Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Physiol Genomics ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007510

ABSTRACT

The prevalence of metabolic syndrome in cardiac diseases such as heart failure with preserved ejection fraction (HFpEF) prompts the scientific community to investigate its adverse effects on cardiac function and remodeling. However, the selection of a preclinical model of obesity-induced cardiac remodeling has proven more challenging with inconsistencies often found in very similar mouse models. Here, we investigated the implication of genetic background as well as diet composition to identify a suitable model of diet-induced cardiac alterations. C57Bl/6J and C57Bl/6N male mice were subjected to distinct obesogenic diets consisting of high-fat and moderate-sucrose content (HF-S) or High-Sucrose and moderate-lipid content (F-HS) versus matching control diets. 5-month dietary intervention with obesogenic diets induced weight gain, adipocyte hypertrophy and increased visceral and subcutaneous fat mass in both substrains. Obese mice showed similar impairment of glucose disposition and insulin tolerance, both strains developing insulin resistance within two months. However, echocardiographic follow-up and histological analysis confirmed that HF-S diet increases cardiac hypertrophy, interstitial fibrosis as well as left atrial area in the C57Bl/6J strain only. On the contrary C57Bl/6N exhibit cardiac eccentric remodeling under control diets, possibly owing to a genetic mutation in the myosin light-chain-kinase 3 (Mylk3) gene, specific to this substrain, which was not further enhanced under obesogenic diets. Altogether, the present results highlight the importance of carefully selecting the suitable mouse strain and diets to model diet-induced cardiac remodeling. In this regard, C57Bl/6J mice develop significant cardiac remodeling in response to HF-S, and seem a suitable model for cardiometabolic disease.

2.
Front Cardiovasc Med ; 9: 854361, 2022.
Article in English | MEDLINE | ID: mdl-35360022

ABSTRACT

Background: Activation of the renin-angiotensin-aldosterone system (RAAS) plays a critical role in the development of hypertension. Published evidence on a putative "memory effect" of AngII on the vascular components is however scarce. Aim: To evaluate the long-term effects of transient exposure to AngII on the mouse heart and the arterial tissue. Methods: Blood pressure, cardiovascular tissue damage and remodeling, and systemic oxidative stress were evaluated in C57/B6/J mice at the end of a 2-week AngII infusion (AngII); 2 and 3 weeks after the interruption of a 2-week AngII treatment (AngII+2W and AngII +3W; so-called "memory" conditions) and control littermate (CTRL). RNAseq profiling of aortic tissues was used to identify potential key regulated genes accounting for legacy effects on the vascular phenotype. RNAseq results were validated by RT-qPCR and immunohistochemistry in a reproduction cohort of mice. Key findings were reproduced in a homotypic cell culture model. Results: The 2 weeks AngII infusion induced cardiac hypertrophy and aortic damage that persisted beyond AngII interruption and despite blood pressure normalization, with a sustained vascular expression of ICAM1, infiltration by CD45+ cells, and cell proliferation associated with systemic oxidative stress. RNAseq profiling in aortic tissue identified robust Acta2 downregulation at transcript and protein levels (α-smooth muscle actin) that was maintained beyond interruption of AngII treatment. Among regulators of Acta2 expression, the transcription factor Myocardin (Myocd), exhibited a similar expression pattern. The sustained downregulation of Acta2 and Myocd was associated with an increase in H3K27me3 in nuclei of aortic sections from mice in the "memory" conditions. A sustained downregulation of ACTA2 and MYOCD was reproduced in the cultured human aortic vascular smooth muscle cells upon transient exposure to Ang II. Conclusion: A transient exposure to Ang II produces prolonged vascular remodeling with robust ACTA2 downregulation, associated with epigenetic imprinting supporting a "memory" effect despite stimulus withdrawal.

4.
Sci Transl Med ; 12(564)2020 10 07.
Article in English | MEDLINE | ID: mdl-33028705

ABSTRACT

Pathological remodeling of the myocardium has long been known to involve oxidant signaling, but strategies using systemic antioxidants have generally failed to prevent it. We sought to identify key regulators of oxidant-mediated cardiac hypertrophy amenable to targeted pharmacological therapy. Specific isoforms of the aquaporin water channels have been implicated in oxidant sensing, but their role in heart muscle is unknown. RNA sequencing from human cardiac myocytes revealed that the archetypal AQP1 is a major isoform. AQP1 expression correlates with the severity of hypertrophic remodeling in patients with aortic stenosis. The AQP1 channel was detected at the plasma membrane of human and mouse cardiac myocytes from hypertrophic hearts, where it colocalized with NADPH oxidase-2 and caveolin-3. We show that hydrogen peroxide (H2O2), produced extracellularly, is necessary for the hypertrophic response of isolated cardiac myocytes and that AQP1 facilitates the transmembrane transport of H2O2 through its water pore, resulting in activation of oxidant-sensitive kinases in cardiac myocytes. Structural analysis of the amino acid residues lining the water pore of AQP1 supports its permeation by H2O2 Deletion of Aqp1 or selective blockade of the AQP1 intrasubunit pore inhibited H2O2 transport in mouse and human cells and rescued the myocyte hypertrophy in human induced pluripotent stem cell-derived engineered heart muscle. Treatment of mice with a clinically approved AQP1 inhibitor, Bacopaside, attenuated cardiac hypertrophy. We conclude that cardiac hypertrophy is mediated by the transmembrane transport of H2O2 by the water channel AQP1 and that inhibitors of AQP1 represent new possibilities for treating hypertrophic cardiomyopathies.


Subject(s)
Aquaporin 1 , Induced Pluripotent Stem Cells , Animals , Humans , Hydrogen Peroxide/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Myocardium/metabolism , Myocytes, Cardiac/metabolism
5.
ESC Heart Fail ; 7(3): 920-932, 2020 06.
Article in English | MEDLINE | ID: mdl-32154661

ABSTRACT

AIMS: The abundance of beta 3-adrenergic receptors (ß3-ARs) is upregulated in diseased human myocardium. We previously showed that cardiac-specific expression of ß3-AR inhibits the hypertrophic response to neurohormonal stimulation. Here, we further analysed signalling pathways involved in the anti-hypertrophic effect of ß3-AR. METHODS AND RESULTS: In vitro hypertrophic responses to phenylephrine (PE) were analysed in neonatal rat ventricular myocytes (NRVM) infected with a recombinant adenovirus expressing the human ß3-AR (AdVhß3). We confirmed results in mice with cardiomyocyte-specific moderate expression of human ß3-AR (ß3-TG) and wild-type (WT) littermates submitted to thoracic transverse aortic constriction (TAC) for 9 weeks. We observed a colocalization of ß3-AR with the AMP-activated protein kinase (AMPK) both in neonatal rat and in adult mouse cardiomyocytes. Treatment of NRVM with PE induced hypertrophy and a decrease in phosphorylation of Thr172-AMPK (/2, P = 0.0487) and phosphorylation of Ser79-acetyl-CoA carboxylase (ACC) (/2.6, P = 0.0317), inducing an increase in phosphorylated Ser235/236 S6 protein (×2.5, P = 0.0367) known to be involved in protein synthesis. These effects were reproduced by TAC in WT mice but restored to basal levels in ß3-AR expressing cells/mice. siRNA targeting of AMPK partly abrogated the anti-hypertrophic effect of ß3-AR in response to PE in NRVM. Concomitant with hypertrophy, autophagy was decreased by PE, as measured by microtubule-associated protein 1 light chain 3 (LC3)-II/LC3-I ratio (/2.6, P = 0.0010) and p62 abundance (×3, P = 0.0016) in NRVM or by TAC in WT mice (LC3-II/LC3-I ratio: /5.4, P = 0.0159), but preserved in human ß3-AR expressing cells and mice, together with reduced hypertrophy. CONCLUSIONS: Cardiac-specific moderate expression of ß3-AR inhibits the hypertrophic response in part through AMPK activation followed by inhibition of protein synthesis and preservation of autophagy. Activation of the cardiac ß3-AR pathway may provide future therapeutic avenues for the modulation of hypertrophic remodelling.


Subject(s)
AMP-Activated Protein Kinases , Autophagy , Animals , Hypertrophy , Mice , Myocardium , Myocytes, Cardiac , Rats
6.
Arterioscler Thromb Vasc Biol ; 38(10): 2345-2357, 2018 10.
Article in English | MEDLINE | ID: mdl-29976767

ABSTRACT

Objective- Members of the microRNA (miR)-199a family, namely miR-199a-5p and miR-199a-3p, have been recently identified as potential regulators of cardiac homeostasis. Also, upregulation of miR-199a expression in cardiomyocytes was reported to influence endothelial cells. Whether miR-199a is expressed by endothelial cells and, if so, whether it directly regulates endothelial function remains unknown. We investigate the implication of miR-199a products on endothelial function by focusing on the NOS (nitric oxide synthase)/NO pathway. Approach and Results- Bovine aortic endothelial cells were transfected with specific miRNA inhibitors (locked-nucleic acids), and potential molecular targets identified with prediction algorithms were evaluated by Western blot or immunofluorescence. Ex vivo experiments were performed with mice treated with antagomiRs targeting miR-199a-3p or -5p. Isolated vessels and blood were used for electron paramagnetic resonance or myograph experiments. eNOS (endothelial NO synthase) activity (through phosphorylations Ser1177/Thr495) is increased by miR-199a-3p/-5p inhibition through an upregulation of the PI3K (phosphoinositide 3-kinase)/Akt (protein kinase B) and calcineurin pathways. SOD1 (superoxide dismutase 1) and PRDX1 (peroxiredoxin 1) upregulation was also observed in locked-nucleic acid-treated cells. Moreover, miR-199a-5p controls angiogenesis and VEGFA (vascular endothelial growth factor A) production and upregulation of NO-dependent relaxation were observed in vessels from antagomiR-treated mice. This was correlated with increased circulated hemoglobin-NO levels and decreased superoxide production. Angiotensin infusion for 2 weeks also revealed an upregulation of miR-199a-3p/-5p in vascular tissues. Conclusions- Our study reveals that miR-199a-3p and miR-199a-5p participate in a redundant network of regulation of the NOS/NO pathway in the endothelium. We highlighted that inhibition of miR-199a-3p and -5p independently increases NO bioavailability by promoting eNOS activity and reducing its degradation, thereby supporting VEGF-induced endothelial tubulogenesis and modulating vessel contractile tone.


Subject(s)
Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , MicroRNAs/metabolism , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Vasodilation , Angiogenesis Inhibitors/pharmacology , Animals , Antagomirs/genetics , Antagomirs/metabolism , Cattle , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Stability , Gene Expression Regulation, Neoplastic , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type III/genetics , Oligonucleotides/genetics , Oligonucleotides/metabolism , Peroxiredoxins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proteolysis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Superoxide Dismutase-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vasodilation/drug effects
7.
Nat Commun ; 9(1): 374, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371602

ABSTRACT

AMP-activated protein kinase (AMPK) has been shown to inhibit cardiac hypertrophy. Here, we show that submaximal AMPK activation blocks cardiomyocyte hypertrophy without affecting downstream targets previously suggested to be involved, such as p70 ribosomal S6 protein kinase, calcineurin/nuclear factor of activated T cells (NFAT) and extracellular signal-regulated kinases. Instead, cardiomyocyte hypertrophy is accompanied by increased protein O-GlcNAcylation, which is reversed by AMPK activation. Decreasing O-GlcNAcylation by inhibitors of the glutamine:fructose-6-phosphate aminotransferase (GFAT), blocks cardiomyocyte hypertrophy, mimicking AMPK activation. Conversely, O-GlcNAcylation-inducing agents counteract the anti-hypertrophic effect of AMPK. In vivo, AMPK activation prevents myocardial hypertrophy and the concomitant rise of O-GlcNAcylation in wild-type but not in AMPKα2-deficient mice. Treatment of wild-type mice with O-GlcNAcylation-inducing agents reverses AMPK action. Finally, we demonstrate that AMPK inhibits O-GlcNAcylation by mainly controlling GFAT phosphorylation, thereby reducing O-GlcNAcylation of proteins such as troponin T. We conclude that AMPK activation prevents cardiac hypertrophy predominantly by inhibiting O-GlcNAcylation.


Subject(s)
AMP-Activated Protein Kinases/genetics , Acetylglucosamine/metabolism , Cardiomegaly/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nitrogenous Group Transferases/genetics , AMP-Activated Protein Kinases/deficiency , Acetylglucosamine/pharmacology , Acylation/drug effects , Animals , Animals, Newborn , Azaserine/pharmacology , Azo Compounds/pharmacology , Biphenyl Compounds , Cardiomegaly/metabolism , Cardiomegaly/pathology , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Gene Expression Regulation , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) , Glycosylation/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Male , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Nitrogenous Group Transferases/antagonists & inhibitors , Nitrogenous Group Transferases/metabolism , Norleucine/analogs & derivatives , Norleucine/pharmacology , Phosphorylation/drug effects , Primary Cell Culture , Pyrones/pharmacology , Rats , Rats, Wistar , Signal Transduction , Thiophenes/pharmacology , Troponin T/genetics , Troponin T/metabolism
8.
Eur Heart J ; 39(10): 888-898, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29106524

ABSTRACT

Aims: Human and mouse cardiac beta3-adrenergic receptors (beta3AR) exert antipathetic effects to those of beta1-2AR stimulation. We examined their role in modulating myocardial remodelling, particularly fibrosis in response to haemodynamic stress. Methods and results: Mice with cardiac myocyte-specific expression of beta3AR (ADRB3-tg) or tamoxifen-inducible homozygous deletion (c-Adrb3-ko, with loxP-targeted Adrb3) were submitted to transaortic constriction. A superfusion assay was used for proteomic analysis of paracrine mediators between beta3AR-expressing cardiac myocytes and cardiac fibroblasts cultured separately. We show that cardiac beta3AR attenuate myocardial fibrosis in response to haemodynamic stress. Interstitial fibrosis and collagen content were reduced in ADRB3-tg, but augmented in c-Adrb3-ko. ADRB3 and collagen (COL1A1) expression were also inversely related in ventricular biopsies of patients with valve disease. Incubation of cardiac fibroblasts with media conditioned by hypertrophic myocytes induced fibroblast proliferation, myo-differentiation, and collagen production. These effects were abrogated upon ADRB3 expression in myocytes. Comparative shotgun proteomic analysis of the myocyte secretomes revealed a number of factors differentially regulated by beta3AR, among which connective tissue growth factor [CTGF (CCN2)] was prominently reduced. CTGF was similarly reduced in stressed hearts from ADRB3-tg, but increased in hearts from c-Adrb3-ko mice. CTGF expression was mediated by reactive oxygen species production which was reduced by ADRB3 expression in vitro and in vivo. This antioxidant and anti-fibrotic effect involved beta3AR coupling to the neuronal isoform of nitric oxide synthase (nNOS) in cardiac myocytes, as both were abrogated upon nNOS inhibition or Nos1 homozygous deletion. Conclusion: Cardiac beta3AR protect from fibrosis in response to haemodynamic stress by modulating nitric oxide and oxidant stress-dependent paracrine signaling to fibroblasts. Specific agonism at beta3AR may offer a new therapeutic modality to prevent cardiac fibrosis.


Subject(s)
Fibrosis , Heart Diseases , Myocytes, Cardiac , Oxidative Stress/physiology , Paracrine Communication/physiology , Receptors, Adrenergic, beta-3/metabolism , Animals , Catecholamines/metabolism , Fibrosis/metabolism , Fibrosis/prevention & control , Heart Diseases/metabolism , Heart Diseases/prevention & control , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism
9.
JCI Insight ; 2(12)2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28614798

ABSTRACT

Adult cardiac progenitor cells (CPCs) display a low capacity to differentiate into cardiomyocytes in injured hearts, strongly limiting the regenerative capacity of the mammalian myocardium. To identify new mechanisms regulating CPC differentiation, we used primary and clonally expanded Sca-1+ CPCs from murine adult hearts in homotypic culture or coculture with cardiomyocytes. Expression kinetics analysis during homotypic culture differentiation showed downregulation of Wnt target genes concomitant with increased expression of the Wnt antagonist, Wnt inhibitory factor 1 (Wif1), which is necessary to stimulate CPC differentiation. We show that the expression of the Wif1 gene is repressed by DNA methylation and regulated by the de novo DNA methyltransferase Dnmt3a. In addition, miR-29a is upregulated early during CPC differentiation and downregulates Dnmt3a expression, thereby decreasing Wif1 gene methylation and increasing the efficiency of differentiation of Sca-1+ CPCs in vitro. Extending these findings in vivo, transient silencing of Dnmt3a in CPCs subsequently injected in the border zone of infarcted mouse hearts improved CPC differentiation in situ and remote cardiac remodeling. In conclusion, miR-29a and Dnmt3a epigenetically regulate CPC differentiation through Wnt inhibition. Remote effects on cardiac remodeling support paracrine signaling beyond the local injection site, with potential therapeutic interest for cardiac repair.

10.
Cardiovasc Res ; 112(1): 478-90, 2016 10.
Article in English | MEDLINE | ID: mdl-27520736

ABSTRACT

AIM: Cardiac progenitor cells (CPC) from adult hearts can differentiate to several cell types composing the myocardium but the underlying molecular pathways are poorly characterized. We examined the role of paracrine nitric oxide (NO) in the specification of CPC to the cardiac lineage, particularly through its inhibition of the canonical Wnt/ß-catenin pathway, a critical step preceding cardiac differentiation. METHODS AND RESULTS: Sca1 + CPC from adult mouse hearts were isolated by magnetic-activated cell sorting and clonally expanded. Pharmacologic NO donors increased their expression of cardiac myocyte-specific sarcomeric proteins in a concentration and time-dependent manner. The optimal time window for NO efficacy coincided with up-regulation of CPC expression of Gucy1a3 (coding the alpha1 subunit of guanylyl cyclase). The effect of paracrine NO was reproduced in vitro upon co-culture of CPC with cardiac myocytes expressing a transgenic NOS3 (endothelial nitric oxide synthase) and in vivo upon injection of CPC in infarcted hearts from cardiac-specific NOS3 transgenic mice. In mono- and co-cultures, this effect was abrogated upon inhibition of soluble guanylyl cyclase or nitric oxide synthase, and was lost in CPC genetically deficient in Gucy1a3. Mechanistically, NO inhibits the constitutive activity of the canonical Wnt/ß-catenin in CPC and in cell reporter assays in a guanylyl cyclase-dependent fashion. This was paralleled with decreased expression of ß-catenin and down-regulation of Wnt target genes in CPC and abrogated in CPC with a stabilized, non-inhibitable ß-catenin. CONCLUSIONS: Exogenous or paracrine sources of NO promote the specification towards the myocyte lineage and expression of cardiac sarcomeric proteins of adult CPC. This is contingent upon the expression and activity of the alpha1 subunit of guanylyl cyclase in CPC that is necessary for NO-mediated inhibition of the canonical Wnt/ß-catenin pathway.


Subject(s)
Adult Stem Cells/metabolism , Cell Differentiation , Cyclic GMP/metabolism , Myocytes, Cardiac/enzymology , Nitric Oxide/metabolism , Paracrine Communication , Sarcomeres/enzymology , Soluble Guanylyl Cyclase/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adult Stem Cells/drug effects , Animals , Antigens, Ly/metabolism , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Coculture Techniques , Dose-Response Relationship, Drug , Female , Immunomagnetic Separation , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Paracrine Communication/drug effects , Sarcomeres/drug effects , Signal Transduction , Soluble Guanylyl Cyclase/deficiency , Soluble Guanylyl Cyclase/genetics , Time Factors , Transfection , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
11.
J Biol Chem ; 291(30): 15841-52, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27231349

ABSTRACT

The study of the mechanisms leading to cardiac hypertrophy is essential to better understand cardiac development and regeneration. Pathological conditions such as ischemia or pressure overload can induce a release of extracellular nucleotides within the heart. We recently investigated the potential role of nucleotide P2Y receptors in cardiac development. We showed that adult P2Y4-null mice displayed microcardia resulting from defective cardiac angiogenesis. Here we show that loss of another P2Y subtype called P2Y6, a UDP receptor, was associated with a macrocardia phenotype and amplified pathological cardiac hypertrophy. Cardiomyocyte proliferation and size were increased in vivo in hearts of P2Y6-null neonates, resulting in enhanced postnatal heart growth. We then observed that loss of P2Y6 receptor enhanced pathological cardiac hypertrophy induced after isoproterenol injection. We identified an inhibitory effect of UDP on in vitro isoproterenol-induced cardiomyocyte hyperplasia and hypertrophy. The present study identifies mouse P2Y6 receptor as a regulator of cardiac development and cardiomyocyte function. P2Y6 receptor could constitute a therapeutic target to regulate cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Receptors, Purinergic P2/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Hyperplasia , Isoproterenol/adverse effects , Isoproterenol/pharmacology , Male , Mice , Mice, Knockout , Myocardial Ischemia/chemically induced , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Receptors, Purinergic P2/genetics
12.
J Cardiovasc Transl Res ; 8(6): 362-71, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26070905

ABSTRACT

We studied intraobserver (n = 24), interobserver (n = 24) and interexperiment (n = 12) reproducibility of left ventricular (LV) mass and volume measurements in mice using an 11.7 T MRI system. The LV systolic function was assessed with a short-axis FLASH-cine sequence in 29 mice, including animals having undergone transverse aortic constriction. Bland-Altman and regression analysis were used to compare the different data sets. Reproducibility was excellent for the LV mass and end-diastolic volume (coefficient of variability (CoV) between 5.4 and 11.8 %), good for end-systolic volume (CoV 15.2-19.4 %) and moderate for stroke volume and ejection fraction (CoV 14.7-20.9 %). We found an excellent correlation between LV mass determined by MRI and ex vivo morphometric data (r = 0.92). In conclusion, LV systolic function can be assessed on an 11.7 T MRI scanner with high reproducibility for most parameters, as needed in longitudinal studies. However, data should be interpreted taking into account the moderate reproducibility of small volumes.


Subject(s)
Magnetic Resonance Imaging, Cine/methods , Ventricular Dysfunction, Left/physiopathology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results , Stroke Volume
13.
J Immunol ; 194(4): 1874-81, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595790

ABSTRACT

Nucleotides are released in the heart under pathological conditions, but little is known about their contribution to cardiac inflammation. The present study defines the P2Y4 nucleotide receptor, expressed on cardiac microvascular endothelial cells and involved in postnatal heart development, as an important regulator of the inflammatory response to cardiac ischemia. P2Y4-null mice displayed smaller infarcts in the left descending artery ligation model, as well as reduced neutrophil infiltration and fibrosis. Gene profiling identified inter alia endothelin-1 (ET-1) as one of the target genes of P2Y4 in ischemic heart. The reduced level of ET-1 was correlated with reduction of microvascular hyperpermeability, neutrophil infiltration, and endothelial adhesion molecule expression, and it could be explained by the decreased number of endothelial cells in P2Y4-null mice. Expression analysis of metalloproteinases and their tissue inhibitors in ischemic heart revealed reduced expression of matrix metalloproteinase (MMP)-9, reported to be potentially regulated by ET-1, and MMP-8, considered as neutrophil collagenase, as well as reduction of tissue inhibitor of MMP-1 and tissue inhibitor of MMP-4 in P2Y4-null mice. Reduction of cardiac permeability and neutrophil infiltration was also observed in P2Y4-null mice in LPS-induced inflammation model. Protection against infarction resulting from loss of P2Y4 brings new therapeutic perspectives for cardiac ischemia and remodeling.


Subject(s)
Endothelin-1/biosynthesis , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Receptors, Purinergic P2/deficiency , Animals , Disease Models, Animal , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Mice , Mice, Knockout , Myocardial Infarction/physiopathology , Real-Time Polymerase Chain Reaction , Transcriptome
14.
J Mol Cell Cardiol ; 74: 32-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24805196

ABSTRACT

Cardiac fibroblasts (CF) are crucial in left ventricular (LV) healing and remodeling after myocardial infarction (MI). They are typically activated into myofibroblasts that express alpha-smooth muscle actin (α-SMA) microfilaments and contribute to the formation of contractile and mature collagen scars that minimize the adverse dilatation of infarcted areas. CF predominantly express the α1 catalytic subunit of AMP-activated protein kinase (AMPKα1), while AMPKα2 is the major catalytic isoform in cardiomyocytes. AMPKα2 is known to protect the heart by preserving the energy charge of cardiac myocytes during injury, but whether AMPKα1 interferes with maladaptative heart responses remains unexplored. In this study, we investigated the role of AMPKα1 in modulating LV dilatation and CF fibrosis during post-MI remodeling. AMPKα1 knockout (KO) and wild type (WT) mice were subjected to permanent ligation of the left anterior descending coronary artery. The absence of AMPKα1 was associated with increased CF proliferation in infarcted areas, while expression of the myodifferentiation marker α-SMA was decreased. Faulty maturation of myofibroblasts might derive from severe down-regulation of the non-canonical transforming growth factor-beta1/p38 mitogen-activated protein kinase (TGF-ß1/p38 MAPK) pathway in KO infarcts. In addition, lysyl oxidase (LOX) protein expression was dramatically reduced in the scar of KO hearts. Although infarct size was similar in AMPK-KO and WT hearts subjected to MI, these changes resulted in compromised scar contractility, defective scar collagen maturation, and exacerbated adverse remodeling, as indicated by increased LV diastolic dimension 30days after MI. Our data genetically demonstrate the centrality of AMPKα1 in post-MI scar formation and highlight the specificity of this catalytic isoform in cardiac fibroblast/myofibroblast biology.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cicatrix/genetics , Myocardial Contraction , Myocardial Infarction/genetics , Myofibroblasts/enzymology , Ventricular Remodeling , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Proliferation , Cicatrix/enzymology , Cicatrix/pathology , Cicatrix/physiopathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation , Mice , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myofibroblasts/pathology , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Circulation ; 129(4): 451-62, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24190960

ABSTRACT

BACKGROUND: ß1-2-adrenergic receptors (AR) are key regulators of cardiac contractility and remodeling in response to catecholamines. ß3-AR expression is enhanced in diseased human myocardium, but its impact on remodeling is unknown. METHODS AND RESULTS: Mice with cardiac myocyte-specific expression of human ß3-AR (ß3-TG) and wild-type (WT) littermates were used to compare myocardial remodeling in response to isoproterenol (Iso) or Angiotensin II (Ang II). ß3-TG and WT had similar morphometric and hemodynamic parameters at baseline. ß3-AR colocalized with caveolin-3, endothelial nitric oxide synthase (NOS) and neuronal NOS in adult transgenic myocytes, which constitutively produced more cyclic GMP, detected with a new transgenic FRET sensor. Iso and Ang II produced hypertrophy and fibrosis in WT mice, but not in ß3-TG mice, which also had less re-expression of fetal genes and transforming growth factor ß1. Protection from Iso-induced hypertrophy was reversed by nonspecific NOS inhibition at low dose Iso, and by preferential neuronal NOS inhibition at high-dose Iso. Adenoviral overexpression of ß3-AR in isolated cardiac myocytes also increased NO production and attenuated hypertrophy to Iso and phenylephrine. Hypertrophy was restored on NOS or protein kinase G inhibition. Mechanistically, ß3-AR overexpression inhibited phenylephrine-induced nuclear factor of activated T-cell activation. CONCLUSIONS: Cardiac-specific overexpression of ß3-AR does not affect cardiac morphology at baseline but inhibits the hypertrophic response to neurohormonal stimulation in vivo and in vitro, through a NOS-mediated mechanism. Activation of the cardiac ß3-AR pathway may provide future therapeutic avenues for the modulation of hypertrophic remodeling.


Subject(s)
Heart Ventricles/pathology , Myocytes, Cardiac/metabolism , Neurotransmitter Agents/pharmacology , Nitric Oxide Synthase/physiology , Receptors, Adrenergic, beta-3/metabolism , Ventricular Remodeling/drug effects , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Cells, Cultured , Cyclic GMP/physiology , Cyclic GMP-Dependent Protein Kinases/physiology , Disease Models, Animal , Heart Ventricles/physiopathology , Humans , Hypertrophy/chemically induced , Hypertrophy/pathology , Hypertrophy/physiopathology , In Vitro Techniques , Isoproterenol/adverse effects , Isoproterenol/pharmacology , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Neurotransmitter Agents/adverse effects , Receptors, Adrenergic, beta-3/genetics , Signal Transduction/physiology , Ventricular Remodeling/physiology
16.
Am J Physiol Heart Circ Physiol ; 304(9): H1267-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23457011

ABSTRACT

The importance of chronic stimulation of ß-adrenoceptors in the development of cardiac dysfunction is the rationale for the use of ß-blockers in the treatment of heart failure. Nebivolol is a third-generation ß-blocker, which has further properties including stimulation of endothelial nitric oxide synthase and/or ß3-adrenoceptors. The aim of this study was to investigate whether nebivolol has additional effects on ß-adrenoceptor-mediated functional responses along with morphologic and molecular determinants of cardiac hypertrophy compared with those of metoprolol, a selective ß1-adrenoceptor blocker. Rats infused by isoprenaline (100 µg·kg(-1)·day(-1), 14 days) were randomized into three groups according to the treatment with metoprolol (30 mg·kg(-1)·day(-1)), nebivolol (10 mg·kg(-1)·day(-1)), or placebo for 13 days starting on day 1 after implantation of minipump. Both metoprolol and nebivolol caused a similar reduction on heart rate. Nebivolol mediated a significant improvement on cardiac mass, coronary flow, mRNA expression levels of sarcoplasmic reticulum Ca(2+) ATPase (SERCA2a) and atrial natriuretic peptide and phospholamban (PLN)/SERCA2a and phospho-PLN/PLN ratio compared with metoprolol and placebo. Nebivolol prevented the detrimental effects of isoprenaline infusion on isoprenaline (68% of control at 30 µM), BRL37344 (63% of control at 0.1 µM), and forskolin (64% of control at 1 µM) responses compared with metoprolol (isoprenaline, 34% of control; BRL37344, no response; forskolin, 26% of control) and placebo (isoprenaline, 33% of control; BRL37344, 28% of control; forskolin, 12% of control). Both ß-blockers improved the changes in mRNA expressions of ß1- and ß3-adrenoceptors. Our results suggest that nebivolol partially protects the responsiveness of ß-adrenoceptor signaling and the development of cardiac hypertrophy independent of its ß1-adrenoceptor blocking effect.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Benzopyrans/pharmacology , Cardiomegaly/prevention & control , Ethanolamines/pharmacology , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Adrenergic beta-Agonists/pharmacology , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Benzopyrans/therapeutic use , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cardiomegaly/metabolism , Coronary Circulation/drug effects , Ethanolamines/therapeutic use , Heart Rate/drug effects , In Vitro Techniques , Isoproterenol/pharmacology , Male , Metoprolol/pharmacology , Metoprolol/therapeutic use , Nebivolol , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...