
- Author
-
Xu X, Lu Z, Fassett J, Zhang P, Hu X, Liu X, Kwak D, Li J, Zhu G, Tao Y, Hou M, Wang H, Guo H, Viollet B, McFalls EO, Bache RJ, and Chen Y
- Subjects
- AMP-Activated Protein Kinases deficiency, AMP-Activated Protein Kinases genetics, Animals, Aorta physiopathology, Disease Models, Animal, Hypertrophy, Left Ventricular physiopathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction physiology, Stroke Volume physiology, Vasoconstriction physiology, AMP-Activated Protein Kinases physiology, Heart Failure, Systolic physiopathology, Heart Failure, Systolic prevention & control, Hypoglycemic Agents therapeutic use, Metformin therapeutic use
- Abstract
Activation of AMP-activated protein kinase (AMPK)-α2 protects the heart against pressure overload-induced heart failure in mice. Although metformin is a known activator of AMPK, it is unclear whether its cardioprotection acts independently of an AMPKα2-dependent pathway. Because the role of AMPKα1 stimulation on remodeling of failing hearts is poorly defined, we first studied the effects of disruption of both the AMPKα1 and AMPKα2 genes on the response to transverse aortic constriction-induced left ventricular (LV) hypertrophy and dysfunction in mice. AMPKα2 gene knockout significantly exacerbated the degree of transverse aortic constriction-induced LV hypertrophy and dysfunction, whereas AMPKα1 gene knockout had no effect on the degree of transverse aortic constriction-induced LV hypertrophy and dysfunction. Administration of metformin was equally effective in attenuating transverse aortic constriction-induced LV remodeling in both wild-type and AMPKα2 knockout mice, as evidenced by reduced LV and lung weights, a preserved LV ejection fraction, and reduced phosphorylation of mammalian target of rapamycin (p-mTOR(Ser2448)) and its downstream target p-p70S6K(Thr389). These data support the notion that activation of AMPKα1 plays a negligible role in protecting the heart against the adverse effects of chronic pressure overload, and that metformin protects against adverse remodeling through a pathway that seems independent of AMPKα2.
- Published
- 2014
- Full Text
- View/download PDF
15. Double-stranded RNA-dependent protein kinase deficiency protects the heart from systolic overload-induced congestive heart failure.
- Author
-
Wang H, Xu X, Fassett J, Kwak D, Liu X, Hu X, Falls TJ, Bell JC, Li H, Bitterman P, Bache RJ, and Chen Y
- Subjects
- 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, Blood Pressure physiology, Heart Failure physiopathology, Heart Failure prevention & control, Hemodynamics physiology, Ventricular Dysfunction, Left prevention & control, eIF-2 Kinase deficiency
- Abstract
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.
- Published
- 2014
- Full Text
- View/download PDF
16. Inducible nitric oxide synthase inhibits oxygen consumption in collateral-dependent myocardium.
- Author
-
Chen Y, Zhang P, Li J, Xu X, and Bache RJ
- Subjects
- Animals, Coronary Circulation, Coronary Vessels metabolism, Coronary Vessels physiology, Dogs, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide Synthase Type II antagonists & inhibitors, Oxygen metabolism, Vasoconstriction, Myocardium metabolism, Nitric Oxide Synthase Type II metabolism, Oxygen Consumption
- Abstract
Following coronary artery occlusion growth of collateral vessels can provide an effective blood supply to the dependent myocardium. The ischemia, which results in growth of collateral vessels, recruits an inflammatory response with expression of cytokines and growth factors, upregulation of endothelial nitric oxide (NO) synthase (eNOS) in vascular endothelial cells, and expression of inducible nitric oxide synthase (iNOS) in both vessels and cardiac myocytes. Because NO is a potent collateral vessel dilator, this study examined whether NO derived from iNOS or constitutive NOS regulates myocardial blood flow (MBF) in the collateral region. Nonselective NOS inhibition with N(G)-nitro-l-arginine (LNA) caused vasoconstriction with a significant decrease in MBF to the collateral region during exercise. In contrast, the highly selective iNOS inhibitor 1400W caused a 21 ± 5% increase of MBF in the collateral region. This increase in MBF following selective iNOS blockade was proportionate to an increase in myocardial O2 consumption (MVo2). The results suggest that NO produced by iNOS inhibits MVo2 in the collateralized region, so that the increase in MBF following iNOS blockade was the result of metabolic vasodilation secondary to an increase in MVo2. Thus the coordinated expression of iNOS to restrain MVo2 and eNOS to maintain collateral vasodilation act to optimize the O2 supply-demand relationship and protect the collateralized myocardium from ischemia.
- Published
- 2014
- Full Text
- View/download PDF
17. Loss of the eukaryotic initiation factor 2α kinase general control nonderepressible 2 protects mice from pressure overload-induced congestive heart failure without affecting ventricular hypertrophy.
- Author
-
Lu Z, Xu X, Fassett J, Kwak D, Liu X, Hu X, Wang H, Guo H, Xu D, Yan S, McFalls EO, Lu F, Bache RJ, and Chen Y
- Subjects
- Animals, Apoptosis, Cells, Cultured, Heart Failure etiology, Lung Diseases etiology, Lung Diseases metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocytes, Cardiac metabolism, Oxidative Stress, Phosphorylation, Protein Serine-Threonine Kinases metabolism, Sarcoplasmic Reticulum Calcium-Transporting ATPases biosynthesis, Heart Failure metabolism, Hypertrophy, Left Ventricular metabolism, Protein Serine-Threonine Kinases biosynthesis
- Abstract
In response to several stresses, including nutrient deprivation, general control nonderepressible 2 kinase (GCN2) attenuates mRNA translation by phosphorylating eukaryotic initiation factor 2α(Ser51). Energy starvation is known to exacerbate congestive heart failure, and eukaryotic initiation factor 2α(Ser51) phosphorylation is increased in the failing heart. However, the effect of GCN2 during the evolution of congestive heart failure has not been tested. In this study, we examined the influence of GCN2 expression in response to a cardiac stress by inducing chronic pressure overload with transverse aortic constriction in wild-type and GCN2 knockout mice. Under basal conditions, GCN2 knockout mice had normal left ventricular structure and function, but after transverse aortic constriction, they demonstrated less contractile dysfunction, less increase in lung weight, less increase in lung inflammation and vascular remodeling, and less myocardial apoptosis and fibrosis compared with wild-type mice, despite an equivalent degree of left ventricular hypertrophy. As expected, GCN2 knockout attenuated transverse aortic constriction-induced cardiac eukaryotic initiation factor 2α(Ser51) phosphorylation and preserved sarcoplasmic reticulum Ca(2+) ATPase expression compared with wild-type mice. Interestingly, the expression of the antiapoptotic protein Bcl-2 was significantly elevated in GCN2 knockout hearts, whereas in isolated neonatal cardiomyocytes, selective knockdown of GCN2 increased Bcl-2 protein expression and enhanced myocyte resistance to an apoptotic stress. Collectively, our data support the notion that GCN2 impairs the ventricular adaptation to chronic pressure overload by reducing Bcl-2 expression and increasing cardiomyocyte susceptibility to apoptotic stimuli. Our findings suggest that strategies to reduce GCN2 activity in cardiac tissue may be a novel approach to attenuate congestive heart failure development.
- Published
- 2014
- Full Text
- View/download PDF
18. DDAH1 deficiency attenuates endothelial cell cycle progression and angiogenesis.
- Author
-
Zhang P, Xu X, Hu X, Wang H, Fassett J, Huo Y, Chen Y, and Bache RJ
- Subjects
- Amidohydrolases genetics, Animals, Blotting, Western, Cell Cycle genetics, Cells, Cultured, Cyclin D1 metabolism, Cyclin E metabolism, Flow Cytometry, Human Umbilical Vein Endothelial Cells metabolism, Humans, Mice, Mice, Knockout, Neovascularization, Physiologic genetics, RNA, Small Interfering, Amidohydrolases deficiency, Amidohydrolases metabolism, Cell Cycle physiology, Endothelial Cells cytology, Endothelial Cells metabolism, Neovascularization, Physiologic physiology
- Abstract
Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide (NO) synthase (NOS). ADMA is eliminated largely by the action of dimethylarginine dimethylaminohydrolase1 (DDAH1). Decreased DDAH activity is found in several pathological conditions and is associated with increased risk of vascular disease. Overexpression of DDAH1 has been shown to augment endothelial proliferation and angiogenesis. To better understand the mechanism by which DDAH1 influences endothelial proliferation, this study examined the effect of DDAH1 deficiency on cell cycle progression and the expression of some cell cycle master regulatory proteins. DDAH1 KO decreased in vivo Matrigel angiogenesis and depressed endothelial repair in a mouse model of carotid artery wire injury. DDAH1 deficiency decreased VEGF expression in HUVEC and increased NF1 expression in both HUVEC and DDAH1 KO mice. The expression of active Ras could overcome the decreased VEGF expression caused by the DDAH1 depletion. The addition of VEGF and knockdown NF1 could both restore proliferation in cells with DDAH1 depletion. Flow cytometry analysis revealed that DDAH1 sRNAi knockdown in HUVEC caused G1 and G2/M arrest that was associated with decreased expression of CDC2, CDC25C, cyclin D1 and cyclin E. MEF cells from DDAH1 KO mice also demonstrated G2/M arrest that was associated with decreased cyclin D1 expression and Akt activity. Our findings indicate that DDAH1 exerts effects on cyclin D1 and cyclin E expression through multiple mechanisms, including VEGF, the NO/cGMP/PKG pathway, the Ras/PI3K/Akt pathway, and NF1 expression. Loss of DDAH1 effects on these pathways results in impaired endothelial cell proliferation and decreased angiogenesis. The findings provide background information that may be useful in the development of therapeutic strategies to manipulate DDAH1 expression in cardiovascular diseases or tumor angiogenesis.
- Published
- 2013
- Full Text
- View/download PDF
19. Microtubule Actin Cross-linking Factor 1 regulates cardiomyocyte microtubule distribution and adaptation to hemodynamic overload.
- Author
-
Fassett JT, Xu X, Kwak D, Wang H, Liu X, Hu X, Bache RJ, and Chen Y
- Subjects
- Animals, Base Sequence, Caveolin 3 physiology, DNA Primers, Echocardiography, Integrin beta1 physiology, Lung physiopathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Kinase C-alpha physiology, Rats, Rats, Sprague-Dawley, Real-Time Polymerase Chain Reaction, Ventricular Dysfunction, Left, Adaptation, Physiological, Hemodynamics, Microfilament Proteins physiology, Microtubules physiology, Myocytes, Cardiac physiology
- Abstract
Aberrant cardiomyocyte microtubule growth is a feature of pressure overload induced cardiac hypertrophy believed to contribute to left ventricular (LV) dysfunction. Microtubule Actin Cross-linking Factor 1 (MACF1/Acf7) is a 600 kd spectraplakin that stabilizes and guides microtubule growth along actin filaments. MACF1 is expressed in the heart, but its impact on cardiac microtubules, and how this influences cardiac structure, function, and adaptation to hemodynamic overload is unknown. Here we used inducible cardiac-specific MACF1 knockout mice (MACF1 KO) to determine the impact of MACF1 on cardiac microtubules and adaptation to pressure overload (transverse aortic constriction (TAC).In adult mouse hearts, MACF1 expression was low under basal conditions, but increased significantly in response to TAC. While MACF1 KO had no observable effect on heart size or function under basal conditions, MACF1 KO exacerbated TAC induced LV hypertrophy, LV dilation and contractile dysfunction. Interestingly, subcellular fractionation of ventricular lysates revealed that MACF1 KO altered microtubule distribution in response to TAC, so that more tubulin was associated with the cell membrane fraction. Moreover, TAC induced microtubule redistribution into this cell membrane fraction in both WT and MACF1 KO mice correlated strikingly with the level of contractile dysfunction (r(2) = 0.786, p<.001). MACF1 disruption also resulted in reduction of membrane caveolin 3 levels, and increased levels of membrane PKCα and β1 integrin after TAC, suggesting MACF1 function is important for spatial regulation of several physiologically relevant signaling proteins during hypertrophy. Together, these data identify for the first time, a role for MACF1 in cardiomyocyte microtubule distribution and in adaptation to hemodynamic overload.
- Published
- 2013
- Full Text
- View/download PDF
20. AMPK attenuates microtubule proliferation in cardiac hypertrophy.
- Author
-
Fassett JT, Hu X, Xu X, Lu Z, Zhang P, Chen Y, and Bache RJ
- Subjects
- AMP-Activated Protein Kinases genetics, Animals, Animals, Newborn, Cells, Cultured, Disease Models, Animal, Mice, Mice, Knockout, Microtubule-Associated Proteins metabolism, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac cytology, RNA, Small Interfering genetics, Rats, Rats, Sprague-Dawley, Tubulin metabolism, Ventricular Pressure physiology, AMP-Activated Protein Kinases metabolism, Cardiomegaly metabolism, Cardiomegaly pathology, Microtubules metabolism, Myocytes, Cardiac enzymology
- Abstract
Cell hypertrophy requires increased protein synthesis and expansion of the cytoskeletal networks that support cell enlargement. AMPK limits anabolic processes, such as protein synthesis, when energy supply is insufficient, but its role in cytoskeletal remodeling is not known. Here, we examined the influence of AMPK in cytoskeletal remodeling during cardiomyocyte hypertrophy, a clinically relevant condition in which cardiomyocytes enlarge but do not divide. In neonatal cardiomyocytes, activation of AMPK with 5-aminoimidazole carboxamide ribonucleotide (AICAR) or expression of constitutively active AMPK (CA-AMPK) attenuated cell area increase by hypertrophic stimuli (phenylephrine). AMPK activation had little effect on intermediate filaments or myofilaments but dramatically reduced microtubule stability, as measured by detyrosinated tubulin levels and cytoskeletal tubulin accumulation. Importantly, low-level AMPK activation limited cell expansion and microtubule growth independent of mTORC1 or protein synthesis repression, identifying a new mechanism by which AMPK regulates cell growth. Mechanistically, AICAR treatment increased Ser-915 phosphorylation of microtubule-associated protein 4 (MAP4), which reduces affinity for tubulin and prevents stabilization of microtubules (MTs). RNAi knockdown of MAP4 confirmed its critical role in cardiomyocyte MT stabilization. In support of a pathophysiological role for AMPK regulation of cardiac microtubules, AMPK α2 KO mice exposed to pressure overload (transverse aortic constriction; TAC) demonstrated reduced MAP4 phosphorylation and increased microtubule accumulation that correlated with the severity of contractile dysfunction. Together, our data identify the microtubule cytoskeleton as a sensitive target of AMPK activity, and the data suggest a novel role for AMPK in limiting accumulation and densification of microtubules that occurs in response to hypertrophic stress.
- Published
- 2013
- Full Text
- View/download PDF
21. Regulation of coronary resistance vessel tone in response to exercise.
- Author
-
Duncker DJ, Bache RJ, and Merkus D
- Subjects
- Animals, Coronary Vessels metabolism, Dogs, Humans, Hyperemia physiopathology, Myocardium metabolism, Nitric Oxide metabolism, Swine, Coronary Vessels physiology, Exercise physiology, Vascular Resistance physiology
- Abstract
Exercise is a primary stimulus for increased myocardial oxygen demand. The ~6-fold increase in oxygen demand of the left ventricle during heavy exercise is met principally by augmenting coronary blood flow (~5-fold), as hemoglobin concentration and oxygen extraction (which is already ~70% at rest) increase only modestly in most species. As a result, coronary blood flow is tightly coupled to myocardial oxygen consumption over a wide range of physical activity. This tight coupling has been proposed to depend on periarteriolar oxygen tension, signals released from cardiomyocytes and the endothelium as well as neurohumoral influences, but the contribution of each of these regulatory pathways, and their interactions, to exercise hyperemia in the heart remain incompletely understood. In humans, nitric oxide, adenosine and K(ATP) channels each appear to contribute to resting coronary resistance vessel tone, but evidence for a critical contribution to exercise hyperemia is lacking. In dogs K(ATP)-channel activation together with adenosine and nitric oxide contribute to exercise hyperemia in a non-linear redundant fashion. In contrast, in swine nitric oxide, adenosine and K(ATP) channels contribute to resting coronary resistance vessel tone control in a linear additive manner, but do not appear to be mandatory for exercise hyperemia. Rather, exercise hyperemia in swine appears to involve β-adrenergic activation in conjunction with exercise-induced blunting of an endothelin-mediated vasoconstrictor influence. In view of these remarkable species differences in coronary vasomotor control during exercise, future studies are required to determine the system of vasodilator components that mediate exercise hyperemia in humans. This article is part of a Special Issue entitled "Coronary Blood Flow"., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
22. Peripheral circulation.
- Author
-
Laughlin MH, Davis MJ, Secher NH, van Lieshout JJ, Arce-Esquivel AA, Simmons GH, Bender SB, Padilla J, Bache RJ, Merkus D, and Duncker DJ
- Subjects
- Blood Pressure physiology, Bone and Bones blood supply, Cardiac Output physiology, Coronary Circulation physiology, Genitalia blood supply, Humans, Muscle, Skeletal blood supply, Oxygen Consumption physiology, Pulmonary Circulation physiology, Renal Circulation physiology, Splanchnic Circulation physiology, Exercise physiology, Regional Blood Flow physiology
- Abstract
Blood flow (BF) increases with increasing exercise intensity in skeletal, respiratory, and cardiac muscle. In humans during maximal exercise intensities, 85% to 90% of total cardiac output is distributed to skeletal and cardiac muscle. During exercise BF increases modestly and heterogeneously to brain and decreases in gastrointestinal, reproductive, and renal tissues and shows little to no change in skin. If the duration of exercise is sufficient to increase body/core temperature, skin BF is also increased in humans. Because blood pressure changes little during exercise, changes in distribution of BF with incremental exercise result from changes in vascular conductance. These changes in distribution of BF throughout the body contribute to decreases in mixed venous oxygen content, serve to supply adequate oxygen to the active skeletal muscles, and support metabolism of other tissues while maintaining homeostasis. This review discusses the response of the peripheral circulation of humans to acute and chronic dynamic exercise and mechanisms responsible for these responses. This is accomplished in the context of leading the reader on a tour through the peripheral circulation during dynamic exercise. During this tour, we consider what is known about how each vascular bed controls BF during exercise and how these control mechanisms are modified by chronic physical activity/exercise training. The tour ends by comparing responses of the systemic circulation to those of the pulmonary circulation relative to the effects of exercise on the regional distribution of BF and mechanisms responsible for control of resistance/conductance in the systemic and pulmonary circulations., (© 2012 American Physiological Society)
- Published
- 2012
- Full Text
- View/download PDF
23. AMP activated protein kinase-α2 regulates expression of estrogen-related receptor-α, a metabolic transcription factor related to heart failure development.
- Author
-
Hu X, Xu X, Lu Z, Zhang P, Fassett J, Zhang Y, Xin Y, Hall JL, Viollet B, Bache RJ, Huang Y, and Chen Y
- Subjects
- AMP-Activated Protein Kinases deficiency, AMP-Activated Protein Kinases genetics, Adult, Aged, Animals, Cells, Cultured, Disease Models, Animal, Energy Metabolism genetics, Energy Metabolism physiology, Female, Heart Failure physiopathology, Heat-Shock Proteins metabolism, Humans, Male, Mice, Mice, Knockout, Middle Aged, Mitochondria, Heart enzymology, Myocardium metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, PPAR gamma metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Rats, Ventricular Dysfunction, Left metabolism, Ventricular Dysfunction, Left physiopathology, ERRalpha Estrogen-Related Receptor, AMP-Activated Protein Kinases metabolism, Heart Failure etiology, Heart Failure metabolism, Receptors, Estrogen metabolism, Transcription Factors metabolism
- Abstract
The normal expression of myocardial mitochondrial enzymes is essential to maintain the cardiac energy reserve and facilitate responses to stress, but the molecular mechanisms to maintain myocardial mitochondrial enzyme expression have been elusive. Here we report that congestive heart failure is associated with a significant decrease of myocardial estrogen-related receptor-α (ERRα), but not peroxisome proliferator-activated receptor-γ coactivator 1α, in human heart failure samples. In addition, chronic pressure overload in mice caused a decrease of ERRα expression that was significantly correlated to the degree of left ventricular dysfunction, pulmonary congestion, and decreases of a group of myocardial energy metabolism-related genes. We found that the metabolic sensor AMP activated protein kinase (AMPK) regulates ERRα expression in vivo and in vitro. AMPKα2 knockout decreased myocardial ERRα (both mRNA and protein) and its downstream targets under basal conditions, with no change in myocardial peroxisome proliferator-activated receptor-γ coactivator 1α expression. Using cultured rat neonatal cardiac myocytes, we found that overexpression of constitutively active AMPKα significantly induced ERRα mRNA, protein, and promoter activity. Conversely, selective gene silencing of AMPKα2 repressed ERRα and its target gene levels, indicating that AMPKα2 is involved in the regulation of ERRα expression. In addition, overexpression of ERRα in AMPKα2 knockout neonatal cardiac myocytes partially rescued the repressed expression of some energy metabolism-related genes. These data support an important role for AMPKα2 in regulating the expression of myocardial ERRα and its downstream mitochondrial enzymes.
- Published
- 2011
- Full Text
- View/download PDF
24. Exacerbated pulmonary arterial hypertension and right ventricular hypertrophy in animals with loss of function of extracellular superoxide dismutase.
- Author
-
Xu D, Guo H, Xu X, Lu Z, Fassett J, Hu X, Xu Y, Tang Q, Hu D, Somani A, Geurts AM, Ostertag E, Bache RJ, Weir EK, and Chen Y
- Subjects
- Animals, Familial Primary Pulmonary Hypertension, Hypertension, Pulmonary chemically induced, Hypertension, Pulmonary genetics, Hypertension, Pulmonary physiopathology, Hypertrophy, Right Ventricular genetics, Hypertrophy, Right Ventricular physiopathology, Hypoxia genetics, Lung physiopathology, Male, Mice, Mice, Knockout, Monocrotaline, Mutation, Rats, Superoxide Dismutase genetics, Hypertension, Pulmonary metabolism, Hypertrophy, Right Ventricular metabolism, Hypoxia metabolism, Lung metabolism, Superoxide Dismutase metabolism
- Abstract
Studies have demonstrated that increased oxidative stress contributes to the pathogenesis and the development of pulmonary artery hypertension (PAH). Extracellular superoxide dismutase (SOD3) is essential for removing extracellular superoxide anions, and it is highly expressed in lung tissue. However, it is not clear whether endogenous SOD3 can influence the development of PAH. Here we examined the effect of SOD3 knockout on hypoxia-induced PAH in mice and a loss-of-function SOD3 gene mutation (SOD3(E124D)) on monocrotaline (40 mg/kg)-induced PAH in rats. SOD3 knockout significantly exacerbated 2 weeks of hypoxia-induced right ventricular (RV) pressure and RV hypertrophy, whereas RV pressure in SOD3 knockout mice under normoxic conditions is similar to wild-type controls. In untreated control rats at age of 8 weeks, there was no significant difference between wild-type and SOD3(E124D) rats in RV pressure and the ratio of RV weight:left ventricular weight (0.25±0.02 in wild-type rats versus 0.25±0.01 in SOD3(E124D) rats). However, monocrotaline caused significantly greater increases of RV pressure in SOD3(E124D) rats (48.6±1.8 mm Hg in wild-type versus 57.5±3.1 mm Hg in SOD3(E124D) rats), of the ratio of RV weight:left ventricular weight (0.41±0.01 versus 0.50±0.09; P<0.05), and of the percentage of fully muscularized small arterioles in SOD3(E124D) rats (55.2±2.3% versus 69.9±2.6%; P<0.05). Together, these findings indicate that the endogenous SOD3 has no role in the development of PAH under control conditions but plays an important role in protecting the lung from the development of PAH under stress conditions.
- Published
- 2011
- Full Text
- View/download PDF
25. Dimethylarginine dimethylaminohydrolase-1 is the critical enzyme for degrading the cardiovascular risk factor asymmetrical dimethylarginine.
- Author
-
Hu X, Atzler D, Xu X, Zhang P, Guo H, Lu Z, Fassett J, Schwedhelm E, Böger RH, Bache RJ, and Chen Y
- Subjects
- Amidohydrolases deficiency, Amidohydrolases genetics, Animals, Arginine blood, Arginine metabolism, Blood Pressure, Cardiovascular Diseases enzymology, Cardiovascular Diseases genetics, Cells, Cultured, Enzyme Inhibitors administration & dosage, Female, Genotype, Hypertension enzymology, Hypertension physiopathology, Infusion Pumps, Implantable, Male, Mice, Mice, 129 Strain, Mice, Knockout, NG-Nitroarginine Methyl Ester administration & dosage, Nitric Oxide metabolism, Nitric Oxide Synthase antagonists & inhibitors, Nitric Oxide Synthase metabolism, Phenotype, RNA Interference, Risk Factors, Substrate Specificity, Time Factors, Transfection, omega-N-Methylarginine metabolism, Amidohydrolases metabolism, Arginine analogs & derivatives, Cardiovascular Diseases etiology, Endothelial Cells enzymology
- Abstract
Objective: The objective of this study was to identify the role of dimethylarginine dimethylaminohydrolase-1 (DDAH1) in degrading the endogenous nitric oxide synthase inhibitors asymmetrical dimethylarginine (ADMA) and N(g)-monomethyl-L-arginine (L-NMMA)., Methods and Results: We generated a global-DDAH1 gene-deficient (DDAH1(-/-)) mouse strain to examine the role of DDAH1 in ADMA and l-NMMA degradation and the physiological consequences of loss of DDAH1. Plasma and tissue ADMA and L-NMMA levels in DDAH1(-/-) mice were several folds higher than in wild-type mice, but growth and development of these DDAH1(-/-) mice were similar to those of their wild-type littermates. Although the expression of DDAH2 was unaffected, DDAH activity was undetectable in all tissues tested. These findings indicate that DDAH1 is the critical enzyme for ADMA and L-NMMA degradation. Blood pressure was ≈ 20 mm Hg higher in the DDAH1(-/-) mice than in wild-type mice, but no other cardiovascular phenotype was found under unstressed conditions. Crossing DDAH1(+/-) male with DDAH1(+/-) female mice yielded DDAH1(+/+), DDAH1(+/-), and DDAH1(-/-) mice at the anticipated ratio of 1:2:1, indicating that DDAH1 is not required for embryonic development in this strain., Conclusions: Our findings indicate that DDAH1 is required for metabolizing ADMA and L-NMMA in vivo, whereas DDAH2 had no detectable role for degrading ADMA and l-NMMA.
- Published
- 2011
- Full Text
- View/download PDF
26. Adenosine kinase regulation of cardiomyocyte hypertrophy.
- Author
-
Fassett JT, Hu X, Xu X, Lu Z, Zhang P, Chen Y, and Bache RJ
- Subjects
- AMP-Activated Protein Kinase Kinases, Adenosine metabolism, Adenosine Kinase antagonists & inhibitors, Animals, Cells, Cultured, Enzyme Inhibitors pharmacology, Hypertrophy metabolism, Hypertrophy pathology, Hypertrophy prevention & control, Models, Animal, Morpholines pharmacology, Myocytes, Cardiac drug effects, Protein Kinases metabolism, Pyrimidines pharmacology, Rats, Rats, Sprague-Dawley, Receptors, Purinergic P1 metabolism, Signal Transduction drug effects, TOR Serine-Threonine Kinases metabolism, raf Kinases metabolism, Adenosine Kinase metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Signal Transduction physiology
- Abstract
There is evidence that extracellular adenosine can attenuate cardiac hypertrophy, but the mechanism by which this occurs is not clear. Here we investigated the role of adenosine receptors and adenosine metabolism in attenuation of cardiomyocyte hypertrophy. Phenylephrine (PE) caused hypertrophy of neonatal rat cardiomyocytes with increases of cell surface area, protein synthesis, and atrial natriuretic peptide (ANP) expression. These responses were attenuated by 5 μM 2-chloroadenosine (CADO; adenosine deaminase resistant adenosine analog) or 10 μM adenosine. While antagonism of adenosine receptors partially blocked the reduction of ANP expression produced by CADO, it did not restore cell size or protein synthesis. In support of a role for intracellular adenosine metabolism in regulating hypertrophy, the adenosine kinase (AK) inhibitors iodotubercidin and ABT-702 completely reversed the attenuation of cell size, protein synthesis, and expression of ANP by CADO or ADO. Examination of PE-induced phosphosignaling pathways revealed that CADO treatment did not reduce AKT(Ser⁴⁷³) phosphorylation but did attenuate sustained phosphorylation of Raf(Ser³³⁸) (24-48 h), mTOR(Ser²⁴⁴⁸) (24-48 h), p70S6k(Thr³⁸⁹) (2.5-48 h), and ERK(Thr²⁰²/Tyr²⁰⁴) (48 h). Inhibition of AK restored activation of these enzymes in the presence of CADO. Using dominant negative and constitutively active Raf adenoviruses, we found that Raf activation is necessary and sufficient for PE-induced mTORC1 signaling and cardiomyocyte hypertrophy. CADO treatment still blocked p70S6k(Thr³⁸⁹) phosphorylation and hypertrophy downstream of constitutively active Raf, however, despite a high level phosphorylation of ERK(Thr202/Tyr204) and AKT(Ser⁴⁷³). Reduction of Raf-induced p70S6k(Thr³⁸⁹) phosphorylation and hypertrophy by CADO was reversed by inhibiting AK. Together, these results identify AK as an important mediator of adenosine attenuation of cardiomyocyte hypertrophy, which acts, at least in part, through inhibition of Raf signaling to mTOR/p70S6k.
- Published
- 2011
- Full Text
- View/download PDF
27. Dimethylarginine dimethylaminohydrolase 1 modulates endothelial cell growth through nitric oxide and Akt.
- Author
-
Zhang P, Hu X, Xu X, Chen Y, and Bache RJ
- Subjects
- Amidohydrolases deficiency, Amidohydrolases genetics, Animals, Arginine analogs & derivatives, Arginine metabolism, Cell Movement, Cells, Cultured, Enzyme Inhibitors pharmacology, Humans, Mice, Mice, Knockout, Neovascularization, Physiologic, Nitric Oxide Synthase Type III antagonists & inhibitors, Nitric Oxide Synthase Type III metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation, RNA Interference, Time Factors, Tissue Culture Techniques, Transfection, ras Proteins antagonists & inhibitors, ras Proteins genetics, ras Proteins metabolism, Amidohydrolases metabolism, Cell Proliferation, Endothelial Cells enzymology, Nitric Oxide metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction
- Abstract
Objective: Dimethylarginine dimethylaminohydrolase 1 (DDAH1) modulates NO production by degrading the endogenous nitric oxide (NO) synthase (NOS) inhibitors asymmetrical dimethylarginine (ADMA) and L-NG-monomethyl arginine (L-NMMA). This study examined whether, in addition to degrading ADMA, DDAH1 exerts ADMA-independent effects that influence endothelial function., Methods and Results: Using selective gene silencing of DDAH1 with small interfering RNA and overexpression of DDAH1 in human umbilical vein endothelial cells, we found that DDAH1 acts to promote endothelial cell proliferation, migration, and tube formation by Akt phosphorylation, as well as through the traditional role of degrading ADMA. Incubation of human umbilical vein endothelial cells with the NOS inhibitors l-NG-nitro-arginine methyl ester (L-NAME) or ADMA, the soluble guanylyl cyclase inhibitor 1H-(1,2,4)oxadiazolo-(4,3-2)quinoxalin-1-one, or the cGMP analog 8-(4-Chlorophenylthio)-cGMP had no effect on phosphorylated (p)-Akt(Ser473), indicating that the increase in p-Akt(Ser473) produced by DDAH1 was independent of the NO-cGMP signaling pathway. DDAH1 formed a protein complex with Ras, and DDAH1 overexpression increased Ras activity. The Ras inhibitor manumycin-A or dominant-negative Ras significantly attenuated the DDAH1-induced increase in p-Akt(Ser473). Furthermore, DDAH1 knockout impaired endothelial sprouting from cultured aortic rings, and overexpression of constitutively active Akt or DDAH1 rescued endothelial sprouting in the aortic rings from these mice., Conclusions: DDAH1 exerts a unique role in activating Akt that affects endothelial function independently of degrading endogenous NOS inhibitors.
- Published
- 2011
- Full Text
- View/download PDF
28. Adenosine regulation of microtubule dynamics in cardiac hypertrophy.
- Author
-
Fassett JT, Xu X, Hu X, Zhu G, French J, Chen Y, and Bache RJ
- Subjects
- 2-Chloroadenosine metabolism, 2-Chloroadenosine pharmacology, 5'-Nucleotidase genetics, Adenosine pharmacology, Animals, Cardiomegaly pathology, Cells, Cultured, Disease Models, Animal, Mice, Mice, Inbred BALB C, Mice, Knockout, Microtubules drug effects, Myocardial Contraction drug effects, Myocardial Contraction physiology, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Rats, Rats, Sprague-Dawley, Tubulin metabolism, Adenosine metabolism, Cardiomegaly drug therapy, Cardiomegaly metabolism, Colchicine pharmacology, Microtubules metabolism, Tubulin Modulators pharmacology
- Abstract
There is evidence that endogenous extracellular adenosine reduces cardiac hypertrophy and heart failure in mice subjected to chronic pressure overload, but the mechanism by which adenosine exerts these protective effects is unknown. Here, we identified a novel role for adenosine in regulation of the cardiac microtubule cytoskeleton that may contribute to its beneficial effects in the overloaded heart. In neonatal cardiomyocytes, phenylephrine promoted hypertrophy and reorganization of the cytoskeleton, which included accumulation of sarcomeric proteins, microtubules, and desmin. Treatment with adenosine or the stable adenosine analog 2-chloroadenosine, which decreased hypertrophy, specifically reduced accumulation of microtubules. In hypertrophied cardiomyocytes, 2-chloroadenosine or adenosine treatment preferentially targeted stabilized microtubules (containing detyrosinated alpha-tubulin). Consistent with a role for endogenous adenosine in reducing microtubule stability, levels of detyrosinated microtubules were elevated in hearts of CD73 knockout mice (deficient in extracellular adenosine production) compared with wild-type mice (195%, P < 0.05). In response to aortic banding, microtubules increased in hearts of wild-type mice; this increase was exaggerated in CD73 knockout mice, with significantly greater amounts of tubulin partitioning into the cold-stable Triton-insoluble fractions. The levels of this stable cytoskeletal fraction of tubulin correlated strongly with the degree of heart failure. In agreement with a role for microtubule stabilization in promoting cardiac dysfunction, colchicine treatment of aortic-banded mice reduced hypertrophy and improved cardiac function compared with saline-treated controls. These results indicate that microtubules contribute to cardiac dysfunction and identify, for the first time, a role for adenosine in regulating cardiomyocyte microtubule dynamics.
- Published
- 2009
- Full Text
- View/download PDF
29. NADPH oxidase contributes to coronary endothelial dysfunction in the failing heart.
- Author
-
Zhang P, Hou M, Li Y, Xu X, Barsoum M, Chen Y, and Bache RJ
- Subjects
- Acetophenones pharmacology, Acetylcholine pharmacology, Aldehydes metabolism, Animals, Antioxidants pharmacology, Cardiac Pacing, Artificial, Coronary Circulation, Coronary Vessels drug effects, Coronary Vessels physiopathology, Disease Models, Animal, Dogs, Dose-Response Relationship, Drug, Endothelium, Vascular drug effects, Endothelium, Vascular physiopathology, Enzyme Inhibitors pharmacology, Female, Heart Failure physiopathology, Hemodynamics, Male, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases genetics, Oxidative Stress, RNA, Messenger metabolism, Tyrosine analogs & derivatives, Tyrosine metabolism, Up-Regulation, Vasodilator Agents pharmacology, Coronary Vessels enzymology, Endothelium, Vascular enzymology, Heart Failure enzymology, NADPH Oxidases metabolism, Superoxides metabolism, Vasodilation drug effects
- Abstract
Increased reactive oxygen species (ROS) produced by the failing heart can react with nitric oxide (NO), thereby decreasing NO bioavailability. This study tested the hypothesis that increased ROS generation contributes to coronary endothelial dysfunction in the failing heart. Congestive heart failure (CHF) was produced in six dogs by ventricular pacing at 240 beats/min for 4 wk. Studies were performed at rest and during treadmill exercise under control conditions and after treatment with the NADPH oxidase inhibitor and antioxidant apocynin (4 mg/kg iv). Apocynin caused no significant changes in heart rate, aortic pressure, left ventricular (LV) systolic pressure, LV end-diastolic pressure, or maximum rate of LV pressure increase at rest or during exercise in normal or CHF dogs. Apocynin caused no change in coronary blood flow (CBF) in normal dogs but increased CBF at rest and during exercise in animals with CHF (P < 0.05). Intracoronary ACh caused dose-dependent increases of CBF that were blunted in CHF. Apocynin had no effect on the response to ACh in normal dogs but augmented the response to ACh in CHF dogs (P < 0.05). The oxidative stress markers nitrotyrosine and 4-hydroxy-2-nonenal were significantly greater in failing than in normal myocardium. Furthermore, coelenterazine chemiluminescence for O(2)(-) was more than twice normal in failing myocardium, and this difference was abolished by apocynin. Western blot analysis of myocardial lysates demonstrated that the p47(phox) and p22(phox) subunits of NADPH were significantly increased in the failing hearts, while real-time PCR demonstrated that Nox2 mRNA was significantly increased. The data indicate that increased ROS generation in the failing heart is associated with coronary endothelial dysfunction and suggest that NADPH oxidase may contribute to this abnormality.
- Published
- 2009
- Full Text
- View/download PDF
30. Xanthine oxidase inhibition with febuxostat attenuates systolic overload-induced left ventricular hypertrophy and dysfunction in mice.
- Author
-
Xu X, Hu X, Lu Z, Zhang P, Zhao L, Wessale JL, Bache RJ, and Chen Y
- Subjects
- Animals, Disease Models, Animal, Febuxostat, Hypertrophy, Left Ventricular physiopathology, Male, Mice, Mice, Inbred C57BL, Systole drug effects, Systole physiology, Thiazoles pharmacology, Ventricular Dysfunction, Left physiopathology, Hypertrophy, Left Ventricular drug therapy, Hypertrophy, Left Ventricular enzymology, Thiazoles therapeutic use, Ventricular Dysfunction, Left drug therapy, Ventricular Dysfunction, Left enzymology
- Abstract
The purine analog xanthine oxidase (XO) inhibitors (XOIs), allopurinol and oxypurinol, have been reported to protect against heart failure secondary to myocardial infarction or rapid ventricular pacing. Because these agents might influence other aspects of purine metabolism that could influence their effect, this study examined the effect of the non-purine XOI, febuxostat, on pressure overload-induced left ventricular (LV) hypertrophy and dysfunction. Transverse aortic constriction (TAC) in mice caused LV hypertrophy and dysfunction and increased myocardial nitrotyrosine at 8 days. TAC also caused increased phosphorylated Akt (p-Akt(Ser473)), p42/44 extracellular signal-regulated kinase (p-Erk(Thr202/Tyr204)), and mammalian target of rapamycin (mTOR) (p-mTOR(Ser2488)). XO inhibition with febuxostat (5 mg/kg/d by gavage for 8 days) beginning approximately 60minutes after TAC attenuated the TAC-induced LV hypertrophy and dysfunction. Febuxostat blunted the TAC-induced increases in nitrotyrosine (indicating reduced myocardial oxidative stress), p-Erk(Thr202/Tyr204), and p-mTOR(Ser2488), with no effect on total Erk or total mTOR. Febuxostat had no effect on myocardial p-Akt(Ser473) or total Akt. The results suggest that XO inhibition with febuxostat reduced oxidative stress in the pressure overloaded LV, thereby diminishing the activation of pathways that result in pathologic hypertrophy and contractile dysfunction.
- Published
- 2008
- Full Text
- View/download PDF
31. AMP activated protein kinase-alpha2 deficiency exacerbates pressure-overload-induced left ventricular hypertrophy and dysfunction in mice.
- Author
-
Zhang P, Hu X, Xu X, Fassett J, Zhu G, Viollet B, Xu W, Wiczer B, Bernlohr DA, Bache RJ, and Chen Y
- Subjects
- AMP-Activated Protein Kinases, Adaptor Proteins, Signal Transducing, Animals, Carrier Proteins metabolism, Cell Cycle Proteins, Cells, Cultured, DNA-Binding Proteins metabolism, Eukaryotic Initiation Factors, Hypertension physiopathology, Hypertrophy, Left Ventricular chemically induced, Hypertrophy, Left Ventricular physiopathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Multienzyme Complexes genetics, Multienzyme Complexes metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Phenylephrine, Phosphoproteins metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Ribosomal Protein S6 metabolism, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Stroke Volume physiology, Transcription Factors metabolism, Hypertension complications, Hypertrophy, Left Ventricular etiology, Multienzyme Complexes deficiency, Protein Serine-Threonine Kinases deficiency, Ventricular Dysfunction, Left metabolism
- Abstract
AMP activated protein kinase (AMPK) plays an important role in regulating myocardial metabolism and protein synthesis. Activation of AMPK attenuates hypertrophy in cultured cardiac myocytes, but the role of AMPK in regulating the development of myocardial hypertrophy in response to chronic pressure overload is not known. To test the hypothesis that AMPKalpha2 protects the heart against systolic overload-induced ventricular hypertrophy and dysfunction, we studied the response of AMPKalpha2 gene deficient (knockout [KO]) mice and wild-type mice subjected to 3 weeks of transverse aortic constriction (TAC). Although AMPKalpha2 KO had no effect on ventricular structure or function under control conditions, AMPKalpha2 KO significantly increased TAC-induced ventricular hypertrophy (ventricular mass increased 46% in wild-type mice compared with 65% in KO mice) while decreased left ventricular ejection fraction (ejection fraction decreased 14% in wild-type mice compared with a 43% decrease in KO mice). AMPKalpha2 KO also significantly exacerbated the TAC-induced increases of atrial natriuretic peptide, myocardial fibrosis, and cardiac myocyte size. AMPKalpha2 KO had no effect on total S6 ribosomal protein (S6), p70 S6 kinase, eukaryotic initiation factor 4E, and 4E binding protein-1 or their phosphorylation under basal conditions but significantly augmented the TAC-induced increases of p-p70 S6 kinase(Thr389), p-S6(Ser235), and p-eukaryotic initiation factor 4E(Ser209). AMPKalpha2 KO also enhanced the TAC-induced increase of p-4E binding protein-1(Thr46) to a small degree and augmented the TAC-induced increase of p-Akt(Ser473). These data indicate that AMPKalpha2 exerts a cardiac protective effect against pressure-overload-induced ventricular hypertrophy and dysfunction.
- Published
- 2008
- Full Text
- View/download PDF
32. Disruption of sarcolemmal ATP-sensitive potassium channel activity impairs the cardiac response to systolic overload.
- Author
-
Hu X, Xu X, Huang Y, Fassett J, Flagg TP, Zhang Y, Nichols CG, Bache RJ, and Chen Y
- Subjects
- ATP-Binding Cassette Transporters antagonists & inhibitors, ATP-Binding Cassette Transporters genetics, Animals, Animals, Newborn, Aorta surgery, Base Sequence, Cell Hypoxia, Cells, Cultured, Constriction, Disease Models, Animal, Energy Metabolism genetics, Forkhead Box Protein O1, Forkhead Transcription Factors metabolism, Hypertrophy, Left Ventricular physiopathology, KATP Channels deficiency, KATP Channels genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Molecular Sequence Data, Mutation, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Potassium Channel Blockers pharmacology, Potassium Channels, Inwardly Rectifying antagonists & inhibitors, Potassium Channels, Inwardly Rectifying deficiency, Potassium Channels, Inwardly Rectifying genetics, Promoter Regions, Genetic, RNA Interference, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Rats, Receptors, Drug antagonists & inhibitors, Receptors, Drug genetics, Sarcolemma drug effects, Severity of Illness Index, Sulfonylurea Receptors, Trans-Activators genetics, Trans-Activators metabolism, Transcription Factors, Transfection, Ventricular Dysfunction, Left physiopathology, ATP-Binding Cassette Transporters metabolism, Hemodynamics, Hypertrophy, Left Ventricular metabolism, KATP Channels metabolism, Myocardium metabolism, Potassium Channels, Inwardly Rectifying metabolism, Receptors, Drug metabolism, Sarcolemma metabolism, Ventricular Dysfunction, Left metabolism
- Abstract
Sarcolemmal ATP-sensitive potassium channels (K(ATP)) act as metabolic sensors that facilitate adaptation of the left ventricle to changes in energy requirements. This study examined the mechanism by which K(ATP) dysfunction impairs the left ventricular response to stress using transgenic mouse strains with cardiac-specific disruption of K(ATP) activity (SUR1-tg mice) or Kir6.2 gene deficiency (Kir6.2 KO). Both SUR1-tg and Kir6.2 KO mice had normal left ventricular mass and function under unstressed conditions. Following chronic transverse aortic constriction, both SUR1-tg and Kir6.2 KO mice developed more severe left ventricular hypertrophy and dysfunction as compared with their corresponding WT controls. Both SUR1-tg and Kir6.2 KO mice had significantly decreased expression of peroxisome proliferator-activated receptor gamma coactivator (PGC)-1alpha and a group of energy metabolism related genes at both protein and mRNA levels. Furthermore, disruption of K(ATP) repressed expression and promoter activity of PGC-1alpha in cultured rat neonatal cardiac myocytes in response to hypoxia, indicating that K(ATP) activity is required to maintain PGC-1alpha expression under stress conditions. PGC-1alpha gene deficiency also exacerbated chronic transverse aortic constriction-induced ventricular hypertrophy and dysfunction, suggesting that depletion of PGC-1alpha can worsen systolic overload induced ventricular dysfunction. Both SUR1-tg and Kir6.2 KO mice had decreased FOXO1 after transverse aortic constriction, in agreement with the reports that a decrease of FOXO1 can repress PGC-1alpha expression. Furthermore, inhibition of K(ATP) caused a decrease of FOXO1 associated with PGC-1alpha promoter. These data indicate that K(ATP) channels facilitate the cardiac response to stress by regulating PGC-1alpha and its target genes, at least partially through the FOXO1 pathway.
- Published
- 2008
- Full Text
- View/download PDF
33. Phosphate metabolite concentrations and ATP hydrolysis potential in normal and ischaemic hearts.
- Author
-
Wu F, Zhang EY, Zhang J, Bache RJ, and Beard DA
- Subjects
- Animals, Biological Transport, Computer Simulation, Dogs, Energy Metabolism, Hydrolysis, Myoglobin metabolism, Oxygen metabolism, Adenosine Triphosphate metabolism, Myocardial Ischemia metabolism, Phosphates metabolism
- Abstract
To understand how cardiac ATP and CrP remain stable with changes in work rate - a phenomenon that has eluded mechanistic explanation for decades - data from (31)phosphate-magnetic resonance spectroscopy ((31)P-MRS) are analysed to estimate cytoplasmic and mitochondrial phosphate metabolite concentrations in the normal state, during high cardiac workstates, during acute ischaemia and reactive hyperaemic recovery. Analysis is based on simulating distributed heterogeneous oxygen transport in the myocardium integrated with a detailed model of cardiac energy metabolism. The model predicts that baseline myocardial free inorganic phosphate (P(i)) concentration in the canine myocyte cytoplasm - a variable not accessible to direct non-invasive measurement - is approximately 0.29 mm and increases to 2.3 mm near maximal cardiac oxygen consumption. During acute ischaemia (from ligation of the left anterior descending artery) P(i) increases to approximately 3.1 mm and ATP consumption in the ischaemic tissue is reduced quickly to less than half its baseline value before the creatine phosphate (CrP) pool is 18% depleted. It is determined from these experiments that the maximal rate of oxygen consumption of the heart is an emergent property and is limited not simply by the maximal rate of ATP synthesis, but by the maximal rate at which ATP can be synthesized at a potential at which it can be utilized. The critical free energy of ATP hydrolysis for cardiac contraction that is consistent with these findings is approximately -63.5 kJ mol(-1). Based on theoretical findings, we hypothesize that inorganic phosphate is both the primary feedback signal for stimulating oxidative phosphorylation in vivo and also the most significant product of ATP hydrolysis in limiting the capacity of the heart to hydrolyse ATP in vivo. Due to the lack of precise quantification of P(i) in vivo, these hypotheses and associated model predictions remain to be carefully tested experimentally.
- Published
- 2008
- Full Text
- View/download PDF
34. Regulation of coronary blood flow during exercise.
- Author
-
Duncker DJ and Bache RJ
- Subjects
- Adaptation, Physiological, Animals, Collateral Circulation, Humans, Models, Cardiovascular, Oxygen Consumption, Coronary Circulation, Coronary Stenosis physiopathology, Exercise, Hemodynamics, Ventricular Function
- Abstract
Exercise is the most important physiological stimulus for increased myocardial oxygen demand. The requirement of exercising muscle for increased blood flow necessitates an increase in cardiac output that results in increases in the three main determinants of myocardial oxygen demand: heart rate, myocardial contractility, and ventricular work. The approximately sixfold increase in oxygen demands of the left ventricle during heavy exercise is met principally by augmenting coronary blood flow (~5-fold), as hemoglobin concentration and oxygen extraction (which is already 70-80% at rest) increase only modestly in most species. In contrast, in the right ventricle, oxygen extraction is lower at rest and increases substantially during exercise, similar to skeletal muscle, suggesting fundamental differences in blood flow regulation between these two cardiac chambers. The increase in heart rate also increases the relative time spent in systole, thereby increasing the net extravascular compressive forces acting on the microvasculature within the wall of the left ventricle, in particular in its subendocardial layers. Hence, appropriate adjustment of coronary vascular resistance is critical for the cardiac response to exercise. Coronary resistance vessel tone results from the culmination of myriad vasodilator and vasoconstrictors influences, including neurohormones and endothelial and myocardial factors. Unraveling of the integrative mechanisms controlling coronary vasodilation in response to exercise has been difficult, in part due to the redundancies in coronary vasomotor control and differences between animal species. Exercise training is associated with adaptations in the coronary microvasculature including increased arteriolar densities and/or diameters, which provide a morphometric basis for the observed increase in peak coronary blood flow rates in exercise-trained animals. In larger animals trained by treadmill exercise, the formation of new capillaries maintains capillary density at a level commensurate with the degree of exercise-induced physiological myocardial hypertrophy. Nevertheless, training alters the distribution of coronary vascular resistance so that more capillaries are recruited, resulting in an increase in the permeability-surface area product without a change in capillary numerical density. Maintenance of alpha- and ss-adrenergic tone in the presence of lower circulating catecholamine levels appears to be due to increased receptor responsiveness to adrenergic stimulation. Exercise training also alters local control of coronary resistance vessels. Thus arterioles exhibit increased myogenic tone, likely due to a calcium-dependent protein kinase C signaling-mediated alteration in voltage-gated calcium channel activity in response to stretch. Conversely, training augments endothelium-dependent vasodilation throughout the coronary microcirculation. This enhanced responsiveness appears to result principally from an increased expression of nitric oxide (NO) synthase. Finally, physical conditioning decreases extravascular compressive forces at rest and at comparable levels of exercise, mainly because of a decrease in heart rate. Impedance to coronary inflow due to an epicardial coronary artery stenosis results in marked redistribution of myocardial blood flow during exercise away from the subendocardium towards the subepicardium. However, in contrast to the traditional view that myocardial ischemia causes maximal microvascular dilation, more recent studies have shown that the coronary microvessels retain some degree of vasodilator reserve during exercise-induced ischemia and remain responsive to vasoconstrictor stimuli. These observations have required reassessment of the principal sites of resistance to blood flow in the microcirculation. A significant fraction of resistance is located in small arteries that are outside the metabolic control of the myocardium but are sensitive to shear and nitrovasodilators. The coronary collateral system embodies a dynamic network of interarterial vessels that can undergo both long- and short-term adjustments that can modulate blood flow to the dependent myocardium. Long-term adjustments including recruitment and growth of collateral vessels in response to arterial occlusion are time dependent and determine the maximum blood flow rates available to the collateral-dependent vascular bed during exercise. Rapid short-term adjustments result from active vasomotor activity of the collateral vessels. Mature coronary collateral vessels are responsive to vasodilators such as nitroglycerin and atrial natriuretic peptide, and to vasoconstrictors such as vasopressin, angiotensin II, and the platelet products serotonin and thromboxane A(2). During exercise, ss-adrenergic activity and endothelium-derived NO and prostanoids exert vasodilator influences on coronary collateral vessels. Importantly, alterations in collateral vasomotor tone, e.g., by exogenous vasopressin, inhibition of endogenous NO or prostanoid production, or increasing local adenosine production can modify collateral conductance, thereby influencing the blood supply to the dependent myocardium. In addition, vasomotor activity in the resistance vessels of the collateral perfused vascular bed can influence the volume and distribution of blood flow within the collateral zone. Finally, there is evidence that vasomotor control of resistance vessels in the normally perfused regions of collateralized hearts is altered, indicating that the vascular adaptations in hearts with a flow-limiting coronary obstruction occur at a global as well as a regional level. Exercise training does not stimulate growth of coronary collateral vessels in the normal heart. However, if exercise produces ischemia, which would be absent or minimal under resting conditions, there is evidence that collateral growth can be enhanced. In addition to ischemia, the pressure gradient between vascular beds, which is a determinant of the flow rate and therefore the shear stress on the collateral vessel endothelium, may also be important in stimulating growth of collateral vessels.
- Published
- 2008
- Full Text
- View/download PDF
35. Ecto-5'-nucleotidase deficiency exacerbates pressure-overload-induced left ventricular hypertrophy and dysfunction.
- Author
-
Xu X, Fassett J, Hu X, Zhu G, Lu Z, Li Y, Schnermann J, Bache RJ, and Chen Y
- Subjects
- Adenosine metabolism, Animals, Blood Pressure physiology, Cell Division physiology, Cells, Cultured, Collagen metabolism, Fibroblasts enzymology, Fibroblasts pathology, Fibrosis, Heart Failure metabolism, Heart Failure pathology, Heart Failure physiopathology, Hypertension metabolism, Hypertension pathology, Hypertrophy, Left Ventricular metabolism, Hypertrophy, Left Ventricular pathology, Mice, Mice, Knockout, Myocytes, Cardiac pathology, Protein Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Rats, Rats, Sprague-Dawley, Ribosomal Protein S6 Kinases, 70-kDa metabolism, TOR Serine-Threonine Kinases, Ventricular Dysfunction, Left metabolism, Ventricular Dysfunction, Left pathology, 5'-Nucleotidase genetics, 5'-Nucleotidase metabolism, Hypertension physiopathology, Hypertrophy, Left Ventricular physiopathology, Myocytes, Cardiac enzymology, Ventricular Dysfunction, Left physiopathology
- Abstract
This study examined whether endogenous extracellular adenosine acts to facilitate the adaptive response of the heart to chronic systolic overload. To examine whether endogenous extracellular adenosine can protect the heart against pressure-overload-induced heart failure, transverse aortic constriction was performed on mice deficient in extracellular adenosine production as the result of genetic deletion of CD73. Although there was no difference in left ventricular size or function between CD73-deficient mice (knockout [KO] mice) and wild-type mice under unstressed conditions, aortic constriction for 2 or 4 weeks induced significantly more myocardial hypertrophy, left ventricular dilation, and left ventricular dysfunction in KO mice compared with wild-type mice. Thus, after 2 weeks of transverse aortic constriction, left ventricular fractional shortening decreased to 27.4+/-2.5% and 21.9+/-1.7% in wild-type and KO mice, respectively (P<0.05). Consistent with a role of adenosine in reducing tissue remodeling, KO mice displayed increased myocardial fibrosis and myocyte hypertrophy compared with wild-type mice. Furthermore, adenosine treatment reduced phenylephrine-induced cardiac myocyte hypertrophy and collagen production in cultured neonatal rat cardiac myocytes and cardiac fibroblasts, respectively. Consistent with a role for adenosine in modulating cardiomyocyte hypertrophy, KO mice demonstrated increased activation of mammalian target of rapamycin signaling, accompanied by higher expression of the hypertrophy marker atrial natriuretic peptide. Conversely, the adenosine analogue 2-chloro-adenosine significantly reduced cell size, mammalian target of rapamycin/p70 ribosomal S6 kinase activation, and atrial natriuretic peptide expression in cultured neonatal cardiomyocytes. These data demonstrate that CD73 helps to preserve cardiac function during chronic systolic overload by preventing maladaptive tissue remodeling.
- Published
- 2008
- Full Text
- View/download PDF
36. Extracellular superoxide dismutase protects the heart against oxidative stress and hypertrophy after myocardial infarction.
- Author
-
van Deel ED, Lu Z, Xu X, Zhu G, Hu X, Oury TD, Bache RJ, Duncker DJ, and Chen Y
- Subjects
- Adenosine Triphosphatases metabolism, Animals, Aorta pathology, Echocardiography methods, Extracellular Matrix metabolism, Fibrosis, Hypertrophy, Male, Mice, Mice, Inbred C57BL, Myocardium pathology, Extracellular Matrix enzymology, Heart physiology, Myocardial Infarction pathology, Oxidative Stress, Superoxide Dismutase metabolism
- Abstract
Extracellular superoxide dismutase (EC-SOD) contributes only a small fraction to total SOD activity in the heart but is strategically located to scavenge free radicals in the extracellular compartment. EC-SOD expression is decreased in myocardial-infarction (MI)-induced heart failure, but whether EC-SOD can abrogate oxidative stress or modify MI-induced ventricular remodeling has not been previously studied. Consequently, the effects of EC-SOD gene deficiency (EC-SOD KO) on left ventricular (LV) oxidative stress, hypertrophy, and fibrosis were studied in EC-SOD KO and wild-type mice under control conditions, and at 4 and 8 weeks after permanent coronary artery ligation. EC-SOD KO had no detectable effect on LV function in normal hearts but caused small but significant increases of LV fibrosis. At 8 weeks after MI, EC-SOD KO mice developed significantly more LV hypertrophy (LV mass increased 1.64-fold in KO mice compared to 1.35-fold in wild-type mice; p<0.01) and more fibrosis and myocyte hypertrophy which was more prominent in the peri-infarct region than in the remote myocardium. EC-SOD KO mice had greater increases of nitrotyrosine in the peri-infarct myocardium, and this was associated with a greater reduction of LV ejection fraction, a greater decrease of sarcoplasmic or endoplasmic reticulum calcium2+ ATPase, and a greater increase of atrial natriuretic peptide in the peri-infarct zone compared to wild-type mice. EC-SOD KO was associated with more increases of phosphorylated p38 (p-p38(Thr180/Tyr182)), p42/44 extracellular signal-regulated kinase (p-Erk(Thr202/Tyr204)), and c-Jun N-terminal kinase (p-JNK(Thr183/Tyr185)) both under control conditions and after MI, indicating that EC-SOD KO increases activation of mitogen-activated protein kinase signaling pathways. These findings demonstrate that EC-SOD plays an important role in protecting the heart against oxidative stress and infarction-induced ventricular hypertrophy.
- Published
- 2008
- Full Text
- View/download PDF
37. Extracellular superoxide dismutase deficiency exacerbates pressure overload-induced left ventricular hypertrophy and dysfunction.
- Author
-
Lu Z, Xu X, Hu X, Zhu G, Zhang P, van Deel ED, French JP, Fassett JT, Oury TD, Bache RJ, and Chen Y
- Subjects
- Animals, Fibrosis etiology, Fibrosis metabolism, Heart Failure metabolism, Heart Ventricles pathology, Heart Ventricles physiopathology, Hypertension complications, Hypertension metabolism, Hypertrophy, Left Ventricular etiology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Contraction physiology, Oxidative Stress physiology, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Ventricular Dysfunction, Left etiology, Hypertrophy, Left Ventricular enzymology, Superoxide Dismutase deficiency, Ventricular Dysfunction, Left enzymology
- Abstract
Extracellular superoxide dismutase (SOD) contributes only a small fraction to total SOD activity in the normal heart but is strategically located to scavenge free radicals in the extracellular compartment. To examine the physiological significance of extracellular SOD in the response of the heart to hemodynamic stress, we studied the effect of extracellular SOD deficiency on transverse aortic constriction (TAC)-induced left ventricular remodeling. Under unstressed conditions extracellular SOD deficiency had no effect on myocardial total SOD activity, the ratio of glutathione:glutathione disulfide, nitrotyrosine content, or superoxide anion production but resulted in small but significant increases in myocardial fibrosis and ventricular mass. In response to TAC for 6 weeks, extracellular SOD-deficient mice developed more severe left ventricular hypertrophy (heart weight increased 2.56-fold in extracellular SOD-deficient mice as compared with 1.99-fold in wild-type mice) and pulmonary congestion (lung weight increased 2.92-fold in extracellular SOD-deficient mice as compared with 1.84-fold in wild-type mice). Extracellular SOD-deficient mice also had more ventricular fibrosis, dilation, and a greater reduction of left ventricular fractional shortening and rate of pressure development after TAC. TAC resulted in greater increases of ventricular collagen I, collagen III, matrix metalloproteinase-2, matrix metalloproteinase-9, nitrotyrosine, and superoxide anion production. TAC also resulted in a greater decrease of the ratio of glutathione:glutathione disulfide in extracellular SOD-deficient mice. The finding that extracellular SOD deficiency had minimal impact on myocardial overall SOD activity but exacerbated TAC induced myocardial oxidative stress, hypertrophy, fibrosis, and dysfunction indicates that the distribution of extracellular SOD in the extracellular space is critically important in protecting the heart against pressure overload.
- Published
- 2008
- Full Text
- View/download PDF
38. Endothelial nitric oxide synthase is dynamically expressed during bone marrow stem cell differentiation into endothelial cells.
- Author
-
Liu Z, Jiang Y, Hao H, Gupta K, Xu J, Chu L, McFalls E, Zweier J, Verfaillie C, and Bache RJ
- Subjects
- Animals, Bone Marrow Cells cytology, Cell Differentiation drug effects, Cells, Cultured, Down-Regulation drug effects, Endothelial Cells physiology, Endothelium, Vascular metabolism, Enzyme Inhibitors pharmacology, Extracellular Signal-Regulated MAP Kinases metabolism, Mice, Mitogen-Activated Protein Kinase 1 metabolism, Multipotent Stem Cells cytology, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide Synthase Type III genetics, Phosphorylation drug effects, Vascular Endothelial Growth Factor A pharmacology, Bone Marrow Cells metabolism, Cell Differentiation physiology, Endothelium, Vascular cytology, Multipotent Stem Cells metabolism, Nitric Oxide Synthase Type III metabolism
- Abstract
This study was designed to investigate the developmental expression of endothelial nitric oxide synthase (eNOS) during stem cell differentiation into endothelial cells and to examine the functional status of the newly differentiated endothelial cells. Mouse adult multipotent progenitor cells (MAPCs) were used as the source of stem cells and were induced to differentiate into endothelial cells with vascular endothelial growth factor (VEGF) in serum-free medium. Expression of eNOS in the cells during differentiation was evaluated with real-time PCR, nitric oxide synthase (NOS) activity, and Western blot analysis. It was found that eNOS, but no other NOS, was present in undifferentiated MAPCs. eNOS expression disappeared in the cells immediately after induction of differentiation. However, eNOS expression reoccurred at day 7 during differentiation. Increasing eNOS mRNA, protein content, and activity were observed in the cells at days 14 and 21 during differentiation. The differentiated endothelial cells formed dense capillary networks on growth factor-reduced Matrigel. VEGF-stimulated phosphorylation of extracellular signal-regulated kinase (ERK)-1 and ERK-2 occurred in these cells, which was inhibited by NOS inhibitor N(G)-nitro-L-arginine methyl ester. In conclusion, these data demonstrate that eNOS is present in MAPCs and is dynamically expressed during the differentiation of MAPCs into endothelial cells in vitro.
- Published
- 2007
- Full Text
- View/download PDF
39. Effect of K+ATP channel and adenosine receptor blockade during rest and exercise in congestive heart failure.
- Author
-
Traverse JH, Chen Y, Hou M, Li Y, and Bache RJ
- Subjects
- Adenosine Triphosphate metabolism, Animals, Anti-Arrhythmia Agents pharmacology, Blood Flow Velocity drug effects, Cardiac Pacing, Artificial, Coronary Circulation drug effects, Disease Models, Animal, Dogs, Exercise Test, Glyburide pharmacology, Heart drug effects, Oxygen Consumption drug effects, Pinacidil pharmacology, Potassium Channels metabolism, Rest, Theophylline analogs & derivatives, Theophylline pharmacology, Vasodilator Agents pharmacology, Heart physiopathology, Heart Failure physiopathology, Physical Exertion drug effects, Potassium Channel Blockers pharmacology, Potassium Channels drug effects, Purinergic P1 Receptor Antagonists
- Abstract
K(+)(ATP) channels are important metabolic regulators of coronary blood flow (CBF) that are activated in the setting of reduced levels of ATP or perfusion pressure. In the normal heart, blockade of K(+)(ATP) channels results in a approximately 20% reduction in resting CBF but does not impair the increase in CBF that occurs during exercise. In contrast, adenosine receptor blockade fails to alter CBF or myocardial oxygen consumption (MVO(2)) in the normal heart but contributes to the increase in CBF during exercise when vascular K(+)(ATP) channels are blocked. Congestive heart failure (CHF) is associated with a decrease in CBF that is matched to a decrease in MVO(2) suggesting downregulation of myocardial energy utilization. Because myocardial ATP levels and coronary perfusion pressure are reduced in CHF, this study was undertaken to examine the role of K(+)(ATP) channels and adenosine in dogs with pacing-induced CHF. Myocardial blood flow (MBF) and MVO(2) were measured during rest and treadmill exercise before and after K(+)(ATP) channel blockade with glibenclamide (50 microg/kg/min ic) or adenosine receptor blockade with 8-phenyltheophylline (8-PT; 5 mg/kg iv). Inhibition of K(+)(ATP) channels resulted in a decrease in CBF and MVO(2) at rest and during exercise without a change in the relationship between CBF and MVO(2). In contrast, adenosine receptor blockade caused a significant increase in CBF that occurred secondary to an increase of MVO(2). These findings demonstrate that coronary K(+)(ATP) channel activity contribute to the regulation of resting MBF in CHF, and that endogenous adenosine may act to inhibit MVO(2) in the failing heart.
- Published
- 2007
- Full Text
- View/download PDF
40. Acute effects of febuxostat, a nonpurine selective inhibitor of xanthine oxidase, in pacing induced heart failure.
- Author
-
Hou M, Hu Q, Chen Y, Zhao L, Zhang J, and Bache RJ
- Subjects
- Animals, Blood Pressure drug effects, Dogs, Febuxostat, Thiazoles pharmacology, Cardiac Pacing, Artificial adverse effects, Heart Diseases etiology, Thiazoles adverse effects, Ventricular Function, Left drug effects, Xanthine Oxidase antagonists & inhibitors
- Abstract
We investigated whether xanthine oxidase inhibition with febuxostat enhances left ventricular (LV) function and improves myocardial high energy phosphates (HEP) in dogs with pacing-induced heart failure (CHF). Febuxostat (2.2 mg/kg over 10 minutes followed by 0.06 mg/kg/min) caused no change of LV function or myocardial oxygen consumption (MVO2) at rest or during treadmill exercise in normal dogs. In dogs with CHF, febuxostat increased LV dP/dtmax at rest and during heavy exercise (P < 0.05), indicating improved LV function with no change of MVO2. Myocardial adenosine triphosphate (ATP) and phosphocreatine (PCr) were examined using 31P nuclear magnetic resonance spectroscopy in the open chest state. In normal dogs, febuxostat increased PCr/ATP during basal conditions and during high workload produced by dobutamine + dopamine (P < 0.05). PCr/ATP was decreased in animals with CHF; in these animals, febuxostat (given after completing basal and high workload measurements with vehicle) tended to increase PCr/ATP during basal conditions with no effect during catecholamine stimulation. Thus, febuxostat improved LV performance in awake dogs with CHF, but caused only a trend toward increased PCr/ATP in the open chest state. It is possible that the antecedent high workload condition prior to drug administration blunted the effect of febuxostat on HEP in the CHF animals. Alternatively, beneficial effects of febuxostat on LV performance in the failing heart may not involve HEP.
- Published
- 2006
- Full Text
- View/download PDF
41. Profound bioenergetic abnormalities in peri-infarct myocardial regions.
- Author
-
Hu Q, Wang X, Lee J, Mansoor A, Liu J, Zeng L, Swingen C, Zhang G, Feygin J, Ochiai K, Bransford TL, From AH, Bache RJ, and Zhang J
- Subjects
- Adenosine Diphosphate metabolism, Adenosine Triphosphatases metabolism, Adenosine Triphosphate metabolism, Animals, Blotting, Western, Citrate (si)-Synthase metabolism, Collagen metabolism, Coronary Circulation physiology, Coronary Vessels physiology, Hemodynamics physiology, Ligation, Magnetic Resonance Spectroscopy, Mitochondria, Heart enzymology, Mitochondria, Heart metabolism, Myocardial Infarction physiopathology, Myocardium chemistry, Myoglobin metabolism, Oxygen Consumption physiology, Phosphocreatine metabolism, Swine, Ventricular Remodeling physiology, Energy Metabolism physiology, Myocardial Infarction metabolism, Myocardium metabolism
- Abstract
Regions of myocardial infarct (MI) are surrounded by a border zone (BZ) of normally perfused but dysfunctional myocardium. Although systolic dysfunction has been attributed to elevated wall stress in this region, there is evidence that intrinsic abnormalities of contractile performance exist in BZ myocardium. This study examined whether decreases of high-energy phosphates (HEP) and mitochondrial F(1)F(0)-ATPase (mtATPase) subunits typical of failing myocardium exist in BZ myocardium of compensated postinfarct remodeled hearts. Eight pigs were studied 6 wk after MI was produced by ligation of the left anterior descending coronary artery (LAD) distal to the second diagonal. Animals developed compensated LV remodeling with a decrease of ejection fraction from 54.6 +/- 5.4% to 31 +/- 2.1% (MRI) 5 wk after LAD occlusion. The remote zone (RZ) myocardium demonstrated modest decreases of ATP and mtATPase components. In contrast, BZ myocardium demonstrated profound abnormalities with ATP levels decreased to 42% of normal, and phosphocreatine-to-ATP ratio ((31)P-magnetic resonance spectroscopy) decreased from 2.06 +/- 0.19 in normal hearts to 1.07 +/- 0.10, with decreases in alpha-, beta-, OSCP, and IF(1) subunits of mtATPase, especially in the subendocardium. The reduction of myocardial creatine kinase isoform protein expression was also more severe in the BZ relative to the RZ myocardium. These abnormalities were independent of a change in mitochondrial content because the mitochondrial citrate synthase protein level was not different between the BZ and RZ. This regional heterogeneity of ATP content and expression of key enzymes in ATP production suggests that energetic insufficiency in the peri-infarct region may contribute to the transition from compensated LV remodeling to congestive heart failure.
- Published
- 2006
- Full Text
- View/download PDF
42. Measurement of myocardial free radical production during exercise using EPR spectroscopy.
- Author
-
Traverse JH, Nesmelov YE, Crampton M, Lindstrom P, Thomas DD, and Bache RJ
- Subjects
- Animals, Carbon Dioxide blood, Coronary Circulation physiology, Cyclic N-Oxides, Data Interpretation, Statistical, Dogs, Electron Spin Resonance Spectroscopy, Heart Rate physiology, Muscle, Skeletal metabolism, Nitrogen Oxides, Oxygen blood, Oxygen Consumption physiology, Rest physiology, Spin Labels, Free Radicals metabolism, Myocardium metabolism, Physical Exertion physiology
- Abstract
Exercise is associated with an increase in oxygen flux through the mitochondrial electron transport chain that has recently been demonstrated to increase the production of reactive oxygen species (ROS) in skeletal muscle. This study examined whether exercise also causes free radical production in the heart. We measured ROS production in seven chronically instrumented dogs during rest and treadmill exercise (6.4 km/h at 10 degrees grade; and heart rate, 204 +/- 3 beats/min) using electron paramagnetic resonance spectroscopy in conjunction with the spin trap alpha-phenyl-tert-butylnitrone (PBN) (0.14 mol/l) in blood collected from the aorta and coronary sinus (CS). To improve signal detection, the free radical adducts were deoxygenated over a nitrogen stream for 15 min and extracted with toluene. The hyperfine splitting constants of the radicals were alpha(N) = 13.7 G and alpha(H) = 1.0 G, consistent with an alkoxyl or carbon-centered radical. Resting aortic and CS PBN adduct concentrations were 6.7 and 6.3 x 10(8) arbitrary units (P = not significant). Both aortic and CS adduct concentrations increased during exercise, but there was no significant difference between the aortic and CS concentrations. Thus, in contrast to skeletal muscle, submaximal treadmill exercise did not result in detectable free radical production by the heart.
- Published
- 2006
- Full Text
- View/download PDF
43. Dimethylarginine dimethylaminohydrolase and endothelial dysfunction in failing hearts.
- Author
-
Chen Y, Li Y, Zhang P, Traverse JH, Hou M, Xu X, Kimoto M, and Bache RJ
- Subjects
- Animals, Arginine analogs & derivatives, Arginine metabolism, Blotting, Western, Cytochromes c metabolism, Dogs, Hemodynamics physiology, Immunohistochemistry, Isoenzymes metabolism, Myocardium enzymology, Myocardium metabolism, Oxygen Consumption physiology, Physical Exertion physiology, Rest physiology, Reverse Transcriptase Polymerase Chain Reaction, Subcellular Fractions enzymology, Vasodilation physiology, Amidohydrolases metabolism, Endothelium, Vascular physiopathology, Heart Failure enzymology, Heart Failure physiopathology
- Abstract
Congestive heart failure (CHF) is associated with impaired endothelium-dependent nitric oxide (NO)-mediated vasodilation (endothelial dysfunction). We hypothesized that coronary endothelial dysfunction in CHF may be due in part to decreased dimethylarginine dimethylaminohydrolase (DDAH), the enzyme that degrades endogenous inhibitors of NO synthase (NOS), including asymmetric dimethylarginine. Coronary blood flow and the endothelium-dependent vasodilator response to acetylcholine were studied in dogs in which CHF was produced by rapid ventricular pacing for 4 wk. Coronary flow and myocardial O2 consumption at rest and during treadmill exercise were decreased after development of CHF, and the vasodilator response to intracoronary acetylcholine (75 microg/min) was decreased by 39 +/- 5%. DDAH activity and DDAH isoform 2 (DDAH-2) protein content were decreased by 53 +/- 13% and 58 +/- 14%, respectively, in hearts with CHF, whereas endothelial NOS and DDAH isoform 1 (DDAH-1) were increased. Caveolin-1 and protein arginine N-methyltransferase 1, the enzyme that produces asymmetric dimethylarginine, were unchanged. Immunohistochemical staining showed DDAH-1 strongly expressed in coronary endothelium and smooth muscle and in the sarcolemma of cardiac myocytes. In cultured human endothelial cells, DDAH-1 was uniformly distributed in the cytosol and nucleus, whereas DDAH-2 was found only in the cytosol. Decreased DDAH activity and DDAH-2 protein expression may cause accumulation of endogenous inhibitors of endothelial NOS, thereby contributing to endothelial dysfunction in the failing heart.
- Published
- 2005
- Full Text
- View/download PDF
44. Nitric oxide regulation of myocardial O2 consumption and HEP metabolism.
- Author
-
Zhang J, Gong G, Ye Y, Guo T, Mansoor A, Hu Q, Ochiai K, Liu J, Wang X, Cheng Y, Iverson N, Lee J, From AH, Ugurbil K, and Bache RJ
- Subjects
- Animals, Dogs, Female, Magnetic Resonance Spectroscopy, Male, Adenosine Diphosphate metabolism, Energy Metabolism physiology, Myocardium metabolism, Nitric Oxide physiology, Oxygen Consumption physiology, Phosphocreatine metabolism
- Abstract
NO and O(2) compete at cytochrome-c oxidase, thus potentially allowing NO to modulate mitochondrial respiration. We previously observed a decrease of myocardial phosphocreatine (PCr)/ATP during very high cardiac work states, corresponding to an increase in cytosolic free ADP. This study tested the hypothesis that NO inhibition of respiration contributes to this increase of ADP. Infusion of dobutamine + dopamine (DbDp, each 20 microg.kg(-1).min(-1) iv) to more than double myocardial oxygen consumption (MVo(2)) in open-chest dogs caused a decrease of myocardial PCr/ATP measured with (31)P NMR from 2.04 +/- 0.09 to 1.85 +/- 0.08 (P < 0.05). Inhibition of NO synthesis with N(omega)-nitro-L-arginine (L-NNA), while catecholamine infusion continued, caused PCr/ATP to increase to the control value. In a second group of animals, L-NNA administered before catecholamine stimulation (reverse intervention of the first group) increased PCr/ATP during basal conditions. In these animals L-NNA did not prevent a decrease of PCr/ATP at the high cardiac work state but, relative to MVo(2), PCr/ATP was significantly higher after L-NNA. In a third group of animals, pharmacological coronary vasodilation with carbochromen was used to prevent changes in coronary flow that might alter endothelial NO production. In these animals L-NNA again restored depressed myocardial PCr/ATP during catecholamine infusion. The finding that inhibition of NO production increased PCr/ATP suggests that during very high work states NO inhibition of mitochondrial respiration requires ADP to increase to drive oxidative phosphorylation.
- Published
- 2005
- Full Text
- View/download PDF
45. Increased superoxide production causes coronary endothelial dysfunction and depressed oxygen consumption in the failing heart.
- Author
-
Chen Y, Hou M, Li Y, Traverse JH, Zhang P, Salvemini D, Fukai T, and Bache RJ
- Subjects
- Acetylcholine pharmacology, Animals, Blood Pressure, Computer Systems, Coronary Circulation drug effects, Coronary Vessels drug effects, Coronary Vessels metabolism, Dogs, Endothelium, Vascular metabolism, Enzyme Inhibitors pharmacology, Heart Failure metabolism, Isoenzymes metabolism, Malondialdehyde metabolism, Myocardium metabolism, Nitroarginine pharmacology, Organometallic Compounds pharmacology, Reverse Transcriptase Polymerase Chain Reaction, Superoxide Dismutase metabolism, Vasodilator Agents pharmacology, Ventricular Function, Left, Coronary Vessels physiopathology, Endothelium, Vascular physiopathology, Heart Failure physiopathology, Oxygen Consumption drug effects, Superoxides metabolism, Vasodilation
- Abstract
This study examined whether increased superoxide (O(2)(-).) production contributes to coronary endothelial dysfunction and decreased coronary blood flow (CBF) in congestive heart failure (CHF). To test this hypothesis, the effects of the low-molecular-weight SOD mimetic M40401 on CBF and myocardial oxygen consumption (MVo(2)) were examined in dogs during normal conditions and after CHF was produced by 4 wk of rapid ventricular pacing. The development of CHF was associated with decreases of left ventricular (LV) systolic pressure, maximum first derivative of LV pressure, MVo(2), and CBF at rest and during treadmill exercise as well as endothelial dysfunction with impaired vasodilation in response to intracoronary acetylcholine. M40401 increased CBF (18 +/- 5%, P < 0.01) and MVo(2) (14 +/- 6%, P < 0.01) in CHF dogs and almost totally reversed the impaired CBF response to acetylcholine. M40401 had no effect on acetylcholine-induced coronary vasodilation, CBF, or MVo(2) in normal dogs. Western blot analysis demonstrated that extracellular SOD (EC-SOD) was significantly decreased in CHF hearts, whereas mitochondrial Mn-containing SOD was increased. Cytosolic Cu/Zn-containing SOD was unchanged. Both increased O(2)(-). production and decreased vascular O(2)(-). scavenging ability by EC-SOD could have contributed to endothelial dysfunction in the failing hearts.
- Published
- 2005
- Full Text
- View/download PDF
46. Activation of p38 MAPK and increased glucose transport in chronic hibernating swine myocardium.
- Author
-
McFalls EO, Hou M, Bache RJ, Best A, Marx D, Sikora J, and Ward HB
- Subjects
- Animals, Biological Transport, Echocardiography, Enzyme Activation, Glucose metabolism, Glucose Transporter Type 4, Heart physiopathology, Myocardium enzymology, Nitric Oxide Synthase metabolism, Swine, Time Factors, p38 Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinases metabolism, Monosaccharide Transport Proteins metabolism, Muscle Proteins, Myocardial Stunning metabolism, Myocardium metabolism
- Abstract
In preconditioned myocardium, activation of the mitogen-activated protein kinase (MAPK) p38 leads to increased glucose uptake via enhanced GLUT-4 translocation. Glucose uptake is also increased in chronic hibernating myocardium, but the role of p38 MAPK and GLUT-4 translocation has not been studied. Nine swine underwent instrumentation of the proximal left anterior descending coronary artery (LAD) with a small, external constrictor. At 3 mo after instrumentation, myocardial glucose uptake by PET imaging was higher in the LAD than in the remote region under basal, fasted conditions (0.08 +/- 0.02 vs. 0.04 +/- 0.01 micromol.min(-1).g(-1), P < 0.05). Compared with the remote region, the LAD region demonstrated increased membrane-bound GLUT-4 relative to total content (61 +/- 04 vs. 45 +/- 06%, P < 0.05), higher glycogen (28.37 +/- 4.41 vs. 19.26 +/- 1.87 mg/g wet wt, P < 0.05), and increased inducible nitric oxide synthase (NOS) activity (1.43 +/- 0.34 vs. 0.51 +/- 0.21 activity/mg protein, P < 0.05). p38 MAPK was 47 +/- 14% higher in the LAD than in the remote region (P < 0.05) and correlated well with the absolute degree of GLUT-4 membrane-bound translocation (r = 0.81, P < 0.01), relative increase in glycogen (r = 0.70, P < 0.05), and total NOS activity (r = 0.68, P < 0.05). In chronic hibernating myocardial tissue, p38 MAPK activation is increased under basal fasted conditions and correlates well with the increased degree of GLUT-4 translocation, glycogen accumulation, and NOS activity. As in preconditioned myocardium, activation of p38 MAPK may play an important role in the metabolic adaptations that characterize chronic hibernating myocardium.
- Published
- 2004
- Full Text
- View/download PDF
47. Autologous stem cell transplantation for myocardial repair.
- Author
-
Liu J, Hu Q, Wang Z, Xu C, Wang X, Gong G, Mansoor A, Lee J, Hou M, Zeng L, Zhang JR, Jerosch-Herold M, Guo T, Bache RJ, and Zhang J
- Subjects
- Animals, Cell Differentiation, Cell Movement, Cells, Cultured, Fibrin, Mesoderm cytology, Myocardial Infarction pathology, Myocytes, Cardiac cytology, Neovascularization, Physiologic, Phenotype, Stem Cells cytology, Stem Cells physiology, Swine, Transduction, Genetic, Transplantation, Autologous, Myocardial Infarction physiopathology, Myocardial Infarction surgery, Stem Cell Transplantation, Wound Healing
- Abstract
Current therapies for heart failure due to transmural left ventricular (LV) infarction are limited. We have developed a novel patch method for delivering autologous bone marrow stem cells to sites of myocardial infarction for the purpose of improving LV function and preventing LV aneurysm formation. The patch consisted of a fibrin matrix seeded with autologous porcine mesenchymal stem cells labeled with lacZ. We applied this patch to a swine model of postinfarction LV remodeling. Myocardial infarction was produced by using a 60-min occlusion of the left anterior descending coronary artery distal to the first diagonal branch followed by reperfusion. Results were compared between eight pigs with stem cell patch transplantation, six pigs with the patch but no stem cells (P), and six pigs with left anterior descending coronary artery ligation alone (L). Magnetic resonance imaging data collected 19 +/- 1 days after the myocardial infarction indicated a significant increase of LV systolic wall thickening fraction in the infarct zone of transplanted hearts compared with P or L hearts. Blue X-gal staining was observed in the infarcted area of transplanted hearts. PCR amplification of specimens from the X-gal-positive area revealed the Ad5 RSV-lacZ vector fragment DNA sequence. Light microscopy demonstrated that transplanted cells had differentiated into cells with myocyte-like characteristics and a robust increase of neovascularization as evidenced by von Willebrand factor-positive angioblasts and capillaries in transplanted hearts. Thus this patch-based autologous stem cell procedure may serve as a therapeutic modality for myocardial repair.
- Published
- 2004
- Full Text
- View/download PDF
48. ET-A receptor activity restrains coronary blood flow in the failing heart.
- Author
-
Hou M, Chen Y, Traverse JH, Li Y, Barsoum M, and Bache RJ
- Subjects
- Animals, Coronary Circulation drug effects, Dogs, Dose-Response Relationship, Drug, Endothelin A Receptor Antagonists, Endothelin-1 pharmacology, Heart Failure metabolism, Oligopeptides pharmacology, Receptor, Endothelin A agonists, Coronary Circulation physiology, Heart Failure physiopathology, Receptor, Endothelin A physiology
- Abstract
Circulating levels of the potent vasoconstrictor peptide endothelin-1 (ET-1) are increased in congestive heart failure (CHF). Coronary blood flow and myocardial oxygen consumption (MVO2) are decreased in some models of CHF. This study tested the hypothesis that ET-1 induced coronary vasoconstriction limits oxygen availability in the failing heart. The effects of selective ET-A receptor blockade with BQ610 (5 microg/min, intracoronary) and selective ET-B receptor blockade with BQ788 (5 microg/min, intracoronary) on coronary blood flow were examined at rest and during graded treadmill exercise in 8 dogs in which congestive heart failure (CHF) had been produced by rapid ventricular pacing for three to four weeks. In animals with CHF, ET-B receptor blockade caused no change in left ventricular (LV) pressure or coronary blood flow. In contrast, ET-A blockade with BQ610 resulted in modest significant increases of coronary blood flow at rest (from 22.4 +/- 2.1 to 27.9 +/- 3.0 mL/min) and during two exercise stages (from 26.9 +/- 2.0 to 30.7 +/- 1.9 during stage 1 exercise and from 28.5 +/- 2.0 to 31.7 +/- 1.3 mL/min during stage 2; all P < 0.05), with an upward shift in the relationship between coronary flow and rate-pressure product. The increase in coronary flow produced by ET-A blockade was not associated with an increase of either myocardial oxygen uptake or LV dP/dt. Thus, although ET-A receptor blockade caused a modest increase in coronary flow, this did not result in an increase of MVO2, implying that ET-A-mediated coronary vasoconstriction did not limit oxygen uptake by the failing heart.
- Published
- 2004
- Full Text
- View/download PDF
49. Genomic profiling of the human heart before and after mechanical support with a ventricular assist device reveals alterations in vascular signaling networks.
- Author
-
Hall JL, Grindle S, Han X, Fermin D, Park S, Chen Y, Bache RJ, Mariash A, Guan Z, Ormaza S, Thompson J, Graziano J, de Sam Lazaro SE, Pan S, Simari RD, and Miller LW
- Subjects
- Adult, Aged, Chemokine CXCL12, Chemokines, CXC genetics, Chemokines, CXC metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Female, GATA4 Transcription Factor, Gene Expression Profiling, Genomics, Humans, Male, Membrane Proteins genetics, Membrane Proteins metabolism, Middle Aged, Molecular Sequence Data, Neuropilin-1 genetics, Neuropilin-1 metabolism, Phosphoproteins genetics, Phosphoproteins metabolism, Signal Transduction, Transcription Factors genetics, Transcription Factors metabolism, Heart-Assist Devices, Myocardium metabolism, RNA, Messenger metabolism
- Abstract
Mechanical unloading of the heart with a left ventricular assist device (LVAD) significantly decreases mortality in patients with heart failure. Moreover, it provides a human model to define the critical regulatory genes governing myocardial remodeling in response to significant reductions in wall stress. Statistical analysis of a gene expression library of 19 paired human heart samples harvested at the time of LVAD implant and again at explant revealed a set of 22 genes that were downregulated and 85 genes that were upregulated in response to mechanical unloading with a false discovery rate of less than 1%. The analysis revealed a high percentage of genes involved in the regulation of vascular networks including neuropilin-1 (a VEGF receptor), FGF9, Sprouty1, stromal-derived factor 1, and endomucin. Taken together these findings suggest that mechanical unloading alters the regulation of vascular organization and migration in the heart. In addition to vascular signaling networks, GATA-4 binding protein, a critical mediator of myocyte hypertrophy, was significantly downregulated following mechanical unloading. In summary, these findings may have important implications for defining the role of mechanical stretch and load on autocrine/paracrine signals directing vascular organization in the failing human heart and the role of GATA-4 in orchestrating reverse myocardial remodeling. This unbiased gene discovery approach in paired human heart samples has the potential to provide critical clues to the next generation of therapeutic treatments aimed at heart failure.
- Published
- 2004
- Full Text
- View/download PDF
50. Adenosine: a modulator of the cardiac response to stress.
- Author
-
Chen Y and Bache RJ
- Subjects
- Animals, Cardiac Output, Low etiology, Models, Cardiovascular, Myocardium metabolism, Rats, Receptors, Purinergic P1 metabolism, Signal Transduction, Adenosine physiology, Cardiomegaly etiology
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.