Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Int J Hematol ; 111(6): 803-811, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32020505

ABSTRACT

The prognosis of cardiac light-chain (AL) amyloidosis is considered to be very poor. We studied the treatment efficacy and outcomes by retrospectively analyzing the clinical results of 45 patients with cardiac AL amyloidosis treated at our hospital between September 2008 and March 2016. The group of patients analyzed included 29 males and 16 females with a median age of 68 years. Their baseline median NT-proBNP, cTnT, and dFLC were 3167 pg/ml, 0.080 ng/ml, and 286.17 mg/l, respectively. Twenty-eight patients were in Cardiac Stage (CS) III and 17 patients were in Revised Prognostic Stage (RPS) IV. At the median follow-up of 10 months, the median overall survival (OS) was 16 months and 3-year OS was 35.9%. The patients in CS III showed significantly poorer survival rate than those in CS I or II (3-year OS: 12.2% vs. 65.8%, p = 0.0115) and the patients in RPS IV showed significantly poorer survival rate than those in RPS I, II, or III (3-year OS: 11.0% vs. 53.3%, p = 0.000914). Regardless of the therapeutic approaches, patients who achieved hematological CR or cardiac organ response demonstrated significantly improved prognosis. Therefore, achievement of hematological and organ responses is important in the treatment of cardiac AL amyloidosis.


Subject(s)
Bortezomib/administration & dosage , Cardiomyopathies/therapy , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Immunoglobulin Light-chain Amyloidosis/therapy , Melphalan/administration & dosage , Peripheral Blood Stem Cell Transplantation , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cardiomyopathies/diagnosis , Cardiomyopathies/mortality , Drug Therapy, Combination , Female , Humans , Immunoglobulin Light-chain Amyloidosis/diagnosis , Immunoglobulin Light-chain Amyloidosis/mortality , Japan , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Prognosis , Retrospective Studies , Retroviridae Proteins, Oncogenic , Severity of Illness Index
2.
Am J Physiol Heart Circ Physiol ; 316(6): H1366-H1377, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30901275

ABSTRACT

Despite the advances in medical therapy, the morbidity and mortality of heart failure (HF) remain unacceptably high. HF results from reduced metabolism-contraction coupling efficiency, so the modulation of cardiac metabolism may be an effective strategy for therapeutic interventions. Tumor suppressor p53 (TP53) and its downstream target TP53-induced glycolysis and apoptosis regulator (TIGAR) are known to modulate cardiac metabolism and cell fate. To investigate TIGAR's function in HF, we compared myocardial, metabolic, and functional outcomes between TIGAR knockout (TIGAR-/-) mice and wild-type (TIGAR+/+) mice subjected to chronic thoracic transverse aortic constriction (TAC), a pressure-overload HF model. In wild-type mice hearts, p53 and TIGAR increased markedly during HF development. Eight weeks after TAC surgery, the left ventricular (LV) dysfunction, fibrosis, oxidative damage, and myocyte apoptosis were significantly advanced in wild-type than in TIGAR-/- mouse heart. Further, myocardial high-energy phosphates in wild-type hearts were significantly decreased compared with those of TIGAR-/- mouse heart. Glucose oxidation and glycolysis rates were also reduced in isolated perfused wild-type hearts following TAC than those in TIGAR-/- hearts, which suggest that the upregulation of TIGAR in HF causes impaired myocardial energetics and function. The effects of TIGAR knockout on LV function were also replicated in tamoxifen (TAM)-inducible cardiac-specific TIGAR knockout mice (TIGARflox/flox/Tg(Myh6-cre/Esr1) mice). The ablation of TIGAR during pressure-overload HF preserves myocardial function and energetics. Thus, cardiac TIGAR-targeted therapy to increase glucose metabolism will be a novel strategy for HF. NEW & NOTEWORTHY The present study is the first to demonstrate that TP53-induced glycolysis and apoptosis regulator (TIGAR) is upregulated in the myocardium during experimental heart failure (HF) in mice and that TIGAR knockout can preserve the heart function and myocardial energetics during HF. Reducing TIGAR to enhance myocardial glycolytic energy production is a promising therapeutic strategy for HF.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Energy Metabolism , Heart Failure/metabolism , Myocardium/metabolism , Myocardium/pathology , Phosphoric Monoester Hydrolases/deficiency , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Autophagy , Disease Models, Animal , Fibrosis , Glycolysis , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Phosphoric Monoester Hydrolases/genetics , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling
3.
Circ Heart Fail ; 10(12)2017 Dec.
Article in English | MEDLINE | ID: mdl-29242353

ABSTRACT

BACKGROUND: Energy starvation and the shift of energy substrate from fatty acids to glucose is the hallmark of metabolic remodeling during heart failure progression. However, ketone body metabolism in the failing heart has not been fully investigated. METHODS AND RESULTS: Microarray data analysis and mitochondrial isobaric tags for relative and absolute quantification proteomics revealed that the expression of D-ß-hydroxybutyrate dehydrogenase I (Bdh1), an enzyme that catalyzes the NAD+/NADH coupled interconversion of acetoacetate and ß-hydroxybutyrate, was increased 2.5- and 2.8-fold, respectively, in the heart after transverse aortic constriction. In addition, ketone body oxidation was upregulated 2.2-fold in transverse aortic constriction hearts, as determined by the amount of 14CO2 released from the metabolism of [1-14C] ß-hydroxybutyrate in isolated perfused hearts. To investigate the significance of this augmented ketone body oxidation, we generated heart-specific Bdh1-overexpressing transgenic mice to recapitulate the observed increase in basal ketone body oxidation. Bdh1 transgenic mice showed a 1.7-fold increase in ketone body oxidation but did not exhibit any differences in other baseline characteristics. When subjected to transverse aortic constriction, Bdh1 transgenic mice were resistant to fibrosis, contractile dysfunction, and oxidative damage, as determined by the immunochemical detection of carbonylated proteins and histone acetylation. Upregulation of Bdh1 enhanced antioxidant enzyme expression. In our in vitro study, flow cytometry revealed that rotenone-induced reactive oxygen species production was decreased by adenovirus-mediated Bdh1 overexpression. Furthermore, hydrogen peroxide-induced apoptosis was attenuated by Bdh1 overexpression. CONCLUSIONS: We demonstrated that ketone body oxidation increased in failing hearts, and increased ketone body utilization decreased oxidative stress and protected against heart failure.


Subject(s)
Gene Expression Regulation , Heart Failure/genetics , Hydroxybutyrate Dehydrogenase/genetics , Mitochondria, Heart/genetics , Oxidative Stress , Ventricular Pressure/physiology , Ventricular Remodeling/genetics , Animals , Disease Models, Animal , Genotype , Heart Failure/enzymology , Heart Failure/physiopathology , Hydroxybutyrate Dehydrogenase/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/metabolism , Polymerase Chain Reaction
4.
Sci Rep ; 7: 43911, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266638

ABSTRACT

D-Amino acids are enantiomers of L-amino acids and have recently been recognized as biomarkers and bioactive substances in mammals, including humans. In the present study, we investigated functions of the novel mammalian mitochondrial protein 9030617O03Rik and showed decreased expression under conditions of heart failure. Genomic sequence analyses showed partial homology with a bacterial aspartate/glutamate/hydantoin racemase. Subsequent determinations of all free amino acid concentrations in 9030617O03Rik-deficient mice showed high accumulations of D-glutamate in heart tissues. This is the first time that a significant amount of D-glutamate was detected in mammalian tissue. Further analysis of D-glutamate metabolism indicated that 9030617O03Rik is a D-glutamate cyclase that converts D-glutamate to 5-oxo-D-proline. Hence, this protein is the first identified enzyme responsible for mammalian D-glutamate metabolism, as confirmed in cloning analyses. These findings suggest that D-glutamate and 5-oxo-D-proline have bioactivities in mammals through the metabolism by D-glutamate cyclase.


Subject(s)
Glutamic Acid/metabolism , Hydro-Lyases/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Animals , Hydro-Lyases/genetics , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Pyrrolidonecarboxylic Acid/metabolism
5.
Am J Physiol Heart Circ Physiol ; 312(2): H305-H313, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28011586

ABSTRACT

Failing heart loses its metabolic flexibility, relying increasingly on glucose as its preferential substrate and decreasing fatty acid oxidation (FAO). Peroxisome proliferator-activated receptor α (PPAR-α) is a key regulator of this substrate shift. However, its role during heart failure is complex and remains unclear. Recent studies reported that heart failure develops in the heart of myosin heavy chain-PPAR-α transgenic mice in a manner similar to that of diabetic cardiomyopathy, whereas cardiac dysfunction is enhanced in PPAR-α knockout mice in response to chronic pressure overload. We created a pressure-overload heart failure model in mice through transverse aortic constriction (TAC) and activated PPAR-α during heart failure using an inducible transgenic model. After 8 wk of TAC, left ventricular (LV) function had decreased with the reduction of PPAR-α expression in wild-type mice. We examined the effect of PPAR-α induction during heart failure using the Tet-Off system. Eight weeks after the TAC operation, LV construction was preserved significantly by PPAR-α induction with an increase in PPAR-α-targeted genes related to fatty acid metabolism. The increase of expression of fibrosis-related genes was significantly attenuated by PPAR-α induction. Metabolic rates measured by isolated heart perfusions showed a reduction in FAO and glucose oxidation in TAC hearts, but the rate of FAO preserved significantly owing to the induction of PPAR-α. Myocardial high-energy phosphates were significantly preserved by PPAR-α induction. These results suggest that PPAR-α activation during pressure-overloaded heart failure improved myocardial function and energetics. Thus activating PPAR-α and modulation of FAO could be a promising therapeutic strategy for heart failure.NEW & NOTEWORTHY The present study demonstrates the role of PPAR-α activation in the early stage of heart failure using an inducible transgenic mouse model. Induction of PPAR-α preserved heart function, and myocardial energetics. Activating PPAR-α and modulation of fatty acid oxidation could be a promising therapeutic strategy for heart failure.


Subject(s)
Energy Metabolism/genetics , Heart Failure/genetics , Myocardial Contraction/genetics , Myocardium/metabolism , PPAR alpha/genetics , Ventricular Dysfunction, Left/genetics , Animals , Aorta/surgery , Blotting, Western , Disease Models, Animal , Echocardiography , Energy Metabolism/drug effects , Fatty Acids/metabolism , Glucose/metabolism , Heart Failure/metabolism , Heart Failure/physiopathology , Male , Mice , Mice, Transgenic , Myocardial Contraction/drug effects , Oxidation-Reduction , PPAR alpha/agonists , Phosphates/metabolism , Pyrimidines/pharmacology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
6.
Am J Physiol Heart Circ Physiol ; 308(8): H951-9, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25659487

ABSTRACT

Pulmonary arterial hypertension (PAH) is a refractory disease characterized by uncontrolled vascular remodeling and elevated pulmonary arterial pressure. Although synthetic inhibitors of some tyrosine kinases have been used to treat PAH, their therapeutic efficacies and safeties remain controversial. Thus, the establishment of novel therapeutic targets based on the molecular pathogenesis underlying PAH is a clinically urgent issue. In the present study, we demonstrated that proline-rich tyrosine kinase 2 (Pyk2), a nonreceptor type protein tyrosine kinase, plays a crucial role in the pathogenesis of pulmonary hypertension (PH) using an animal model of hypoxia-induced PH. Resistance to hypoxia-induced PH was markedly higher in Pyk2-deficient mice than in wild-type mice. Pathological investigations revealed that medial thickening of the pulmonary arterioles, which is a characteristic of hypoxia-induced PH, was absent in Pyk2-deficient mice, suggesting that Pyk2 is involved in the hypoxia-induced aberrant proliferation of vascular smooth muscle cells in hypoxia-induced PH. In vitro experiments using human pulmonary smooth muscle cells showed that hypoxic stress increased the proliferation and migration of cells in a Pyk2-dependent manner. We also demonstrated that Pyk2 plays a crucial role in ROS generation during hypoxic stress and that this Pyk2-dependent generation of ROS is necessary for the activation of hypoxia-inducible factor-1α, a key molecule in the pathogenesis of hypoxia-induced PH. In summary, the results of the present study reveal that Pyk2 plays an important role in the pathogenesis of hypoxia-induced PH. Therefore, Pyk2 may represent a promising therapeutic target for PAH in a clinical setting.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Arterioles/cytology , Arterioles/metabolism , Arterioles/physiology , Cell Hypoxia , Cell Movement , Cell Proliferation , Cells, Cultured , Focal Adhesion Kinase 2/genetics , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung/blood supply , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Reactive Oxygen Species/metabolism
7.
Biochem Biophys Res Commun ; 456(1): 250-6, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25446079

ABSTRACT

Alpha-adrenergic agonists is known to be protective in cardiac myocytes from apoptosis induced by beta-adrenergic stimulation. Although there has been a recent focus on the role of cardiac autophagy in heart failure, its role in heart failure with adrenergic overload has not yet been elucidated. In the present study, we investigated the contribution of autophagy to cardiac failure during adrenergic overload both in vitro and in vivo. Neonatal rat cardiac myocytes overexpressing GFP-tagged LC3 were prepared and stimulated with the alpha1-adrenergic agonist, phenylephrine (PE), the beta-adrenergic agonist, isoproterenol (ISO), or norepinephrine (NE) in order to track changes in the formation of autophagosomes in vitro. All adrenergic stimulators increased cardiac autophagy by stimulating autophagic flux. Blocking autophagy by the knockdown of autophagy-related 5 (ATG5) exacerbated ISO-induced apoptosis and negated the anti-apoptotic effects of PE, which indicated the cardioprotective role of autophagy during adrenergic overload. PE-induced cardiac autophagy was mediated by the PI3-kinase/Akt pathway, but not by MEK/ERK, whereas both pathways mediated the anti-apoptotic effects of PE. Knock down of Akt1 was the most essential among the three Akt family members examined for the induction of cardiac autophagy. The four-week administration of PE kept the high level of cardiac autophagy without heart failure in vivo, whereas autophagy levels in a myocardium impaired by four-week persistent administration of ISO or NE were the same with the control state. These present study indicated that cardiac autophagy played a protective role during adrenergic overload and also that the Akt pathway could mediate cardiac autophagy for the anti-apoptotic effects of the alpha-adrenergic pathway.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Autophagy , Heart Failure/metabolism , Heart/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Animals , Animals, Newborn , Apoptosis , Cardiotonic Agents/pharmacology , Green Fluorescent Proteins/metabolism , Heart Failure/prevention & control , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Norepinephrine/pharmacology , Phenylephrine/pharmacology , Rats , TOR Serine-Threonine Kinases/metabolism
8.
Circ Heart Fail ; 7(3): 500-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24740269

ABSTRACT

BACKGROUND: Mitochondrial compromise is a fundamental contributor to heart failure. Recent studies have revealed that several surveillance systems maintain mitochondrial integrity. The present study evaluated the role of mitochondrial AAA+ protease in a mouse model of pressure overload heart failure. METHODS AND RESULTS: The fluorescein isothiocyanate casein assay and immunoblotting for endogenous mitochondrial proteins revealed a marked reduction in ATP-dependent proteolytic activity in failing heart mitochondria. The level of reduced cysteine was decreased, and tyrosine nitration and protein carbonylation were promoted in Lon protease homolog (LONP1), the most abundant mitochondrial AAA+ protease, in heart failure. Comprehensive analysis revealed that electron transport chain protein levels were increased even with a reduction in the expression of their corresponding mRNAs in heart failure, which indicated decreased protein turnover and resulted in the accumulation of oxidative damage in the electron transport chain. The induction of mitochondria-targeted human catalase ameliorated proteolytic activity and protein homeostasis in the electron transport chain, leading to improvements in mitochondrial energetics and cardiac contractility even during the late stage of pressure overload. Moreover, the infusion of mitoTEMPO, a mitochondria-targeted superoxide dismutase mimetic, recovered oxidative modifications of LONP1 and improved mitochondrial respiration capacity and cardiac function. The in vivo small interfering RNA repression of LONP1 partially canceled the protective effects of mitochondria-targeted human catalase induction and mitoTEMPO infusion. CONCLUSIONS: Oxidative post-translational modifications attenuate mitochondrial AAA+ protease activity, which is involved in impaired electron transport chain protein homeostasis, mitochondrial respiration deficiency, and left ventricular contractile dysfunction. Oxidatively inactivated proteases may be an endogenous target for mitoTEMPO treatment in pressure overload heart failure.


Subject(s)
Heart Failure/metabolism , Mitochondria, Heart/metabolism , Protease La/metabolism , Protein Processing, Post-Translational , Animals , Cysteine/metabolism , Disease Models, Animal , Heart Failure/drug therapy , Mice , Mice, Inbred C57BL , Organophosphorus Compounds/therapeutic use , Oxidation-Reduction , Piperidines/therapeutic use , RNA, Messenger/metabolism
9.
Proc Natl Acad Sci U S A ; 111(8): 3116-21, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24516131

ABSTRACT

Mitochondrial compromise is a fundamental contributor to pancreatic ß-cell failure in diabetes. Previous studies have demonstrated a broader role for tumor suppressor p53 that extends to the modulation of mitochondrial homeostasis. However, the role of islet p53 in glucose homeostasis has not yet been evaluated. Here we show that p53 deficiency protects against the development of diabetes in streptozotocin (STZ)-induced type 1 and db/db mouse models of type 2 diabetes. Glucolipotoxicity stimulates NADPH oxidase via receptor for advanced-glycation end products and Toll-like receptor 4. This oxidative stress induces the accumulation of p53 in the cytosolic compartment of pancreatic ß-cells in concert with endoplasmic reticulum stress. Cytosolic p53 disturbs the process of mitophagy through an inhibitory interaction with Parkin and induces mitochondrial dysfunction. The occurrence of mitophagy is maintained in STZ-treated p53(-/-) mice that exhibit preserved glucose oxidation capacity and subsequent insulin secretion signaling, leading to better glucose tolerance. These protective effects are not observed when Parkin is deleted. Furthermore, pifithrin-α, a specific inhibitor of p53, ameliorates mitochondrial dysfunction and glucose intolerance in both STZ-treated and db/db mice. Thus, an intervention with cytosolic p53 for a mitophagy deficiency may be a therapeutic strategy for the prevention and treatment of diabetes.


Subject(s)
Diabetes Mellitus/physiopathology , Insulin-Secreting Cells/metabolism , Mitophagy/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Benzothiazoles/pharmacology , Cell Line , Endoplasmic Reticulum Stress/physiology , Flow Cytometry , Fluorescent Antibody Technique , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/physiology , Mice , Mice, Knockout , Microscopy, Electron , Oxidative Stress/physiology , RNA Interference , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
10.
Nat Commun ; 4: 2308, 2013.
Article in English | MEDLINE | ID: mdl-23917356

ABSTRACT

Cumulative evidence indicates that mitochondrial dysfunction has a role in heart failure progression, but whether mitochondrial quality control mechanisms are involved in the development of cardiac dysfunction remains unclear. Here we show that cytosolic p53 impairs autophagic degradation of damaged mitochondria and facilitates mitochondrial dysfunction and heart failure in mice. Prevalence and induction of mitochondrial autophagy is attenuated by senescence or doxorubicin treatment in vitro and in vivo. We show that cytosolic p53 binds to Parkin and disturbs its translocation to damaged mitochondria and their subsequent clearance by mitophagy. p53-deficient mice show less decline of mitochondrial integrity and cardiac functional reserve with increasing age or after treatment with doxorubicin. Furthermore, overexpression of Parkin ameliorates the functional decline in aged hearts, and is accompanied by decreased senescence-associated ß-galactosidase activity and proinflammatory phenotypes. Thus, p53-mediated inhibition of mitophagy modulates cardiac dysfunction, raising the possibility that therapeutic activation of mitophagy by inhibiting cytosolic p53 may ameliorate heart failure and symptoms of cardiac ageing.


Subject(s)
Cytosol/metabolism , Mitochondria/metabolism , Mitophagy , Myocardium/metabolism , Myocardium/pathology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Aging/pathology , Animals , Cellular Senescence/drug effects , Doxorubicin/adverse effects , Embryo, Mammalian/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , HCT116 Cells , Heterozygote , Humans , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/ultrastructure , Mitophagy/drug effects , Myocardium/ultrastructure , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Ubiquitin-Protein Ligases/chemistry
12.
J Mol Cell Cardiol ; 52(1): 175-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22044588

ABSTRACT

Inhibition of tumor suppressor p53 is cardioprotective against ischemic injury and provides resistance to subsequent cardiac remodeling. We investigated p53-mediated expansion of ischemic damage with a focus on mitochondrial integrity in association with autophagy and apoptosis. p53(-/-) heart showed that autophagic flux was promoted under ischemia without a change in cardiac tissue ATP content. Electron micrographs revealed that ischemic border zone in p53(-/-) mice had 5-fold greater numbers of autophagic vacuoles containing mitochondria, indicating the occurrence of mitophagy, with an apparent reduction of abnormal mitochondria compared with those in WT mice. Analysis of autophagic mediators acting downstream of p53 revealed that TIGAR (TP53-induced glycolysis and apoptosis regulator) was exclusively up-regulated in ischemic myocardium. TIGAR(-/-) mice exhibited the promotion of mitophagy followed by decrease of abnormal mitochondria and resistance to ischemic injury, consistent with the phenotype of p53(-/-) mice. In p53(-/-) and TIGAR(-/-) ischemic myocardium, ROS production was elevated and followed by Bnip3 activation which is an initiator of mitophagy. Furthermore, the activation of Bnip3 and mitophagy due to p53/TIGAR inhibition were reversed with antioxidant N-acetyl-cysteine, indicating that this adaptive response requires ROS signal. Inhibition of mitophagy using chloroquine in p53(-/-) or TIGAR(-/-) mice exacerbated accumulation of damaged mitochondria to the level of wild-type mice and attenuated cardioprotective action. These findings indicate that p53/TIGAR-mediated inhibition of myocyte mitophagy is responsible for impairment of mitochondrial integrity and subsequent apoptosis, the process of which is closely involved in p53-mediated ventricular remodeling after myocardial infarction.


Subject(s)
Myocardial Ischemia/metabolism , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins , Autophagy/genetics , Gene Expression Regulation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Ischemia/genetics , Oxidative Stress , Phosphoric Monoester Hydrolases , Proteins/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Ventricular Remodeling/genetics
13.
Circ Heart Fail ; 5(1): 106-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22075967

ABSTRACT

BACKGROUND: Diabetic cardiomyopathy is characterized by energetic dysregulation caused by glucotoxicity, lipotoxicity, and mitochondrial alterations. p53 and its downstream mitochondrial assembly protein, synthesis of cytochrome c oxidase 2 (SCO2), are important regulators of mitochondrial respiration, whereas the involvement in diabetic cardiomyopathy remains to be determined. METHODS AND RESULTS: The role of p53 and SCO2 in energy metabolism was examined in both type I (streptozotocin [STZ] administration) and type II diabetic (db/db) mice. Cardiac expressions of p53 and SCO2 in 4-week STZ diabetic mice were upregulated (185% and 152% versus controls, respectively, P<0.01), with a marked decrease in cardiac performance. Mitochondrial oxygen consumption was increased (136% versus control, P<0.01) in parallel with augmentation of mitochondrial cytochrome c oxidase (complex IV) activity. Reactive oxygen species (ROS)-damaged myocytes and lipid accumulation were increased in association with membrane-localization of fatty acid translocase protein FAT/CD36. Antioxidant tempol reduced the increased expressions of p53 and SCO2 in STZ-diabetic hearts and normalized alterations in mitochondrial oxygen consumption, lipid accumulation, and cardiac dysfunction. Similar results were observed in db/db mice, whereas in p53-deficient or SCO2-deficient diabetic mice, the cardiac and metabolic abnormalities were prevented. Overexpression of SCO2 in cardiac myocytes increased mitochondrial ROS and fatty acid accumulation, whereas knockdown of SCO2 ameliorated them. CONCLUSIONS: Myocardial p53/SCO2 signal is activated by diabetes-mediated ROS generation to increase mitochondrial oxygen consumption, resulting in excessive generation of mitochondria-derived ROS and lipid accumulation in association with cardiac dysfunction.


Subject(s)
Diabetes Complications/complications , Diabetes Mellitus, Experimental/complications , Diabetic Cardiomyopathies/etiology , Lipid Metabolism/physiology , Mitochondria, Heart/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , CD36 Antigens/metabolism , Diabetes Complications/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Disease Models, Animal , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fatty Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Molecular Chaperones , Myocytes, Cardiac/metabolism , Oxygen Consumption/physiology , Streptozocin/adverse effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
14.
Am J Physiol Heart Circ Physiol ; 299(6): H1908-16, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20935145

ABSTRACT

Bioenergetic homeostasis is altered in heart failure and may play an important role in pathogenesis. p53 has been implicated in heart failure, and although its role in regulating tumorigenesis is well characterized, its activities on cellular metabolism are just beginning to be understood. We investigated the role of p53 and its transcriptional target gene TP53-induced glycolysis and apoptosis regulator (TIGAR) in myocardial energy metabolism under conditions simulating ischemia that can lead to heart failure. Expression of p53 and TIGAR was markedly upregulated after myocardial infarction, and apoptotic myocytes were decreased by 42% in p53-deficient mouse hearts compared with those in wild-type mice. To examine the effect of p53 on energy metabolism, cardiac myocytes were exposed to hypoxia. Hypoxia induced p53 and TIGAR expression in a p53-dependent manner. Knockdown of p53 or TIGAR increased glycolysis with elevated fructose-2,6-bisphosphate levels and reduced myocyte apoptosis. Hypoxic stress decreased phosphocreatine content and the mitochondrial membrane potential of myocytes without changes in ATP content, the effects of which were prevented by the knockdown of TIGAR. Inhibition of glycolysis by 2-deoxyglucose blocked these bioenergetic effects and TIGAR siRNA-mediated prevention of apoptosis, and, in contrast, overexpression of TIGAR reduced glucose utilization and increased apoptosis. Our data demonstrate that p53 and TIGAR inhibit glycolysis in hypoxic myocytes and that inhibition of glycolysis is closely involved in apoptosis, suggesting that p53 and TIGAR are significant mediators of cellular energy homeostasis and cell death under ischemic stress.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Energy Metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Proteins/metabolism , Stress, Physiological , Tumor Suppressor Protein p53/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Cell Hypoxia , Cells, Cultured , Deoxyglucose/metabolism , Disease Models, Animal , Glycolysis , Homeostasis , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Phosphocreatine/metabolism , Phosphofructokinase-2/metabolism , Phosphoric Monoester Hydrolases , Proteins/genetics , RNA Interference , Rats , Time Factors , Transfection , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...