301
|
Chen M, Wang Y, Qu A. PGC-1 alpha accelerates cytosolic Ca2+ clearance without disturbing Ca2+ homeostasis in cardiac myocytes. Biochem Biophys Res Commun 2010; 396:894-900. [PMID: 20457122 DOI: 10.1016/j.bbrc.2010.05.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Accepted: 05/05/2010] [Indexed: 10/19/2022]
Abstract
Energy metabolism and Ca(2+) handling serve critical roles in cardiac physiology and pathophysiology. Peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1 alpha) is a multi-functional coactivator that is involved in the regulation of cardiac mitochondrial functional capacity and cellular energy metabolism. However, the regulation of PGC-1 alpha in cardiac Ca(2+) signaling has not been fully elucidated. To address this issue, we combined confocal line-scan imaging with off-line imaging processing to characterize calcium signaling in cultured adult rat ventricular myocytes expressing PGC-1 alpha via adenoviral transduction. Our data shows that overexpressing PGC-1 alpha improved myocyte contractility without increasing the amplitude of Ca(2+) transients, suggesting that myofilament sensitivity to Ca(2+) increased. Interestingly, the decay kinetics of global Ca(2+) transients and Ca(2+) waves accelerated in PGC-1 alpha-expressing cells, but the decay rate of caffeine-elicited Ca(2+) transients showed no significant change. This suggests that sarcoplasmic reticulum (SR) Ca(2+)-ATPase (SERCA2a), but not Na(+)/Ca(2+) exchange (NCX) contribute to PGC-1 alpha-induced cytosolic Ca(2+) clearance. Furthermore, PGC-1 alpha induced the expression of SERCA2a in cultured cardiac myocytes. Importantly, overexpressing PGC-1 alpha did not disturb cardiac Ca(2+) homeostasis, because SR Ca(2+) load and the propensity for Ca(2+) waves remained unchanged. These data suggest that PGC-1 alpha can ameliorate cardiac Ca(2+) cycling and improve cardiac work output in response to physiological stress. Unraveling the PGC-1 alpha-calcium handling pathway sheds new light on the role of PGC-1 alpha in the therapy of cardiac diseases.
Collapse
Affiliation(s)
- Min Chen
- Institute of Molecular Medicine, State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking University, Beijing 100871, China.
| | | | | |
Collapse
|
302
|
Faerber G, Barreto-Perreia F, Schoepe M, Gilsbach R, Schrepper A, Schwarzer M, Mohr FW, Hein L, Doenst T. Induction of heart failure by minimally invasive aortic constriction in mice: reduced peroxisome proliferator-activated receptor γ coactivator levels and mitochondrial dysfunction. J Thorac Cardiovasc Surg 2010; 141:492-500, 500.e1. [PMID: 20447656 DOI: 10.1016/j.jtcvs.2010.03.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 02/22/2010] [Accepted: 03/20/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Mitochondrial dysfunction has been suggested as a potential cause for heart failure. Pressure overload is a common cause for heart failure. However, implementing pressure overload in mice is considered a model for compensated hypertrophy but not for heart failure. We assessed the suitability of minimally invasive transverse aortic constriction to induce heart failure in C57BL/6 mice and assessed mitochondrial biogenesis and function. METHODS Minimally invasive transverse aortic constriction was performed through a ministernotomy without intubation (minimally invasive transverse aortic constriction, n = 68; sham operation, n = 43). Hypertrophy was assessed based on heart weight/body weight ratios and histologic analyses, and contractile function was assessed based on intracardiac Millar pressure measurements. Expression of selected metabolic genes was assessed with reverse transcription-polymerase chain reaction and Western blotting. Maximal respiratory capacity (state 3) of isolated mitochondria was measured with a Clark-type electrode. RESULTS Survival was 62%. Within 7 weeks, minimally invasive transverse aortic constriction induced significant hypertrophy (heart weight/body weight ratio: 10.08±0.28 mg/g for minimally invasive transverse aortic constriction vs 4.66±0.07 mg/g for sham operation; n=68; P<.01). Fifty-seven percent of mice undergoing minimally invasive transverse aortic constriction displayed signs of heart failure (pleural effusions, dyspnea, weight loss, and dp/dtmax of 3114±422 mm Hg/s, P<.05). All of them had heart weight/body weight ratios of greater than 10. Mice undergoing minimally invasive transverse aortic constriction with heart weight/body weight ratios of less than 10 had normal contractile function (dp/dtmax of 6471±292 mm Hg/s vs dp/dtmax of 6933±205 mmHg/s in sham mice) and no clinical signs of heart failure. The mitochondrial coactivator peroxisome proliferator-activated receptor γ coactivator alpha (PGC-1α) was downregulated in failing hearts only. PGC-1α and fatty acid oxidation gene expression were also decreased in failing hearts. State 3 respiration of isolated mitochondria was significantly reduced in all hearts subjected to pressure overload. CONCLUSIONS Contractile dysfunction and heart failure can be induced in wild-type mice by means of minimally invasive aortic constriction. Pressure overload-induced heart failure in mice is associated with mitochondrial dysfunction, as characterized by downregulation of PGC-1α and reduced oxidative capacity.
Collapse
Affiliation(s)
- Gloria Faerber
- Department of Cardiac Surgery, University of Leipzig Heart Center, Leipzig, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
303
|
|
304
|
Lin RCY, Weeks KL, Gao XM, Williams RBH, Bernardo BC, Kiriazis H, Matthews VB, Woodcock EA, Bouwman RD, Mollica JP, Speirs HJ, Dawes IW, Daly RJ, Shioi T, Izumo S, Febbraio MA, Du XJ, McMullen JR. PI3K(p110 alpha) protects against myocardial infarction-induced heart failure: identification of PI3K-regulated miRNA and mRNA. Arterioscler Thromb Vasc Biol 2010; 30:724-32. [PMID: 20237330 DOI: 10.1161/atvbaha.109.201988] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Myocardial infarction (MI) is a serious complication of atherosclerosis associated with increasing mortality attributable to heart failure. Activation of phosphoinositide 3-kinase [PI3K(p110 alpha)] is considered a new strategy for the treatment of heart failure. However, whether PI3K(p110 alpha) provides protection in a setting of MI is unknown, and PI3K(p110 alpha) is difficult to target because it has multiple actions in numerous cell types. The goal of this study was to assess whether PI3K(p110 alpha) is beneficial in a setting of MI and, if so, to identify cardiac-selective microRNA and mRNA that mediate the protective properties of PI3K(p110 alpha). METHODS AND RESULTS Cardiomyocyte-specific transgenic mice with increased or decreased PI3K(p110 alpha) activity (caPI3K-Tg and dnPI3K-Tg, respectively) were subjected to MI for 8 weeks. The caPI3K-Tg subjected to MI had better cardiac function than nontransgenic mice, whereas dnPI3K-Tg had worse function. Using microarray analysis, we identified PI3K-regulated miRNA and mRNA that were correlated with cardiac function, including growth factor receptor-bound 14. Growth factor receptor-bound 14 is highly expressed in the heart and positively correlated with PI3K(p110 alpha) activity and cardiac function. Mice deficient in growth factor receptor-bound 14 have cardiac dysfunction. CONCLUSIONS Activation of PI3K(p110 alpha) protects the heart against MI-induced heart failure. Cardiac-selective targets that mediate the protective effects of PI3K(p110 alpha) represent new drug targets for heart failure.
Collapse
Affiliation(s)
- Ruby C Y Lin
- Ramaciotti Centre for Gene Function Analysis, University of New South Wales, Randwick, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
305
|
Sun CK, Chen CH, Kao YH, Yuen CM, Sheu JJ, Lee FY, Chen YT, Kung CT, Yip HK. Bone marrow cells reduce fibrogenesis and enhance regeneration in fibrotic rat liver. J Surg Res 2010; 169:e15-26. [PMID: 20605605 DOI: 10.1016/j.jss.2010.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 02/03/2010] [Accepted: 03/09/2010] [Indexed: 01/07/2023]
Abstract
BACKGROUND This study aimed at investigating the cellular and molecular impacts of bone marrow-derived mononuclear cells (BMCs) on regeneration of fibrotic liver in rats. MATERIALS AND METHODS Thirty male adult Fisher rats were randomized into three groups: group 1 (normal controls, n = 10); group 2 (carbon tetrachloride-induced liver fibrosis, n = 10), and group 3 (liver fibrosis with portal venous infusion of autologous BMCs, 1 × 10(6), n = 10). After 7-d culturing, BMCs were characterized by flow cytometry. Groups 2 and 3 received BMC aspiration through bilateral femurs 5 d before hepatectomy. All animals received 70% hepatectomy, whereas only group 3 received a bolus of intra-portal BMC infusion 48 h after hepatectomy. Liver-to-body weight ratio, degree of fibrosis (Masson trichrome staining), oxidative stress, peroxisome proliferator activated receptor-γ coactivator-1α (PGC-1α), collagen I, tumor necrosis factor-α (TNF-α), and transforming growth factor- β (TGF-β) expressions were analyzed 14 d after hepatectomy. Immunohistochemical staining for albumin, α-smooth muscle actin, and CD31 was performed for tracing cellular differentiation. RESULTS Cellular phenotypes typical of hepatocyte, hepatic stellate cell (HSC), and endothelial cell were identified in the engrafted BMCs. Liver-to-body weight ratio was enhanced with PGC-1α significantly preserved, whereas oxidative index, collagen I, α-SMA, TNF-α, and TGF-β expressions were all decreased in group 3 compared with group 2 (all P < 0.05). CONCLUSIONS This study demonstrated a positive impact of intra-portal BMC infusion in enhancing regeneration and reducing fibrosis of the regenerating fibrotic liver in rats through suppressing HSC activation and inflammatory cytokine expressions, preserving mitochondrial function and reducing oxidative stress.
Collapse
Affiliation(s)
- Cheuk-Kwan Sun
- Division of General Surgery, Chang Gung Memorial Hospital, Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung Hsien, Taiwan, ROC
| | | | | | | | | | | | | | | | | |
Collapse
|
306
|
Zhu L, Wang Q, Zhang L, Fang Z, Zhao F, Lv Z, Gu Z, Zhang J, Wang J, Zen K, Xiang Y, Wang D, Zhang CY. Hypoxia induces PGC-1α expression and mitochondrial biogenesis in the myocardium of TOF patients. Cell Res 2010; 20:676-87. [PMID: 20368732 DOI: 10.1038/cr.2010.46] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PGC-1alpha, a potent transcriptional coactivator, is the major regulator of mitochondrial biogenesis and activity in the cardiac muscle. The dysregulation of PGC-1alpha and its target genes has been reported to be associated with congenital and acquired heart diseases. By examining myocardium samples from patients with Tetralogy of Fallot, we show here that PGC-1alpha expression levels are markedly increased in patients compared with healthy controls and positively correlated with the severity of cyanosis. Furthermore, hypoxia significantly induced the expression of PGC-1alpha and mitochondrial biogenesis in cultured cardiac myocytes. Mechanistic studies suggest that hypoxia-induced PGC-1alpha expression is regulated through the AMPK signaling pathway. Together, our data indicate that hypoxia can stimulate the expression of PGC-1alpha and mitochondrial biogenesis in the cardiac myocytes, and this process might provide a potential adaptive mechanism for cardiac myocytes to increase ATP output and minimize hypoxic damage to the heart.
Collapse
Affiliation(s)
- Lingyun Zhu
- Jiangsu Diabetes Center, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
307
|
Ahuja P, Zhao P, Angelis E, Ruan H, Korge P, Olson A, Wang Y, Jin ES, Jeffrey FM, Portman M, Maclellan WR. Myc controls transcriptional regulation of cardiac metabolism and mitochondrial biogenesis in response to pathological stress in mice. J Clin Invest 2010; 120:1494-505. [PMID: 20364083 DOI: 10.1172/jci38331] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 01/27/2010] [Indexed: 12/29/2022] Open
Abstract
In the adult heart, regulation of fatty acid oxidation and mitochondrial genes is controlled by the PPARgamma coactivator-1 (PGC-1) family of transcriptional coactivators. However, in response to pathological stressors such as hemodynamic load or ischemia, cardiac myocytes downregulate PGC-1 activity and fatty acid oxidation genes in preference for glucose metabolism pathways. Interestingly, despite the reduced PGC-1 activity, these pathological stressors are associated with mitochondrial biogenesis, at least initially. The transcription factors that regulate these changes in the setting of reduced PGC-1 are unknown, but Myc can regulate glucose metabolism and mitochondrial biogenesis during cell proliferation and tumorigenesis in cancer cells. Here we have demonstrated that Myc activation in the myocardium of adult mice increases glucose uptake and utilization, downregulates fatty acid oxidation by reducing PGC-1alpha levels, and induces mitochondrial biogenesis. Inactivation of Myc in the adult myocardium attenuated hypertrophic growth and decreased the expression of glycolytic and mitochondrial biogenesis genes in response to hemodynamic load. Surprisingly, the Myc-orchestrated metabolic alterations were associated with preserved cardiac function and improved recovery from ischemia. Our data suggest that Myc directly regulates glucose metabolism and mitochondrial biogenesis in cardiac myocytes and is an important regulator of energy metabolism in the heart in response to pathologic stress.
Collapse
Affiliation(s)
- Preeti Ahuja
- Department of Medicine, The Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1760,, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
308
|
Sepsis-induced myocardial depression is associated with transcriptional changes in energy metabolism and contractile related genes: a physiological and gene expression-based approach. Crit Care Med 2010; 38:894-902. [PMID: 20101178 DOI: 10.1097/ccm.0b013e3181ce4e50] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Increased nitric oxide production and altered mitochondrial function have been implicated in sepsis-induced cardiac dysfunction. The molecular mechanisms underlying myocardial depression in sepsis and the contribution of nitric oxide in this process however, are incompletely understood. OBJECTIVES To assess the transcriptional profile associated with sepsis-induced myocardial depression in a clinically relevant mouse model, and specifically test the hypothesis that critical transcriptional changes are inducible nitric oxide synthase-dependent. DESIGN Laboratory investigation. SETTING University affiliated research laboratory. SUBJECTS C57/BL6 wild type and congenic B6 129P2-Nos2tm1Lau/J (iNOS) mice. INTERVENTIONS Assessment of myocardial function after 48 hrs of induction of polymicrobial sepsis by caecal ligation and perforation. MEASUREMENTS AND RESULTS We compared the myocardial transcriptional profile in C57/BL6 wild type mice and congenic B6 129P2-Nos2tm1Lau/J litter mates after 48 hrs of polymicrobial sepsis induced by caecal ligation and perforation. Profiling of 22,690 expressed sequence tags by gene set enrichment analysis demonstrated that inducible nitric oxide synthase -/- failed to down regulate critical bioenergy and metabolism related genes including the gene for peroxisome proliferator-activated receptor gamma coactivator 1. Bioinformatics analysis identified a striking concordance in down regulation of transcriptional activity of proliferator-activated receptor gamma coactivator 1-related transcription factors resulting in sepsis associated myocardial remodeling as shown by isoform switching in the expression of contractile protein myosin heavy chain. In inducible nitric oxide synthase -/- deficient mice, contractile depression was minimal, and the transcriptional switch was absent. CONCLUSIONS Metabolic and myosin isoform gene expression switch in sepsis-induced myocardial depression is inducible nitric oxide synthase-dependent. Furthermore, we suggest that the molecular switch favoring the expression of fetal isoforms of contraction related proteins is associated with regulation of proliferator-activated receptor gamma coactivator 1 and related transcription factors in an inducible nitric oxide synthase-dependent manner.
Collapse
|
309
|
Guzmán-Ruiz R, Somoza B, Gil-Ortega M, Merino B, Cano V, Attané C, Castan-Laurell I, Valet P, Fernández-Alfonso MS, Ruiz-Gayo M. Sensitivity of cardiac carnitine palmitoyltransferase to malonyl-CoA is regulated by leptin: similarities with a model of endogenous hyperleptinemia. Endocrinology 2010; 151:1010-8. [PMID: 20056820 DOI: 10.1210/en.2009-1170] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute leptin increase as well as endogenous hyperleptinemia evoked by high-fat diets (HF) activate fatty acid metabolism in nonadipose tissues. This supports the notion that hyperleptinemia is pivotal to prevent/delay steatosis during periods of positive energy balance. We have previously shown that long-term HF spares ectopic accumulation of lipids specifically in the miocardium. Because carnitine palmitoyltransferase I (CPT-I) allows mitochondrial uptake/oxidation of fatty acids, we have hypothesized that leptin drives cardiac CPT-I activity. In the current study, hyperleptinemia was induced in C57BL/6J mice either by exogenous leptin administration or by means of HF, and the ability of malonyl-coenzyme A (malonyl-CoA) (the main endogenous inhibitor of CPT-I) to inhibit cardiac CPT was analyzed. IC(50) values of malonyl-CoA were 8.1 +/- 1.5 micromol/liter in controls vs. 69.3 +/- 5.2 micromol/liter (P < 0.01) in leptin-treated mice. This effect was also observed in cardiac explants incubated with leptin and was blocked by triciribine, a compound shown to inhibit protein kinase B (Akt) phosphorylation (pAkt). In accordance, acute leptin evoked an increase of cardiac pAkt levels, which correlated with CPT sensitivity to malonyl-CoA. Otherwise, the inhibitory effect of malonyl-CoA was hindered in HF hyperleptinemic mice, and in this case, pAkt levels also correlated with CPT sensitivity to malonyl-CoA. Our data show that leptin reduces the sensitivity of cardiac CPT-I to malonyl-CoA and suggest the involvement of an Akt-related signaling pathway in this effect. This mechanism appears to be sensitive to both acute and chronic hyperleptinemia. We conclude that this action of leptin is pivotal to drive cardiac metabolism under situations associated to hyperleptinemia.
Collapse
Affiliation(s)
- Rocío Guzmán-Ruiz
- Departamento de Ciencias Farmacéuticas y de la Alimentación, Facultad de Farmacia, Universidad Ceu-San Pablo, Urbanización Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
310
|
Torre-Villalvazo I, Gonzalez F, Aguilar-Salinas CA, Tovar AR, Torres N. Dietary soy protein reduces cardiac lipid accumulation and the ceramide concentration in high-fat diet-fed rats and ob/ob mice. J Nutr 2009; 139:2237-43. [PMID: 19828684 DOI: 10.3945/jn.109.109769] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Obesity is an epidemic condition strongly associated with cardiovascular morbidity and mortality. Heart disease secondary to obesity is associated with myocardial steatosis, leading to ceramide synthesis and cell dysfunction in a process known as lipotoxicity. Soy protein has been demonstrated to reduce lipotoxicity in the liver and pancreas in different rodent models of obesity. Thus, our purpose in the present work was to assess the effect of dietary soy protein on cardiac lipid accumulation and ceramide formation during obesity and to evaluate its effect in the following 2 rodent models of obesity: 1) a diet-induced obesity model in Sprague-Dawley rats was produced by feeding rats a control or a high-fat casein or soy protein diet for 180 d; and 2) wild-type and ob/ob mice were fed a casein or soy protein diet for 90 d. Soy protein intake led to lower cholesterol and triglyceride concentrations in the hearts of rats and ob/ob mice in association with a greater PPARalpha mRNA concentration and a lower level of sterol regulatory element binding protein-1 mRNA than those fed casein. The ceramide concentration was also lower in hearts of rats and ob/ob mice that were fed soy protein in association with lower serine palmitoyl transferase (SPT)-1 and tumor necrosis factor-alpha mRNA concentrations. These results indicate that dietary soy protein can reduce the heart ceramide concentration by reducing the expression of SPT-1, a key enzyme in the formation of this sphingolipid in the heart of obese rodents, and by reducing lipid accumulation. Thus, soy protein consumption may be considered as a dietary therapeutic approach for lipotoxic cardiomyopathy prevention.
Collapse
Affiliation(s)
- Ivan Torre-Villalvazo
- Department Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, DF, México
| | | | | | | | | |
Collapse
|
311
|
Energetic myocardial metabolism and oxidative stress: let's make them our friends in the fight against heart failure. Biomed Pharmacother 2009; 64:203-7. [PMID: 19954925 DOI: 10.1016/j.biopha.2009.10.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 10/19/2009] [Indexed: 01/31/2023] Open
Abstract
Heart failure (HF) is a syndrome causing a huge burden in morbidity and mortality worldwide. Current medical therapies for HF are aimed at suppressing the neurohormonal activation. However, novel therapies are needed for HF, independent of the neurohormonal axis, that can improve cardiac performance and prevent the progression of heart dysfunction. The modulation of cardiac metabolism may represent a new approach to the treatment of HF. The healthy heart converts chemical energy stored in fatty acids (FA) and glucose. Utilization of FA costs more oxygen per unit of ATP generated than glucose, and the heart gets 60-90% of its energy for oxidative phosphorylation from FA oxidation. The failing heart has been demonstrated to be metabolically abnormal, in both animal models and in patients, showing a shift toward an increased glucose uptake and utilization. The manipulation of myocardial substrate oxidation toward greater carbohydrate oxidation and less FA oxidation may improve ventricular performance and slow the progression of heart dysfunction. Impaired mitochondrial function and oxidative phosphorylation can reduce cardiac function by providing an insufficient supply of ATP to cardiomyocytes and by increasing myocardial oxidative stress. Although there are no effective stimulators of oxidative phosphorylation, several classes of drugs have been shown to open mitochondrial K(ATP) channels and, indirectly, to improve cardiac protection against oxidative stress. This article focuses on the energetic myocardial metabolism and oxidative status in the normal and failing heart, and briefly, it overviews the therapeutic potential strategies to improve cardiac energy and oxidative status in HF patients.
Collapse
|
312
|
Burelle Y, Khairallah M, Ascah A, Allen BG, Deschepper CF, Petrof BJ, Des Rosiers C. Alterations in mitochondrial function as a harbinger of cardiomyopathy: lessons from the dystrophic heart. J Mol Cell Cardiol 2009; 48:310-21. [PMID: 19769982 DOI: 10.1016/j.yjmcc.2009.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 09/03/2009] [Accepted: 09/09/2009] [Indexed: 10/20/2022]
Abstract
While compelling evidence supports the central role of mitochondrial dysfunction in the pathogenesis of heart failure, there is comparatively less information available on mitochondrial alterations that occur prior to failure. Building on our recent work with the dystrophin-deficient mdx mouse heart, this review focuses on how early changes in mitochondrial functional phenotype occur prior to overt cardiomyopathy and may be a determinant for the development of adverse cardiac remodelling leading to failure. These include alterations in energy substrate utilization and signalling of cell death through increased permeability of mitochondrial membranes, which may result from abnormal calcium handling, and production of reactive oxygen species. Furthermore, we will discuss evidence supporting the notion that these alterations in the dystrophin-deficient heart may represent an early "subclinical" signature of a defective nitric oxide/cGMP signalling pathway, as well as the potential benefit of mitochondria-targeted therapies. While the mdx mouse is an animal model of Duchenne muscular dystrophy (DMD), changes in the structural integrity of dystrophin, the mutated cytoskeletal protein responsible for DMD, have also recently been implicated as a common mechanism for contractile dysfunction in heart failure. In fact, altogether our findings support a critical role for dystrophin in maintaining optimal coupling between metabolism and contraction in the heart.
Collapse
Affiliation(s)
- Yan Burelle
- Department of Kinesiology, Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
313
|
High-fat feeding in cardiomyocyte-restricted PPARdelta knockout mice leads to cardiac overexpression of lipid metabolic genes but fails to rescue cardiac phenotypes. J Mol Cell Cardiol 2009; 47:536-43. [PMID: 19595695 DOI: 10.1016/j.yjmcc.2009.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 07/01/2009] [Accepted: 07/01/2009] [Indexed: 11/21/2022]
Abstract
Peroxisome proliferator-activated receptor delta (PPARdelta) is an essential determinant of basal myocardial fatty acid oxidation (FAO) and bioenergetics. We wished to determine whether increased lipid loading affects the PPARdelta deficient heart in transcriptional regulation of FAO and in the development of cardiac pathology. Cardiomyocyte-restricted PPARdelta knockout (CR-PPARdelta(-/-)) and control (alpha-MyHC-Cre) mice were subjected to 48 h of fasting and to a long-term maintenance on a (28 weeks) high-fat diet (HFD). The expression of key FAO proteins in heart was examined. Serum lipid profiles, cardiac pathology, and changes of various transduction signaling pathways were also examined. Mice subjected to fasting exhibited upregulated transcript expression of FAO genes in the CR-PPARdelta(-/-) hearts. Moreover, long-term HFD in CR-PPARdelta(-/-) mice induced a strikingly greater transcriptional response. After HFD, genes encoding key FAO enzymes were expressed remarkably more in CR-PPARdelta(-/-) hearts than in those of control mice. Despite the marked rise of FAO gene expression, corresponding protein expression remained low in the CR-PPARdelta(-/-) heart, accompanied by abnormalities in sarcomere structures and mitochondria that were similar to those of CR-PPARdelta(-/-) hearts with regular chow feeding. The CR-PPARdelta(-/-) mice displayed increased expression of PPARgamma co-activator-1alpha (PGC-1alpha) and PPARalpha in the heart with deactivated Akt and p42/44 MAPK signaling in response to HFD. We conclude that PPARdelta is an essential determinant of myocardial FAO. Increased lipid intake activates cardiac expression of FAO genes via PPARalpha/PGC-1alpha pathway, albeit it is not sufficient to improve cardiac pathology due to PPARdelta deficiency.
Collapse
|
314
|
Finley LW, Haigis MC. The coordination of nuclear and mitochondrial communication during aging and calorie restriction. Ageing Res Rev 2009; 8:173-88. [PMID: 19491041 DOI: 10.1016/j.arr.2009.03.003] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 02/27/2009] [Accepted: 03/04/2009] [Indexed: 12/24/2022]
Abstract
Mitochondria are dynamic organelles that integrate environmental signals to regulate energy production, apoptosis and Ca(2+) homeostasis. Not surprisingly, mitochondrial dysfunction is associated with aging and the pathologies observed in age-related diseases. The vast majority of mitochondrial proteins are encoded in the nuclear genome, and so communication between the nucleus and mitochondria is essential for maintenance of appropriate mitochondrial function. Several proteins have emerged as major regulators of mitochondrial gene expression, capable of increasing transcription of mitochondrial genes in response to the physiological demands of the cell. In this review, we will focus on PGC-1alpha, SIRT1, AMPK and mTOR and discuss how these proteins regulate mitochondrial function and their potential involvement in aging, calorie restriction and age-related disease. We will also discuss the pathways through which mitochondria signal to the nucleus. Although such retrograde signaling is not well studied in mammals, there is growing evidence to suggest that it may be an important area for future aging research. Greater understanding of the mechanisms by which mitochondria and the nucleus communicate will facilitate efforts to slow or reverse the mitochondrial dysfunction that occurs during aging.
Collapse
|
315
|
Meng C, Jin X, Xia L, Shen SM, Wang XL, Cai J, Chen GQ, Wang LS, Fang NY. Alterations of mitochondrial enzymes contribute to cardiac hypertrophy before hypertension development in spontaneously hypertensive rats. J Proteome Res 2009; 8:2463-75. [PMID: 19265432 DOI: 10.1021/pr801059u] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mitochondrial dysfunction is recently thought to be tightly associated with the development of cardiac hypertrophy as well as hypertension. However, the detailed molecular events in mitochondria at early stages of hypertrophic pathogenesis are still unclear. Applying two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) combined with MALDI-TOF/TOF tandem mass spectrometry, here we identified the changed mitochondrial proteins of left ventricular mitochondria in prehypertensive/hypertensive stages of cardiac hypertrophy through comparing spontaneously hypertensive rats (SHR) and the age-matched normotensive Wistar Kyoto (WKY) rats. The results revealed that in the hypertrophic left ventricle of SHR as early as 4 weeks old with normal blood pressure, 33 mitochondrial protein spots presented significant alterations, with 17 down-regulated and 16 up-regulated. Such alterations were much greater than those in 20-week-old SHR with elevated blood pressure. Of the total alterations, the expression of two mitochondrial enzymes, trifunctional enzyme alpha subunit (Hadha) and NADH dehydrogenase 1 alpha subcomplex 10 (Ndufa10), were found to have special expression modification patterns in SHR strain. These data would provide new clues to investigate the potential contribution of mitochondrial dysfunction to the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Chao Meng
- The Department of Geriatrics, Ren-Ji Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai 200001, China
| | | | | | | | | | | | | | | | | |
Collapse
|
316
|
Regulation of myocardial ketone body metabolism by the gut microbiota during nutrient deprivation. Proc Natl Acad Sci U S A 2009; 106:11276-81. [PMID: 19549860 DOI: 10.1073/pnas.0902366106] [Citation(s) in RCA: 198] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Studies in mice indicate that the gut microbiota promotes energy harvest and storage from components of the diet when these components are plentiful. Here we examine how the microbiota shapes host metabolic and physiologic adaptations to periods of nutrient deprivation. Germ-free (GF) mice and mice who had received a gut microbiota transplant from conventionally raised donors were compared in the fed and fasted states by using functional genomic, biochemical, and physiologic assays. A 24-h fast produces a marked change in gut microbial ecology. Short-chain fatty acids generated from microbial fermentation of available glycans are maintained at higher levels compared with GF controls. During fasting, a microbiota-dependent, Ppar alpha-regulated increase in hepatic ketogenesis occurs, and myocardial metabolism is directed to ketone body utilization. Analyses of heart rate, hydraulic work, and output, mitochondrial morphology, number, and respiration, plus ketone body, fatty acid, and glucose oxidation in isolated perfused working hearts from GF and colonized animals (combined with in vivo assessments of myocardial physiology) revealed that the fasted GF heart is able to sustain its performance by increasing glucose utilization, but heart weight, measured echocardiographically or as wet mass and normalized to tibial length or lean body weight, is significantly reduced in both fasted and fed mice. This myocardial-mass phenotype is completely reversed in GF mice by consumption of a ketogenic diet. Together, these results illustrate benefits provided by the gut microbiota during periods of nutrient deprivation, and emphasize the importance of further exploring the relationship between gut microbes and cardiovascular health.
Collapse
|
317
|
Golomb E, Nyska A, Schwalb H. Occult Cardiotoxicity—Toxic Effects on Cardiac Ischemic Tolerance. Toxicol Pathol 2009; 37:572-93. [DOI: 10.1177/0192623309339503] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The outcome of cardiac ischemic events depends not only on the extent and duration of the ischemic stimulus but also on the myocardial intrinsic tolerance to ischemic injury. Cardiac ischemic tolerance reflects myocardial functional reserves that are not always used when the tissue is appropriately oxygenated. Ischemic tolerance is modulated by ubiquitous signal transduction pathways, transcription factors and cellular enzymes, converging on the mitochondria as the main end effector. Therefore, drugs and toxins affecting these pathways may impair cardiac ischemic tolerance without affecting myocardial integrity or function in oxygenated conditions. Such effect would not be detected by current toxicological studies but would considerably influence the outcome of ischemic events. The authors refer to such effect as “occult cardiotoxicity.” In this review, the authors summarize current knowledge about main mechanisms that determine cardiac ischemic tolerance, methods to assess it, and the effects of drugs and toxins on it. The authors offer a view that low cardiac ischemic tolerance is a premorbid status and, therefore, that occult cardiotoxicity is a significant potential source of cardiac morbidity. The authors propose that toxicologic assessment of compounds would include the assessment of their effect on cardiac ischemic tolerance.
Collapse
Affiliation(s)
- Eliahu Golomb
- Department of Pathology, Shaare Zedek Medical Center, Jerusalem 91031, Israel
| | - Abraham Nyska
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 69978, Israel
| | - Herzl Schwalb
- The Joseph Lunenfeld Cardiac Surgery Research Center, Department of Cardiothoracic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| |
Collapse
|
318
|
Handschin C. The biology of PGC-1α and its therapeutic potential. Trends Pharmacol Sci 2009; 30:322-9. [PMID: 19446346 DOI: 10.1016/j.tips.2009.03.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 03/31/2009] [Accepted: 03/31/2009] [Indexed: 02/06/2023]
Abstract
In eukaryotes, cellular and systemic metabolism is primarily controlled by mitochondrial activity. The peroxisome proliferator-activated receptor gamma coactivator 1alpha (PGC-1alpha) is an important regulator of mitochondrial biogenesis and function. Furthermore, PGC-1alpha controls many of the phenotypic adaptations of oxidative tissues to external and internal perturbations. By contrast, dysregulated metabolic plasticity is involved in the etiology of numerous diseases. Accordingly, modulation of PGC-1alpha levels and activity has recently been proposed as a therapeutic option for several pathologies. However, pharmacological interventions aimed at PGC-1alpha have to overcome inherent limitations of targeting a coactivator protein. Here, I focus on the recent breakthroughs in the identification of physiological and pathophysiological contexts involving PGC-1alpha. In addition, perspectives regarding the therapeutic importance of PGC-1alpha-controlled cellular and systemic metabolism are outlined.
Collapse
Affiliation(s)
- Christoph Handschin
- Biozentrum, University of Basel, Klingelbergstrasse 50-70, CH-4056 Basel, Switzerland.
| |
Collapse
|
319
|
Bone marrow–derived mononuclear cell therapy alleviates left ventricular remodeling and improves heart function in rat-dilated cardiomyopathy*. Crit Care Med 2009; 37:1197-205. [DOI: 10.1097/ccm.0b013e31819c0667] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
320
|
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]
|
321
|
Cresci S, Jones PG, Sucharov CC, Marsh S, Lanfear DE, Garsa A, Courtois M, Weinheimer CJ, Wu J, Province MA, Kelly DP, McLeod HL, Spertus JA. Interaction between PPARA genotype and beta-blocker treatment influences clinical outcomes following acute coronary syndromes. Pharmacogenomics 2009; 9:1403-17. [PMID: 18855529 DOI: 10.2217/14622416.9.10.1403] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS beta-blockers (BB) are strongly recommended after an acute coronary syndrome (ACS), although all patients may not benefit. Causes for variable patient responses to BB are unknown. Given that myocardial ischemia and BB influence metabolic processes regulated by peroxisome proliferator-activated receptor alpha (PPARalpha), we hypothesized that interactions between polymorphisms of the PPARalpha gene (PPARA) and BB treatment would influence clinical outcome following ACS. PATIENTS & METHODS Patients were prospectively enrolled into an ACS registry. A total of 735 ACS patients were genotyped. Mortality and cardiac rehospitalization through 1 year were analyzed in relation to PPARA genotype and BB prescription (597 BB; 138 no BB) at discharge. RESULTS Significantly different outcomes associated with BB therapy were observed according to PPARA IVS7 2498 genotype (p = 0.002 for interaction). PPARA IVS7 2498 GG homozygous patients discharged on BB had decreased cardiac rehospitalization (hazard ratio [HR]: 0.52; 95% CI: 0.32-0.86; p = 0.011), while C allele carriers discharged on BB had nearly threefold increased cardiac rehospitalization (HR: 2.92; 95% CI: 1.32-6.92; p = 0.015; genotype interaction p = 0.0005) compared with patients not on BB. PPARA genotype was also associated with differences in PPARalpha expression, with significantly increased mRNA levels in myocardial samples from normal hearts among GC heterozygotes compared with GG homozygotes (p = 0.04). Transgenic mice with cardiac-specific overexpression of PPARalpha showed significantly reduced myocardial contractile and chronotropic responses to the beta-sympathomimetic dobutamine (p < 0.05) compared with wild-type littermates, supporting the hypothesis that increased PPARalpha levels result in a blunted beta-adrenergic response. CONCLUSIONS PPARA IVS7 2498 genotype is associated with heterogeneity in 1-year outcome in response to BB among patients following ACS, and may predict which patients benefit from BB therapy, putatively related to the effect of myocardial PPARalpha expression on beta-adrenergic responsiveness.
Collapse
Affiliation(s)
- Sharon Cresci
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8086, Saint Louis, MO 63110-1093, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
Sihag S, Li AY, Cresci S, Sucharov CC, Lehman JJ. PGC-1alpha and ERRalpha target gene downregulation is a signature of the failing human heart. J Mol Cell Cardiol 2009; 46:201-12. [PMID: 19061896 PMCID: PMC2681265 DOI: 10.1016/j.yjmcc.2008.10.025] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Revised: 10/20/2008] [Accepted: 10/21/2008] [Indexed: 12/25/2022]
Abstract
Heart failure is a cause of significant morbidity and mortality in developed nations, and results from a complex interplay between genetic and environmental factors. To discover gene regulatory networks underlying heart failure, we analyzed DNA microarray data based on left ventricular free-wall myocardium from 59 failing (32 ischemic cardiomyopathy, 27 idiopathic dilated cardiomyopathy) and 33 non-failing explanted human hearts from the Cardiogenomics Consortium. In particular, we sought to investigate cardiac gene expression changes at the level of individual genes, as well as biological pathways which contain groups of functionally related genes. Utilizing a combination of computational techniques, including Comparative Marker Selection and Gene Set Enrichment Analysis, we identified a subset of downstream gene targets of the master mitochondrial transcriptional regulator, peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha), whose expression is collectively decreased in failing human hearts. We also observed decreased expression of the key PGC-1alpha regulatory partner, estrogen-related receptor alpha (ERRalpha), as well as ERRalpha target genes which may participate in the downregulation of mitochondrial metabolic capacity. Gene expression of the antiapoptotic Raf-1/extracellular signal-regulated kinase (ERK) pathway was decreased in failing hearts. Alterations in PGC-1alpha and ERRalpha target gene sets were significantly correlated with an important clinical parameter of disease severity - left ventricular ejection fraction, and were predictive of failing vs. non-failing phenotypes. Overall, our results implicate PGC-1alpha and ERRalpha in the pathophysiology of human heart failure, and define dynamic target gene sets sharing known interrelated regulatory mechanisms capable of contributing to the mitochondrial dysfunction characteristic of this disease process.
Collapse
Affiliation(s)
- Smita Sihag
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Allie Y. Li
- Center for Cardiovascular Research, Department of Medicine, Genetics, Molecular Biology & Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Sharon Cresci
- Center for Cardiovascular Research, Department of Medicine, Genetics, Molecular Biology & Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Carmen C. Sucharov
- Division of Cardiology, School of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | - John J. Lehman
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
323
|
Lee SH, Yang DK, Choi BY, Lee YH, Kim SY, Jeong D, Hajjar RJ, Park WJ. The transcription factor Eya2 prevents pressure overload-induced adverse cardiac remodeling. J Mol Cell Cardiol 2009; 46:596-605. [PMID: 19272299 DOI: 10.1016/j.yjmcc.2008.12.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Revised: 12/30/2008] [Accepted: 12/31/2008] [Indexed: 10/21/2022]
Abstract
Eyes absent 2 (Eya2) is a transcription factor involved in a number of cellular and developmental processes. We have previously shown that Eya2 is up-regulated during regression of cardiac hypertrophy and blocks phenylephrine-induced development of cardiomyocyte hypertrophy in vitro, suggesting that Eya2 is a negative regulator of cardiac hypertrophy. In this study, we generated transgenic mice with cardiac-specific overexpression of Eya2 to elucidate the in vivo function of Eya2 in cardiac remodeling. Mild cardiac hypertrophy developed in Eya2 transgenic mice under baseline conditions with no obvious structural or functional defects. Eya2 overexpression inhibited development of cardiac hypertrophy as judged by the abrogation of increases in heart weight and cross-sectional cell surface areas and re-activation of fetal genes under pressure overload (4 weeks). Eya2 overexpression also prevented wall thinning, ventricular dilation, and deterioration of cardiac function as well as fibrosis and inflammation in the heart under long-term pressure overload (12 weeks). Gene expression profiling using the parametric analysis of gene set enrichment (PAGE) method revealed that gene sets related to mitochondrial biogenesis and metabolism were elevated in the Eya2 transgenic mice. We also observed that the PI3K/Akt/mTOR signaling cascade was preserved in the Eya2 transgenic mice, while it was significantly reduced in the wild type littermates under pressure overload. These results demonstrate that Eya2 prevents adverse cardiac remodeling under pressure overload partly through altering metabolic gene expression and preserving PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Seung Hee Lee
- Department of Life Science, Global Research Lab, Gwangju Institute of Science and Technology, Buk-gu, Gwangju, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
324
|
|
325
|
Vornanen M, Stecyk JA, Nilsson GE. Chapter 9 The Anoxia-Tolerant Crucian Carp (Carassius Carassius L.). FISH PHYSIOLOGY 2009. [DOI: 10.1016/s1546-5098(08)00009-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
326
|
Palomer X, Alvarez-Guardia D, Rodríguez-Calvo R, Coll T, Laguna JC, Davidson MM, Chan TO, Feldman AM, Vázquez-Carrera M. TNF-alpha reduces PGC-1alpha expression through NF-kappaB and p38 MAPK leading to increased glucose oxidation in a human cardiac cell model. Cardiovasc Res 2008; 81:703-12. [PMID: 19038972 DOI: 10.1093/cvr/cvn327] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Inflammatory responses in the heart that are driven by sustained increases in cytokines have been associated with several pathological processes, including cardiac hypertrophy and heart failure. Emerging data suggest a link between cardiomyopathy and myocardial metabolism dysregulation. To further elucidate the relationship between a pro-inflammatory profile and cardiac metabolism dysregulation, a human cell line of cardiac origin, AC16, was treated with tumour necrosis factor-alpha (TNF-alpha). METHODS AND RESULTS Exposure of AC16 cells to TNF-alpha inhibited the expression of peroxisome proliferator-activated receptor coactivator 1alpha (PGC-1alpha), an upstream regulator of lipid and glucose oxidative metabolism. Studies performed with cardiac-specific transgenic mice (Mus musculus) overexpressing TNF-alpha, which have been well characterized as a model of cytokine-induced cardiomyopathy, also displayed reduced PGC-1alpha expression in the heart compared with that of control mice. The mechanism by which TNF-alpha reduced PGC-1alpha expression in vitro appeared to be largely mediated via both p38 mitogen-activated protein kinase and nuclear factor-kappaB pathways. PGC-1alpha downregulation resulted in an increase in glucose oxidation rate, which involved a reduction in pyruvate dehydrogenase kinase 4 expression and depended on the DNA-binding activity of both peroxisome proliferator-activated receptor beta/delta and estrogen-related receptor alpha transcription factors. CONCLUSION These results point to PGC-1alpha downregulation as a potential contributor to cardiac dysfunction and heart failure in metabolic disorders with an inflammatory background.
Collapse
Affiliation(s)
- Xavier Palomer
- Pharmacology Unit, Department of Pharmacology and Therapeutic Chemistry, IBUB and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
327
|
Lin JD. Minireview: the PGC-1 coactivator networks: chromatin-remodeling and mitochondrial energy metabolism. Mol Endocrinol 2008; 23:2-10. [PMID: 19008463 DOI: 10.1210/me.2008-0344] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Transcriptional coactivators and corepressors are emerging as important regulators of energy metabolism and other biological processes. These factors exert their effects on the transcription of target genes through interaction with selective transcription factors and the recruitment of chromatin-remodeling complexes. Recent genetic and biochemical analyses of the peroxisomal proliferator-activated receptor-gamma coactivator 1 networks provide novel mechanistic insights regarding their role in the control of mitochondrial oxidative metabolism. These coactivators integrate tissue metabolic functions in response to nutritional signals as well as circadian timing cues. In contrast to coactivators, transcriptional corepressors have been demonstrated to play an opposite role in the control of mitochondrial biogenesis and respiration. The balance of these coactivator and corepressor proteins and, more importantly, their access to specific transcriptional partners are predicted to dictate the epigenetic states of target genes as well as the metabolic phenotype of the cells. This review highlights the biological role and mechanistic basis of the peroxisomal proliferator-activated receptor-gamma coactivator 1 networks in the regulation of chromatin-remodeling and mitochondrial oxidative metabolism.
Collapse
Affiliation(s)
- Jiandie D Lin
- Life Sciences Institute, Department of Cell and Developmental Biology, University of Michigan Medical Center, 210 Washtenaw Avenue, Ann Arbor, Michigan 48109, USA.
| |
Collapse
|
328
|
Stecyk JAW, Galli GL, Shiels HA, Farrell AP. Cardiac survival in anoxia-tolerant vertebrates: An electrophysiological perspective. Comp Biochem Physiol C Toxicol Pharmacol 2008; 148:339-54. [PMID: 18589002 DOI: 10.1016/j.cbpc.2008.05.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Revised: 05/31/2008] [Accepted: 05/31/2008] [Indexed: 11/24/2022]
Abstract
Certain vertebrates, such as freshwater turtles of the genus Chrysemys and Trachemys and crucian carp (Carassius carassius), have anoxia-tolerant hearts that continue to function throughout prolonged periods of anoxia (up to many months) due to successful balancing of cellular ATP supply and demand. In the present review, we summarize the current and limited understanding of the cellular mechanisms underlying this cardiac anoxia tolerance. What emerges is that cold temperature substantially modifies cardiac electrophysiology to precondition the heart for winter anoxia. Intrinsic heart rate is slowed and density of sarcolemmal ion currents substantially modified to alter cardiac action potential (AP) characteristics. These changes depress cardiac activity and reduce the energetic costs associated with ion pumping. In contrast, anoxia per se results in limited changes to cardiac AP shape or ion current densities in turtle and crucian carp, suggesting that anoxic modifications of cardiac electrophysiology to reduce ATP demand are not extensive. Additionally, as knowledge of cellular physiology in non-mammalian vertebrates is still in its infancy, we briefly discuss the cellular defense mechanisms towards the acidosis that accompanies anoxia as well as mammalian cardiac models of hypoxia/ischemia tolerance. By examining if fundamental cellular mechanisms have been conserved during the evolution of anoxia tolerance we hope to have provided a framework for the design of future experiments investigating cardiac cellular mechanisms of anoxia survival.
Collapse
Affiliation(s)
- Jonathan A W Stecyk
- Physiology Programme, Department of Molecular Biosciences, University of Oslo, PO Box 1041, N-0316, Oslo, Norway.
| | | | | | | |
Collapse
|
329
|
Abstract
Transcriptional control of cellular energy metabolic pathways is achieved by the coordinated action of numerous transcription factors and associated coregulators. Several members of the nuclear receptor superfamily have been shown to play important roles in this process because they can translate hormonal, nutrient, and metabolite signals into specific gene expression networks to satisfy energy demands in response to distinct physiological cues. Estrogen-related receptor (ERR) alpha, ERRbeta, and ERRgamma are nuclear receptors that have yet to be associated with a natural ligand and are thus considered as orphan receptors. However, the transcriptional activity of the ERRs is exquisitely sensitive to the presence of coregulatory proteins known to be essential for the control of energy homeostasis, and for all intents and purposes, these coregulators function as protein ligands for the ERRs. In particular, functional genomics and biochemical studies have shown that ERRalpha and ERRgamma operate as the primary conduits for the activity of members of the family of PGC-1 coactivators. As transcription factors, the ERRs control vast gene networks involved in all aspects of energy homeostasis, including fat and glucose metabolism as well as mitochondrial biogenesis and function. Phenotypic analyses of knockout mouse models have shown that all three ERRs are indispensable for proper development and/or survival of the organism when subjected to a variety of physiological challenges. The focus of this review is on the recent and rapid advances in understanding the functions of the ERRs in regulating bioenergetic pathways, with an emphasis on their roles in the specification of energetic properties required for cell- and tissue-specific functions.
Collapse
Affiliation(s)
- Vincent Giguère
- The Rosalind and Morris Goodman Cancer Centre, Cancer Pavilion, 1160 Pine Avenue West, Montreal, Quebec, Canada H3A 1A3.
| |
Collapse
|
330
|
Elas M, Bielanska J, Pustelny K, Plonka PM, Drelicharz L, Skorka T, Tyrankiewicz U, Wozniak M, Heinze-Paluchowska S, Walski M, Wojnar L, Fortin D, Ventura-Clapier R, Chlopicki S. Detection of mitochondrial dysfunction by EPR technique in mouse model of dilated cardiomyopathy. Free Radic Biol Med 2008; 45:321-8. [PMID: 18466775 DOI: 10.1016/j.freeradbiomed.2008.04.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Revised: 03/21/2008] [Accepted: 04/09/2008] [Indexed: 11/28/2022]
Abstract
Tgalphaq44 mice with targeted overexpression of activated Galphaq protein in cardiomyocytes mimic many of the phenotypic characteristics of dilated cardiomyopathy in humans. However, it is not known whether the phenotype of Tgalphaq44 mice would also involve dysfunction of cardiac mitochondria. The aim of the present work was to examine changes in EPR signals of semiquinones and iron in Fe-S clusters, as compared to classical biochemical indices of mitochondrial function in hearts from Tgalphaq44 mice in relation to the progression of heart failure. Tgalphaq44 mice at the age of 14 months displayed pulmonary congestion, increased heart/body ratio and impairment of cardiac function as measured in vivo by MRI. However, in hearts from Tgalphaq44 mice already at the age of 10 months EPR signals of semiquinones, as well as cyt c oxidase activity were decreased, suggesting alterations in mitochondrial electron flow. Furthermore, in 14-months old Tgalphaq44 mice loss of iron in Fe-S clusters, impaired citrate synthase activity, and altered mitochondrial ultrastructure were observed, supporting mitochondrial dysfunction in Tgalphaq44 mice. In conclusion, the assessment of semiquinones content and Fe(III) analysis by EPR represents a rational approach to detect dysfunction of cardiac mitochondria. Decreased contents of semiquinones detected by EPR and a parallel decrease in cyt c oxidase activity occurs before hemodynamic decompensation of heart failure in Tgalphaq44 mice suggesting that alterations in function of cardiac mitochondria contribute to the development of the overt heart failure in this model.
Collapse
Affiliation(s)
- Martyna Elas
- Department of Biophysics, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
331
|
Affiliation(s)
- Romain Harmancey
- Department of Internal Medicine, Division of Cardiology, University of Texas Medical School at Houston, 6431 Fannin, MSB 1.246, Houston, TX 77030, USA
| | | | | |
Collapse
|
332
|
Lehman JJ, Boudina S, Banke NH, Sambandam N, Han X, Young DM, Leone TC, Gross RW, Lewandowski ED, Abel ED, Kelly DP. The transcriptional coactivator PGC-1alpha is essential for maximal and efficient cardiac mitochondrial fatty acid oxidation and lipid homeostasis. Am J Physiol Heart Circ Physiol 2008; 295:H185-96. [PMID: 18487436 DOI: 10.1152/ajpheart.00081.2008] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High-capacity mitochondrial ATP production is essential for normal function of the adult heart, and evidence is emerging that mitochondrial derangements occur in common myocardial diseases. Previous overexpression studies have shown that the inducible transcriptional coactivator peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1alpha is capable of activating postnatal cardiac myocyte mitochondrial biogenesis. Recently, we generated mice deficient in PGC-1alpha (PGC-1alpha(-/-) mice), which survive with modestly blunted postnatal cardiac growth. To determine if PGC-1alpha is essential for normal cardiac energy metabolic capacity, mitochondrial function experiments were performed on saponin-permeabilized myocardial fibers from PGC-1alpha(-/-) mice. These experiments demonstrated reduced maximal (state 3) palmitoyl-l-carnitine respiration and increased maximal (state 3) pyruvate respiration in PGC-1alpha(-/-) mice compared with PGC-1alpha(+/+) controls. ATP synthesis rates obtained during maximal (state 3) respiration in permeabilized myocardial fibers were reduced for PGC-1alpha(-/-) mice, whereas ATP produced per oxygen consumed (ATP/O), a measure of metabolic efficiency, was decreased by 58% for PGC-1alpha(-/-) fibers. Ex vivo isolated working heart experiments demonstrated that PGC-1alpha(-/-) mice exhibited lower cardiac power, reduced palmitate oxidation, and increased reliance on glucose oxidation, with the latter likely a compensatory response. (13)C NMR revealed that hearts from PGC-1alpha(-/-) mice exhibited a limited capacity to recruit triglyceride as a source for lipid oxidation during beta-adrenergic challenge. Consistent with reduced mitochondrial fatty acid oxidative enzyme gene expression, the total triglyceride content was greater in hearts of PGC-1alpha(-/-) mice relative to PGC-1alpha(+/+) following a fast. Overall, these results demonstrate that PGC-1alpha is essential for the maintenance of maximal, efficient cardiac mitochondrial fatty acid oxidation, ATP synthesis, and myocardial lipid homeostasis.
Collapse
Affiliation(s)
- John J Lehman
- Center for Cardiovascular Research, Washington Univ. School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
333
|
Burkart EM, Sambandam N, Han X, Gross RW, Courtois M, Gierasch CM, Shoghi K, Welch MJ, Kelly DP. Nuclear receptors PPARbeta/delta and PPARalpha direct distinct metabolic regulatory programs in the mouse heart. J Clin Invest 2008; 117:3930-9. [PMID: 18037994 DOI: 10.1172/jci32578] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Accepted: 09/26/2007] [Indexed: 11/17/2022] Open
Abstract
In the diabetic heart, chronic activation of the PPARalpha pathway drives excessive fatty acid (FA) oxidation, lipid accumulation, reduced glucose utilization, and cardiomyopathy. The related nuclear receptor, PPARbeta/delta, is also highly expressed in the heart, yet its function has not been fully delineated. To address its role in myocardial metabolism, we generated transgenic mice with cardiac-specific expression of PPARbeta/delta, driven by the myosin heavy chain (MHC-PPARbeta/delta mice). In striking contrast to MHC-PPARalpha mice, MHC-PPARbeta/delta mice had increased myocardial glucose utilization, did not accumulate myocardial lipid, and had normal cardiac function. Consistent with these observed metabolic phenotypes, we found that expression of genes involved in cellular FA transport were activated by PPARalpha but not by PPARbeta/delta. Conversely, cardiac glucose transport and glycolytic genes were activated in MHC-PPARbeta/delta mice, but repressed in MHC-PPARalpha mice. In reporter assays, we showed that PPARbeta/delta and PPARalpha exerted differential transcriptional control of the GLUT4 promoter, which may explain the observed isotype-specific effects on glucose uptake. Furthermore, myocardial injury due to ischemia/reperfusion injury was significantly reduced in the MHC-PPARbeta/delta mice compared with control or MHC-PPARalpha mice, consistent with an increased capacity for myocardial glucose utilization. These results demonstrate that PPARalpha and PPARbeta/delta drive distinct cardiac metabolic regulatory programs and identify PPARbeta/delta as a potential target for metabolic modulation therapy aimed at cardiac dysfunction caused by diabetes and ischemia.
Collapse
Affiliation(s)
- Eileen M Burkart
- Center for Cardiovascular Research, Department of Medicine,Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
334
|
Chicco AJ, McCune SA, Emter CA, Sparagna GC, Rees ML, Bolden DA, Marshall KD, Murphy RC, Moore RL. Low-intensity exercise training delays heart failure and improves survival in female hypertensive heart failure rats. Hypertension 2008; 51:1096-102. [PMID: 18259016 DOI: 10.1161/hypertensionaha.107.107078] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Exercise training improves functional capacity and quality of life in patients with heart failure. However, the long-term effects of exercise on mortality associated with hypertensive heart disease have not been well defined. In the present study, we investigated the effect of low-intensity exercise training on disease progression and survival in female spontaneously hypertensive heart failure rats. Animals with severe hypertension (16 months old) were treadmill trained (14.5 m/min, 45 min/d, 3 d/wk) until they developed terminal heart failure or were euthanized because of age-related complications. Exercise delayed mortality resulting from heart failure (P<0.001) and all causes (P<0.05) and transiently attenuated the systolic hypertension and contractile dysfunction observed in the sedentary animals but had no effect on cardiac morphology or contractile function in end-stage heart failure. Training had no effect on terminal myocardial protein expression of antioxidant enzymes, calcium handling proteins, or myosin heavy chain isoforms but was associated with higher cytochrome oxidase activity in cardiac mitochondria (P<0.05) and a greater mitochondrial content of cardiolipin, a phospholipid that is essential for optimal mitochondrial energy metabolism. In conclusion, low-intensity exercise training significantly delays the onset of heart failure and improves survival in female hypertensive heart failure rats without eliciting sustained improvements in blood pressure, cardiac function, or expression of several myocardial proteins associated with the cardiovascular benefits of exercise. The effects of exercise on cytochrome oxidase and cardiolipin provide novel evidence that training may improve prognosis in hypertensive heart disease by preserving mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Adam J Chicco
- Department of Integrative Physiology, University of Colorado Cardiovascular Research Institute, University of Colorado, Boulder, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
335
|
Persistent Regional Downregulation in Mitochondrial Enzymes and Upregulation of Stress Proteins in Swine With Chronic Hibernating Myocardium. Circ Res 2008; 102:103-12. [DOI: 10.1161/circresaha.107.155895] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hibernating myocardium is accompanied by a downregulation in energy utilization that prevents the immediate development of ischemia during stress at the expense of an attenuated level of regional contractile function. We used a discovery based proteomic approach to identify novel regional molecular adaptations responsible for this phenomenon in subendocardial samples from swine instrumented with a chronic LAD stenosis. After 3 months (n=8), hibernating myocardium was present as reflected by reduced resting LAD flow (0.75±0.14 versus 1.19±0.14 mL · min
−1
· g
−1
in remote) and wall thickening (1.93±0.46 mm versus 5.46±0.41 mm in remote,
P
<0.05). Regionally altered proteins were quantified with 2D Differential-in-Gel Electrophoresis (2D-DIGE) using normal myocardium as a reference with identification of candidates using MALDI-TOF mass spectrometry. Hibernating myocardium developed a significant downregulation of many mitochondrial proteins and an upregulation of stress proteins. Of particular note, the major entry points to oxidative metabolism (eg, pyruvate dehydrogenase complex and Acyl-CoA dehydrogenase) and enzymes involved in electron transport (eg, complexes I, III, and V) were reduced (
P
<0.05). Multiple subunits within an enzyme complex frequently showed a concordant downregulation in abundance leading to an amplification of their cumulative effects on activity (eg, “total” LAD PDC activity was 21.9±3.1 versus 42.8±1.9 mU,
P
<0.05). After 5-months (n=10), changes in mitochondrial and stress proteins persisted whereas cytoskeletal proteins (eg, desmin and vimentin) normalized. These data indicate that the proteomic phenotype of hibernating myocardium is dynamic and has similarities to global changes in energy substrate metabolism and function in the advanced failing heart. These proteomic changes may limit oxidative injury and apoptosis and impact functional recovery after revascularization.
Collapse
|
336
|
Shaik ZP, Fifer EK, Nowak G. Akt activation improves oxidative phosphorylation in renal proximal tubular cells following nephrotoxicant injury. Am J Physiol Renal Physiol 2007; 294:F423-32. [PMID: 18077599 DOI: 10.1152/ajprenal.00463.2007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previously, we showed that protein kinase B (Akt) activation increases intracellular ATP levels and decreases necrosis in renal proximal tubular cells (RPTC) injured by the nephrotoxicant S-(1, 2-dichlorovinyl)-l-cysteine (DCVC) (Shaik ZP, Fifer EK, Nowak G. Am J Physiol Renal Physiol 292: F292-F303, 2007). This study examined the role of Akt in improving mitochondrial function in DCVC-injured RPTC. Our data show a novel observation that phosphorylated (active) Akt is localized in mitochondria of noninjured RPTC, both in mitoplasts and the mitochondrial outer membrane. Mitochondrial levels of active Akt decreased in nephrotoxicant-injured RPTC, and this decrease was associated with mitochondrial dysfunction. DCVC decreased basal, uncoupled, and state 3 respirations; ATP production; activities of complexes I, II, and III; the mitochondrial membrane potential (DeltaPsi(m)); and F(0)F(1)-ATPase activity. Expressing constitutively active Akt in DCVC-injured RPTC increased the levels of phosphorylated Akt in mitochondria, reduced the decreases in basal and uncoupled respirations, increased complex I-coupled state 3 respiration and ATP production, enhanced activities of complex I, complex III, and F(0)F(1)-ATPase, and improved DeltaPsi(m). In contrast, inhibiting Akt activation by expressing dominant negative (inactive) Akt or using 20 microM LY294002 exacerbated decreases in electron transport rate, state 3 respiration, ATP production, DeltaPsi(m), and activities of complex I, complex III, and F(0)F(1)-ATPase. In conclusion, our data show that Akt activation promotes mitochondrial respiration and ATP production in toxicant-injured RPTC by 1) improving integrity of the respiratory chain and maintaining activities of complex I and complex III, 2) reducing decreases in DeltaPsi(m), and 3) restoring F(0)F(1)-ATPase activity.
Collapse
Affiliation(s)
- Zabeena P Shaik
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | | |
Collapse
|
337
|
Abstract
We derive an analytical model of the potential differences induced across plasma and internal organelle membranes in suspended cells exposed to oscillatory electric fields. Multiple shells are modeled using iterative applications of the single-shell calculation with mobile charges. This work is motivated, in part, by recent results suggesting the ability to use alternating current (ac) fields to noninvasively monitor enzyme activity within internal membranes, particularly the mitochondrial electron transport chain. Previous work, on induced transmembrane voltages in cells subjected to ac fields, has mainly been limited to oscillatory potentials across the plasma membrane. Here we first develop a three-membrane model, consisting of a plasma membrane surrounding inner and outer membranes representing an internal organelle, such as a mitochondrion. Frequency-dependent transmembrane potentials are modeled for spherical, weakly conducting membrane shells enclosing a conductive cytoplasm surrounding an idealized internal organelle. We then use a two-shell model to simulate induced ac membrane potentials of a suspended isolated mitochondrion in which the outer membrane is usually much more permeable than the inner membrane.
Collapse
|
338
|
Jucker BM, Doe CP, Schnackenberg CG, Olzinski AR, Maniscalco K, Williams C, Hu TCC, Lenhard SC, Costell M, Bernard R, Sarov-Blat L, Steplewski K, Willette RN. PPARdelta activation normalizes cardiac substrate metabolism and reduces right ventricular hypertrophy in congestive heart failure. J Cardiovasc Pharmacol 2007; 50:25-34. [PMID: 17666912 DOI: 10.1097/fjc.0b013e31804b4163] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Previously, it was shown that selective deletion of peroxisome proliferator activated receptor delta (PPARdelta) in the heart resulted in a cardiac lipotoxicity, hypertrophy, and heart failure. The aim of the present study was to determine the effects of chronic and selective pharmacological activation of PPARdelta in a model of congestive heart failure. PPARdelta-specific agonist treatment (GW610742X at 30 and 100 mg/kg/day for 6-9 weeks) was initiated immediately postmyocardial infarction (MI) in Sprague-Dawley rats. Magnetic resonance imaging/spectroscopy was used to assess cardiac function and energetics. A 1-(13)C glucose clamp was performed to assess relative cardiac carbohydrate versus fat oxidation. Additionally, cardiac hemodynamics and reverse-transcription polymerase chain reaction gene expression analysis was performed. MI rats had significantly reduced left ventricle (LV) ejection fractions and whole heart phosphocreatine/adenosine triphosphate ratio compared with Sham animals (reduction of 43% and 14%, respectively). However, GW610742X treatment had no effect on either parameter. In contrast, the decrease in relative fat oxidation rate observed in both LV and right ventricle (RV) following MI (decrease of 58% and 54%, respectively) was normalized in a dose-dependent manner following treatment with GW610742X. These metabolic changes were associated with an increase in lipid transport/metabolism target gene expression (eg, CD36, CPT1, UCP3). Although there was no difference between groups in LV weight or infarct size measured upon necropsy, there was a dramatic reduction in RV hypertrophy and lung congestion (decrease of 22-48%, P<0.01) with treatment which was associated with a >7-fold decrease (P<0.05) in aterial natriuretic peptide gene expression in RV. Diuretic effects were not observed with GW610742X. In conclusion, chronic treatment with a selective PPARdelta agonist normalizes cardiac substrate metabolism and reduces RV hypertrophy and pulmonary congestion consistent with improvement in congestive heart failure.
Collapse
Affiliation(s)
- Beat M Jucker
- Cardiovascular and Urogenital Center of Excellence for Drug Discovery, and Genomics Research, GlaxoSmithKline, King of Prussia, PA 19406, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
339
|
Gramolini AO, Kislinger T, Alikhani-Koopaei R, Fong V, Thompson NJ, Isserlin R, Sharma P, Oudit GY, Trivieri MG, Fagan A, Kannan A, Higgins DG, Huedig H, Hess G, Arab S, Seidman JG, Seidman CE, Frey B, Perry M, Backx PH, Liu PP, MacLennan DH, Emili A. Comparative proteomics profiling of a phospholamban mutant mouse model of dilated cardiomyopathy reveals progressive intracellular stress responses. Mol Cell Proteomics 2007; 7:519-33. [PMID: 18056057 DOI: 10.1074/mcp.m700245-mcp200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Defective mobilization of Ca2+ by cardiomyocytes can lead to cardiac insufficiency, but the causative mechanisms leading to congestive heart failure (HF) remain unclear. In the present study we performed exhaustive global proteomics surveys of cardiac ventricle isolated from a mouse model of cardiomyopathy overexpressing a phospholamban mutant, R9C (PLN-R9C), and exhibiting impaired Ca2+ handling and death at 24 weeks and compared them with normal control littermates. The relative expression patterns of 6190 high confidence proteins were monitored by shotgun tandem mass spectrometry at 8, 16, and 24 weeks of disease progression. Significant differential abundance of 593 proteins was detected. These proteins mapped to select biological pathways such as endoplasmic reticulum stress response, cytoskeletal remodeling, and apoptosis and included known biomarkers of HF (e.g. brain natriuretic peptide/atrial natriuretic factor and angiotensin-converting enzyme) and other indicators of presymptomatic functional impairment. These altered proteomic profiles were concordant with cognate mRNA patterns recorded in parallel using high density mRNA microarrays, and top candidates were validated by RT-PCR and Western blotting. Mapping of our highest ranked proteins against a human diseased explant and to available data sets indicated that many of these proteins could serve as markers of disease. Indeed we showed that several of these proteins are detectable in mouse and human plasma and display differential abundance in the plasma of diseased mice and affected patients. These results offer a systems-wide perspective of the dynamic maladaptions associated with impaired Ca2+ homeostasis that perturb myocyte function and ultimately converge to cause HF.
Collapse
Affiliation(s)
- Anthony O Gramolini
- Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario M5G 1L6, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
340
|
Berthiaume JM, Wallace KB. Persistent alterations to the gene expression profile of the heart subsequent to chronic Doxorubicin treatment. Cardiovasc Toxicol 2007; 7:178-91. [PMID: 17901561 DOI: 10.1007/s12012-007-0026-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Revised: 11/30/1999] [Accepted: 07/02/2007] [Indexed: 10/23/2022]
Abstract
Doxorubicin (DOX, Adriamycin) is a potent antineoplastic agent used to treat a number of cancers. Despite its utility, DOX causes a cumulative, irreversible cardiomyopathy that may become apparent shortly after treatment or years subsequent to therapy. Numerous studies have been conducted to elucidate the basis of DOX cardiotoxicity, but the precise mechanism responsible remains elusive. This investigation was designed to assess global gene expression using microarrays in order to identify the full spectrum of potential molecular targets of DOX cardiotoxicity to further delineate the underlying pathological mechanism(s) responsible for this dose-limiting cardiomyopathy. Male, Sprague-Dawley rats received 6 weekly injections of 2 mg/kg (s.c.) DOX followed by a 5 week drug-free period prior to analysis of cardiac tissue transcripts. Ontological evaluation in terms of subcellular targets identified gene products involved in mitochondrial processes are significantly suppressed, consistent with the well-established persistent mitochondrial dysfunction. Further classification of genes into biochemical networks revealed several pathways modulated by DOX, including glycolysis and fatty acid metabolism, supporting the notion that mitochondria are key targets in DOX toxicity. In conclusion, this comprehensive transcript profile provides important insights into critical targets and molecular adaptations that characterize the persistent cardiomyopathy associated with long-term exposure to DOX.
Collapse
Affiliation(s)
- Jessica M Berthiaume
- Toxicology Graduate Program, Department of Biochemistry and Molecular Biology, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | | |
Collapse
|
341
|
Abstract
The prevalence of Type 2 diabetes is increasing at an alarming rate in most parts of the world. Effective therapeutic drugs are urgently needed, not only to control the disease but also to prevent or delay its progression. Therapies that target the underlying pathogenesis could, in theory, hold such potential. Recent evidence strongly suggests that impaired mitochondrial function is part of the underlying pathogenesis of insulin resistance and Type 2 diabetes. Peroxisome proliferator-activated receptor gamma co-activator-1 alpha (PGC-1alpha) is a transcription co-activator that plays a key role in regulating mitochondrial biogenesis and energy metabolism in multiple tissues. Thus, improvement and restoration of mitochondrial function and oxidative capacity through activation of PGC-1alpha could provide new treatments for metabolic diseases. A diverse array of proteins has been shown to regulate PGC-1alpha transcription and/or activity, some of which represent promising targets for pharmaceutical intervention.
Collapse
Affiliation(s)
- Zhidan Wu
- Novartis Institutes for BioMedical Research, Inc., 100 Technology Square, Cambridge, MA 02139, USA.
| | | |
Collapse
|
342
|
Doss MX, Winkler J, Chen S, Hippler-Altenburg R, Sotiriadou I, Halbach M, Pfannkuche K, Liang H, Schulz H, Hummel O, Hübner N, Rottscheidt R, Hescheler J, Sachinidis A. Global transcriptome analysis of murine embryonic stem cell-derived cardiomyocytes. Genome Biol 2007; 8:R56. [PMID: 17428332 PMCID: PMC1896009 DOI: 10.1186/gb-2007-8-4-r56] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 02/16/2007] [Accepted: 04/11/2007] [Indexed: 12/12/2022] Open
Abstract
Microarray analysis reveals that the specific pattern of gene expression in cardiomyocytes derived from embryonic stem cells reflects the biological, physiological and functional processes occurring in mature cardiomyocytes. Background Characterization of gene expression signatures for cardiomyocytes derived from embryonic stem cells will help to define their early biologic processes. Results A transgenic α-myosin heavy chain (MHC) embryonic stem cell lineage was generated, exhibiting puromycin resistance and expressing enhanced green fluorescent protein (EGFP) under the control of the α-MHC promoter. A puromycin-resistant, EGFP-positive, α-MHC-positive cardiomyocyte population was isolated with over 92% purity. RNA was isolated after electrophysiological characterization of the cardiomyocytes. Comprehensive transcriptome analysis of α-MHC-positive cardiomyocytes in comparison with undifferentiated α-MHC embryonic stem cells and the control population from 15-day-old embryoid bodies led to identification of 884 upregulated probe sets and 951 downregulated probe sets in α-MHC-positive cardiomyocytes. A subset of upregulated genes encodes cytoskeletal and voltage-dependent channel proteins, and proteins that participate in aerobic energy metabolism. Interestingly, mitosis, apoptosis, and Wnt signaling-associated genes were downregulated in the cardiomyocytes. In contrast, annotations for genes upregulated in the α-MHC-positive cardiomyocytes are enriched for the following Gene Ontology (GO) categories: enzyme-linked receptor protein signaling pathway (GO:0007167), protein kinase activity (GO:0004672), negative regulation of Wnt receptor signaling pathway (GO:0030178), and regulation of cell size (O:0008361). They were also enriched for the Biocarta p38 mitogen-activated protein kinase signaling pathway and Kyoto Encyclopedia of Genes and Genomes (KEGG) calcium signaling pathway. Conclusion The specific pattern of gene expression in the cardiomyocytes derived from embryonic stem cells reflects the biologic, physiologic, and functional processes that take place in mature cardiomyocytes. Identification of cardiomyocyte-specific gene expression patterns and signaling pathways will contribute toward elucidating their roles in intact cardiac function.
Collapse
Affiliation(s)
- Michael Xavier Doss
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Johannes Winkler
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Shuhua Chen
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Rita Hippler-Altenburg
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Isaia Sotiriadou
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Marcel Halbach
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Kurt Pfannkuche
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Huamin Liang
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Herbert Schulz
- Max-Delbrueck-Center for Molecular Medicine - MDC, Robert-Rössle Str., 13092 Berlin, Germany
| | - Oliver Hummel
- Max-Delbrueck-Center for Molecular Medicine - MDC, Robert-Rössle Str., 13092 Berlin, Germany
| | - Norbert Hübner
- Max-Delbrueck-Center for Molecular Medicine - MDC, Robert-Rössle Str., 13092 Berlin, Germany
| | - Ruth Rottscheidt
- Institute for Genetics, Department of Evolutionary Genetics, University of Cologne, Zülpicher Str., 50674 Cologne, Germany
| | - Jürgen Hescheler
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert Koch Str., 50931 Cologne, Germany
| |
Collapse
|
343
|
Abstract
PURPOSE OF REVIEW Energetic abnormalities in cardiac and skeletal muscle occur in heart failure and correlate with clinical symptoms and mortality. It is likely that the cellular mechanism leading to energetic failure involves mitochondrial dysfunction. Therefore, it is crucial to elucidate the causes of mitochondrial myopathy, in order to improve cardiac and skeletal muscle function, and hence quality of life, in heart failure patients. RECENT FINDINGS Recent studies identified several potential stresses that lead to mitochondrial dysfunction in heart failure. Chronically elevated plasma free fatty acid levels in heart failure are associated with decreased metabolic efficiency and cellular insulin resistance. Tissue hypoxia, resulting from low cardiac output and endothelial impairment, can lead to oxidative stress and mitochondrial DNA damage, which in turn causes dysfunction and loss of mitochondrial mass. Therapies aimed at protecting mitochondrial function have shown promise in patients and animal models with heart failure. SUMMARY Despite current therapies, which provide substantial benefit to patients, heart failure remains a relentlessly progressive disease, and new approaches to treatment are necessary. Novel pharmacological agents are needed that optimize substrate metabolism and maintain mitochondrial integrity, improve oxidative capacity in heart and skeletal muscle, and alleviate many of the clinical symptoms associated with heart failure.
Collapse
Affiliation(s)
- Andrew J Murray
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK. [corrected]
| | | | | |
Collapse
|
344
|
Abstract
The goal of pharmacogenetics is to define the genetic determinants of individual drug responsiveness, and thereby provide personalized treatment to each individual. The peroxisome proliferator-activated receptors (PPARs) are polypeptide products of a set of related genes functioning to regulate several cellular processes that are central to cardiovascular health and disease. Given their pleiotropic roles in lipid and glucose homeostasis, cardiac energy balance and regulation of adipocyte release of circulating inflammatory factors, it is not surprising that PPARs represent an attractive target for clinical investigation and intervention in disease states, such as diabetes, obesity, atherosclerosis, cardiomyopathy, cardiac hypertrophy and heart failure. Research into the manipulation of PPAR function by pharmacologic agents has already resulted in important advances in the treatment of diabetes mellitus and cardiovascular disease. It follows that PPAR pharmacogenetics promises important advances in the personalized treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Sharon Cresci
- Washington University School of Medicine, Department of Medicine, Saint Louis, Missouri, 660 South Euclid Avenue, Campus Box 8086 Saint Louis, MO 63110-1093, USA
| |
Collapse
|
345
|
Mancuso DJ, Sims HF, Han X, Jenkins CM, Guan SP, Yang K, Moon SH, Pietka T, Abumrad NA, Schlesinger PH, Gross RW. Genetic ablation of calcium-independent phospholipase A2gamma leads to alterations in mitochondrial lipid metabolism and function resulting in a deficient mitochondrial bioenergetic phenotype. J Biol Chem 2007; 282:34611-22. [PMID: 17923475 DOI: 10.1074/jbc.m707795200] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously, we identified a novel calcium-independent phospholipase, designated calcium-independent phospholipase A(2) gamma (iPLA(2)gamma), which possesses dual mitochondrial and peroxisomal subcellular localization signals. To identify the roles of iPLA(2)gamma in cellular bioenergetics, we generated mice null for the iPLA(2)gamma gene by eliminating the active site of the enzyme through homologous recombination. Mice null for iPLA(2)gamma display multiple bioenergetic dysfunctional phenotypes, including 1) growth retardation, 2) cold intolerance, 3) reduced exercise endurance, 4) greatly increased mortality from cardiac stress after transverse aortic constriction, 5) abnormal mitochondrial function with a 65% decrease in ascorbate-induced Complex IV-mediated oxygen consumption, and 6) a reduction in myocardial cardiolipin content accompanied by an altered cardiolipin molecular species composition. We conclude that iPLA(2)gamma is essential for maintaining efficient bioenergetic mitochondrial function through tailoring mitochondrial membrane lipid metabolism and composition.
Collapse
Affiliation(s)
- David J Mancuso
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
346
|
O’Neill BT, Kim J, Wende AR, Theobald HA, Tuinei J, Buchanan J, Guo A, Zaha VG, Davis DK, Schell JC, Boudina S, Wayment B, Litwin SE, Shioi T, Izumo S, Birnbaum MJ, Abel ED. A conserved role for phosphatidylinositol 3-kinase but not Akt signaling in mitochondrial adaptations that accompany physiological cardiac hypertrophy. Cell Metab 2007; 6:294-306. [PMID: 17908558 PMCID: PMC2084219 DOI: 10.1016/j.cmet.2007.09.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2006] [Revised: 03/16/2007] [Accepted: 09/11/2007] [Indexed: 11/27/2022]
Abstract
Physiological cardiac hypertrophy is associated with mitochondrial adaptations that are characterized by activation of PGC-1alpha and increased fatty acid oxidative (FAO) capacity. It is widely accepted that phosphatidylinositol 3-kinase (PI3K) signaling to Akt1 is required for physiological cardiac growth. However, the signaling pathways that coordinate physiological hypertrophy and metabolic remodeling are incompletely understood. We show here that activation of PI3K is sufficient to increase myocardial FAO capacity and that inhibition of PI3K signaling prevents mitochondrial adaptations in response to physiological hypertrophic stimuli despite increased expression of PGC-1alpha. We also show that activation of the downstream kinase Akt is not required for the mitochondrial adaptations that are secondary to PI3K activation. Thus, in physiological cardiac growth, PI3K is an integrator of cellular growth and metabolic remodeling. Although PI3K signaling to Akt1 is required for cellular growth, Akt-independent pathways mediate the accompanying mitochondrial adaptations.
Collapse
Affiliation(s)
- Brian T. O’Neill
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Jaetaek Kim
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Adam R. Wende
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Heather A. Theobald
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Joseph Tuinei
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Jonathan Buchanan
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Aili Guo
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Vlad G. Zaha
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Don K. Davis
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - John C. Schell
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | - Sihem Boudina
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| | | | | | - Tetsuo Shioi
- Department of Internal Medicine and Cardiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Seigo Izumo
- Novartis Institutes for Biomedical Research, Cambridge, MA
| | | | - E. Dale Abel
- Program in Human Molecular Biology and Genetics and Division of Endocrinology, Metabolism, and Diabetes; University of Utah, School of Medicine, Salt Lake City, UT 84112
| |
Collapse
|
347
|
Boudina S, Sena S, Theobald H, Sheng X, Wright JJ, Hu XX, Aziz S, Johnson JI, Bugger H, Zaha VG, Abel ED. Mitochondrial energetics in the heart in obesity-related diabetes: direct evidence for increased uncoupled respiration and activation of uncoupling proteins. Diabetes 2007; 56:2457-66. [PMID: 17623815 DOI: 10.2337/db07-0481] [Citation(s) in RCA: 451] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE In obesity and diabetes, myocardial fatty acid utilization and myocardial oxygen consumption (MVo(2)) are increased, and cardiac efficiency is reduced. Mitochondrial uncoupling has been proposed to contribute to these metabolic abnormalities but has not been directly demonstrated. RESEARCH DESIGN AND METHODS Oxygen consumption and cardiac function were determined in db/db hearts perfused with glucose or glucose and palmitate. Mitochondrial function was determined in saponin-permeabilized fibers and proton leak kinetics and H(2)O(2) generation determined in isolated mitochondria. RESULTS db/db hearts exhibited reduced cardiac function and increased MVo(2). Mitochondrial reactive oxygen species (ROS) generation and lipid and protein peroxidation products were increased. Mitochondrial proliferation was increased in db/db hearts, oxidative phosphorylation capacity was impaired, but H(2)O(2) production was increased. Mitochondria from db/db mice exhibited fatty acid-induced mitochondrial uncoupling that is inhibitable by GDP, suggesting that these changes are mediated by uncoupling proteins (UCPs). Mitochondrial uncoupling was not associated with an increase in UCP content, but fatty acid oxidation genes and expression of electron transfer flavoproteins were increased, whereas the content of the F1 alpha-subunit of ATP synthase was reduced. CONCLUSIONS These data demonstrate that mitochondrial uncoupling in the heart in obesity and diabetes is mediated by activation of UCPs independently of changes in expression levels. This likely occurs on the basis of increased delivery of reducing equivalents from beta-oxidation to the electron transport chain, which coupled with decreased oxidative phosphorylation capacity increases ROS production and lipid peroxidation.
Collapse
Affiliation(s)
- Sihem Boudina
- Division of Endocrinology, Metabolism, and Diabetes, Program in Human Molecular Biology and Genetics, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
348
|
Sebastiani M, Giordano C, Nediani C, Travaglini C, Borchi E, Zani M, Feccia M, Mancini M, Petrozza V, Cossarizza A, Gallo P, Taylor RW, d'Amati G. Induction of mitochondrial biogenesis is a maladaptive mechanism in mitochondrial cardiomyopathies. J Am Coll Cardiol 2007; 50:1362-9. [PMID: 17903636 DOI: 10.1016/j.jacc.2007.06.035] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 05/11/2007] [Accepted: 06/03/2007] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The purpose of this study was to clarify the molecular mechanisms linking human mitochondrial deoxyribonucleic acid (mtDNA) dysfunction to cardiac remodeling. BACKGROUND Defects of the mitochondrial genome cause a heterogeneous group of clinical disorders, including mitochondrial cardiomyopathies (MIC). The molecular events linking mtDNA defects to cardiac remodeling are unknown. Energy derangements and increase of mitochondrial-derived reactive oxygen species (ROS) could both play a role in the development of cardiac dysfunction in MIC. In addition, mitochondrial proliferation could interfere with sarcomere alignment and contraction. METHODS We performed a detailed morphologic and molecular analysis on failing hearts from 3 patients with MIC, failing human hearts due to ischemic heart disease (IHD) or dilated cardiomyopathies (DCM), and nonfailing hearts. RESULTS The MIC hearts showed marked mitochondrial proliferation with myofibril displacement. Consistent with morphologic features, increase in mtDNA content per cell and induction of genes involved in mitochondrial biogenesis, fatty acid metabolism, and glucose transport were observed. Down-regulation of these genes characterized DCM and IHD hearts. A pronounced increase in mitochondrial-derived ROS was observed in MIC hearts compared with failing hearts due to other causes. This was paralleled by up-regulation of genes encoding for uncoupling proteins and antioxidant enzymes. However, there was not a significant increase in antioxidant enzyme activity. CONCLUSIONS Our results suggest that besides energy deficiency, mitochondrial biogenesis per se is a maladaptive response in MIC and, possibly, in other metabolic cardiomyopathies.
Collapse
Affiliation(s)
- Mariangela Sebastiani
- Dipartimento di Medicina Sperimentale, Polo Pontino, Sapienza, Università di Roma, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
349
|
Abozguia K, Nallur Shivu G, Phan TT, Ahmed I, Maher AR, Frenneaux M. Potential of metabolic agents as adjunct therapies in heart failure. Future Cardiol 2007; 3:525-35. [DOI: 10.2217/14796678.3.5.525] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Heart failure continues to have a significant morbidity and mortality rate despite several recent advances in treatment such as additional neurohumoral blockades and cardiac resynchronisation therapy. There is emerging evidence that, irrespective of etiology, heart failure is associated with an energetic disorder and that this may contribute to the pathogenesis of the syndrome. Recently, a number of studies have suggested that some metabolic agents may have potential as adjunctive therapy in patients with heart failure. These agents cause a shift of myocardial-substrate utilization away from free fatty acids toward glucose. Free fatty acid utilization consumes more oxygen to generate an equivalent amount of energy compared with glucose. Some of these agents are also effective antianginals, presumably by reducing the myocardial oxygen requirement. In this review we will discuss some of the current issues and progresses relating to metabolic manipulation in heart failure.
Collapse
Affiliation(s)
- Khalid Abozguia
- BHF Research Fellow, University of Birmingham, Department of Cardiovascular Medicine, Medical School, Edgbaston, Birmingham B15 2TT, UK
| | - Ganesh Nallur Shivu
- BHF Research Fellow, University of Birmingham, Department of Cardiovascular Medicine, Birmingham B15 2TT, UK
| | - Thanh Trung Phan
- BHF Research Fellow, University of Birmingham, Department of Cardiovascular Medicine, Medical School, Edgbaston, Birmingham B15 2TT, UK
| | - Ibrar Ahmed
- BHF Research Fellow, University of Birmingham, Department of Cardiovascular Medicine, Medical School, Edgbaston, Birmingham B15 2TT, UK
| | - Abdul R Maher
- BHF Research Fellow, University of Birmingham, Department of Cardiovascular Medicine, Medical School, Edgbaston, Birmingham B15 2TT, UK
| | - Michael Frenneaux
- BHF Chair of Cardiology, University of Birmingham, Department of Cardiovascular Medicine, Medical School, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
350
|
Sano M, Tokudome S, Shimizu N, Yoshikawa N, Ogawa C, Shirakawa K, Endo J, Katayama T, Yuasa S, Ieda M, Makino S, Hattori F, Tanaka H, Fukuda K. Intramolecular control of protein stability, subnuclear compartmentalization, and coactivator function of peroxisome proliferator-activated receptor gamma coactivator 1alpha. J Biol Chem 2007; 282:25970-80. [PMID: 17620342 DOI: 10.1074/jbc.m703634200] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator (PGC)-1 is a critical transcriptional regulator of energy metabolism. Here we found that PGC-1alpha is a short lived and aggregation-prone protein. PGC-1alpha localized throughout the nucleoplasm and was rapidly destroyed via the ubiquitin-proteasome pathway. Upon proteasome inhibition, PGC-1alpha formed insoluble polyubiquitinated aggregates. Ubiquitination of PGC-1alpha depended on the integrity of the C terminus-containing arginine-serine-rich domains and an RNA recognition motif. Interestingly, ectopically expressed C-terminal fragment of PGC-1alpha was autonomously ubiquitinated and aggregated with promyelocytic leukemia protein. Cooperation of the N-terminal region containing two PEST-like motifs was required for prevention of aggregation and targeting of the polyubiquitinated PGC-1alpha for degradation. This region thereby negatively controlled the aggregation properties of the C-terminal region to regulate protein turnover and intranuclear compartmentalization of PGC-1alpha. Exogenous expression of the PGC-1alpha C-terminal fragment interfered with degradation of full-length PGC-1alpha and enhanced its coactivation properties. We concluded that PGC-1alpha function is critically regulated at multiple steps via intramolecular cooperation among several distinct structural domains of the protein.
Collapse
Affiliation(s)
- Motoaki Sano
- Department of Regenerative Medicine and Advanced Cardiac Therapeutics, Keio University School of Medicine, and Department of Rheumatology and Allergy, Research Hospital, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|