Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Apoptosis ; 29(7-8): 935-937, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38642320

ABSTRACT

This study explores how 14-15 h fasting or acute exercise affects immune cell epigenetics, specifically focusing on miRNAs in mononuclear cells. Findings suggest fasting significantly impacts microRNAs associated with endothelial metabolism compared to exercise, but does not directly connect these changes to cell apoptosis or autophagy. This enhances comprehension of cellular self-consumption under health-promoting interventions.


Subject(s)
Fasting , Leukocytes, Mononuclear , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Exercise/physiology , Healthy Volunteers , Apoptosis/genetics , Male , Endothelial Cells/metabolism , Autophagy/genetics , Adult , Female , Epigenesis, Genetic
2.
Apoptosis ; 27(9-10): 730-739, 2022 10.
Article in English | MEDLINE | ID: mdl-35852665

ABSTRACT

The concomitant investigation of apoptosis (a regulated cell death) and autophagy (a conserved cell survival mechanism) in immune cells is rare. More detailed knowledge of these two types of self-consumption in circulating lymphocytes and monocytes would be important, since conditions such as fasting and acute exercise could promote health by a coordinated/linked modulation of autophagy and apoptosis in these mononuclear cells. In this study we performed flow cytometry to quantify numbers of apoptotic and autophagic mononuclear cells, lymphocytes and monocytes in fasting, standardized fed, and exercise conditions, using Annexin V, LC3B, and p62, respectively. We show that within total mononuclear cells lymphocytes are less apoptotic and autophagic than monocytes during fasting (p < 0.001, p < 0.05, respectively) and after acute exercise (p < 0.01, p < 0.05, respectively). Fasting increased circulating autophagic monocyte concentrations, but not lymphocytes compared to the fed control condition. Acute exercise elevated circulating autophagic lymphocyte concentrations, but not monocytes. Interestingly, Western blotting analysis of the fasting samples showed that higher LC3BII/I ratios were correlated with lower numbers of autophagic mononuclear cells (r = - 0.74, p = 0.02, n = 8), which could be attributed to the monocyte subgroup, but not lymphocytes. These results extend the current knowledge of the two types of self-consumption in circulating immune cells and underline their possible importance in pro-inflammatory monocytes during fasting and exercise as health promoting interventions.


Subject(s)
Fasting , Health Promotion , Annexin A5 , Apoptosis/physiology , Autophagy , Exercise/physiology
4.
Biochim Biophys Acta Mol Cell Res ; 1867(3): 118562, 2020 03.
Article in English | MEDLINE | ID: mdl-31669265

ABSTRACT

During stress conditions such as pressure overload and acute ischemia, the myocardial endothelium releases neuregulin-1ß (NRG-1), which acts as a cardioprotective factor and supports recovery of the heart. Recently, we demonstrated that recombinant human (rh)NRG-1 enhances glucose uptake in neonatal rat ventricular myocytes via the ErbB2/ErbB4 heterodimer and PI3Kα. The present study aimed to further elucidate the mechanism whereby rhNRG-1 activates glucose uptake in comparison to the well-established insulin and to extend the findings to adult models. Combinations of rhNRG-1 with increasing doses of insulin did not yield any additive effect on glucose uptake measured as 3H-deoxy-d-glucose incorporation, indicating that the mechanisms of the two stimuli are similar. In c-Myc-GLUT4-mCherry-transfected neonatal rat cardiomyocytes, rhNRG-1 increased sarcolemmal GLUT4 by 16-fold, similar to insulin. In contrast to insulin, rhNRG-1 did not phosphorylate IRS-1 at Tyr612, indicating that IRS-1 is not implicated in the signal transmission. Treatment of neonatal rats with rhNRG-1 induced a signaling response comparable with that observed in vitro, including increased ErbB4-pTyr1284, Akt-pThr308 and Erk1/2-pThr202/Tyr204. In contrast, in adult cardiomyocytes rhNRG-1 only increased the phosphorylation of Erk1/2 without having any significant effect on Akt and AS160 phosphorylation and glucose uptake, suggesting that rhNRG-1 function in neonatal cardiomyocytes differs from that in adult cardiomyocytes. In conclusion, our results show that similar to insulin, rhNRG-1 can induce glucose uptake by activating the PI3Kα-Akt-AS160 pathway and GLUT4 translocation. Unlike insulin, the rhNRG-1-induced effect is not mediated by IRS proteins and is observed in neonatal, but not in adult rat cardiomyocytes.


Subject(s)
Glucose Transporter Type 4/genetics , Myocytes, Cardiac/metabolism , Neuregulin-1/genetics , Receptor, ErbB-3/genetics , Animals , Animals, Newborn , Glucose/metabolism , Humans , Myocardium/metabolism , Myocardium/pathology , Neuregulin-1/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Rats , Receptor, Insulin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Substrate Specificity
5.
Biochim Biophys Acta Mol Cell Res ; 1867(3): 118561, 2020 03.
Article in English | MEDLINE | ID: mdl-31655100

ABSTRACT

The development of functional and reliable in vitro cardiac models composed of fully mature cardiomyocytes is essential for improving drug screening test quality, therefore, the success of clinical trial outcomes. In their lifespan, cardiomyocytes undergo a dynamic maturation process from the fetal to adult stage, radically changing their metabolism, morphology, contractility and electrical properties. Before employing cells of human origin, in vitro models often use neonatal rat cardiomyocytes (NRCM) to obtain key proof-of-principles. Nevertheless, NRCM monolayers are prone to de-differentiate when maintained in culture. Supplementation of free fatty acids (FFA), the main energy source for mature cardiomyocytes, and co-culture with fibroblasts are each by itself known to promote the shift from fetal to adult cardiomyocytes. Using a co-culture system, our study investigates the effects of FFA on the cardiomyocyte phenotype in comparison to glucose as typical fetal energy source, and to 10% serum used as standard control condition. NRCM decreased their differentiation status and fibroblasts increased in number after 7days of culture in the control condition. On the contrary, both glucose- and FFA-supplementation better preserved protein expression of myosin-light-chain-2v, a marker of mature cardiomyocytes, and the fibroblast number at levels similar to those found in freshly isolated NRCM. Nevertheless, compared to glucose, FFA resulted in a significant increase in sarcomere striation and organization. Our findings constitute an important step forward towards the definition of the optimal culture conditions, highlighting the possible benefits of a further supplementation of specific FFA to promote CM maturation in a co-culture system with FB.


Subject(s)
Cell Differentiation/genetics , Fatty Acids/metabolism , Heart/growth & development , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cell Culture Techniques , Coculture Techniques , Fibroblasts/drug effects , Humans , Rats
6.
Circ Heart Fail ; 12(10): e006288, 2019 10.
Article in English | MEDLINE | ID: mdl-31607147

ABSTRACT

Heart failure is a complex syndrome whose phenotypic presentation and disease progression depends on a complex network of adaptive and maladaptive responses. One of these responses is the endothelial release of NRG (neuregulin)-1-a paracrine growth factor activating ErbB2 (erythroblastic leukemia viral oncogene homolog B2), ErbB3, and ErbB4 receptor tyrosine kinases on various targets cells. NRG-1 features a multitasking profile tuning regenerative, inflammatory, fibrotic, and metabolic processes. Here, we review the activities of NRG-1 on different cell types and organs and their implication for heart failure progression and its comorbidities. Although, in general, effects of NRG-1 in heart failure are compensatory and beneficial, translation into therapies remains unaccomplished both because of the complexity of the underlying pathways and because of the challenges in the development of therapeutics (proteins, peptides, small molecules, and RNA-based therapies) for tyrosine kinase receptors. Here, we give an overview of the complexity to be faced and how it may be tackled.


Subject(s)
Endothelial Cells/metabolism , Heart Failure/metabolism , Neuregulin-1/metabolism , Animals , Cardiovascular Agents/therapeutic use , Chronic Disease , Endothelial Cells/drug effects , ErbB Receptors/metabolism , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Ligands , Molecular Targeted Therapy , Neuregulin-1/therapeutic use , Signal Transduction
7.
Front Physiol ; 9: 242, 2018.
Article in English | MEDLINE | ID: mdl-29618980

ABSTRACT

Aging is associated with a decline in cardiac function due to a decreased myocardial reserve. This adverse cardiac remodeling comprises of a variety of changes, including a reduction in mitochondrial function and a decline in the expression of the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a central regulator of mitochondrial biogenesis and metabolic adaptation in the myocardium. To study the etiological involvement of PGC-1α in cardiac aging, we used mouse models mimicking the modest down- and upregulation of this coactivator in the old and the exercised heart, respectively. Young mice with reduced cardiac expression of PGC-1α recapitulated part of the age-related impairment in mitochondrial gene expression, but otherwise did not aggravate the aging process. Inversely however, moderate overexpression of PGC-1α counteracts numerous key age-related remodeling changes, e.g., by improving blood pressure, age-associated apoptosis, and collagen accumulation, as well as in the expression of many, but not all cardiac genes involved in mitochondrial biogenesis, dynamics, metabolism, calcium handling and contractility. Thus, while the reduction of PGC-1α in the heart is insufficient to cause an aging phenotype, moderate overexpression reduces pathological remodeling of older hearts and could thereby contribute to the beneficial effects of exercise on cardiac function in aging.

8.
Sci Signal ; 9(435): ra68, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27382027

ABSTRACT

Malignant hyperthermia is a potentially fatal hypermetabolic disorder triggered by halogenated anesthetics and the myorelaxant succinylcholine in genetically predisposed individuals. About 50% of susceptible individuals carry dominant, gain-of-function mutations in RYR1 [which encodes ryanodine receptor type 1 (RyR1)], though they have normal muscle function and no overt clinical symptoms. RyR1 is predominantly found in skeletal muscle but also at lower amounts in immune and smooth muscle cells, suggesting that RYR1 mutations may have a wider range of effects than previously suspected. Mild bleeding abnormalities have been described in patients with malignant hyperthermia carrying gain-of-function RYR1 mutations. We sought to determine the frequency and molecular basis for this symptom. We found that some patients with specific RYR1 mutations had abnormally high bleeding scores, whereas their healthy relatives did not. Knock-in mice with the malignant hyperthermia susceptibility RYR1 mutation Y522S (MHS RYR1Y522S) had longer bleeding times than their wild-type littermates. Primary vascular smooth muscle cells from RYR1Y522S knock-in mice exhibited a higher frequency of subplasmalemmal Ca(2+) sparks, leading to a more negative resting membrane potential. The bleeding defect of RYR1Y522S mice and of one patient was reversed by treatment with the RYR1 antagonist dantrolene, and Ca(2+) sparks in primary vascular smooth muscle cells from the MHS RYR1Y522S mice were blocked by ryanodine or dantrolene. Thus, RYR1 mutations may lead to prolonged bleeding by altering vascular smooth muscle cell function. The reversibility of the bleeding phenotype emphasizes the potential therapeutic value of dantrolene in the treatment of such bleeding disorders.


Subject(s)
Blood Coagulation Disorders/metabolism , Calcium Signaling , Malignant Hyperthermia/metabolism , Muscle, Smooth, Vascular/metabolism , Mutation, Missense , Myocytes, Smooth Muscle/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Substitution , Animals , Blood Coagulation Disorders/genetics , Blood Coagulation Disorders/pathology , Dantrolene/pharmacology , Female , Humans , Male , Malignant Hyperthermia/genetics , Malignant Hyperthermia/pathology , Mice , Mice, Mutant Strains , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Ryanodine Receptor Calcium Release Channel/genetics
9.
Am J Physiol Endocrinol Metab ; 310(9): E782-94, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26979522

ABSTRACT

Nrg1ß is critically involved in cardiac development and also maintains function of the adult heart. Studies conducted in animal models showed that it improves cardiac performance under a range of pathological conditions, which led to its introduction in clinical trials to treat heart failure. Recent work also implicated Nrg1ß in the regenerative potential of neonatal and adult hearts. The molecular mechanisms whereby Nrg1ß acts in cardiac cells are still poorly understood. In the present study, we analyzed the effects of Nrg1ß on glucose uptake in neonatal rat ventricular myocytes and investigated to what extent mTOR/Akt signaling pathways are implicated. We show that Nrg1ß enhances glucose uptake in cardiomyocytes as efficiently as IGF-I and insulin. Nrg1ß causes phosphorylation of ErbB2 and ErbB4 and rapidly induces the phosphorylation of FAK (Tyr(861)), Akt (Thr(308) and Ser(473)), and its effector AS160 (Thr(642)). Knockdown of ErbB2 or ErbB4 reduces Akt phosphorylation and blocks the glucose uptake. The Akt inhibitor VIII and the PI3K inhibitors LY-294002 and Byl-719 abolish Nrg1ß-induced phosphorylation and glucose uptake. Finally, specific mTORC2 inactivation after knockdown of rictor blocks the Nrg1ß-induced increases in Akt-p-Ser(473) but does not modify AS160-p-Thr(642) or the glucose uptake responses to Nrg1ß. In conclusion, our study demonstrates that Nrg1ß enhances glucose uptake in cardiomyocytes via ErbB2/ErbB4 heterodimers, PI3Kα, and Akt. Furthermore, although Nrg1ß activates mTORC2, the resulting Akt-Ser(473) phosphorylation is not essential for glucose uptake induction. These new insights into pathways whereby Nrg1ß regulates glucose uptake in cardiomyocytes may contribute to the understanding of its regenerative capacity and protective function in heart failure.


Subject(s)
Glucose/metabolism , Heart Ventricles/cytology , Multiprotein Complexes/metabolism , Myocytes, Cardiac/drug effects , Neuregulin-1/pharmacology , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Blotting, Western , Gene Knockdown Techniques , Hypoglycemic Agents/pharmacology , Immunoprecipitation , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Rats , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-4/drug effects , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism
10.
Biochim Biophys Acta ; 1863(7 Pt B): 1894-903, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26775585

ABSTRACT

Mammalian target of rapamycin (mTOR) is an evolutionary conserved kinase that senses the nutrient and energy status of cells, the availability of growth factors, stress stimuli and other cellular and environmental cues. It responds by regulating a range of cellular processes related to metabolism and growth in accordance with the available resources and intracellular needs. mTOR has distinct functions depending on its assembly in the structurally distinct multiprotein complexes mTORC1 or mTORC2. Active mTORC1 enhances processes including glycolysis, protein, lipid and nucleotide biosynthesis, and it inhibits autophagy. Reported functions for mTORC2 after growth factor stimulation are very diverse, are tissue and cell-type specific, and include insulin-stimulated glucose transport and enhanced glycogen synthesis. In accordance with its cellular functions, mTOR has been demonstrated to regulate cardiac growth in response to pressure overload and is also known to regulate cells of the immune system. The present manuscript presents recently obtained insights into mechanisms whereby mTOR may change anabolic, catabolic and stress response pathways in cardiomocytes and discusses how mTOR may affect inflammatory cells in the heart during hemodynamic stress. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Cardiomegaly/enzymology , Inflammation Mediators/metabolism , Myocarditis/enzymology , Myocytes, Cardiac/enzymology , TOR Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiovascular Agents/pharmacology , Humans , Myocarditis/drug therapy , Myocarditis/genetics , Myocarditis/pathology , Myocarditis/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Protein Biosynthesis , Protein Kinase Inhibitors/pharmacology , Proteolysis , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Ventricular Remodeling
11.
Cardiovasc Res ; 109(1): 103-14, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26598511

ABSTRACT

AIMS: Mammalian target of rapamycin (mTOR), a central regulator of growth and metabolism, has tissue-specific functions depending on whether it is part of mTOR complex 1 (mTORC1) or mTORC2. We have previously shown that mTORC1 is required for adaptive cardiac hypertrophy and maintenance of function under basal and pressure-overload conditions. In the present study, we aimed to identify functions of mTORC2 in the heart. METHODS AND RESULTS: Using tamoxifen-inducible cardiomyocyte-specific gene deletion, we generated mice deficient for cardiac rapamycin-insensitive companion of mTOR (rictor), an essential and specific component of mTORC2. Under basal conditions, rictor deficiency did not affect cardiac growth and function in young mice and also had no effects in adult mice. However, transverse aortic constriction caused dysfunction in the rictor-deficient hearts, whereas function was maintained in controls after 1 week of pressure overload. Adaptive increases in cardiac weight and cardiomyocyte cross-sectional area, fibrosis, and hypertrophic and metabolic gene expression were not different between the rictor-deficient and control mice. In control mice, maintained function was associated with increased protein levels of rictor, protein kinase C (PKC)ßII, and PKCδ, whereas rictor ablation abolished these increases. Rictor deletion also significantly decreased PKCε at baseline and after pressure overload. Our data suggest that reduced PKCε and the inability to increase PKCßII and PKCδ abundance are, in accordance with their known function, responsible for decreased contractile performance of the rictor-deficient hearts. CONCLUSION: Our study demonstrates that mTORC2 is implicated in maintaining contractile function of the pressure-overloaded male mouse heart.


Subject(s)
Cardiomegaly/physiopathology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Ventricular Function/physiology , Animals , Apoptosis , Carrier Proteins/physiology , Fibrosis , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Myocardium/pathology , Phosphoproteins/physiology , Phosphorylation , Protein Kinase C/analysis , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction
12.
PLoS One ; 7(5): e38331, 2012.
Article in English | MEDLINE | ID: mdl-22675453

ABSTRACT

Cardiac Na(+) channels encoded by the SCN5A gene are essential for initiating heart beats and maintaining a regular heart rhythm. Mutations in these channels have recently been associated with atrial fibrillation, ventricular arrhythmias, conduction disorders, and dilated cardiomyopathy (DCM).We investigated a young male patient with a mixed phenotype composed of documented conduction disorder, atrial flutter, and ventricular tachycardia associated with DCM. Further family screening revealed DCM in the patient's mother and sister and in three of the mother's sisters. Because of the complex clinical phenotypes, we screened SCN5A and identified a novel mutation, R219H, which is located on a highly conserved region on the fourth helix of the voltage sensor domain of Na(v)1.5. Three family members with DCM carried the R219H mutation.The wild-type (WT) and mutant Na(+) channels were expressed in a heterologous expression system, and intracellular pH (pHi) was measured using a pH-sensitive electrode. The biophysical characterization of the mutant channel revealed an unexpected selective proton leak with no effect on its biophysical properties. The H(+) leak through the mutated Na(v)1.5 channel was not related to the Na(+) permeation pathway but occurred through an alternative pore, most probably a proton wire on the voltage sensor domain.We propose that acidification of cardiac myocytes and/or downstream events may cause the DCM phenotype and other electrical problems in affected family members. The identification of this clinically significant H(+) leak may lead to the development of more targeted treatments.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Cardiomyopathy, Dilated/physiopathology , Protons , Sodium Channels/metabolism , Adult , Amino Acid Sequence , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/genetics , Base Sequence , Cardiomyopathy, Dilated/genetics , Cell Line , Humans , Hydrogen-Ion Concentration , Male , Molecular Sequence Data , Mutation , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Oocytes/metabolism , Pedigree , Phenotype , Sodium Channels/genetics , Xenopus
13.
Free Radic Biol Med ; 51(3): 609-18, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21620958

ABSTRACT

ß(1)-Integrin mediates cardiomyocyte growth and survival and its proper regulation is essential for the structural and functional integrity of the heart. ß(1)-Integrin expression is enhanced in hypertrophy, but the mechanism and significance of its up-regulation are unknown. Because reactive oxygen species (ROS) are important mediators of myocardial remodeling we examined their role in regulated ß(1)-integrin expression. Hypertrophy was induced in neonatal cardiomyocytes by endothelin-1 (ET-1), which activated the regulatory NADPH oxidase subunit Rac1, evoked ROS, and enhanced fetal gene expression and cardiomyocyte size. ET-1 also enhanced cell adhesion and FAK phosphorylation and inhibited oxidative stress-induced cardiomyocyte apoptosis. Further, ET-1 increased ß(1)-integrin mRNA and protein expression via Rac1-ROS-dependent MEK/ERK and EGF receptor-PI3K/Akt activation as shown by adenoviral dominant-negative Rac1 or overexpression of copper/zinc-superoxide dismutase. The relevance of regulated ß(1)-integrin expression was examined in cardiomyocytes, in which targeting siRNA impeded the ET-1-induced ß(1)-integrin up-regulation. In these cells, ET-1-induced cell adhesion, FAK phosphorylation, and hypertrophic response were significantly blunted, whereas its antiapoptotic effect was predominantly unchanged, suggesting at least partial dissociation of prohypertrophic and prosurvival signaling elicited by ET-1. In conclusion, ß(1)-integrin up-regulation in response to ET-1 is mediated via Rac1-ROS-dependent activation of prohypertrophic pathways and is mandatory for ET-1-induced FAK activation, cell adhesion, and hypertrophic response.


Subject(s)
Hyperplasia/metabolism , Integrin beta1/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , Endothelin-1/pharmacology , Gene Expression Regulation, Developmental/drug effects , Hyperplasia/genetics , Integrin beta1/genetics , Mutation/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Signal Transduction/drug effects , Transgenes/genetics , Up-Regulation/drug effects , Ventricular Remodeling/genetics , rac1 GTP-Binding Protein/genetics
14.
Circulation ; 123(10): 1073-82, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21357822

ABSTRACT

BACKGROUND: Cardiac hypertrophy involves growth responses to a variety of stimuli triggered by increased workload. It is an independent risk factor for heart failure and sudden death. Mammalian target of rapamycin (mTOR) plays a key role in cellular growth responses by integrating growth factor and energy status signals. It is found in 2 structurally and functionally distinct multiprotein complexes called mTOR complex (mTORC) 1 and mTORC2. The role of each of these branches of mTOR signaling in the adult heart is currently unknown. METHODS AND RESULTS: We generated mice with deficient myocardial mTORC1 activity by targeted ablation of raptor, which encodes an essential component of mTORC1, during adulthood. At 3 weeks after the deletion, atrial and brain natriuretic peptides and ß-myosin heavy chain were strongly induced, multiple genes involved in the regulation of energy metabolism were altered, but cardiac function was normal. Function deteriorated rapidly afterward, resulting in dilated cardiomyopathy and high mortality within 6 weeks. Aortic banding-induced pathological overload resulted in severe dilated cardiomyopathy already at 1 week without a prior phase of adaptive hypertrophy. The mechanism involved a lack of adaptive cardiomyocyte growth via blunted protein synthesis capacity, as supported by reduced phosphorylation of ribosomal S6 kinase 1 and 4E-binding protein 1. In addition, reduced mitochondrial content, a shift in metabolic substrate use, and increased apoptosis and autophagy were observed. CONCLUSIONS: Our results demonstrate an essential function for mTORC1 in the heart under physiological and pathological conditions and are relevant for the understanding of disease states in which the insulin/insulin-like growth factor signaling axis is affected such as diabetes mellitus and heart failure or after cancer therapy.


Subject(s)
Cardiomegaly/genetics , Cardiomegaly/physiopathology , Carrier Proteins/genetics , Carrier Proteins/physiology , Heart Failure/etiology , Heart Rate/physiology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis , Atrial Natriuretic Factor/analysis , Atrial Natriuretic Factor/metabolism , Autophagy , Carrier Proteins/metabolism , Cell Cycle Proteins , Energy Metabolism/genetics , Energy Metabolism/physiology , Eukaryotic Initiation Factors , Gene Expression/physiology , Heart Failure/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/physiology , Myosin Heavy Chains/analysis , Myosin Heavy Chains/metabolism , Natriuretic Peptide, Brain/analysis , Natriuretic Peptide, Brain/metabolism , Nonmuscle Myosin Type IIB/analysis , Nonmuscle Myosin Type IIB/metabolism , Phosphoproteins/metabolism , Phosphorylation , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
15.
J Proteomics ; 74(5): 672-82, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21338724

ABSTRACT

Dahl salt-sensitive (DS) and salt-resistant (DR) inbred rat strains represent a well established animal model for cardiovascular research. Upon prolonged administration of high-salt-containing diet, DS rats develop systemic hypertension, and as a consequence they develop left ventricular hypertrophy, followed by heart failure. The aim of this work was to explore whether this animal model is suitable to identify biomarkers that characterize defined stages of cardiac pathophysiological conditions. The work had to be performed in two stages: in the first part proteomic differences that are attributable to the two separate rat lines (DS and DR) had to be established, and in the second part the process of development of heart failure due to feeding the rats with high-salt-containing diet has to be monitored. This work describes the results of the first stage, with the outcome of protein expression profiles of left ventricular tissues of DS and DR rats kept under low salt diet. Substantial extent of quantitative and qualitative expression differences between both strains of Dahl rats in heart tissue was detected. Using Principal Component Analysis, Linear Discriminant Analysis and other statistical means we have established sets of differentially expressed proteins, candidates for further molecular analysis of the heart failure mechanisms.


Subject(s)
Gene Expression Regulation , Heart Failure/metabolism , Heart Ventricles/metabolism , Muscle Proteins/biosynthesis , Proteome/biosynthesis , Animals , Disease Models, Animal , Heart Failure/chemically induced , Heart Failure/pathology , Heart Failure/physiopathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Proteomics/methods , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary/adverse effects , Sodium Chloride, Dietary/pharmacology
16.
Eur J Heart Fail ; 12(8): 797-804, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20388649

ABSTRACT

AIMS: To test acute effects of the corticotropin-releasing factor-related peptide urocortin 2 (Ucn2) on left ventricular (LV) function and the propensity for ventricular arrhythmias in the isolated heart of an animal model of hypertension-induced heart failure. METHODS AND RESULTS: Hearts from Dahl salt-sensitive rats with severe LV dysfunction were perfused according to Langendorff. Left ventricular developed pressure and the positive and negative derivatives of LV pressure were analysed before and after perfusion with Ucn2 (n = 15) or normal perfusion solution (control, n = 9). Intracellular calcium cycling parameters were assessed by surface fluorometry. Furthermore, monophasic action potential duration (MAPD) and ventricular fibrillation threshold (VFT) were determined, the latter by a train-of-pulses method at increasing voltage to scan the vulnerable period of repolarization. Urocortin 2 significantly affected intracellular calcium cycling and improved LV contractile function and relaxation. Compared with baseline values, Ucn2 significantly decreased MAPD at 30, 50, and 90% repolarization and significantly increased VFT compared with baseline values. No changes were observed in control experiments. CONCLUSION: Administration of Ucn2 rapidly improves LV function and increases VF threshold in failing, isolated rat hearts with increased propensity for ventricular arrhythmias. These observations suggest a potential use of Ucn2 as a safe and novel agent for the treatment of acute heart failure.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Myocardium/pathology , Urocortins/pharmacology , Acute Disease , Animals , Disease Models, Animal , Fluorometry , Heart Failure/diagnostic imaging , Hemodynamics , Hypertrophy, Left Ventricular/diagnostic imaging , Intracellular Calcium-Sensing Proteins , Male , Perfusion , Rats , Rats, Inbred Dahl , Sodium, Dietary , Ultrasonography , Urocortins/analysis , Urocortins/therapeutic use , Ventricular Dysfunction, Left/physiopathology , Ventricular Fibrillation/diagnostic imaging , Ventricular Fibrillation/physiopathology
17.
Biochem J ; 424(1): 119-27, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19708858

ABSTRACT

Ucn2 (urocortin 2) has been shown to exert potent beneficial effects in the cardiovascular system, including inhibition of apoptosis, improvement of cardiomyocyte contractility and decrease of oxidative stress. The mechanisms that contribute to the regulation of hUcn2 (human Ucn2) expression in cardiovascular pathologies are not known. In the present study, we analysed the mechanism by which hypoxia, a major stimulus in ischaemic heart disease, regulates Ucn2 gene expression. Hypoxia and CPX (ciclopirox olamine), which prevents proteolytic degradation of HIF (hypoxia-inducible factor), significantly increased hUcn2 mRNA levels in TE-671 cells. Gene silencing of endogenous HIF1alpha abolishes this increase. Hypoxia and CPX activated a luciferase-linked fragment of the 3'FLR (3'-flanking region) of the hUcn2 gene containing two putative HREs (hypoxia-response elements), HRE1 and HRE2. Site-directed mutagenesis experiments demonstrated that HRE1 is required for HIF1alpha-dependent luciferase activation. This activation was conserved in constructs with the 3'FLR fragment placed upstream of the luciferase gene, indicating an enhancer function for HRE1. Competition assays revealed direct binding between HRE1 and HIF1alpha. Regulation of Ucn2 by hypoxia was confirmed in rat neonatal cardiomyocytes and in cardiac-derived H9c2 cells transfected with constructs of the 3'FLR of the hUcn2 gene. In conclusion, our study demonstrates that hypoxia induces hUcn2 expression via a specific HRE in the 3'FLR of the hUcn2 gene, which interacts with the transcription factor HIF1alpha. Hypoxia-mediated stimulation of cardioprotective Ucn2 may help to preserve cardiac function and prevent apoptosis in ischaemic conditions in the heart.


Subject(s)
Cell Hypoxia/physiology , Response Elements/genetics , Urocortins/genetics , Urocortins/metabolism , Animals , Animals, Newborn , Antifungal Agents/pharmacology , Cell Hypoxia/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Ciclopirox , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Immunoblotting , Mutagenesis, Site-Directed , Pyridones/pharmacology , Rats , Rats, Wistar , Response Elements/physiology
18.
Can J Cardiol ; 25(8): 455-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19668779

ABSTRACT

BACKGROUND: Long QT syndrome (LQTS) is characterized by corrected QT interval prolongation leading to torsades de pointes and sudden cardiac death. LQTS type 2 (LQTS2) is caused by mutations in the KCNH2 gene, leading to a reduction of the rapidly activating delayed rectifier K+ current and loss of human ether-à-go-go-related gene (hERG) channel function by different mechanisms. Triggers for life-threatening arrhythmias in LQTS2 are often auditory stimuli. OBJECTIVES: To screen KCNH2 for mutations in patients with LQTS2 on an electrocardiogram and auditory-induced syncope interpreted as seizures and sudden cardiac death, and to analyze their impact on the channel function in vitro. METHODS: The KCNH2 gene was screened for mutations in the index patients of three families. The novel mutations were reproduced in vitro using site-directed mutagenesis and characterized using the Xenopus oocyte expression system in voltage clamp mode. RESULTS: Novel KCNH2 mutations (Y493F, A429P and del234-241) were identified in the index patients with mostly typical LQTS2 features on their electrocardiograms. The biochemical data revealed a trafficking defect. The biophysical data revealed a loss of function when mutated hERG channels were coexpressed with the wild type. CONCLUSIONS: In all families, at least one patient carrying the mutation had a history of seizures after auditory stimuli, which is a major trigger for arrhythmic events in LQTS2. Seizures are likely due to cardiac syncope as a consequence of mutation-induced loss of function of the rapidly activating delayed rectifier K+ current.


Subject(s)
Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/complications , Long QT Syndrome/genetics , Seizures/etiology , Adult , Aged , ERG1 Potassium Channel , Electrocardiography , Female , Humans , Long QT Syndrome/diagnosis , Male , Middle Aged , Mutation
19.
Hypertension ; 53(4): 739-44, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19204182

ABSTRACT

Recently, novel corticotropin-releasing factor-related peptides, named urocortin 1, 2, and 3, and a distinct cardiac and peripheral vascular receptor (corticotropin-releasing factor receptor 2) were described being part of a peripheral corticotropin-releasing factor system modulating cardiovascular function in response to stress. Vasorelaxation and blood pressure lowering have been reported after acute administration of these peptides. No data are available on the acute and chronic effects of urocortin 2 on blood pressure in models of arterial hypertension. To test these effects, hypertensive salt-sensitive and normotensive salt-resistant Dahl rats were randomly assigned to twice-daily applications of urocortin 2 or vehicle for 5 weeks. Blood pressure, heart rate, and left ventricular dimension and function were recorded at baseline, after initial application, and, together with cardiac and aortic expression of urocortin 2 and its receptor, after 5 weeks of treatment. Urocortin 2 significantly reduced blood pressure in hypertensive rats without affecting heart rate. Long-term urocortin 2 treatment in hypertensive rats induced sustained blood pressure reduction and diminished the development of hypertension-induced left ventricular hypertrophy and the deterioration of left ventricular contractile function. Corticotropin-releasing factor receptor 2 expression was preserved despite chronic stimulation by urocortin 2. In conclusion, our study shows that, in an animal model of arterial hypertension, urocortin 2 has immediate and sustained blood pressure-lowering effects. Beneficial effects on blood pressure, left ventricular dimension, and function, together with preserved receptor expression, suggest that corticotropin-releasing factor receptor 2 stimulation by urocortin 2 may represent a novel approach to the treatment of arterial hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Corticotropin-Releasing Hormone/pharmacology , Hypertension/drug therapy , Hypertension/metabolism , Urocortins/pharmacology , Animals , Corticotropin-Releasing Hormone/genetics , Disease Models, Animal , Echocardiography , Gene Expression/drug effects , Heart Rate/drug effects , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/drug therapy , Hypertrophy, Left Ventricular/metabolism , Male , Rats , Rats, Inbred Dahl , Receptors, Corticotropin-Releasing Hormone/genetics , Urocortins/genetics
20.
Cell Metab ; 8(5): 411-24, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19046572

ABSTRACT

Mammalian target of rapamycin (mTOR) is a central controller of cell growth. mTOR assembles into two distinct multiprotein complexes called mTOR complex 1 (mTORC1) and mTORC2. Here we show that the mTORC1 component raptor is critical for muscle function and prolonged survival. In contrast, muscles lacking the mTORC2 component rictor are indistinguishable from wild-type controls. Raptor-deficient muscles become progressively dystrophic, are impaired in their oxidative capacity, and contain increased glycogen stores, but they express structural components indicative of oxidative muscle fibers. Biochemical analysis indicates that these changes are probably due to loss of activation of direct downstream targets of mTORC1, downregulation of genes involved in mitochondrial biogenesis, including PGC1alpha, and hyperactivation of PKB/Akt. Finally, we show that activation of PKB/Akt does not require mTORC2. Together, these results demonstrate that muscle mTORC1 has an unexpected role in the regulation of the metabolic properties and that its function is essential for life.


Subject(s)
Carrier Proteins/physiology , Mitochondria/physiology , Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Enzyme Activation , Gene Expression Regulation , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Oncogene Protein v-akt/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Proteins , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , TOR Serine-Threonine Kinases , Trans-Activators/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL