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










Publication year range
1.
Nutrients ; 12(12)2020 Nov 29.
Article in English | MEDLINE | ID: mdl-33260293

ABSTRACT

(1) Background: It is recommended that an athlete, in order to ensure correct nutrition and performance, should consume between 1.2 and 2.0 g/kg/day of protein, while the daily recommended protein intake for a non-athlete is 0.8and 0.9 mg/kg/day. It is unclear if athletes living in Mediterranean countries are able to meet protein requirements without supplementation, since Mediterranean diet de-emphasizes meat and meat products. (2) Methods: 166 athletes (125 males) enrolled between 2017 and 2019 were required to keep a dietary journal for three consecutive days (2 workdays and 1 weekend day). Athletes had to be >18 years old, train in a particular sport activity more than 3 h a week and compete at least at an amateur level. Journal data were collected and then translated into macro-nutrient content (grams of protein, carbohydrates, and lipids) by a nutritionist. (3) Results: The protein intake reported by this specific population vary slightly from the Academy of Nutrition and Dietetics (AND), Dietitians of Canada (DC), and the American College of Sports Medicine (ACSM) joint statement recommendation level. Average protein levels without protein supplementation fell within the protein guidelines. Counterintuitively, the intake among those who supplemented their diet with protein was higher compared with those who did not, even when excluding the contribution of supplements. Although the majority of subjects participating in the study were able to meet protein intake recommended for athletes without protein supplementation, 27% of athletes were below the guideline range. (4) Conclusions: these data suggest that athletes' nutrition should be more often evaluated by a nutritionist and that they will benefit from increasing their nutritional knowledge in order to make better food choices, resorting to protein supplementation only when effectively needed.


Subject(s)
Athletes , Diet, Mediterranean , Dietary Proteins/administration & dosage , Dietary Supplements , Sports , Body Weight , Diet Records , Eating , Energy Intake , Female , Humans , Italy , Male , Nutritional Requirements , Nutritional Status , Physical Conditioning, Human , Recommended Dietary Allowances , Young Adult
2.
Circulation ; 142(22): 2138-2154, 2020 12.
Article in English | MEDLINE | ID: mdl-32933333

ABSTRACT

BACKGROUND: Concentric and eccentric cardiac hypertrophy are associated with pressure and volume overload, respectively, in cardiovascular disease both conferring an increased risk of heart failure. These contrasting forms of hypertrophy are characterized by asymmetrical growth of the cardiac myocyte in mainly width or length, respectively. The molecular mechanisms determining myocyte preferential growth in width versus length remain poorly understood. Identification of the mechanisms governing asymmetrical myocyte growth could provide new therapeutic targets for the prevention or treatment of heart failure. METHODS: Primary adult rat ventricular myocytes, adeno-associated virus (AAV)-mediated gene delivery in mice, and human tissue samples were used to define a regulatory pathway controlling pathological myocyte hypertrophy. Chromatin immunoprecipitation assays with sequencing and precision nuclear run-on sequencing were used to define a transcriptional mechanism. RESULTS: We report that asymmetrical cardiac myocyte hypertrophy is modulated by SRF (serum response factor) phosphorylation, constituting an epigenomic switch balancing the growth in width versus length of adult ventricular myocytes in vitro and in vivo. SRF Ser103 phosphorylation is bidirectionally regulated by RSK3 (p90 ribosomal S6 kinase type 3) and PP2A (protein phosphatase 2A) at signalosomes organized by the scaffold protein mAKAPß (muscle A-kinase anchoring protein ß), such that increased SRF phosphorylation activates AP-1 (activator protein-1)-dependent enhancers that direct myocyte growth in width. AAV are used to express in vivo mAKAPß-derived RSK3 and PP2A anchoring disruptor peptides that block the association of the enzymes with the mAKAPß scaffold. Inhibition of RSK3 signaling prevents concentric cardiac remodeling induced by pressure overload, while inhibition of PP2A signaling prevents eccentric cardiac remodeling induced by myocardial infarction, in each case improving cardiac function. SRF Ser103 phosphorylation is significantly decreased in dilated human hearts, supporting the notion that modulation of the mAKAPß-SRF signalosome could be a new therapeutic approach for human heart failure. CONCLUSIONS: We have identified a new molecular switch, namely mAKAPß signalosome-regulated SRF phosphorylation, that controls a transcriptional program responsible for modulating changes in cardiac myocyte morphology that occur secondary to pathological stressors. Complementary AAV-based gene therapies constitute rationally-designed strategies for a new translational modality for heart failure.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Enlargement , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Serum Response Factor/metabolism , A Kinase Anchor Proteins/genetics , Adenoviridae/genetics , Animals , Animals, Newborn , Cells, Cultured , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Heart Failure/genetics , Heart Failure/pathology , Humans , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley
3.
Circulation ; 142(10): 948-962, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32611257

ABSTRACT

BACKGROUND: The Ca2+/calmodulin-dependent phosphatase calcineurin is a key regulator of cardiac myocyte hypertrophy in disease. An unexplained paradox is how the ß isoform of the calcineurin catalytic A-subunit (CaNAß) is required for induction of pathological myocyte hypertrophy, despite calcineurin Aα expression in the same cells. It is unclear how the pleiotropic second messenger Ca2+ drives excitation-contraction coupling while not stimulating hypertrophy by calcineurin in the normal heart. Elucidation of the mechanisms conferring this selectivity in calcineurin signaling should reveal new strategies for targeting the phosphatase in disease. METHODS: Primary adult rat ventricular myocytes were studied for morphology and intracellular signaling. New Förster resonance energy transfer reporters were used to assay Ca2+ and calcineurin activity in living cells. Conditional gene deletion and adeno-associated virus-mediated gene delivery in the mouse were used to study calcineurin signaling after transverse aortic constriction in vivo. RESULTS: CIP4 (Cdc42-interacting protein 4)/TRIP10 (thyroid hormone receptor interactor 10) was identified as a new polyproline domain-dependent scaffold for CaNAß2 by yeast 2-hybrid screen. Cardiac myocyte-specific CIP4 gene deletion in mice attenuated pressure overload-induced pathological cardiac remodeling and heart failure. Blockade of CaNAß polyproline-dependent anchoring using a competing peptide inhibited concentric hypertrophy in cultured myocytes; disruption of anchoring in vivo using an adeno-associated virus gene therapy vector inhibited cardiac hypertrophy and improved systolic function after pressure overload. Live cell Förster resonance energy transfer biosensor imaging of cultured myocytes revealed that Ca2+ levels and calcineurin activity associated with the CIP4 compartment were increased by neurohormonal stimulation, but minimally by pacing. Conversely, Ca2+ levels and calcineurin activity detected by nonlocalized Förster resonance energy transfer sensors were induced by pacing and minimally by neurohormonal stimulation, providing functional evidence for differential intracellular compartmentation of Ca2+ and calcineurin signal transduction. CONCLUSIONS: These results support a structural model for Ca2+ and CaNAß compartmentation in cells based on an isoform-specific mechanism for calcineurin protein-protein interaction and localization. This mechanism provides an explanation for the specific role of CaNAß in hypertrophy and its selective activation under conditions of pathologic stress. Disruption of CaNAß polyproline-dependent anchoring constitutes a rational strategy for therapeutic targeting of CaNAß-specific signaling responsible for pathological cardiac remodeling in cardiovascular disease deserving of further preclinical investigation.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/enzymology , Heart Ventricles/enzymology , Myocytes, Cardiac/enzymology , Animals , Calcineurin/genetics , Cardiomegaly/genetics , Cardiomegaly/pathology , Heart Ventricles/pathology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Myocytes, Cardiac/pathology
4.
J Mol Cell Cardiol ; 93: 98-105, 2016 04.
Article in English | MEDLINE | ID: mdl-26940993

ABSTRACT

Noonan syndrome (NS) is a congenital disorder resulting from mutations of the Ras-Raf signaling pathway. Hypertrophic cardiomyopathy associated with RAF1 "RASopathy" mutations is a major risk factor for heart failure and death in NS and has been attributed to activation of MEK1/2-ERK1/2 mitogen-activated protein kinases. We recently discovered that type 3 p90 ribosomal S6 kinase (RSK3) is an ERK effector that is required, like ERK1/2, for concentric myocyte hypertrophy in response to pathological stress such as pressure overload. In order to test whether RSK3 also contributes to NS-associated hypertrophic cardiomyopathy, RSK3 knock-out mice were crossed with mice bearing the Raf1(L613V) human NS mutation. We confirmed that Raf1(L613V) knock-in confers a NS-like phenotype, including cardiac hypertrophy. Active RSK3 was increased in Raf1(L613V) mice. Constitutive RSK3 gene deletion prevented the Raf1(L613V)-dependent concentric growth in width of the cardiac myocyte and attenuated cardiac hypertrophy in female mice. These results are consistent with RSK3 being an important mediator of ERK1/2-dependent growth in RASopathy. In conjunction with previously published data showing that RSK3 is important for pathological remodeling of the heart, these data suggest that targeting of this downstream MAP-kinase pathway effector should be considered in the treatment of RASopathy-associated hypertrophic cardiomyopathy.


Subject(s)
Cardiomyopathy, Hypertrophic/etiology , Noonan Syndrome/complications , Noonan Syndrome/genetics , Proto-Oncogene Proteins c-raf/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Animals , Cardiomyopathy, Hypertrophic/diagnosis , Cardiomyopathy, Hypertrophic/metabolism , Disease Models, Animal , Echocardiography , Enzyme Activation , Female , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Noonan Syndrome/metabolism , Phenotype , Proto-Oncogene Proteins c-raf/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ventricular Remodeling
5.
IUBMB Life ; 67(5): 331-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25988524

ABSTRACT

The family of p90 ribosomal S6 kinases (RSKs) are pleiotropic effectors for extracellular signal-regulated kinase signaling pathways. Recently, RSK3 was shown to be important for pathological remodeling of the heart. Although cardiac myocyte hypertrophy can be compensatory for increased wall stress, in chronic heart diseases, this nonmitotic cell growth is usually associated with interstitial fibrosis, increased cell death, and decreased cardiac function. Although RSK3 is less abundant in the cardiac myocyte than other RSK family members, RSK3 appears to serve a unique role in cardiac myocyte stress responses. A potential mechanism conferring the unique function of RSK3 in the heart is anchoring by the scaffold protein muscle A-kinase anchoring protein ß (mAKAPß). Recent findings suggest that RSK3 should be considered as a therapeutic target for the prevention of heart failure, a clinical syndrome of major public health significance.


Subject(s)
Myocytes, Cardiac/pathology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Humans , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Stress, Physiological
6.
J Cardiovasc Pharmacol ; 65(3): 218-25, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25551320

ABSTRACT

Cardiac remodeling is regulated by an extensive intracellular signal transduction network. Each of the many signaling pathways in this network contributes uniquely to the control of cellular adaptation. In the last few years, it has become apparent that multimolecular signaling complexes or "signalosomes" are important for fidelity in intracellular signaling and for mediating crosstalk between the different signaling pathways. These complexes integrate upstream signals and control downstream effectors. In the cardiac myocyte, the protein mAKAPß serves as a scaffold for a large signalosome that is responsive to cAMP, calcium, hypoxia, and mitogen-activated protein kinase signaling. The main function of mAKAPß signalosomes is to modulate stress-related gene expression regulated by the transcription factors NFATc, MEF2, and HIF-1α and type II histone deacetylases that control pathological cardiac hypertrophy.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cardiomegaly/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Ventricular Remodeling , Animals , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Humans , Myocytes, Cardiac/pathology
7.
Circ Heart Fail ; 7(4): 663-72, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24812305

ABSTRACT

BACKGROUND: Cardiac myocyte hypertrophy is regulated by an extensive intracellular signal transduction network. In vitro evidence suggests that the scaffold protein muscle A-kinase anchoring protein ß (mAKAPß) serves as a nodal organizer of hypertrophic signaling. However, the relevance of mAKAPß signalosomes to pathological remodeling and heart failure in vivo remains unknown. METHODS AND RESULTS: Using conditional, cardiac myocyte-specific gene deletion, we now demonstrate that mAKAPß expression in mice is important for the cardiac hypertrophy induced by pressure overload and catecholamine toxicity. mAKAPß targeting prevented the development of heart failure associated with long-term transverse aortic constriction, conferring a survival benefit. In contrast to 29% of control mice (n=24), only 6% of mAKAPß knockout mice (n=31) died in the 16 weeks of pressure overload (P=0.02). Accordingly, mAKAPß knockout inhibited myocardial apoptosis and the development of interstitial fibrosis, left atrial hypertrophy, and pulmonary edema. This improvement in cardiac status correlated with the attenuated activation of signaling pathways coordinated by the mAKAPß scaffold, including the decreased phosphorylation of protein kinase D1 and histone deacetylase 4 that we reveal to participate in a new mAKAP signaling module. Furthermore, mAKAPß knockout inhibited pathological gene expression directed by myocyte-enhancer factor-2 and nuclear factor of activated T-cell transcription factors that associate with the scaffold. CONCLUSIONS: mAKAPß orchestrates signaling that regulates pathological cardiac remodeling in mice. Targeting of the underlying physical architecture of signaling networks, including mAKAPß signalosome formation, may constitute an effective therapeutic strategy for the prevention and treatment of pathological remodeling and heart failure.


Subject(s)
A Kinase Anchor Proteins/genetics , Cardiomegaly/genetics , Gene Expression Regulation , Heart Failure/genetics , Myocardium/pathology , RNA/genetics , Ventricular Remodeling , A Kinase Anchor Proteins/biosynthesis , Animals , Apoptosis , Cardiomegaly/metabolism , Cardiomegaly/pathology , Disease Models, Animal , Heart Failure/metabolism , Heart Failure/physiopathology , Mice , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Signal Transduction
8.
Am J Physiol Heart Circ Physiol ; 305(7): H1010-9, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23913705

ABSTRACT

Myocardial interstitial fibrosis is an important contributor to the development of heart failure. Type 3 p90 ribosomal S6 kinase (RSK3) was recently shown to be required for concentric myocyte hypertrophy under in vivo pathological conditions. However, the role of RSK family members in myocardial fibrosis remains uninvestigated. Transgenic expression of α-tropomyosin containing a Glu180Gly mutation (TM180) in mice of a mixed C57BL/6:FVB/N background induces a cardiomyopathy characterized by a small left ventricle, interstitial fibrosis, and diminished systolic and diastolic function. Using this mouse model, we now show that RSK3 is required for the induction of interstitial fibrosis in vivo. TM180 transgenic mice were crossed to RSK3 constitutive knockout (RSK3(-/-)) mice. Although RSK3 knockout did not affect myocyte growth, the decreased cardiac function and mild pulmonary edema associated with the TM180 transgene were attenuated by RSK3 knockout. The improved cardiac function was consistent with reduced interstitial fibrosis in the TM180;RSK3(-/-) mice as shown by histology and gene expression analysis, including the decreased expression of collagens. The specific inhibition of RSK3 should be considered as a potential novel therapeutic strategy for improving cardiac function and the prevention of sudden cardiac death in diseases in which interstitial fibrosis contributes to the development of heart failure.


Subject(s)
Cardiomyopathies/enzymology , Myocardium/enzymology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Tropomyosin/metabolism , Ventricular Dysfunction, Left/enzymology , Ventricular Function, Left , Animals , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cardiomyopathies/prevention & control , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Genotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Myocardial Contraction , Myocardium/pathology , Phenotype , Pulmonary Edema/enzymology , Pulmonary Edema/genetics , Pulmonary Edema/prevention & control , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Stroke Volume , Tropomyosin/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control
9.
Circ Res ; 112(1): 128-39, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-22997248

ABSTRACT

RATIONALE: Cardiac myocyte hypertrophy is the main compensatory response to chronic stress on the heart. p90 ribosomal S6 kinase (RSK) family members are effectors for extracellular signal-regulated kinases that induce myocyte growth. Although increased RSK activity has been observed in stressed myocytes, the functions of individual RSK family members have remained poorly defined, despite being potential therapeutic targets for cardiac disease. OBJECTIVE: To demonstrate that type 3 RSK (RSK3) is required for cardiac myocyte hypertrophy. METHODS AND RESULTS: RSK3 contains a unique N-terminal domain that is not conserved in other RSK family members. We show that this domain mediates the regulated binding of RSK3 to the muscle A-kinase anchoring protein scaffold, defining a novel kinase anchoring event. Disruption of both RSK3 expression using RNA interference and RSK3 anchoring using a competing muscle A-kinase anchoring protein peptide inhibited the hypertrophy of cultured myocytes. In vivo, RSK3 gene deletion in the mouse attenuated the concentric myocyte hypertrophy induced by pressure overload and catecholamine infusion. CONCLUSIONS: Taken together, these data demonstrate that anchored RSK3 transduces signals that modulate pathologic myocyte growth. Targeting of signaling complexes that contain select kinase isoforms should provide an approach for the specific inhibition of cardiac myocyte hypertrophy and for the development of novel strategies for the prevention and treatment of heart failure.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cardiomegaly/enzymology , Myocytes, Cardiac/enzymology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , A Kinase Anchor Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Binding Sites , COS Cells , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Chlorocebus aethiops , Disease Models, Animal , Female , Genotype , HEK293 Cells , Humans , Immunoprecipitation , Isoproterenol , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Phenotype , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA Interference , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction , Transduction, Genetic , Transfection
10.
Biochim Biophys Acta ; 1823(4): 800-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22230191

ABSTRACT

The responses of AMP-activated protein kinase (AMPK) and Ornithine decarboxylase (ODC) to isoproterenol have been examined in H9c2 cardiomyoblasts, AMPK represents the link between cell growth and energy availability whereas ODC, the key enzyme in polyamine biosynthesis, is essential for all growth processes and it is thought to have a role in the development of cardiac hypertrophy. Isoproterenol rapidly induced ODC activity in H9c2 cardiomyoblasts by promoting the synthesis of the enzyme protein and this effect was counteracted by inhibitors of the PI3K/Akt pathway. The increase in enzyme activity became significant between 15 and 30min after the treatment. At the same time, isoproterenol stimulated the phosphorylation of AMPKα catalytic subunits (Thr172), that was associated to an increase in acetyl coenzyme A carboxylase (Ser72) phosphorylation. Downregulation of both α1 and α2 isoforms of the AMPK catalytic subunit by siRNA to knockdown AMPK enzymatic activity, led to superinduction of ODC in isoproterenol-treated cardiomyoblasts. Downregulation of AMPKα increased ODC activity even in cells treated with other adrenergic agonists and in control cells. Analogue results were obtained in SH-SY5Y neuroblastoma cells transfected with a shRNA construct against AMPKα. In conclusion, isoproterenol quickly activates in H9c2 cardiomyoblasts two events that seem to contrast one another. The first one, an increase in ODC activity, is linked to cell growth, whereas the second, AMPK activation, is a homeostatic mechanism that negatively modulates the first. The modulation of ODC activity by AMPK represents a mechanism that may contribute to control cell growth processes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Myoblasts, Cardiac/enzymology , Ornithine Decarboxylase/metabolism , Animals , Cardiomegaly/enzymology , Cardiomegaly/pathology , Down-Regulation/drug effects , Eflornithine/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Isoproterenol/pharmacology , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/pathology , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Signal Transduction/drug effects
11.
J Pediatr Endocrinol Metab ; 24(7-8): 419-25, 2011.
Article in English | MEDLINE | ID: mdl-21932575

ABSTRACT

The purpose of this study was to evaluate the associations of visfatin and adiponectin concentrations with insulin resistance and body composition in regularly physically active pubertal girls. In 129 girls, aged 13-15 years (pubertal stages 3-5), visfatin, adiponectin, insulin resistance measured by homeostasis model assessment (HOMA), and body composition measured by dual-energy X-ray absorptiometry were evaluated. Visfatin concentration was related to HOMA and overall adiposity (body mass index, fat mass) markers, whereas adiponectin concentration was related to overall adiposity (fat mass), central adiposity (trunk fat) and fat free mass values. These relationships remained significant (p < 0.05) after adjusting for pubertal stage. Visfatin was independently related to body mass index (beta = 0.936; p = 0.0001) and HOMA (beta = 0.444; p = 0.039) indices, whereas adiponectin was independently related to fat free mass (beta = 0.889; p = 0.003) and trunk fat (beta = -0.468; p = 0.042) values. In conclusion, visfatin could be related to insulin resistance and overall adiposity indices, whereas adiponectin was related to different body composition values in regularly physically active pubertal girls.


Subject(s)
Athletes , Body Composition , Cytokines/blood , Insulin Resistance , Nicotinamide Phosphoribosyltransferase/blood , Abdominal Fat/growth & development , Absorptiometry, Photon , Adiponectin/blood , Adiposity , Adolescent , Body Mass Index , Female , Humans , Motor Activity , Puberty/blood
12.
Biochem Biophys Res Commun ; 407(3): 512-6, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21414296

ABSTRACT

The sirtuin SIRT1 is an ubiquitous NAD(+) dependent deacetylase that plays a role in biological processes such as longevity and stress response. In cardiac models, SIRT1 is associated to protection against many stresses. However, the link between SIRT1 and heart hypertrophy is complex and not fully understood. This study focuses specifically on the response of SIRT1 to the α-adrenergic agonist phenylephrine in H9c2 cardiac myoblasts, a cell model of cardiac hypertrophy. After 24 and 48h of phenylephrine treatment, SIRT1 expression and deacetylase activity were significantly increased. SIRT1 upregulation by phenylephrine was not associated to changes in NAD(+) levels, but was blocked by inhibitors of AMP-activated Protein Kinase (AMPK) or by AMPK knockdown by siRNA. When SIRT1 was inhibited with sirtinol or downregulated by siRNA, H9c2 cell viability was significantly decreased following phenylephrine treatment, showing that SIRT1 improves cell survival under hypertrophic stress. We so then propose that the increase in SIRT1 activity and expression in H9c2 cells treated with phenylephrine is an adaptive response to the hypertrophic stress, suggesting that adrenergic stimulation of heart cells activates hypertrophic programming and at the same time also promotes a self-protecting and self-regulating mechanism.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/pharmacology , Cardiomegaly/enzymology , Myoblasts, Cardiac/drug effects , Phenylephrine/pharmacology , Sirtuin 1/biosynthesis , Stress, Physiological/drug effects , AMP-Activated Protein Kinase Kinases , Animals , Cell Line , Cell Survival/drug effects , Cytoprotection/drug effects , Gene Knockdown Techniques , Myoblasts, Cardiac/enzymology , Protein Biosynthesis/drug effects , Protein Kinases/genetics , Protein Kinases/metabolism , Rats , Up-Regulation
13.
Am J Hum Biol ; 22(6): 813-8, 2010.
Article in English | MEDLINE | ID: mdl-20878967

ABSTRACT

UNLABELLED: Recently, two single nucleotide polymorphisms at position 45 and 276 on the adiponectin gene (ADIPOQ) have been recognized as determinants of total adiponectin levels, insulin resistance, and risk for diabetes in various obese populations. OBJECTIVES: The aim of this study was to determine whether these two polymorphisms are indeed determinants in the development of metabolic disorders or whether they are secondary to other confounding factors. METHODS: To do so, we have selected 170 physically active adolescent girls (mean age, 14.03 ± 1.5 years and mean body mass index, 19.98 ± 2.5 kg/m²) devoid of any metabolic diseases or confounding factors, to better attribute any findings to genotype effects. Concentration of adiponectin, insulin, and glucose were determined from blood samples with appropriate kits. Body fat parameters were evaluated with dual-energy X-ray absorptiometry, and genotype was analyzed with DNA extracted from whole blood samples followed by polymerase chain reaction and electrophoresis to separate alleles. RESULTS: Neither single nucleotide polymorphism +45T/G nor +276G/T was related to homeostasis model assessment index or adiponectin levels; however, the presence of the G allele on site 45 favored a significant decrease in lean body mass compared with those who were T homozygous (TG:36.90/TT:41.07 kg, P < 0.05). CONCLUSIONS: Results suggest that the reported increase in the risk of diabetes in subjects that were G allele carriers at site 45 in obese populations compared with normal-weight populations can be linked instead to a change in muscle mass or the muscle itself present in this genotype group.


Subject(s)
Body Weights and Measures , Exercise , Polymorphism, Single Nucleotide , Adiponectin/blood , Adiponectin/genetics , Adolescent , Adolescent Development , Blood Glucose , Female , Genetic Association Studies , Genotype , Humans , Insulin/blood
14.
Chem Biol Interact ; 181(3): 409-16, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19576191

ABSTRACT

Methoctramine and its analogues are polymethylene tetramines that selectively bind to a variety of receptor sites. Although these compounds are widely used as pharmacological tools for receptor characterization, the toxicological properties of these polyamine-based structures are largely unknown. We have evaluated the cytotoxic effects of methoctramine and related symmetrical analogues differing in polymethylene chain length between the inner nitrogens against a panel of cell lines. Methoctramine caused cell death only at high micromolar concentrations, whereas its pharmacological action is exerted at nanomolar level. Increasing the spacing between the inner nitrogen atoms resulted in a significative increase in cytotoxicity. In particular, an elevated cytotoxicity is associated to a methylene chain length of 12 units dividing the inner amine functions (compound 5). H9c2 cardiomyoblasts were the most sensitive cells, followed by SH-SY5Y neuroblastoma, whereas HL60 leukaemia cells were much more resistant. Methoctramine and related compounds down-regulated ornithine decarboxylase, the first enzyme of polyamine biosynthesis even at non-toxic concentration. Further, methoctramine and compound 5 caused a limited up-regulation of spermine/spermidine N-acetyltransferase, suggesting that interference in polyamine metabolism is not a primary mechanism of toxicity. Methoctramine and its analogues bound to DNA with a higher affinity than spermine, but the correlation with their toxic effect was poor. The highly toxic compound 5 killed the cells in the absence of caspase activation and caused an increase in p53 expression and ERK1/2 phosphorylation. Compound 5 was directly oxidized by cell homogenates producing hydrogen peroxide and its toxic effect was partially subdued by the inhibition of its uptake, by the NMDA ligand MK-801, and by the antioxidant N-acetylcysteine, suggesting that compound 5 can act at different cellular levels and lead to oxidative stress.


Subject(s)
Cell Death/drug effects , Diamines/pharmacology , Polyamines/pharmacology , Blotting, Western , Cell Line, Tumor , Diamines/chemistry , Humans , Polyamines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...