Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 12(9): 824, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34471096

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is a master regulator of mitochondrial metabolism but its precise mechanism of action yet remains unclear. Here, we found that a dietary saturated fatty acid (FA), palmitate increased intracellular cAMP synthesis through the palmitoylation of soluble adenylyl cyclase in cardiomyocytes. cAMP further induced exchange protein directly activated by cyclic AMP 1 (Epac1) activation, which was upregulated in the myocardium of obese patients. Epac1 enhanced the activity of a key enzyme regulating mitochondrial FA uptake, carnitine palmitoyltransferase 1. Consistently, pharmacological or genetic Epac1 inhibition prevented lipid overload, increased FA oxidation (FAO), and protected against mitochondrial dysfunction in cardiomyocytes. In addition, analysis of Epac1 phosphoproteome led us to identify two key mitochondrial enzymes of the the ß-oxidation cycle as targets of Epac1, the long-chain FA acyl-CoA dehydrogenase (ACADL) and the 3-ketoacyl-CoA thiolase (3-KAT). Epac1 formed molecular complexes with the Ca2+/calmodulin-dependent protein kinase II (CaMKII), which phosphorylated ACADL and 3-KAT at specific amino acid residues to decrease lipid oxidation. The Epac1-CaMKII axis also interacted with the α subunit of ATP synthase, thereby further impairing mitochondrial energetics. Altogether, these findings indicate that Epac1 disrupts the balance between mitochondrial FA uptake and oxidation leading to lipid accumulation and mitochondrial dysfunction, and ultimately cardiomyocyte death.


Subject(s)
Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Palmitic Acid/toxicity , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Catecholamines/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Lipoylation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Phosphoproteins/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Solubility , Stress, Physiological/drug effects
2.
Biochimie ; 178: 69-80, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32835733

ABSTRACT

Recent works reported the relevance of cellular exosomes in the evolution of different pathologies. However, most of these studies focused on the ability of exosomes to convey mi-RNA from cell to cell. The level of knowledge concerning the transport of lipid mediators by these nanovesicles is more than fragmented. The role of lipid mediators in the inflammatory signaling is fairly well described, in particular concerning the derivatives of the arachidonic acid (AA), called eicosanoïds or lipid mediators. The aim of the present work was to study the transport of these lipids within the extracellular vesicles of rat bone marrow mesenchymal stem cells (BM-MSC) and the cardiomyoblast cell line H9c2. We were able to characterize, for the first time, complete profiles of oxilipins within these nanovesicles. We studied also the impact on these profiles, of the polyunsaturated fatty acids (PUFAs) know to be precursors of the inflammatory signaling molecules (AA, eicosapentaenoic acid EPA and Docosahexaenoic acid DHA), at physiological concentrations. By growing the progenitor cells under PUFAs supplementation, we provide a comprehensive assessment of the beneficial effect of ω-3 PUFA therapy. Actually, our results tend to support the resolving role of the inflammation that stromal cell-derived extracellular vesicles can have within the cardiac microenvironment.


Subject(s)
Eicosanoids/chemistry , Eicosanoids/metabolism , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism , Myoblasts, Cardiac/chemistry , Myoblasts, Cardiac/metabolism , Animals , Bone Marrow/chemistry , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Line , Extracellular Vesicles/drug effects , Humans , Inflammation/metabolism , Inflammation Mediators/chemistry , Inflammation Mediators/metabolism , Lipid Metabolism , Mesenchymal Stem Cells/drug effects , Myoblasts, Cardiac/drug effects , Oxylipins/chemistry , Oxylipins/metabolism , Rats
3.
Cell Death Differ ; 27(6): 1907-1923, 2020 06.
Article in English | MEDLINE | ID: mdl-31819159

ABSTRACT

Chronic remodeling postmyocardial infarction consists in various maladaptive changes including interstitial fibrosis, cardiomyocyte death and mitochondrial dysfunction that lead to heart failure (HF). Reactive aldehydes such as 4-hydroxynonenal (4-HNE) are critical mediators of mitochondrial dysfunction but the sources of mitochondrial 4-HNE in cardiac diseases together with its mechanisms of action remain poorly understood. Here, we evaluated whether the mitochondrial enzyme monoamine oxidase-A (MAO-A), which generates H2O2 as a by-product of catecholamine metabolism, is a source of deleterious 4-HNE in HF. We found that MAO-A activation increased mitochondrial ROS and promoted local 4-HNE production inside the mitochondria through cardiolipin peroxidation in primary cardiomyocytes. Deleterious effects of MAO-A/4-HNE on cardiac dysfunction were prevented by activation of mitochondrial aldehyde dehydrogenase 2 (ALDH2), the main enzyme for 4-HNE metabolism. Mechanistically, MAO-A-derived 4-HNE bound to newly identified targets VDAC and MCU to promote ER-mitochondria contact sites and MCU higher-order complex formation. The resulting mitochondrial Ca2+ accumulation participated in mitochondrial respiratory dysfunction and loss of membrane potential, as shown with the protective effects of the MCU inhibitor, RU360. Most interestingly, these findings were recapitulated in a chronic model of ischemic remodeling where pharmacological or genetic inhibition of MAO-A protected the mice from 4-HNE accumulation, MCU oligomer formation and Ca2+ overload, thus mitigating ventricular dysfunction. To our knowledge, these are the first evidences linking MAO-A activation to mitoCa2+ mishandling through local 4-HNE production, contributing to energetic failure and postischemic remodeling.


Subject(s)
Aldehydes/metabolism , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Monoamine Oxidase/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac , Animals , Calcium/metabolism , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Ventricular Remodeling
4.
Cardiovasc Res ; 114(1): 123-137, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29136112

ABSTRACT

Aims: Tenascin-C (TNC) is an endogenous danger signal molecule strongly associated with inflammatory diseases and with poor outcome in patients with cardiomyopathies. Its function within pathological cardiac tissue during pressure overload remains poorly understood. Methods and results: We showed that TNC accumulates after 1 week of transverse aortic constriction (TAC) in the heart of 12-week-old male mice. By cross bone marrow transplantation experiments, we determined that TNC deposition relied on cardiac cells and not on haematopoietic cells. The expression of TNC induced by TAC, or by administration of a recombinant lentivector coding for TNC, triggered a pro-inflammatory cardiac microenvironment, monocyte/macrophage (MO/MΦ) accumulation, and systolic dysfunction. TNC modified macrophage polarization towards the pro-inflammatory phenotype and stimulated RhoA/Rho-associated protein kinase (ROCK) pathways to promote mesenchymal to amoeboid transition that enhanced macrophage migration into fibrillar collagen matrices. The amplification of inflammation and MO/MΦ recruitment by TNC was abrogated by genetic invalidation of TNC in knockout mice. These mice showed less ventricular remodelling and an improved cardiac function after TAC as compared with wild-type mice. Conclusions: By promoting a pro-inflammatory microenvironment and macrophage migration, TNC appears to be a key factor to enable the MO/MΦ accumulation within fibrotic hearts leading to cardiac dysfunction. As TNC is highly expressed during inflammation and sparsely during the steady state, its inhibition could be a promising therapeutic strategy to control inflammation and immune cell infiltration in heart disease.


Subject(s)
Cell Movement , Hypertrophy, Left Ventricular/metabolism , Macrophages/metabolism , Monocytes/metabolism , Myocardium/metabolism , Tenascin/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Cellular Microenvironment , Chemokines/metabolism , Disease Models, Animal , Fibrosis , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Phenotype , Signal Transduction , Tenascin/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
5.
Circ Res ; 120(4): 645-657, 2017 Feb 17.
Article in English | MEDLINE | ID: mdl-28096195

ABSTRACT

RATIONALE: Although the second messenger cyclic AMP (cAMP) is physiologically beneficial in the heart, it largely contributes to cardiac disease progression when dysregulated. Current evidence suggests that cAMP is produced within mitochondria. However, mitochondrial cAMP signaling and its involvement in cardiac pathophysiology are far from being understood. OBJECTIVE: To investigate the role of MitEpac1 (mitochondrial exchange protein directly activated by cAMP 1) in ischemia/reperfusion injury. METHODS AND RESULTS: We show that Epac1 (exchange protein directly activated by cAMP 1) genetic ablation (Epac1-/-) protects against experimental myocardial ischemia/reperfusion injury with reduced infarct size and cardiomyocyte apoptosis. As observed in vivo, Epac1 inhibition prevents hypoxia/reoxygenation-induced adult cardiomyocyte apoptosis. Interestingly, a deleted form of Epac1 in its mitochondrial-targeting sequence protects against hypoxia/reoxygenation-induced cell death. Mechanistically, Epac1 favors Ca2+ exchange between the endoplasmic reticulum and the mitochondrion, by increasing interaction with a macromolecular complex composed of the VDAC1 (voltage-dependent anion channel 1), the GRP75 (chaperone glucose-regulated protein 75), and the IP3R1 (inositol-1,4,5-triphosphate receptor 1), leading to mitochondrial Ca2+ overload and opening of the mitochondrial permeability transition pore. In addition, our findings demonstrate that MitEpac1 inhibits isocitrate dehydrogenase 2 via the mitochondrial recruitment of CaMKII (Ca2+/calmodulin-dependent protein kinase II), which decreases nicotinamide adenine dinucleotide phosphate hydrogen synthesis, thereby, reducing the antioxidant capabilities of the cardiomyocyte. CONCLUSIONS: Our results reveal the existence, within mitochondria, of different cAMP-Epac1 microdomains that control myocardial cell death. In addition, our findings suggest Epac1 as a promising target for the treatment of ischemia-induced myocardial damage.


Subject(s)
Guanine Nucleotide Exchange Factors/biosynthesis , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cell Death/physiology , Cells, Cultured , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Membrane Microdomains/metabolism , Membrane Microdomains/pathology , Mice , Mice, Knockout , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Rats
6.
Med Sci (Paris) ; 31(10): 881-8, 2015 Oct.
Article in French | MEDLINE | ID: mdl-26481027

ABSTRACT

Physical exercise or hypertension requires that the heart increases its hemodynamic work. However, this adaptation is based on distinct cardiac remodelling according to the physiological or pathological origin of the stress. As shown here with two examples, understanding the molecular events leading to cardiac remodeling may offer new opportunities for the development of therapies for heart failure. The recently described Epac1 protein is an effector of the second messenger cAMP. Following a pathological stress, the cAMP-binding protein Epac1 induces cardiac hypertrophy and fibrosis as well as alteration of calcium cycling suggesting that Epac1 pharmacological inhibition may be of therapeutic value. Furthermore, the protein carabin is an important regulator of several effectors of pathological cardiac remodelling. Experimental manipulation of carabin expression profoundly alters the development of heart failure.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Guanine Nucleotide Exchange Factors/physiology , Heart Failure/genetics , Ventricular Remodeling/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , GTPase-Activating Proteins , Guanine Nucleotide Exchange Factors/genetics , Heart Failure/pathology , Humans , Signal Transduction/genetics
7.
Am J Hematol ; 90(1): 15-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25242620

ABSTRACT

Peripheral serotonin (5-HT) has been involved in adverse cardiac remodeling and valve fibrosis. The peripheral levels of 5-HT mainly depend on its release from activated platelets and degradation by monoamine oxidase A (MAO-A). The SERAOPI study investigated the relationship between arterial serotoninergic system, degree of platelet activation and cardiac remodeling, in patients with aortic valve stenosis (AS). Thirty patients with severe AS and 15 control subjects underwent transthoracic echocardiography, radial, and aortic arterial blood sampling. Measurements of 5-HT and its MAO-A-dependent degradation product, 5-HIAA, were performed by HPLC. Arterial platelet activation was assessed by flow cytometry analysis of platelet surface expression of P-selectin and activated integrin GPIIb/IIIa. Activated platelets and arterial plasma 5-HT increased in AS patients as compared to control subjects (P-selectin 1.08 ± 0.2MFI vs. 0.49 ± 0.1MFI, P = 0.04; GPIIb/IIIa 0.71 ± 0.1MFI vs. 0.35 ± 0.1MFI; P = 0.0015 and arterial plasma 5-HT 11.55 ± 1.6 nM vs. 6.18 ± 0.7 nM, P = 0.028, respectively). Moreover, 5-HT was strongly correlated to left ventricular hypertrophy assessed by echocardiography. The correlation was independent of cardiovascular risk comorbidities and others echocardiographic AS parameters. Finally, plasma 5-HIAA increased in AS patients (74.64 ± 9.7 nM vs. 37.16 ± 4.1 nM; P = 0.0002) indicating a higher 5-HT degradation rate by MAO-A. Platelet activation, arterial circulating serotonin, and serotonin degradation increased in patients with AS. These observations suggest that the serotoninergic system may contribute to the pathogenesis of AS including valve fibrosis and adverse ventricular remodeling.


Subject(s)
Aortic Valve Stenosis/blood , Blood Platelets/metabolism , Platelet Activation , Serotonin/blood , Ventricular Remodeling , Aortic Valve Stenosis/pathology , Case-Control Studies , Chromatography, High Pressure Liquid , Echocardiography , Echocardiography, Doppler , Flow Cytometry , Humans , Hydroxyindoleacetic Acid/blood , Middle Aged , Radial Artery , von Willebrand Factor/analysis
8.
Circulation ; 131(4): 390-400; discussion 400, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25369805

ABSTRACT

BACKGROUND: Cardiac hypertrophy is an early hallmark during the clinical course of heart failure and is regulated by various signaling pathways. However, the molecular mechanisms that negatively regulate these signal transduction pathways remain poorly understood. METHODS AND RESULTS: Here, we characterized Carabin, a protein expressed in cardiomyocytes that was downregulated in cardiac hypertrophy and human heart failure. Four weeks after transverse aortic constriction, Carabin-deficient (Carabin(-/-)) mice developed exaggerated cardiac hypertrophy and displayed a strong decrease in fractional shortening (14.6±1.6% versus 27.6±1.4% in wild type plus transverse aortic constriction mice; P<0.0001). Conversely, compensation of Carabin loss through a cardiotropic adeno-associated viral vector encoding Carabin prevented transverse aortic constriction-induced cardiac hypertrophy with preserved fractional shortening (39.9±1.2% versus 25.9±2.6% in control plus transverse aortic constriction mice; P<0.0001). Carabin also conferred protection against adrenergic receptor-induced hypertrophy in isolated cardiomyocytes. Mechanistically, Carabin carries out a tripartite suppressive function. Indeed, Carabin, through its calcineurin-interacting site and Ras/Rab GTPase-activating protein domain, functions as an endogenous inhibitor of calcineurin and Ras/extracellular signal-regulated kinase prohypertrophic signaling. Moreover, Carabin reduced Ca(2+)/calmodulin-dependent protein kinase II activation and prevented nuclear export of histone deacetylase 4 after adrenergic stimulation or myocardial pressure overload. Finally, we showed that Carabin Ras-GTPase-activating protein domain and calcineurin-interacting domain were both involved in the antihypertrophic action of Carabin. CONCLUSIONS: Our study identifies Carabin as a negative regulator of key prohypertrophic signaling molecules, calcineurin, Ras, and Ca(2+)/calmodulin-dependent protein kinase II and implicates Carabin in the development of cardiac hypertrophy and failure.


Subject(s)
Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , GTPase-Activating Proteins/biosynthesis , Genes, ras/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Rats , Signal Transduction/physiology
9.
Cardiovasc Res ; 105(1): 55-64, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25411381

ABSTRACT

AIMS: Stimulation of ß-adrenergic receptors (ß-AR) increases cAMP production and contributes to the pathogenesis of cardiac hypertrophy and failure through poorly understood mechanisms. We previously demonstrated that Exchange protein directly activated by cAMP 1 (Epac1)-induced hypertrophy in primary cardiomyocytes. Among the mechanisms triggered by cardiac stress, autophagy has been highlighted as a protective or harmful response. Here, we investigate whether Epac1 promotes cardiac autophagy and how altered autophagy has an impact on Epac1-induced cardiomyocyte hypertrophy. METHODS AND RESULTS: We reported that direct stimulation of Epac1 with the agonist, Sp-8-(4-chlorophenylthio)-2'-O-methyl-cAMP (Sp-8-pCPT) promoted autophagy activation in neonatal cardiomyocytes. Stimulation of ß-AR with isoprenaline (ISO) mimicked the effect of Epac1 on autophagy markers. Conversely, the induction of autophagy flux following ISO treatment was prevented in cardiomyocytes pre-treated with a selective inhibitor of Epac1, CE3F4. Importantly, we found that Epac1 deletion in mice protected against ß-AR-induced cardiac remodelling and prevented the induction of autophagy. The signalling mechanisms underlying Epac1-induced autophagy involved a Ca(2+)/calmodulin-dependent kinase kinase ß (CaMKKß)/AMP-dependent protein kinase (AMPK) pathway. Finally, we provided evidence that pharmacological inhibition of autophagy using 3-methyladenine (3-MA) or down-regulation of autophagy-related protein 5 (Atg5) significantly potentiated Epac1-promoted cardiomyocyte hypertrophy. CONCLUSION: Altogether, these findings demonstrate that autophagy is an adaptive response to antagonize Epac1-promoted cardiomyocyte hypertrophy.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Autophagy/drug effects , Autophagy/physiology , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Enlargement , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Female , Guanine Nucleotide Exchange Factors/agonists , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Quinolines/pharmacology , Rats , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Thionucleotides/pharmacology
10.
Circulation ; 129(21): 2111-24, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24657994

ABSTRACT

BACKGROUND: The mechanisms by which the heart adapts to chronic pressure overload, producing compensated hypertrophy and eventually heart failure (HF), are still not well defined. We aimed to investigate the involvement of T cells in the progression to HF using a transverse aortic constriction (TAC) model. METHODS AND RESULTS: Chronic HF was associated with accumulation of T lymphocytes and activated/effector CD4(+) T cells within cardiac tissue. After TAC, enlarged heart mediastinal draining lymph nodes showed a high density of both CD4(+) and CD8(+) T-cell subsets. To investigate the role of T cells in HF, TAC was performed on mice deficient for recombination activating gene 2 expression (RAG2KO) lacking B and T lymphocytes. Compared with wild-type TAC mice, RAG2KO mice did not develop cardiac dilation and showed improved contractile function and blunted adverse remodeling. Reconstitution of the T-cell compartment into RAG2KO mice before TAC enhanced contractile dysfunction, fibrosis, collagen accumulation, and cross-linking. To determine the involvement of a specific T-cell subset, we performed TAC on mice lacking CD4(+) (MHCIIKO) and CD8(+) T-cell subsets (CD8KO). In contrast to CD8KO mice, MHCIIKO mice did not develop ventricular dilation and dysfunction. MHCIIKO mice also displayed very low fibrosis, collagen accumulation, and cross-linking within cardiac tissue. Interestingly, mice with transgenic CD4(+) T-cell receptor specific for ovalbumin failed to develop HF and adverse remodeling. CONCLUSIONS: These results demonstrate for the first time a crucial role of CD4(+) T cells and specific antigen recognition in the progression from compensated cardiac hypertrophy to HF.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Cardiomegaly/pathology , Disease Progression , Heart Failure/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , Cardiomegaly/immunology , Heart Failure/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
11.
Antioxid Redox Signal ; 18(1): 5-18, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22738191

ABSTRACT

AIMS: Oxidative stress and mitochondrial dysfunction participate together in the development of heart failure (HF). mRNA levels of monoamine oxidase-A (MAO-A), a mitochondrial enzyme that produces hydrogen peroxide (H(2)O(2)), increase in several models of cardiomyopathies. Therefore, we hypothesized that an increase in cardiac MAO-A could cause oxidative stress and mitochondrial damage, leading to cardiac dysfunction. In the present study, we evaluated the consequences of cardiac MAO-A augmentation on chronic oxidative damage, cardiomyocyte survival, and heart function, and identified the intracellular pathways involved. RESULTS: We generated transgenic (Tg) mice with cardiac-specific MAO-A overexpression. Tg mice displayed cardiac MAO-A activity levels similar to those found in HF and aging. As expected, Tg mice showed a significant decrease in the cardiac amounts of the MAO-A substrates serotonin and norepinephrine. This was associated with enhanced H(2)O(2) generation in situ and mitochondrial DNA oxidation. As a consequence, MAO-A Tg mice demonstrated progressive loss of cardiomyocytes by necrosis and ventricular failure, which were prevented by chronic treatment with the MAO-A inhibitor clorgyline and the antioxidant N-acetyl-cystein. Interestingly, Tg hearts exhibited p53 accumulation and downregulation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a master regulator of mitochondrial function. This was concomitant with cardiac mitochondrial ultrastructural defects and ATP depletion. In vitro, MAO-A adenovirus transduction of neonatal cardiomyocytes mimicked the results in MAO-A Tg mice, triggering oxidative stress-dependent p53 activation, leading to PGC-1α downregulation, mitochondrial impairment, and cardiomyocyte necrosis. INNOVATION AND CONCLUSION: We provide the first evidence that MAO-A upregulation in the heart causes oxidative mitochondrial damage, p53-dependent repression of PGC-1α, cardiomyocyte necrosis, and chronic ventricular dysfunction.


Subject(s)
Mitochondria, Heart/enzymology , Monoamine Oxidase/metabolism , Myocytes, Cardiac/pathology , Necrosis/enzymology , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Ventricular Dysfunction, Left/enzymology , Animals , Cardiomyopathy, Dilated/enzymology , Cells, Cultured , Chronic Disease , Enzyme Induction , Fibrosis , Heart Ventricles/enzymology , Heart Ventricles/pathology , Hypertrophy, Left Ventricular/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monoamine Oxidase/genetics , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Sprague-Dawley , Transcription Factors , Up-Regulation , Ventricular Dysfunction, Left/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...