101
|
Depre C, Park JY, Shen YT, Zhao X, Qiu H, Yan L, Tian B, Vatner SF, Vatner DE. Molecular mechanisms mediating preconditioning following chronic ischemia differ from those in classical second window. Am J Physiol Heart Circ Physiol 2010; 299:H752-62. [PMID: 20581088 DOI: 10.1152/ajpheart.00147.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A major difference between experimental ischemic preconditioning (IPC), induced by brief ischemic episodes, and the clinical situation is that patients generally have repetitive episodes of ischemia. We used a swine model to examine differences in genes regulated by classical second-window IPC (SWOP) [two 10-min episodes of coronary artery occlusion (CAO) followed by 24 h reperfusion] compared with repetitive CAO/reperfusion (RCO), i.e., two 10-min CAO 12 h apart, and to repetitive coronary stenosis (RCS), six episodes of 90 min coronary stenosis 12 h apart (n = 5/group). All three models reduced infarct size by 60-85%, which was mediated by nitric oxide in SWOP but not in the other two models. We employed microarray analyses to discover additional molecular pathways intrinsic to models of repetitive ischemia and different from classical SWOP. There was an 85% homology in gene response between the RCO and RCS models, whereas SWOP was qualitatively different. Both RCO and RCS, but not SWOP, showed downregulation of genes encoding proteins involved in oxidative metabolism and upregulation of genes involved in protein synthesis, unfolded protein response, autophagy, heat shock response, protein secretion, and an activation of the NF-kappaB signaling pathway. Therefore, the regulated genes mediating IPC with repetitive ischemia differ radically from SWOP both quantitatively and qualitatively, showing that a repetitive pattern of ischemia, rather than the difference between no-flow vs. low-flow ischemia, dictates the genomic response of the heart. These findings illustrate new cardioprotective mechanisms developed by repetitive IPC, which are potentially more relevant to patients with chronic ischemic heart disease, who are subjected to repetitive episodes of ischemia.
Collapse
Affiliation(s)
- Christophe Depre
- Dept. of Cell Biology and Molecular Medicine, Univ. of Medicine and Dentistry of New Jersey, New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Affiliation(s)
- Linda R Peterson
- Cardiovascular Division, Department of Medicine, Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
103
|
Swynghedauw B, Delcayre C, Samuel JL, Mebazaa A, Cohen-Solal A. Molecular mechanisms in evolutionary cardiology failure. Ann N Y Acad Sci 2010; 1188:58-67. [PMID: 20201887 DOI: 10.1111/j.1749-6632.2009.05084.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Integration of the relevant evolutionary paradigm in cardiology has not yet been fully achieved: In the past, heart failure (HF) was mainly ascribed to infections, and the origins of cardiac hypertrophy (CH) were regarded as mechanical. Recent changes in lifestyle have both reduced the incidence of infections and increased lifespan, and HF is now seen as a complex disease--one that is still caused by mechanical disorder, but also associated with ischemia and senescence. The long-held view that CH serves to restore myocardial economy back to normal is still valid. The adaptive process is characterized by a quantitative and a qualitative fetal gene reprogramming, which is now being confirmed by recent advances in microRNA research. It underscores the fact CH is the physiologic reaction of the heart to a pathologic stimulus. The goal for therapy is economic, not inotropic. Another major issue is myocardial fibrosis, a major determinant of diastolic function and arrhythmias. Recent changes in lifestyle have crucially modified the context in which HF occurs.
Collapse
Affiliation(s)
- Bernard Swynghedauw
- Institut National de la Santé et de la Recherche Médicale, INSERM U942, Hôpital Lariboisière, Paris, France.
| | | | | | | | | |
Collapse
|
104
|
Taegtmeyer H, Sen S, Vela D. Return to the fetal gene program: a suggested metabolic link to gene expression in the heart. Ann N Y Acad Sci 2010; 1188:191-8. [PMID: 20201903 DOI: 10.1111/j.1749-6632.2009.05100.x] [Citation(s) in RCA: 305] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A hallmark of cardiac metabolism before birth is the predominance of carbohydrate use for energy provision. After birth, energy substrate metabolism rapidly switches to the oxidation of fatty acids. This switch accompanies the expression of "adult" isoforms of metabolic enzymes and other proteins. However, in a variety of pathophysiologic conditions, including hypoxia, ischemia, hypertrophy, atrophy, diabetes, and hypothyroidism, the postnatal heart returns to the "fetal" gene program. These adaptive mechanisms are also a feature of the failing heart muscle, where at a certain point this fetal-like reprogramming no longer suffices to support cardiac structure and function. We advance the hypothesis that in the postnatal heart, metabolic remodeling triggers the process through glycosylation of transcription factors, potentially protecting the stressed heart from irreversible functional impairment and programmed cell death. In other words, we propose a metabolic link to gene expression in the heart.
Collapse
Affiliation(s)
- Heinrich Taegtmeyer
- The University of Texas-Houston Medical School, Department of Internal Medicine, Division of Cardiology, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
105
|
Hearts of dystonia musculorum mice display normal morphological and histological features but show signs of cardiac stress. PLoS One 2010; 5:e9465. [PMID: 20209123 PMCID: PMC2830884 DOI: 10.1371/journal.pone.0009465] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Accepted: 02/08/2010] [Indexed: 12/31/2022] Open
Abstract
Dystonin is a giant cytoskeletal protein belonging to the plakin protein family and is believed to crosslink the major filament systems in contractile cells. Previous work has demonstrated skeletal muscle defects in dystonin-deficient dystonia musculorum (dt) mice. In this study, we show that the dystonin muscle isoform is localized at the Z-disc, the H zone, the sarcolemma and intercalated discs in cardiac tissue. Based on this localization pattern, we tested whether dystonin-deficiency leads to structural defects in cardiac muscle. Desmin intermediate filament, microfilament, and microtubule subcellular organization appeared normal in dt hearts. Nevertheless, increased transcript levels of atrial natriuretic factor (ANF, 66%) β-myosin heavy chain (beta-MHC, 95%) and decreased levels of sarcoplasmic reticulum calcium pump isoform 2A (SERCA2a, 26%), all signs of cardiac muscle stress, were noted in dt hearts. Hearts from two-week old dt mice were assessed for the presence of morphological and histological alterations. Heart to body weight ratios as well as left ventricular wall thickness and left chamber volume measurements were similar between dt and wild-type control mice. Hearts from dt mice also displayed no signs of fibrosis or calcification. Taken together, our data provide new insights into the intricate structure of the sarcomere by situating dystonin in cardiac muscle fibers and suggest that dystonin does not significantly influence the structural organization of cardiac muscle fibers during early postnatal development.
Collapse
|
106
|
Koike M, Kojima H, Fujimiya M, Matsubayashi K, Aimi Y, Kimura H, Asai T. Transfer of bone marrow progenitors prevents coronary insufficiency and systolic dysfunction in the mechanical unloaded heart in mice. J Surg Res 2010; 171:47-57. [PMID: 20451917 DOI: 10.1016/j.jss.2010.01.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 01/21/2010] [Accepted: 01/25/2010] [Indexed: 12/19/2022]
Abstract
BACKGROUND Left ventricular-assist device (LVAD) can lead to improvement of cardiac performance in a subset of patients, but chronic mechanical unloading in this fashion may result in left ventricular (LV)-atrophy and impaired functional recovery. Here, we evaluate the efficacy of transferring bone-marrow KSL cells (Lin-/c-kit+/Sca1+), a fraction containing endothelial progenitor cells, for preventing LV-atrophy and malfunction in a mouse model of mechanical unloading of the heart. MATERIALS AND METHODS Recipients of an isogenic heart transplant received intramyocardial isogenic KSL cells or PBS in three different locations of the left ventricle (LV). Coronary blood flow and LV systolic function were evaluated by echocardiography, and morphologic changes were analyzed on d 7 and 56. RESULTS PBS-treated mice showed severe systolic dysfunction and large thrombi in LV at both time points. In contrast, KSL cell transfer markedly reduced systolic dysfunction and thrombus size. Furthermore, in comparison with PBS control, KSL recipients had increased coronary blood flow (3-fold, P < 0.01) accompanied by increased LV capillary density and muscle mass. CONCLUSIONS These results indicate that intramyocardial transfer of bone marrow KSL cells significantly protects against coronary insufficiency and systolic dysfunction in the chronic LV-unloading heart, suggesting that this approach may have clinical potential as a combination therapy with LVAD.
Collapse
Affiliation(s)
- Masato Koike
- Department of Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | | | | | | | | | | |
Collapse
|
107
|
Ito K, Kagaya Y, Shimokawa H. Thyroid hormone and chronically unloaded hearts. Vascul Pharmacol 2009; 52:138-41. [PMID: 19879960 DOI: 10.1016/j.vph.2009.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 10/09/2009] [Indexed: 11/25/2022]
Abstract
The heart is subjected to chronic mechanical unloading during prolonged spaceflight and microgravity. The heart in patients with end-stage heart failure is also unloaded in prolonged duration after left ventricular assist devices (LVAD) are implanted. Heterotopic heart transplantation in rats is an established model of chronic cardiac unloading, and has been used to investigate the effects of chronic cardiac unloading on the heart. Observations that have been found using this experimental model are as follow. Chronic cardiac unloading induces time-dependent depressions of Ca2+ handling and myocyte contractility, which are associated with the shift of myosin heavy chain (MHC) isozymes and altered expressions of Ca2+ cycling-related proteins. Treatment with the physiological treatment dose of thyroid hormone restores the expression levels of Ca2+ cycling-related proteins, Ca2+ handling, and contractile function of cardiac myocytes in chronically unloaded hearts. Although future studies are required to determine precise mechanisms of the beneficial effects of thyroid hormone on chronically unloaded hearts, these observations may have clinical implications in the future for chronic cardiac unloading in the space industry as well as in the treatment of patients with end-stage heart failure supported by LVAD.
Collapse
Affiliation(s)
- Kenta Ito
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | |
Collapse
|
108
|
Bugger H, Schwarzer M, Chen D, Schrepper A, Amorim PA, Schoepe M, Nguyen TD, Mohr FW, Khalimonchuk O, Weimer BC, Doenst T. Proteomic remodelling of mitochondrial oxidative pathways in pressure overload-induced heart failure. Cardiovasc Res 2009; 85:376-84. [DOI: 10.1093/cvr/cvp344] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
109
|
Kassiotis C, Ballal K, Wellnitz K, Vela D, Gong M, Salazar R, Frazier OH, Taegtmeyer H. Markers of autophagy are downregulated in failing human heart after mechanical unloading. Circulation 2009; 120:S191-7. [PMID: 19752367 DOI: 10.1161/circulationaha.108.842252] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Autophagy is a molecular process that breaks down damaged cellular organelles and yields amino acids for de novo protein synthesis or energy provision. Mechanical unloading with a left ventricular assist device (LVAD) decreases the energy demand of the failing human heart. We tested the hypothesis that LVAD support reverses activation of autophagy. METHODS AND RESULTS Paired biopsy samples of left ventricular myocardium were obtained from 9 patients with idiopathic dilated cardiomyopathy (mean duration of LVAD support, 214 days) at the time of implantation and explantation of the LVAD. Transcript and protein levels of markers and mediators of autophagy and apoptosis were measured by quantitative reverse-transcription polymerase chain reaction and Western blotting. TUNEL assays, C9 immunohistochemistry, and 20S proteasome activity assays were also performed. Mechanical unloading significantly decreased mRNA transcript levels of Beclin-1, autophagy-related gene 5 (Atg5), and microtubule-associated protein-1 light chain-3 (MAP1-LC3 or LC3; P<0.02). Protein levels of Beclin-1, Atg5-Atg12 conjugate, and LC3-II were also significantly reduced after LVAD support (P<0.05). A significant increase in 20S proteasome activity was observed with unloading, in parallel to the decrease in autophagic markers. Although BNIP3 and the ratio of activated caspase 3 to procaspase 3 increased after LVAD support, Bcl-2 and TUNEL-positive nuclei were not significantly different between samples. CONCLUSIONS Mechanical unloading of the failing human heart decreases markers of autophagy. These findings suggest that autophagy may be an adaptive mechanism in the failing heart, and this phenomenon is attenuated by LVAD support.
Collapse
|
110
|
Micronutrient Deficiencies. J Am Coll Cardiol 2009; 54:1660-73. [DOI: 10.1016/j.jacc.2009.08.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 08/24/2009] [Accepted: 08/25/2009] [Indexed: 01/08/2023]
|
111
|
Hu CL, Chandra R, Ge H, Pain J, Yan L, Babu G, Depre C, Iwatsubo K, Ishikawa Y, Sadoshima J, Vatner SF, Vatner DE. Adenylyl cyclase type 5 protein expression during cardiac development and stress. Am J Physiol Heart Circ Physiol 2009; 297:H1776-82. [PMID: 19734365 DOI: 10.1152/ajpheart.00050.2009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adenylyl cyclase (AC) types 5 and 6 (AC5 and AC6) are the two major AC isoforms expressed in the mammalian heart that mediate signals from beta-adrenergic receptor stimulation. Because of the unavailability of isoform-specific antibodies, it is difficult to ascertain the expression levels of AC5 protein in the heart. Here we demonstrated the successful generation of an AC5 isoform-specific mouse monoclonal antibody and studied the expression of AC5 protein during cardiac development in different mammalian species. The specificity of the antibody was confirmed using heart and brain tissues from AC5 knockout mice and from transgenic mice overexpressing AC5. In mice, the AC5 protein was highest in the brain but was also detectable in all organs studied, including the heart, brain, lung, liver, stomach, kidney, skeletal muscle, and vascular tissues. Western blot analysis showed that AC5 was most abundant in the neonatal heart and declined to basal levels in the adult heart. AC5 protein increased in the heart with pressure-overload left ventricular hypertrophy. Thus this new AC5 antibody demonstrated that this AC isoform behaves similarly to fetal type genes, such as atrial natriuretic peptide; i.e., it declines with development and increases with pressure-overload hypertrophy.
Collapse
Affiliation(s)
- Che-Lin Hu
- Department of Cell Biology and Molecular Medicine and the Cardiovascular Research Institute at the University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Nielsen JM, Kristiansen SB, Nørregaard R, Andersen CL, Denner L, Nielsen TT, Flyvbjerg A, Bøtker HE. Blockage of receptor for advanced glycation end products prevents development of cardiac dysfunction in db/db type 2 diabetic mice. Eur J Heart Fail 2009; 11:638-47. [PMID: 19502378 DOI: 10.1093/eurjhf/hfp070] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Activation of the receptor for advanced glycation end products (RAGE) is associated with long-term complications in diabetes mellitus. In this study, we tested whether RAGE activation in the diabetic myocardium is implicated in the development of cardiac dysfunction. METHODS AND RESULTS Using MRI and conductance catheter techniques, we evaluated cardiac function in a type 2 diabetic mouse model (db/db), and assessed the effect of blocking RAGE with a RAGE antibody. Gene expressions were evaluated in samples of myocardial tissue. Diabetic db/db mice demonstrated an accelerated age-dependent deterioration in cardiac function associated with altered expression of genes related to cardiac structure and function. Blockage of RAGE signalling prevented the reduction in systolic function (preload recruitable stroke work: 109.8 +/- 13.8 vs. 94.5 +/- 14.9 mmHg/microL, P = 0.04) and development of increased LV diastolic chamber stiffness (0.18 +/- 0.05 vs. 0.27 +/- 0.07 mmHg, P = 0.01). The cardiac expression of collagen (col1a1) was reduced by approximately 45% and the expression of myosin was switched from the foetal isoform (MHCbeta) to the adult isoform (MHCalpha). CONCLUSION Activation of RAGE is a significant pathogenetic mechanism for the development of cardiac dysfunction in type 2 diabetes. The underlying mechanisms involve not only the passive biophysical properties of the myocardium but also myocyte function.
Collapse
Affiliation(s)
- Jan M Nielsen
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus N, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
113
|
Russell RR. Myocardial metabolic imaging: Viability and beyond. CURRENT CARDIOVASCULAR IMAGING REPORTS 2009. [DOI: 10.1007/s12410-009-0027-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
114
|
LACHANCE DOMINIC, CHAMPETIER SERGE, PLANTE ÉRIC, BOUCHARD-THOMASSIN ANDRÉEANNE, ROUSSEL ÉLISE, COUET JACQUES, ARSENAULT MARIE. Effects of Exercise in Volume Overload. Med Sci Sports Exerc 2009; 41:1230-8. [DOI: 10.1249/mss.0b013e318195b938] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
115
|
Brinks H, Tevaearai H, Mühlfeld C, Bertschi D, Gahl B, Carrel T, Giraud MN. Contractile function is preserved in unloaded hearts despite atrophic remodeling. J Thorac Cardiovasc Surg 2009; 137:742-6. [PMID: 19258100 DOI: 10.1016/j.jtcvs.2008.09.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 07/31/2008] [Accepted: 09/06/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Recent studies have shown that mechanically unloading a failing heart may induce reverse remodeling and functional improvement. However, these benefits may be balanced by an unloading-related remodeling including myocardial atrophy that might lead to decrease in function. Using a model of heterotopic heart transplantation, we aimed to characterize the myocardial changes induced by long-term unloading. MATERIAL AND METHODS Macroscopic as well as cellular and functional changes were followed in normal hearts unloaded for a 3-month period. Microscopic parameters were evaluated with stereologic methodology. Myocardial contractile function was quantified with a Langendorff isolated, perfused heart technique. RESULTS Atrophy was macroscopically obvious and accompanied by a 67% reduction of the myocyte volume and a 43% reduction of the interstitial tissue volume, thus accounting for a shift of the myocyte/connective tissue ratio in favor of noncontractile tissue. The absolute number of cardiomyocyte nuclei decreased from 64.7 +/- 5.1 x 10(7) in controls to 22.6 +/- 3.7 x 10(7) (30 days) and 21.6 +/- 3.1 x 10(7) (90 days) after unloading (P < .05). The numeric nucleic density in the unloaded myocardium, as well as the mean cardiomyocyte volume per cardiomyocyte nucleus, remained constant throughout the 90 days of observation. Functional data indicated an increase in ventricular stiffness, although contractile function was preserved, as confirmed by unaltered maximal developed pressure and increased contractility (maximum rate of left ventricular pressure development) and relaxation (minimum rate of left ventricular pressure development). CONCLUSION Atrophic remodeling involves both the myocyte and interstitial tissue compartment. These data suggest that although there is decreased myocardial volume and increased stiffness, contractile capacity is preserved in the long-term unloaded heart.
Collapse
Affiliation(s)
- Henriette Brinks
- Department of Cardiovascular Surgery, Inselspital, Berne University Hospital and University of Berne, Berne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
116
|
Gallego-Delgado J, Connolly SB, Lázaro A, Sadlier D, Kieran NE, Sugrue DD, Doran P, Brady HR, Osende J, Egido J. Transcriptome of hypertension-induced left ventricular hypertrophy and its regression by antihypertensive therapies. Hypertens Res 2009; 32:347-57. [DOI: 10.1038/hr.2009.27] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
117
|
Bashir A, Gropler RJ. Translation of myocardial metabolic imaging concepts into the clinics. Cardiol Clin 2009; 27:291-310, Table of Contents. [PMID: 19306771 DOI: 10.1016/j.ccl.2008.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Flexibility in myocardial substrate metabolism for energy production is fundamental to cardiac health. This loss in plasticity or flexibility leads to overdependence on the metabolism of an individual category of substrates, with the predominance in fatty acid metabolism characteristic of diabetic heart disease and the accelerated glucose use associated with pressure-overload left ventricular hypertrophy being prime examples. There is a strong demand for accurate noninvasive imaging approaches of myocardial substrate metabolism that can facilitate the crosstalk between the bench and the bedside, leading to improved patient management paradigms. In this article potential future applications of metabolic imaging, particularly radionuclide approaches, for assessment of cardiovascular disease are discussed.
Collapse
Affiliation(s)
- Adil Bashir
- Division of Radiological Sciences, Cardiovascular Imaging Laboratory, Edward Mallinckrodt Institute of Radiology, St Louis, MO 63110, USA
| | | |
Collapse
|
118
|
Abstract
The development of heart failure is associated with alterations in the expression of a wide variety of structural, signaling, and metabolic proteins. One such effect is the downregulation of uncoupling proteins in the setting of heart failure. This group of proteins regulates the mitochondrial membrane potential and therefore plays a role in mitochondrial energy metabolism as well as reactive oxygen species generation by the mitochondria. This review discusses the role of uncoupling proteins in regulating oxidant stress and implications with respect to the pathogenesis of heart failure and potential treatments.
Collapse
|
119
|
Refaat M, Chemaly E, Lebeche D, Gwathmey JK, Hajjar RJ. Ventricular arrhythmias after left ventricular assist device implantation. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2008; 31:1246-52. [PMID: 18811803 DOI: 10.1111/j.1540-8159.2008.01173.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Left ventricular assist devices (LVADs) have been used as a bridge to cardiac transplantation and as destination therapy in patients with advanced heart failure. The period after LVAD support is associated with ventricular arrhythmias (VAs) despite ventricular unloading and such VAs can have a detrimental effect on survival. Despite the increasing use of LVAD, little is known regarding post-LVAD VAs at the molecular level and in vivo. METHODS Forty-two patients who received LVAD over a 24-month period were evaluated and grouped on the basis of the presence or absence of VAs during LVAD support. We completed a comparative microarray analyses between six patients who developed ventricular tachycardia (VT) or ventricular fibrillation (VF) after LVAD support and six patients who did not develop VAs after LVAD. RESULTS VAs occurred in 15 patients (35.7%) during LVAD support at a median post-LVAD day of 25.2. VAs were strongly associated with nonusage of a beta-blocker post-LVAD (odds ratio of 7.04, P-value = 0.001). Analysis of a subset of patients who had VT or VF after LVAD placement showed a decrease in the expression of connexin 43 (0.48 +/- 0.07), Na+/K+-ATPase (0.60 +/- 0.05), and voltage-gated K+ channel Kv4.3 (0.42 +/- 0.04), and an increase in Na+/Ca2+ exchanger (2.2 +/- 0.4) and the structural genes: Titin (2.1 +/- 0.2), laminin (1.7 +/- 0.4), calsequestrin (1.8 +/- 0.5), skeletal muscle isoform of troponin T (5.1 +/- 0.9), and skeletal muscle isoform of troponin I (3.9 +/- 0.7). CONCLUSION After LVAD, the increased risk of VAs is strongly associated with nonusage of beta-blocker postoperatively.
Collapse
Affiliation(s)
- Marwan Refaat
- Divisions of Cardiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | |
Collapse
|
120
|
Mass spectrometric demonstration of the presence of liver carnitine palmitoyltransferase-I (CPT-I) in heart mitochondria of adult rats. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2008; 1794:431-7. [PMID: 19111953 DOI: 10.1016/j.bbapap.2008.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 10/17/2008] [Accepted: 11/07/2008] [Indexed: 01/03/2023]
Abstract
The carnitine palmitoyltransferase-I (CPT-I) enzymes catalyze the regulated step in overall mitochondrial fatty acid oxidation. The liver and muscle isoforms are expressed in liver and skeletal muscle respectively with the isoforms exhibiting different kinetic properties and apparent molecular weight masses. In contrast, the heart expresses both isoforms at the mRNA level. However, for the expression of the liver isoform at the protein level only indirect evidence is available, such as tagging with radiolabeled CPT-I inhibitors followed by SDS-PAGE separation and kinetic analysis using inhibitors. The importance of fatty acid oxidation in the heart and the potential regulation via the liver isoform of CPT-I demands proof of the liver isoform in the heart. Using a proteomic approach in the present study we demonstrate that rat heart mitochondria (a) contain both the muscle and liver isoforms; (b) both proteins retain their C- and N-termini; (c) the N-terminal alanine residues are acetylated; (d) and in rat heart mitochondria the liver isoform is phosphorylated on tyrosine 281. By providing amino acid sequence information this is the first unequivocal demonstration that the liver isoform of CPT-I is expressed at the protein level in adult rat heart mitochondria and that the apparent smaller molecular size of the muscle isoform is not due to proteolytic truncation.
Collapse
|
121
|
Thakker GD, Frangogiannis NG, Zymek PT, Sharma S, Raya JL, Barger PM, Taegtmeyer H, Entman ML, Ballantyne CM. Increased myocardial susceptibility to repetitive ischemia with high-fat diet-induced obesity. Obesity (Silver Spring) 2008; 16:2593-600. [PMID: 18833212 PMCID: PMC3049112 DOI: 10.1038/oby.2008.414] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity and diabetes are frequently associated with cardiovascular disease. When a normal heart is subjected to brief/sublethal repetitive ischemia and reperfusion (I/R), adaptive responses are activated to preserve cardiac structure and function. These responses include but are not limited to alterations in cardiac metabolism, reduced calcium responsiveness, and induction of antioxidant enzymes. In a model of ischemic cardiomyopathy inducible by brief repetitive I/R, we hypothesized that dysregulation of these adaptive responses in diet-induced obese (DIO) mice would contribute to enhanced myocardial injury. DIO C57BL/6J mice were subjected to 15 min of daily repetitive I/R while under short-acting anesthesia, a protocol that results in the development of fibrotic cardiomyopathy. Cardiac lipids and candidate gene expression were analyzed at 3 days, and histology at 5 days of repetitive I/R. Total free fatty acids (FFAs) in the cardiac extracts of DIO mice were significantly elevated, reflecting primarily the dietary fatty acid (FA) composition. Compared with lean controls, cardiac FA oxidation (FAO) capacity of DIO mice was significantly higher, concurrent with increased expression of FA metabolism gene transcripts. Following 15 min of daily repetitive I/R for 3 or 5 days, DIO mice exhibited increased susceptibility to I/R and, in contrast to lean mice, developed microinfarction, which was associated with an exaggerated inflammatory response. Repetitive I/R in DIO mice was associated with more profound significant downregulation of FA metabolism gene transcripts and elevated FFAs and triglycerides. Maladaptive metabolic changes of FA metabolism contribute to enhanced myocardial injury in diet-induced obesity.
Collapse
Affiliation(s)
- Geeta D. Thakker
- Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, Houston, TX
| | | | - Pawel T. Zymek
- Section of Cardiovascular Sciences, Baylor College of Medicine, Houston, TX
| | - Saumya Sharma
- Division of Cardiology, Department of Internal Medicine, The University of Texas Medical School at Houston, Houston, TX
| | - Joe L. Raya
- Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, Houston, TX
| | - Philip M. Barger
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, The University of Texas Medical School at Houston, Houston, TX
| | - Mark L. Entman
- Section of Cardiovascular Sciences, Baylor College of Medicine, Houston, TX
| | - Christie M. Ballantyne
- Section of Atherosclerosis and Vascular Medicine, Baylor College of Medicine, Houston, TX
- Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart Center, Houston, TX
| |
Collapse
|
122
|
Mearini G, Schlossarek S, Willis MS, Carrier L. The ubiquitin–proteasome system in cardiac dysfunction. Biochim Biophys Acta Mol Basis Dis 2008; 1782:749-63. [DOI: 10.1016/j.bbadis.2008.06.009] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 06/12/2008] [Accepted: 06/18/2008] [Indexed: 12/31/2022]
|
123
|
Schwoerer AP, Neuber C, Schmechel A, Melnychenko I, Mearini G, Boknik P, Kirchhefer U, Schmitz W, Ehmke H, Eschenhagen T, El-Armouche A. Mechanical unloading of the rat heart involves marked changes in the protein kinase–phosphatase balance. J Mol Cell Cardiol 2008; 45:846-52. [DOI: 10.1016/j.yjmcc.2008.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 09/08/2008] [Accepted: 09/08/2008] [Indexed: 02/06/2023]
|
124
|
Alterations in circulating activin A, GDF-15, TGF-beta3 and MMP-2, -3, and -9 during one year of left ventricular reverse remodelling in patients operated for severe aortic stenosis. Eur J Heart Fail 2008; 10:1201-7. [PMID: 18996047 DOI: 10.1016/j.ejheart.2008.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2008] [Accepted: 09/25/2008] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Patients with aortic stenosis (AS) develop left ventricular remodelling with cardiomyocyte hypertrophy and increased fibrosis. Following aortic valve replacement (AVR) reverse remodelling usually takes place. AIMS To examine circulating levels of members of the transforming growth factor (TGF) beta superfamily and matrix metalloproteinases (MMP), known to have important effects on hypertrophy and extracellular matrix, in patients operated for AS. METHODS Circulating levels of activin A, GDF-15, TGF-beta3, MMP-2, -3, and -9 were measured in twenty-two patients undergoing AVR preoperatively, and 2 days, six months and 12 months postoperatively. Echocardiography and a six minute walking test evaluated reverse remodelling and physical performance. RESULTS Activin A increased at six (1081.00+/-98.05 pg/ml, p<0.05) and twelve months (1263.09+/-141.43 pg/ml, p<0.05) compared to the preoperative value (855.00+/-76.30 pg/ml) and correlated negatively to physical performance. The preoperative value was also increased compared to controls (639.54+/-63.05 pg/ml, p<0.05). GDF-15, MMP-3 and -9 were all increased at two days postoperatively (p<0.05). MMP-3 correlated with left ventricular end diastolic dimension (p<0.05). MMP-2 did not change during the study period. TGF-beta3 was only slightly reduced at six months postoperatively. CONCLUSION The observed alteration in circulating levels of members of the TGF-beta superfamily and MMPs might play a role in the reverse remodelling process following AVR for AS.
Collapse
|
125
|
Schwoerer AP, Melnychenko I, Goltz D, Hedinger N, Broichhausen I, El-Armouche A, Eschenhagen T, Volk T, Ehmke H. Unloaded rat hearts in vivo express a hypertrophic phenotype of cardiac repolarization. J Mol Cell Cardiol 2008; 45:633-41. [DOI: 10.1016/j.yjmcc.2008.02.271] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 02/11/2008] [Accepted: 02/12/2008] [Indexed: 11/25/2022]
|
126
|
van Bilsen M, van Nieuwenhoven FA, van der Vusse GJ. Metabolic remodelling of the failing heart: beneficial or detrimental? Cardiovasc Res 2008; 81:420-8. [PMID: 18854380 DOI: 10.1093/cvr/cvn282] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The failing heart is characterized by alterations in energy metabolism, including mitochondrial dysfunction and a reduction in fatty acid (FA) oxidation rate, which is partially compensated by an increase in glucose utilization. Together, these changes lead to an impaired capacity to convert chemical energy into mechanical work. This has led to the concept that supporting cardiac energy conversion through metabolic interventions provides an important adjuvant therapy for heart failure. The potential success of such a therapy depends on whether the shift from FA towards glucose utilization should be considered beneficial or detrimental, a question still incompletely resolved. In this review, the current status of the literature is evaluated and possible causes of observed discrepancies are discussed. It is cautiously concluded that for the failing heart, from a therapeutic point of view, it is preferable to further stimulate glucose oxidation rather than to normalize substrate metabolism by stimulating FA utilization. Whether this also applies to the pre-stages of cardiac failure remains to be established.
Collapse
Affiliation(s)
- Marc van Bilsen
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, PO Box 616, 6200 MD Maastricht, the Netherlands.
| | | | | |
Collapse
|
127
|
Kassiotis C, Rajabi M, Taegtmeyer H. Metabolic reserve of the heart: the forgotten link between contraction and coronary flow. Prog Cardiovasc Dis 2008; 51:74-88. [PMID: 18634919 DOI: 10.1016/j.pcad.2007.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myocardial energy substrate metabolism entails a complex system of enzyme catalyzed reactions, in which the heart efficiently converts chemical to mechanical energy. The system is highly regulated and responsive to changes in workload as well as in substrate and hormone supply to the heart. Akin to the terms "contractile reserve" and "coronary flow reserve" we propose the term "metabolic reserve" to reflect the heart's capacity to respond to increases in workload. The heart's metabolic response to inotropic stimulation involves the ability to increase oxidative metabolism over a wide range, by activating the oxidation of glycogen and carbohydrate substrates. Here we review the known biochemical mechanisms responsible for those changes. Specifically, we explore the notion that disturbances in the metabolic reserve result in contractile dysfunction of the stressed heart.
Collapse
Affiliation(s)
- Christos Kassiotis
- Department of Internal Medicine, Division of Cardiology, The University of Texas Houston Medical School, Houston, TX 77030, USA
| | | | | |
Collapse
|
128
|
|
129
|
Cardiovascular genetic medicine: genomic assessment of prognosis and diagnosis in patients with cardiomyopathy and heart failure. J Cardiovasc Transl Res 2008; 1:225-31. [PMID: 20559924 DOI: 10.1007/s12265-008-9044-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 06/12/2008] [Indexed: 12/22/2022]
Abstract
In the last half century, epidemiologic studies and basic science investigations revealed that hypertension (Kannel et al., Ann Intern Med 55:33-50, 1961), hyperlipidemia (Dawber et al., Am J Public Health Nations Health 49:1349-1356, 1959), diabetes (Kannel et al., Am J Cardiol 34(1):29-34, 1974), smoking (Dawber et al., Am J Public Health Nations Health 49:1349-1356, 1959), and inflammation (Rossmann et al., Exp Gerontol 43(3):229-237, 2008) posed increased risk for cardiovascular disease. These associations served both as risk factors and offered insight into disease pathophysiology. Currently, it is increasingly appreciated that polygenic factors may also play a role as etiologic or risk factors (Chakravarti and Little, Nature 421(6921):412-414, 2003; Dorn and Molkentin, Circulation 109(2):150-158, 2004). Recent technologic advances in genomic screening make the search for these factors possible, and robust technologies are now available for both entire genome screening for expression or single nucleotide polymorphisms. In this paper, we review the basic principles of gene expression and molecular signature analysis in the context of potential clinical applications of transcriptomics.
Collapse
|
130
|
Hedhli N, Lizano P, Hong C, Fritzky LF, Dhar SK, Liu H, Tian Y, Gao S, Madura K, Vatner SF, Depre C. Proteasome inhibition decreases cardiac remodeling after initiation of pressure overload. Am J Physiol Heart Circ Physiol 2008; 295:H1385-93. [PMID: 18676687 DOI: 10.1152/ajpheart.00532.2008] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We tested the possibility that proteasome inhibition may reverse preexisting cardiac hypertrophy and improve remodeling upon pressure overload. Mice were submitted to aortic banding and followed up for 3 wk. The proteasome inhibitor epoxomicin (0.5 mg/kg) or the vehicle was injected daily, starting 2 wk after banding. At the end of the third week, vehicle-treated banded animals showed significant (P<0.05) increase in proteasome activity (PA), left ventricle-to-tibial length ratio (LV/TL), myocyte cross-sectional area (MCA), and myocyte apoptosis compared with sham-operated animals and developed signs of heart failure, including increased lung weight-to-TL ratio and decreased ejection fraction. When compared with that group, banded mice treated with epoxomicin showed no increase in PA, a lower LV/TL and MCA, reduced apoptosis, stabilized ejection fraction, and no signs of heart failure. Because overload-mediated cardiac remodeling largely depends on the activation of the proteasome-regulated transcription factor NF-kappaB, we tested whether epoxomicin would prevent this activation. NF-kappaB activity increased significantly upon overload, which was suppressed by epoxomicin. The expression of NF-kappaB-dependent transcripts, encoding collagen types I and III and the matrix metalloprotease-2, increased (P<0.05) after banding, which was abolished by epoxomicin. The accumulation of collagen after overload, as measured by histology, was 75% lower (P<0.05) with epoxomicin compared with vehicle. Myocyte apoptosis increased by fourfold in hearts submitted to aortic banding compared with sham-operated hearts, which was reduced by half upon epoxomicin treatment. Therefore, we propose that proteasome inhibition after the onset of pressure overload rescues ventricular remodeling by stabilizing cardiac function, suppressing further progression of hypertrophy, repressing collagen accumulation, and reducing myocyte apoptosis.
Collapse
Affiliation(s)
- Nadia Hedhli
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Heidecker B, Kasper EK, Wittstein IS, Champion HC, Breton E, Russell SD, Kittleson MM, Baughman KL, Hare JM. Transcriptomic biomarkers for individual risk assessment in new-onset heart failure. Circulation 2008; 118:238-46. [PMID: 18591436 DOI: 10.1161/circulationaha.107.756544] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Prediction of prognosis remains a major unmet need in new-onset heart failure (HF). Although several clinical tests are in use, none accurately distinguish between patients with poor versus excellent survival. We hypothesized that a transcriptomic signature, generated from a single endomyocardial biopsy, could serve as a novel prognostic biomarker in HF. METHODS AND RESULTS Endomyocardial biopsy samples and clinical data were collected from all patients presenting with new-onset HF from 1997 to 2006. Among a total of 350 endomyocardial biopsy samples, 180 were identified as idiopathic dilated cardiomyopathy. Patients with phenotypic extremes in survival were selected: good prognosis (event-free survival for at least 5 years; n=25) and poor prognosis (events [death, requirement for left ventricular assist device, or cardiac transplant] within the first 2 years of presentation with HF symptoms; n=18). We used human U133 Plus 2.0 microarrays (Affymetrix) and analyzed the data with significance analysis of microarrays and prediction analysis of microarrays. We identified 46 overexpressed genes in patients with good versus poor prognosis, of which 45 genes were selected by prediction analysis of microarrays for prediction of prognosis in a train set (n=29) with subsequent validation in test sets (n=14 each). The biomarker performed with 74% sensitivity (95% CI 69% to 79%) and 90% specificity (95% CI 87% to 93%) after 50 random partitions. CONCLUSIONS These findings suggest the potential of transcriptomic biomarkers to predict prognosis in patients with new-onset HF from a single endomyocardial biopsy sample. In addition, our findings offer potential novel therapeutic targets for HF and cardiomyopathy.
Collapse
Affiliation(s)
- Bettina Heidecker
- Miller School of Medicine, University of Miami Division of Cardiology, CRB, 1120 NW 14th St, Suite 1124, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Taegtmeyer H, Harinstein ME, Gheorghiade M. More than bricks and mortar: comments on protein and amino acid metabolism in the heart. Am J Cardiol 2008; 101:3E-7E. [PMID: 18514624 DOI: 10.1016/j.amjcard.2008.02.064] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Few cardiologists have considered that constituents of the heart muscle cells are in a continuous state of flux. The proteins of sarcomeres, mitochondria, membranes, the cytosol, and even ribosomes and the cell nucleus, are continuously degraded and remade. Schoenheimer's concept of the "dynamic state of body constituents," has received relatively little attention in the world of cardiovascular research, at least not until recently. We propose that the term nutrition of the heart extends well beyond the supply of energy-providing substrates and includes the supply of amino acids, micronutrients, and regulators of protein synthesis and degradation. This short article is written to make the reader think in broad concepts.
Collapse
Affiliation(s)
- Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical School at Houston, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
133
|
Qiu H, Dai H, Jain K, Shah R, Hong C, Pain J, Tian B, Vatner DE, Vatner SF, Depre C. Characterization of a novel cardiac isoform of the cell cycle-related kinase that is regulated during heart failure. J Biol Chem 2008; 283:22157-65. [PMID: 18508765 DOI: 10.1074/jbc.m710459200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myocardial infarction (MI) is often followed by heart failure (HF), but the mechanisms precipitating the transition to HF remain largely unknown. A genomic profile was performed in a monkey model of MI, from the myocardium adjacent to chronic (2-month) MI followed by 3 weeks of pacing to develop HF. The transcript of the gene encoding the cell cycle-related kinase (CCRK) was down-regulated by 50% in HF heart compared with control (p<0.05), which was confirmed by quantitative PCR. The CCRK sequence cloned from a heart library showed a conservation of the N-terminal kinase domain when compared with the "generic" isoform cloned previously but a different C-terminal half due to alternative splicing with frameshift. The homology of the cardiac sequence was 100% between mice and humans. Expression of the corresponding protein, measured upon generation of a monoclonal antibody, was limited to heart, liver, and kidney. Upon overexpression in cardiac myocytes, both isoforms promote cell growth and reduce apoptosis by chelerythrine (p<0.05 versus control). Using a yeast two-hybrid screening, we found an interaction of the generic but not the cardiac CCRK with cyclin H and casein kinase 2. In addition, only the generic CCRK phosphorylates the cyclin-dependent kinase 2, which was accompanied by a doubling of myocytes in the S and G(2) phases of the cell cycle (p < 0.05 versus control). Therefore, the heart expresses a splice variant of CCRK, which promotes cardiac cell growth and survival; differs from the generic isoform in terms of protein-protein interactions, substrate specificity and regulation of the cell cycle; and is down-regulated significantly in HF.
Collapse
Affiliation(s)
- Hongyu Qiu
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey 07103, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Khairallah RJ, Khairallah M, Gélinas R, Bouchard B, Young ME, Allen BG, Lopaschuk GD, Deschepper CF, Des Rosiers C. Cyclic GMP signaling in cardiomyocytes modulates fatty acid trafficking and prevents triglyceride accumulation. J Mol Cell Cardiol 2008; 45:230-9. [PMID: 18590915 DOI: 10.1016/j.yjmcc.2008.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Revised: 05/05/2008] [Accepted: 05/20/2008] [Indexed: 12/21/2022]
Abstract
While the balance between carbohydrates and fatty acids for energy production appears to be crucial for cardiac homeostasis, much remains to be learned about the molecular mechanisms underlying this relationship. Given the reported benefits of cGMP signaling on the myocardium, we investigated the impact of its chronic activation on cardiac energy metabolism using mice overexpressing a constitutively active cytoplasmic guanylate cyclase (GC(+/0)) in cardiomyocytes. Ex vivo working GC(+/0) heart perfusions with (13)C-labeled substrates revealed an altered pattern of exogenous substrate fuel selection compared to controls, namely a 38+/-9% lower contribution of exogenous fatty acids to acetyl-CoA formation, while that of carbohydrates remains unchanged despite a two-fold increase in glycolysis. The lower contribution of exogenous fatty acids to energy production is not associated with changes in energy demand or supply (contractile function, oxygen consumption, tissue acetyl-CoA or CoA levels, citric acid cycle flux rate) or in the regulation of beta-oxidation (acetyl-CoA carboxylase activity, tissue malonyl-CoA levels). However, GC(+/0) hearts show a two-fold increase in the incorporation of exogenous oleate into triglycerides. Furthermore, the following molecular data are consistent with a concomitant increase in triglyceride hydrolysis: (i) increased abundance of hormone sensitive lipase (HSL) protein (24+/-11%) and mRNA (22+/-4%) as well as (ii) several phosphorylation events related to HSL inhibitory (AMPK) and activation (ERK 1/2) sites, which should contribute to enhance its activity. These changes in exogenous fatty acid trafficking in GC(+/0) hearts appear to be functionally relevant, as demonstrated by their resistance to fasting-induced triglyceride accumulation. While the documented metabolic profile of GC(+/0) mouse hearts is partly reminiscent of hypertrophied hearts, the observed changes in lipid trafficking have not been previously documented, and may be part of the molecular mechanism underlying the benefits of cGMP signaling on the myocardium.
Collapse
Affiliation(s)
- Ramzi J Khairallah
- Montreal Heart Institute, University of Montreal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Ambalavanan N, Nicola T, Hagood J, Bulger A, Serra R, Murphy-Ullrich J, Oparil S, Chen YF. Transforming growth factor-beta signaling mediates hypoxia-induced pulmonary arterial remodeling and inhibition of alveolar development in newborn mouse lung. Am J Physiol Lung Cell Mol Physiol 2008; 295:L86-95. [PMID: 18487357 DOI: 10.1152/ajplung.00534.2007] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxia causes abnormal neonatal pulmonary artery remodeling (PAR) and inhibition of alveolar development (IAD). Transforming growth factor (TGF)-beta is an important regulator of lung development and repair from injury. We tested the hypothesis that inhibition of TGF-beta signaling attenuates hypoxia-induced PAR and IAD. Mice with an inducible dominant-negative mutation of the TGF-beta type II receptor (DNTGFbetaRII) and nontransgenic wild-type (WT) mice were exposed to hypoxia (12% O(2)) or air from birth to 14 days of age. Expression of DNTGFbetaRII was induced by 20 microg/g ZnSO(4) given intraperitoneally daily from birth. PAR, IAD, cell proliferation, and expression of extracellular matrix (ECM) proteins were assessed. In WT mice, hypoxia led to thicker, more muscularized resistance pulmonary arteries and impaired alveolarization, accompanied by increases in active TGF-beta and phosphorylated Smad2. Hypoxia-induced PAR and IAD were greatly attenuated in DNTGFbetaRII mice given ZnSO(4) compared with WT control mice and DNTGFbetaRII mice not given ZnSO(4). The stimulatory effects of hypoxic exposure on pulmonary arterial cell proliferation and lung ECM proteins were abrogated in DNTGFbetaRII mice given ZnSO(4). These data support the conclusion that TGF-beta plays an important role in hypoxia-induced pulmonary vascular adaptation and IAD in the newborn animal model.
Collapse
Affiliation(s)
- Namasivayam Ambalavanan
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama 35233, USA.
| | | | | | | | | | | | | | | |
Collapse
|
136
|
Essop MF, Camp HS, Choi CS, Sharma S, Fryer RM, Reinhart GA, Guthrie PH, Bentebibel A, Gu Z, Shulman GI, Taegtmeyer H, Wakil SJ, Abu-Elheiga L. Reduced heart size and increased myocardial fuel substrate oxidation in ACC2 mutant mice. Am J Physiol Heart Circ Physiol 2008; 295:H256-65. [PMID: 18487439 DOI: 10.1152/ajpheart.91489.2007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cardiac-enriched isoform of acetyl-CoA carboxylase (ACC2) is a key regulator of mitochondrial fatty acid (FA) uptake via carnitine palmitoyltransferase 1 (CPT1). To test the hypothesis that oxidative metabolism is upregulated in hearts from animals lacking ACC2 (employing a transgenic Acc2-mutant mouse), we assessed cardiac function in vivo and determined rates of myocardial substrate oxidation ex vivo. When examined by echocardiography, there was no difference in systolic function, but left ventricular mass of the Acc2-mutant (MUT) mouse was significantly reduced ( approximately 25%) compared with wild-types (WT). Reduced activation of the mammalian target of rapamycin (mTOR) and its downstream target p70S6K was found in MUT hearts. Exogenous oxidation rates of oleate were increased approximately 22%, and, unexpectedly, exogenous glucose oxidation rates were also increased in MUT hearts. Using a hyperinsulinemic-euglycemic clamp, we found that glucose uptake in MUT hearts was increased by approximately 83%. Myocardial triglyceride levels were significantly reduced in MUT vs. WT while glycogen content was the same. In parallel, transcript levels of PPARalpha and its target genes, pyruvate dehydrogenase kinase-4 (PDK-4), malonyl-CoA decarboxylase (MCD), and mCPT1, were downregulated in MUT mice. In summary, we report that 1) Acc2-mutant hearts exhibit a marked preference for the oxidation of both glucose and FAs coupled with greater utilization of endogenous fuel substrates (triglycerides), 2) attenuated mTOR signaling may result in reduced heart sizes observed in Acc2-mutant mice, and 3) Acc2-mutant hearts displayed normal functional parameters despite a significant decrease in size.
Collapse
Affiliation(s)
- M Faadiel Essop
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Maejima Y, Adachi S, Ito H, Hirao K, Isobe M. Induction of premature senescence in cardiomyocytes by doxorubicin as a novel mechanism of myocardial damage. Aging Cell 2008; 7:125-36. [PMID: 18031568 DOI: 10.1111/j.1474-9726.2007.00358.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cellular senescence is an important phenomenon in decreased cellular function. Recently, it was shown that cellular senescence is induced in proliferating cells within a short period of time by oxidative stresses. This phenomenon is known as premature senescence. However, it is still unknown whether premature senescence can be also induced in cardiomyocytes. The aim of the present study was to investigate whether a senescence-like phenotype can be induced in cardiomyocytes by oxidative stress. In cardiomyocytes obtained from aged rats (24 months of age), the staining for senescence-associated beta-galactosidase increased significantly and the protein or RNA levels of cyclin-dependent kinase inhibitors increased compared to those of young rats. Decreased cardiac troponin I phosphorylation and telomerase activity were also observed in aged cardiomyocytes. Treatment of cultured neonatal rat cardiomyocytes with a low concentration of doxorubicin (DOX) (10(-7) mol L(-1)) did not induce apoptosis but did induce oxidative stress, which was confirmed by 2',7'-dichlorofluorescin diacetate staining. In DOX-treated neonatal cardiomyocytes, increased positive staining for senescence-associated beta-galactosidase, cdk-I expression, decreased cardiac troponin I phosphorylation, and decreased telomerase activity were observed, as aged cardiomyocytes. Alterations in mRNA expression typically seen in aged cells were observed in DOX-treated neonatal cardiomyocytes. We also found that promyelocytic leukemia protein and acetylated p53, key proteins involved in stress-induced premature senescence in proliferating cells, were associated with cellular alterations of senescence in DOX-treated cardiomyocytes. In conclusion, cardiomyocytes treated with DOX showed characteristic changes similar to cardiomyocytes of aged rats. promyelocytic leukemia-related p53 acetylation may be an underlying mechanism of senescence-like alterations in cardiomyocytes. These findings indicate a novel mechanism of myocardial dysfunction induced by oxidative stress.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
138
|
Garcia RAP, Afeche SC, Scialfa JH, do Amaral FG, dos Santos SHJ, Lima FB, Young ME, Cipolla-Neto J. Insulin modulates norepinephrine-mediated melatonin synthesis in cultured rat pineal gland. Life Sci 2008; 82:108-14. [DOI: 10.1016/j.lfs.2007.10.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 09/17/2007] [Accepted: 10/22/2007] [Indexed: 10/22/2022]
|
139
|
Gallego-Delgado J, Lazaro A, Osende JI, Esteban V, Barderas MG, Gomez-Guerrero C, Vega R, Vivanco F, Egido J. Proteomic analysis of early left ventricular hypertrophy secondary to hypertension: modulation by antihypertensive therapies. J Am Soc Nephrol 2007; 17:S159-64. [PMID: 17130255 DOI: 10.1681/asn.2006080937] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Untreated or poorly controlled arterial hypertension induced development of pathologic left ventricular hypertrophy (LVH), a common finding in hypertensive patients and a strong predictor of cardiovascular morbidity and mortality. The proteomic approach is a powerful technique to analyze a complex mixture of proteins in various settings. An experimental model of hypertension-induced early LVH was performed in spontaneously hypertensive rats, and the cardiac protein pattern compared with the normotensive Wistar Kyoto counterpart was analyzed. Fifteen altered protein spots were shown in the early stage of LVH. Compared with a previous animal model of established and regressed LVH, three protein spots were common in both models. These three altered protein spots corresponded to two unique proteins that were identified as Calsarcin-1 (CS-1) and ubiquinone biosynthesis protein COQ7 homolog. CS-1 is a negative regulator of the calcineurin/NF-AT pathway. Because upregulation in the expression levels of this protein was observed, the activation level of NF-kappaB by oxidative stress as an alternative pathway was investigated. It was found that antihypertensive therapies partially decreased oxidative stress and normalized the activation of NF-kappaB in the kidneys and aorta NF-kappaB activation but just moderately in the heart. This could be due to the interaction of any specific cardiac protein with any component of the NF-kappaB pathway. In this sense, CS-1 could be a good candidate because it is expressed preferentially in heart, to a lesser extent in smooth muscle cells, but not in kidney. Further investigations are necessary to elucidate the exact role of CS-1 and ubiquinone biosynthesis protein COQ7 in the setting of hypertension-induced LVH.
Collapse
Affiliation(s)
- Julio Gallego-Delgado
- Renal and Vascular Pathology Laboratory, Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Hickson-Bick DLM, Jones C, Buja LM. Stimulation of mitochondrial biogenesis and autophagy by lipopolysaccharide in the neonatal rat cardiomyocyte protects against programmed cell death. J Mol Cell Cardiol 2007; 44:411-8. [PMID: 18062988 DOI: 10.1016/j.yjmcc.2007.10.013] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 10/02/2007] [Accepted: 10/17/2007] [Indexed: 02/05/2023]
Abstract
Adult rat cardiomyocytes in culture respond to sub-lethal doses of lipopolysaccharides (LPS) by activation of pathways including the production of TNF-alpha and increased apoptosis. We and others have demonstrated a protective phenotype for neonatal rat cardiomyocytes to LPS. Concentrations of LPS far exceeding those necessary to induce TNF-alpha release do not induce apoptosis in the neonatal cells, although these cells are fully capable or inducing apoptosis in response to multiple other stimuli. In neonatal cells, we demonstrate that LPS treatment leads to a loss of mitochondrial membrane potential (Deltapsi) which is temporally associated with an increase in the level of uncoupling protein 3 (UCP3). Cells remain viable with no measurable increase in apoptotic or necrotic cell death. Many markers of mitochondrial biogenesis are also activated. LPS treatment stimulates an increase in the (i) transcription of mitochondrial transcription factor A (Tfam), (ii) nuclear accumulation of redox-sensitive nuclear respiratory factor 1 (NRF-1), and (iii) expression of peroxisome proliferator-activated receptor gamma co-activator 1 (PGC-1). We also observed that LPS increased intracellular autophagy. Autophagy was assessed by monitoring the levels of a mammalian protein specifically associated with autophagosomes, microtubule-associated light chain 3 (LC3). Furthermore, inhibition of autophagy in the presence of LPS stimulates markers of apoptosis. Our data suggest that the protective response of neonatal cells to LPS is multi-faceted at the level of the mitochondrion. Viable cells replace dysfunctional mitochondria by mitochondrial biogenesis and the extent of the damage limited by the rapid removal of damaged organelles by the stimulation of autophagy.
Collapse
Affiliation(s)
- Diane L M Hickson-Bick
- Department of Pathology and Laboratory Medicine, University of Texas Medical School, Houston, TX 77030, USA.
| | | | | |
Collapse
|
141
|
Young ME, Yan J, Razeghi P, Cooksey RC, Guthrie PH, Stepkowski SM, McClain DA, Tian R, Taegtmeyer H. Proposed regulation of gene expression by glucose in rodent heart. GENE REGULATION AND SYSTEMS BIOLOGY 2007; 1:251-62. [PMID: 19936093 PMCID: PMC2759127 DOI: 10.4137/grsb.s222] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background During pressure overload-induced hypertrophy, unloading-induced atrophy, and diabetes mellitus, the heart induces ‘fetal’ genes (e.g. myosin heavy chain β; mhcβ). Hypothesis We propose that altered glucose homeostasis within the cardiomyocyte acts as a central mechanism for the regulation of gene expression in response to environmental stresses. The evidence is as follows. Methods and Results Forced glucose uptake both ex vivo and in vivo results in mhc isoform switching. Restricting dietary glucose prevents mhc isoform switching in hearts of both GLUT1-Tg mice and rats subjected to pressure overload-induced hypertrophy. Thus, glucose availability correlates with mhc isoform switching under all conditions investigated. A potential mechanism by which glucose affects gene expression is through O-linked glycosylation of specific transcription factors. Glutamine:fructose-6-phosphate amidotransferase (GFAT) catalyzes the flux generating step in UDP-N-acetylglucosamine biosynthesis, the rate determining metabolite in protein glycosylation. Ascending aortic constriction increased intracellular levels of UDP-N-acetylglucosamine, and the expression of gfat2, but not gfat1, in the rat heart. Conclusions Collectively, the results strongly suggest glucose-regulated gene expression in the heart, and the involvement of glucose metabolites in isoform switching of sarcomeric proteins characteristic for the fetal gene program.
Collapse
Affiliation(s)
- Martin E Young
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Wilson C, Tran M, Salazar K, Young M, Taegtmeyer H. Western diet, but not high fat diet, causes derangements of fatty acid metabolism and contractile dysfunction in the heart of Wistar rats. Biochem J 2007; 406:457-67. [PMID: 17550347 PMCID: PMC2049036 DOI: 10.1042/bj20070392] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Obesity and diabetes are associated with increased fatty acid availability in excess of muscle fatty acid oxidation capacity. This mismatch is implicated in the pathogenesis of cardiac contractile dysfunction and also in the development of skeletal-muscle insulin resistance. We tested the hypothesis that 'Western' and high fat diets differentially cause maladaptation of cardiac- and skeletal-muscle fatty acid oxidation, resulting in cardiac contractile dysfunction. Wistar rats were fed on low fat, 'Western' or high fat (10, 45 or 60% calories from fat respectively) diet for acute (1 day to 1 week), short (4-8 weeks), intermediate (16-24 weeks) or long (32-48 weeks) term. Oleate oxidation in heart muscle ex vivo increased with high fat diet at all time points investigated. In contrast, cardiac oleate oxidation increased with Western diet in the acute, short and intermediate term, but not in the long term. Consistent with fatty acid oxidation maladaptation, cardiac power decreased with long-term Western diet only. In contrast, soleus muscle oleate oxidation (ex vivo) increased only in the acute and short term with either Western or high fat feeding. Fatty acid-responsive genes, including PDHK4 (pyruvate dehydrogenase kinase 4) and CTE1 (cytosolic thioesterase 1), increased in heart and soleus muscle to a greater extent with feeding a high fat diet compared with a Western diet. In conclusion, we implicate inadequate induction of a cassette of fatty acid-responsive genes, and impaired activation of fatty acid oxidation, in the development of cardiac dysfunction with Western diet.
Collapse
Affiliation(s)
- Christopher R. Wilson
- *Department of Internal Medicine, Division of Cardiology, University of Texas Medical School at Houston, University of Texas Health Science Center at Houston, 6431 Fannin, MSB 1.246, Houston, TX 77030, U.S.A
| | - Mai K. Tran
- *Department of Internal Medicine, Division of Cardiology, University of Texas Medical School at Houston, University of Texas Health Science Center at Houston, 6431 Fannin, MSB 1.246, Houston, TX 77030, U.S.A
| | - Katrina L. Salazar
- *Department of Internal Medicine, Division of Cardiology, University of Texas Medical School at Houston, University of Texas Health Science Center at Houston, 6431 Fannin, MSB 1.246, Houston, TX 77030, U.S.A
| | - Martin E. Young
- †Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, U.S.A
| | - Heinrich Taegtmeyer
- *Department of Internal Medicine, Division of Cardiology, University of Texas Medical School at Houston, University of Texas Health Science Center at Houston, 6431 Fannin, MSB 1.246, Houston, TX 77030, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
143
|
Essop MF, Chan WYA, Taegtmeyer H. Metabolic gene switching in the murine female heart parallels enhanced mitochondrial respiratory function in response to oxidative stress. FEBS J 2007; 274:5278-84. [PMID: 17892491 DOI: 10.1111/j.1742-4658.2007.06051.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The mechanisms underlying increased cardioprotection in younger female mice are unclear. We hypothesized that serine-threonine protein kinase (protein kinase B; Akt) triggers a metabolic gene switch (decreased fatty acids, increased glucose) in female hearts to enhance mitochondrial bioenergetic capacity, conferring protection against oxidative stress. Here, we employed male and female control (db/+) and obese (db/db) mice. We found diminished transcript levels of peroxisome proliferator-activated receptor-alpha, muscle-type carnitine palmitoyltransferase 1 and pyruvate dehydrogenase kinase 4 in female control hearts versus male hearts. Moreover, females displayed improved recovery of cardiac mitochondrial respiratory function and higher ATP levels versus males in response to acute oxygen deprivation. All these changes were reversed in female db/db hearts. However, we found no significant gender-based differences in levels of Akt, suggesting that Akt-independent signaling mechanisms are responsible for the resilient mitochondrial phenotype observed in female mouse hearts. As glucose is a more energetically efficient fuel substrate when oxygen is limiting, this gene program may be a crucial component that enhances tolerance to oxygen deprivation in female hearts.
Collapse
Affiliation(s)
- M Faadiel Essop
- Department of Physiological Sciences, Stellenbosch University, South Africa.
| | | | | |
Collapse
|
144
|
Birkenfeld AL, Boschmann M, Jordan J. Metabolic regulation: effects of natriuretic peptide interactions. Expert Rev Endocrinol Metab 2007; 2:607-614. [PMID: 30736123 DOI: 10.1586/17446651.2.5.607] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In addition to their well-established effects on blood pressure and volume homeostasis, natriuretic peptides have complex effects on carbohydrate and lipid metabolism. In vivo, pharmacological and physiological concentrations of atrial natriuretic peptides induce lipolysis in a concentration-dependent manner and increase the lipid oxidation rate. The response appears to be mediated through the stimulation of natriuretic peptide receptor-A. More recent studies suggest that natriuretic peptides also affect the production of several adipokines. These mechanisms may be relevant, as natriuretic peptide availability is altered in numerous physiological and pathological conditions, including physical exercise, congestive heart failure and obesity.
Collapse
Affiliation(s)
- Andreas L Birkenfeld
- a Franz-Volhard Clinical Research Center, Haus 129, Charité Campus Buch, Wiltbergstr. 50, 13125 Berlin, Germany
| | - Michael Boschmann
- a Franz-Volhard Clinical Research Center, Haus 129, Charité Campus Buch, Wiltbergstr. 50, 13125 Berlin, Germany
| | - Jens Jordan
- b Franz-Volhard Clinical Research Center, Haus 129, Charité Campus Buch, Wiltbergstr. 50, 13125 Berlin, Germany.
| |
Collapse
|
145
|
Qiu H, Depre C, Ghosh K, Resuello RG, Natividad FF, Rossi F, Peppas A, Shen YT, Vatner DE, Vatner SF. Mechanism of gender-specific differences in aortic stiffness with aging in nonhuman primates. Circulation 2007; 116:669-76. [PMID: 17664374 DOI: 10.1161/circulationaha.107.689208] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Our hypothesis was that the changes in vascular properties responsible for aortic stiffness with aging would be greater in old male monkeys than old female monkeys. METHODS AND RESULTS We analyzed the effects of gender differences in aging on in vivo measurements of aortic pressure and diameter and on extracellular matrix of the thoracic aorta in young adult (age, 6.6+/-0.5 years) versus old adult (age, 21.2+/-0.2 years) monkeys (Macaca fascicularis). Aortic stiffness, as represented by the pressure strain elastic modulus (Ep), increased more in old male monkeys (5.08+/-0.81; P<0.01) than in old females (3.06+/-0.52). In both genders, collagen density was maintained, collagen-bound glycation end products increased, and collagen type 1 decreased. However, elastin density decreased significantly (from 22+/-1.5% to 15+/-1.2%) with aging (P<0.05) only in males. Furthermore, only old males were characterized by a decrease (P<0.05) in collagen type 3 (an isoform that promotes elasticity) and an increase in collagen type 8 (an isoform that promotes the neointimal migration of vascular smooth muscle cells). In contrast to the data in monkeys, collagen types 1 and 3 both increased significantly in aging rats. CONCLUSIONS There are major species differences in the effects of aging on aortic collagen types 1 and 3. Furthermore, because alterations in collagen density, collagen content, hydroxyproline, and collagen advanced glycation end products were similar in both old male and female monkeys, these factors cannot be responsible for the greater increase in stiffness in old males. However, changes in collagen isoforms and the decrease in elastin observed only in old males likely account for the greater increase in aortic stiffness.
Collapse
Affiliation(s)
- Hongyu Qiu
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, 185 S Orange Ave, Newark, NJ 07103, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Rajabi M, Kassiotis C, Razeghi P, Taegtmeyer H. Return to the fetal gene program protects the stressed heart: a strong hypothesis. Heart Fail Rev 2007; 12:331-43. [PMID: 17516164 DOI: 10.1007/s10741-007-9034-1] [Citation(s) in RCA: 318] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A common feature of the hemodynamically or metabolically stressed heart is the return to a pattern of fetal metabolism. A hallmark of fetal metabolism is the predominance of carbohydrates as substrates for energy provision in a relatively hypoxic environment. When the normal heart is exposed to an oxygen rich environment after birth, energy substrate metabolism is rapidly switched to oxidation of fatty acids. This switch goes along with the expression of "adult" isoforms of metabolic enzymes and other proteins. However, the heart retains the ability to return to the "fetal" gene program. Specifically, the fetal gene program is predominant in a variety of pathophysiologic conditions including hypoxia, ischemia, hypertrophy, and atrophy. A common feature of all of these conditions is extensive remodeling, a decrease in the rate of aerobic metabolism in the cardiomyocyte, and an increase in cardiac efficiency. The adaptation is associated with a whole program of cell survival under stress. The adaptive mechanisms are prominently developed in hibernating myocardium, but they are also a feature of the failing heart muscle. We propose that in failing heart muscle at a certain point the fetal gene program is no longer sufficient to support cardiac structure and function. The exact mechanisms underlying the transition from adaptation to cardiomyocyte dysfunction are still not completely understood.
Collapse
Affiliation(s)
- Mitra Rajabi
- Department of Internal Medicine, Division of Cardiology, University of Texas-Houston Medical School, 6431 Fannin, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
147
|
Sharma S, Guthrie P, Chan S, Haq S, Taegtmeyer H. Glucose phosphorylation is required for insulin-dependent mTOR signalling in the heart. Cardiovasc Res 2007; 76:71-80. [PMID: 17553476 PMCID: PMC2257479 DOI: 10.1016/j.cardiores.2007.05.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 04/26/2007] [Accepted: 05/07/2007] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Insulin regulates both glucose uptake and postnatal cardiac growth. The anabolic effects of insulin are mediated by the mammalian target of rapamycin (mTOR), an evolutionarily conserved kinase which is also a convergence point between nutrient sensing and cell growth. We postulated that mTOR signalling in the heart requires the metabolism of glucose. METHODS We interrogated the insulin-mediated mTOR signalling pathway in response to different metabolic interventions regulating substrate metabolism in the isolated working rat heart and in isolated cardiomyocytes. RESULTS Although insulin enhanced Akt activity, phosphorylation of mTOR and its downstream targets (p70S6K and 4EBP1) required the addition of glucose. Glucose-dependent p70S6K phosphorylation was independent of the hexosamine biosynthetic pathway, the AMP kinase pathway, and the pentose phosphate pathway. However, inhibition of glycolysis downstream of hexokinase markedly enhanced p70S6K phosphorylation. Furthermore, 2-deoxyglucose activated p70S6K suggesting that phosphorylation of glucose is required for carbohydrate-mediated mTOR signalling in the heart. Lastly, we also found enhanced p70S6K phosphorylation in the hearts of diabetic rats. CONCLUSION Phosphorylation of glucose is necessary for insulin-dependent mTOR activity in the heart, suggesting a link between intermediary metabolism and cardiac growth.
Collapse
Affiliation(s)
- Saumya Sharma
- Division of Cardiology, The University of Texas Houston Medical School, Houston, Texas
| | - Patrick Guthrie
- Division of Cardiology, The University of Texas Houston Medical School, Houston, Texas
| | - Suzanne Chan
- Division of Cardiology, The University of Texas Houston Medical School, Houston, Texas
| | - Syed Haq
- Molecular Cardiology Research Institute, Tufts-New England Medical Center and Department of Medicine, Tufts University
| | - Heinrich Taegtmeyer
- Division of Cardiology, The University of Texas Houston Medical School, Houston, Texas
| |
Collapse
|
148
|
Ding L, Liang XG, Lou YJ. Time-dependence of cardiomyocyte differentiation disturbed by peroxisome proliferator-activated receptor alpha inhibitor GW6471 in murine embryonic stem cells in vitro. Acta Pharmacol Sin 2007; 28:634-42. [PMID: 17439719 DOI: 10.1111/j.1745-7254.2007.00558.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AIM To investigate the possible roles of peroxisome proliferator-activated receptor alpha(PPAR alpha) and the signal pathway regulating the transcription of PPAR alpha in the cardiomyocyte differentiation course of murine embryonic stem (ES) cells in vitro. METHODS The expression of PPAR alpha during cardiomyocyte differentiation was analyzed using both Western blotting and immunofluorescence. Cardiac specific genes and sarcomeric proteins were evaluated when embryoid bodies were challenged with PPAR alpha specific inhibitor GW6471 at different time courses. The phosphorylation of p38 mitogen-activated protein kinase (MAPK) was studied in the differentiation process, and its specific inhibitor SB203580 was employed to study the function of p38 MAPK on cardiac differentiation and the expression of PPAR alpha. RESULTS The expression of PPAR alpha was observed to be at a low level in undifferentiated ES cells and markedly induced with the appearance of beating clusters. The inhibition of PPAR alpha by its specific inhibitor GW6471 (1X10(-5) mol/L) significantly prevented cardiomyocyte differentiation and resulted in the reduced expression of cardiac sarcomeric proteins (ie alpha-actinin, troponin-T) and specific genes (ie alpha-MHC, MLC2v) in a time-dependent manner. In the differentiation course, p-p38 MAPK was maintained at a high level from d 3 followed by a decrease from d 10. The inhibition of the p38 MAPK pathway by SB203580 between d 3 and d 7 efficiently prevented cardiomyocyte differentiation and resulted in the capture of the upregulation of PPAR alpha. CONCLUSION Taken together, these results showed a close association between PPAR alpha and cardiomyocyte differentiation in vitro, and p38 MAPK was partly responsible for the regulation of PPAR alpha.
Collapse
Affiliation(s)
- Ling Ding
- Institute of Pharmacology and Toxicology and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | |
Collapse
|
149
|
Chen SY, Tang WHW. Emerging drugs for acute and chronic heart failure: current and future developments. Expert Opin Emerg Drugs 2007; 12:75-95. [PMID: 17355215 DOI: 10.1517/14728214.12.1.75] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Heart failure continues to be a major public health issue. Although angiotensin-converting enzyme inhibitors and beta-adrenergic blockers have been broadly used as evidence-based therapies in heart failure, morbidity and mortality remains high. Furthermore, treatment for acute decompensated heart failure and diastolic heart failure (or 'heart failure with preserved ejection fraction') is far from perfect. This review provides a broad overview of some of the novel compounds under investigation for the treatment of heart failure. Novel strategies include drugs that aim to alleviate congestion and improve hemodynamics, drugs that preserve renal function, drugs that reduce arterial and myocardial stiffness, drugs that module myocardial contractility, drugs that affect metabolic and hormonal balance, and drugs that act on existing and novel physiologic targets.
Collapse
Affiliation(s)
- Stephen Y Chen
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
150
|
Sorokina N, O'Donnell JM, McKinney RD, Pound KM, Woldegiorgis G, LaNoue KF, Ballal K, Taegtmeyer H, Buttrick PM, Lewandowski ED. Recruitment of compensatory pathways to sustain oxidative flux with reduced carnitine palmitoyltransferase I activity characterizes inefficiency in energy metabolism in hypertrophied hearts. Circulation 2007; 115:2033-41. [PMID: 17404155 DOI: 10.1161/circulationaha.106.668665] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Transport rates of long-chain free fatty acids into mitochondria via carnitine palmitoyltransferase I relative to overall oxidative rates in hypertrophied hearts remain poorly understood. Furthermore, the extent of glucose oxidation, despite increased glycolysis in hypertrophy, remains controversial. The present study explores potential compensatory mechanisms to sustain tricarboxylic acid cycle flux that resolve the apparent discrepancy of reduced fatty acid oxidation without increased glucose oxidation through pyruvate dehydrogenase complex in the energy-poor, hypertrophied heart. METHODS AND RESULTS We studied flux through the oxidative metabolism of intact adult rat hearts subjected to 10 weeks of pressure overload (hypertrophied; n=9) or sham operation (sham; n=8) using dynamic 13C-nuclear magnetic resonance. Isolated hearts were perfused with [2,4,6,8,10,12,14,16-(13)C8] palmitate (0.4 mmol/L) plus glucose (5 mmol/L) in a 14.1-T nuclear magnetic resonance magnet. At similar tricarboxylic acid cycle rates, flux through carnitine palmitoyltransferase I was 23% lower in hypertrophied (P<0.04) compared with sham hearts and corresponded to a shift toward increased expression of the L-carnitine palmitoyltransferase I isoform. Glucose oxidation via pyruvate dehydrogenase complex did not compensate for reduced palmitate oxidation rates. However, hypertrophied rats displayed an 83% increase in anaplerotic flux into the tricarboxylic acid cycle (P<0.03) that was supported by glycolytic pyruvate, coincident with increased mRNA transcript levels for malic enzyme. CONCLUSIONS In cardiac hypertrophy, fatty acid oxidation rates are reduced, whereas compensatory increases in anaplerosis maintain tricarboxylic acid cycle flux and account for a greater portion of glucose oxidation than previously recognized. The shift away from acetyl coenzyme A production toward carbon influx via anaplerosis bypasses energy, yielding reactions contributing to a less energy-efficient heart.
Collapse
Affiliation(s)
- Natalia Sorokina
- Center for Cardiovascular Research, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|