Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
EBioMedicine ; 46: 264-273, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31345784

ABSTRACT

BACKGROUND: Termination of acute inflammation is an active process orchestrated by lipid mediators (LM) derived from polyunsaturated fatty acids, referred to as specialized pro-resolving mediators (SPM). These mediators also provide novel therapeutic opportunities for treating inflammatory disease. However, the regulation of these molecules following acute myocardial infarction (MI) remains of interest. METHODS: In this prospective observational study we aimed to profile plasma levels of SPMs in ST-elevation MI (STEMI) patients during the first week following MI. Plasma LM concentrations were measured in patients with STEMI (n = 15) at three time points and compared with stable coronary artery disease (CAD; n = 10) and healthy controls (n = 10). FINDINGS: Our main findings were: (i) Immediately after onset of MI and before peak troponin T levels, STEMI patients had markedly increased levels of SPMs as compared with healthy controls and stable CAD patients, with levels of these mediators declining during follow-up. (ii) The increase in SPMs primarily reflected an increase in docosapentaenoic acid- and docosahexaenoic acid-derived protectins. (iii) Several individual protectins were correlated with the rapid increase in neutrophil counts, but not with CRP. (iv) A shift in 5-LOX activity from the leukotriene B4 pathway to the pro-resolving RvTs was observed. INTERPRETATION: The temporal regulation of SPMs indicates that resolution mechanisms are activated early during STEMI as part of an endogenous mechanism to initiate repair. Thus strategies to boost the activity and/or efficacy of these endogenous mechanisms may represent novel therapeutic opportunities for treatment of patients with MI. FUND: This work was supported by grants from the South-Eastern Norwegian regional health authority, the European Research Council under the European Union's Horizon 2020 research and innovation program, a Sir Henry Dale Fellowship jointly funded by the Wellcome Trust and the Royal Society, and the Barts Charity.


Subject(s)
Inflammation Mediators/blood , Lipids/blood , ST Elevation Myocardial Infarction/blood , Aged , Biomarkers , Case-Control Studies , Cytokines/blood , Female , Humans , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Myocardial Infarction/etiology , Prospective Studies , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/etiology
2.
Front Immunol ; 10: 1285, 2019.
Article in English | MEDLINE | ID: mdl-31244838

ABSTRACT

Background: IL-1ß is a highly potent pro-inflammatory cytokine and its secretion is tightly regulated. Inactive pro-IL-1ß is transcribed in response to innate immune receptors activating NFκB. If tissue damage occurs, danger signals released from necrotic cells, such as ATP, can activate NLRP3-inflammasomes (multiprotein complexes consisting of NLRP3, ASC, and active caspase-1) which cleaves and activates pro-IL-1ß. NLRP3 activation also depends on NEK7 and mitochondrial ROS-production. Thus, IL-1ß secretion may be regulated at the level of each involved component. We have previously shown that NLRP3-dependent IL-1ß release can be induced in cardiac fibroblasts by pro-inflammatory stimuli. However, anti-inflammatory mechanisms targeting IL-1ß release in cardiac cells have not been investigated. mTOR is a key regulator of protein metabolism, including autophagy and proteasome activity. In this study we explored whether autophagy or proteasomal degradation are regulators of NLRP3 inflammasome activation and IL-1ß release from cardiac fibroblasts. Methods and Results: Serum starvation selectively reduced LPS/ATP-induced IL-1ß secretion from cardiac fibroblasts. However, no other inflammasome components, nor mitochondrial mass, were affected. The mTOR inhibitor rapamycin restored pro-IL-1ß protein levels as well as LPS/ATP-induced IL-1ß release from serum starved cells. However, neither serum starvation nor rapamycin induced autophagy in cardiac fibroblasts. Conversely, chloroquine and bafilomycin A (inhibitors of autophagy) and betulinic acid (a proteasome activator) effectively reduced LPS-induced pro-IL-1ß protein levels. Key findings were reinvestigated in human monocyte-derived macrophages. Conclusion: In cardiac fibroblasts, mTOR inhibition selectively favors pro-IL-1ß synthesis while proteasomal degradation and not autophagy is the major catabolic anti-inflammatory mechanism for degradation of this cytokine.


Subject(s)
Fibroblasts/metabolism , Gene Expression , Inflammasomes/metabolism , Interleukin-1beta/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Biomarkers , Cell Line , Cells, Cultured , Chloroquine , Cytokines , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mitochondria/metabolism , Proteolysis , Reactive Oxygen Species/metabolism
3.
J Mol Med (Berl) ; 95(7): 767-777, 2017 07.
Article in English | MEDLINE | ID: mdl-28357477

ABSTRACT

Wnt signaling is dysregulated in heart failure (HF) and may promote cardiac hypertrophy, fibrosis, and inflammation. Blocking the Wnt ligand Wnt5a prevents HF in animal models. However, the role of Wnt5a in human HF and its functions in cardiac cells remain unclear. Here, we investigated Wnt5a regulation in HF patients and its effects on primary mouse and human cardiac fibroblasts. Serum Wnt5a was elevated in HF patients and associated with hemodynamic, neurohormonal, and clinical measures of disease severity. In failing human hearts, Wnt5a protein correlated with interleukin (IL)-6 and tissue inhibitor of metalloproteinase (TIMP)-1. Wnt5a messenger RNA (mRNA) levels were markedly upregulated in failing myocardium and both mRNA and protein levels declined following left ventricular assist device therapy. In primary mouse and human cardiac fibroblasts, recombinant Wnt5a dose-dependently upregulated mRNA and protein release of IL-6 and TIMP-1. Wnt5a did not affect ß-catenin levels, but activated extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. Importantly, inhibition of ERK1/2 activation attenuated Wnt5a-induced release of IL-6 and TIMP-1. In conclusion, our results show that Wnt5a is elevated in the serum and myocardium of HF patients and is associated with measures of progressive HF. Wnt5a induces IL-6 and TIMP-1 in cardiac fibroblasts, which might promote myocardial inflammation and fibrosis, and thereby contribute to HF progression. KEY MESSAGES: • Wnt5a is elevated in serum and myocardium of HF patients and is associated with measures of progressive HF. • In cardiac fibroblasts, Wnt5a upregulates interleukin (IL)-6 and tissue inhibitor of metalloproteinase (TIMP)-1 through the ERK pathway. • Wnt5a-mediated effects might promote myocardial inflammation and fibrosis, and thereby contribute to HF progression.


Subject(s)
Fibroblasts/metabolism , Heart Failure/metabolism , Wnt-5a Protein/metabolism , Adult , Aged , Animals , Female , Fibroblasts/pathology , Heart Failure/blood , Heart Failure/pathology , Humans , Interleukin-6/metabolism , MAP Kinase Signaling System , Male , Mice , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Wnt Signaling Pathway , Wnt-5a Protein/blood
4.
J Card Fail ; 21(11): 916-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26231517

ABSTRACT

BACKGROUND: Adipose tissue has endocrine properties, secreting a wide range of mediators into the circulation, including factors involved in cardiovascular disease. However, little is known about the potential role of adipose tissue in heart failure (HF), and the aim of this study was to investigate epicardial (EAT) and subcutaneous (SAT) adipose tissue in HF patients. METHODS AND RESULTS: Thirty patients with systolic HF and 30 patients with normal systolic function undergoing thoracic surgery were included in the study. Plasma was sampled and examined with the use of enzyme-linked immunosorbent assays, whereas SAT and EAT biopsies were collected and examined by means of reverse-transcription polymerase chain reaction and gas chromatography. Significantly higher expressions of mRNA encoding interleukin-6, adrenomedullin, peroxisome proliferator-activated receptor α, and fatty acid (FA)-binding protein 3, as well as higher levels of monounsaturated FA and palmitoleic acid, were seen in the EAT of HF patients, whereas the levels of docosahexaenoic acid were lower. Palmitoleic acid levels in EAT were correlated with 2 parameters of cardiac remodeling: increasing left ventricular end-diastolic diameter and N-terminal pro-B-type natriuretic peptide. CONCLUSIONS: Our results demonstrate adipose tissue depot-specific alterations of synthesis of FA and inflammatory and metabolic mediators in systolic HF patients. EAT may be a source of increased circulatory and myocardial levels of these mediators through endocrine actions.


Subject(s)
C-Reactive Protein/metabolism , Fatty Acids/metabolism , Heart Failure, Systolic/metabolism , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Adipose Tissue/metabolism , Adult , Aged , Biomarkers/analysis , Cardiac Surgical Procedures/methods , Case-Control Studies , Chi-Square Distribution , Elective Surgical Procedures , Enzyme-Linked Immunosorbent Assay , Female , Heart Failure, Systolic/diagnostic imaging , Heart Failure, Systolic/surgery , Humans , Linear Models , Male , Middle Aged , Pericardium/metabolism , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction/methods , Statistics, Nonparametric , Subcutaneous Fat/metabolism , Ultrasonography
5.
Am J Physiol Heart Circ Physiol ; 303(1): H66-74, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22542621

ABSTRACT

Transgenic mice with cardiac-specific expression of a peptide inhibitor of G protein-coupled receptor kinase (GRK)3 [transgenic COOH-terminal GRK3 (GRK3ct) mice] display myocardial hypercontractility without hypertrophy and enhanced α(1)-adrenergic receptor signaling. A role for GRK3 in the pathogenesis of heart failure (HF) has not been investigated, but inhibition of its isozyme, GRK2, has been beneficial in several HF models. Here, we tested whether inhibition of GRK3 modulated evolving cardiac hypertrophy and dysfunction after pressure overload. Weight-matched male GRK3ct transgenic and nontransgenic littermate control (NLC) mice subjected to chronic pressure overload by abdominal aortic banding (AB) were compared with sham-operated (SH) mice. At 6 wk after AB, a significant increase of cardiac mass consistent with induction of hypertrophy was found, but no differences between GRK3ct-AB and NLC-AB mice were discerned. Simultaneous left ventricular (LV) pressure-volume analysis of electrically paced, ex vivo perfused working hearts revealed substantially reduced systolic and diastolic function in NLC-AB mice (n = 7), which was completely preserved in GRK3ct-AB mice (n = 7). An additional cohort was subjected to in vivo cardiac catheterization and LV pressure-volume analysis at 12 wk after AB. NLC-AB mice (n = 11) displayed elevated end-diastolic pressure (8.5 ± 3.1 vs. 2.9 ± 1.2 mmHg, P < 0.05), reduced cardiac output (3,448 ± 323 vs. 4,488 ± 342 µl/min, P < 0.05), and reduced dP/dt(max) and dP/dt(min) (both P < 0.05) compared with GRK3ct-AB mice (n = 16), corroborating the preserved cardiac structure and function observed in GRK3ct-AB hearts assessed ex vivo. Increased cardiac mass and myocardial mRNA expression of ß-myosin heavy chain confirmed the similar induction of cardiac hypertrophy in both AB groups, but only NLC-AB hearts displayed significantly elevated mRNA levels of brain natriuretic peptide and myocardial collagen contents as well as reduced ß(1)-adrenergic receptor responsiveness to isoproterenol, indicating increased LV wall stress and the transition to HF. Inhibition of cardiac GRK3 in mice does not alter the hypertrophic response but attenuates cardiac dysfunction and HF after chronic pressure overload.


Subject(s)
G-Protein-Coupled Receptor Kinase 3/physiology , Heart Diseases/drug therapy , Hypertension/complications , Myocytes, Cardiac/physiology , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cardiomegaly/etiology , Cardiomegaly/pathology , Endomyocardial Fibrosis/pathology , G-Protein-Coupled Receptor Kinase 3/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 3/genetics , Heart Diseases/etiology , Heart Diseases/physiopathology , Heart Failure/prevention & control , Immunohistochemistry , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Myocardium/enzymology , Myocardium/metabolism , Myocytes, Cardiac/enzymology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Ventricular Function, Left/physiology
6.
Circulation ; 125(17): 2108-18, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22496128

ABSTRACT

BACKGROUND: G-protein-coupled receptor kinase 2 (GRK2) is a primary regulator of ß-adrenergic signaling in the heart. G-protein-coupled receptor kinase 2 ablation impedes heart failure development, but elucidation of the cellular mechanisms has not been achieved, and such elucidation is the aim of this study. METHODS AND RESULTS: Myocyte contractility, Ca(2+) handling and excitation-contraction coupling were studied in isolated cardiomyocytes from wild-type and GRK2 knockout (GRK2KO) mice without (sham) or with myocardial infarction (MI). In cardiac myocytes isolated from unstressed wild-type and GRK2KO hearts, myocyte contractions and Ca(2+) transients were similar, but GRK2KO myocytes had lower sarcoplasmic reticulum (SR) Ca(2+) content because of increased sodium-Ca(2+) exchanger activity and inhibited SR Ca(2+) ATPase by local protein kinase A-mediated activation of phosphodiesterase 4 resulting in hypophosphorylated phospholamban. This Ca(2+) handling phenotype is explained by a higher fractional SR Ca(2+) release induced by increased L-type Ca(2+) channel currents. After ß-adrenergic stimulation, GRK2KO myocytes revealed significant increases in contractility and Ca(2+) transients, which were not mediated through cardiac L-type Ca(2+) channels but through an increased SR Ca(2+). Interestingly, post-MI GRK2KO mice showed better cardiac function than post-MI control mice, which is explained by an improved Ca(2+) handling phenotype. The SR Ca(2+) content was better maintained in post-MI GRK2KO myocytes than in post-MI control myocytes because of better-maintained L-type Ca(2+) channel current density and no increase in sodium-Ca(2+) exchanger in GRK2KO myocytes. An L-type Ca(2+) channel blocker, verapamil, reversed some beneficial effects of GRK2KO. CONCLUSIONS: These data argue for novel differential regulation of L-type Ca(2+) channel currents and SR load by GRK2. G-protein-coupled receptor kinase 2 ablation represents a novel beneficial Ca(2+) handling phenotype resisting adverse remodeling after MI.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Excitation Contraction Coupling/physiology , G-Protein-Coupled Receptor Kinase 2/physiology , Myocardial Infarction/complications , Sodium-Calcium Exchanger/metabolism , Ventricular Remodeling/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , G-Protein-Coupled Receptor Kinase 2/deficiency , G-Protein-Coupled Receptor Kinase 2/genetics , Heart Failure/etiology , Heart Failure/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocardial Infarction/enzymology , Myocytes, Cardiac/metabolism , Phenotype , Protein Structure, Tertiary , Receptors, Adrenergic, beta/physiology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Verapamil/pharmacology
7.
J Appl Physiol (1985) ; 112(8): 1372-82, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22345433

ABSTRACT

Chemokines have been suggested to play a role during development of left ventricular failure, but little is known about their role during right ventricular (RV) remodeling and dysfunction. We have previously shown that the chemokine (C-X-C motif) ligand 13 (CXCL13) regulates small leucine-rich proteoglycans (SLRPs). We hypothesized that chemokines are upregulated in the pressure-overloaded RV, and that they regulate SLRPs. Mice with RV pressure overload following pulmonary banding (PB) had a significant increase in RV weight and an increase in liver weight after 1 wk. Microarray analysis (Affymetrix) of RV tissue from mice with PB revealed that CXCL10, CXCL6, chemokine (C-X3-C motif) ligand 1 (CX3CL1), chemokine (C-C motif) ligand 5 (CCL5), CXCL16, and CCL2 were the most upregulated chemokines. Stimulation of cardiac fibroblasts with these same chemokines showed that CXCL16 increased the expression of the four SLRPs: decorin, lumican, biglycan, and fibromodulin. CCL5 increased the same SLRPs, except decorin, whereas CX3CL1 increased the expression of decorin and lumican. CXCL16, CX3CL1, and CCL5 were also shown to increase the levels of glycosylated decorin and lumican in the medium after stimulation of fibroblasts. In the pressure-overloaded RV tissue, Western blotting revealed an increase in the total protein level of lumican and a glycosylated form of decorin with a higher molecular weight compared with control mice. Both mice with PB and patients with pulmonary stenosis had significantly increased circulating levels of CXCL16 compared with healthy controls measured by enzyme immunoassay. In conclusion, we have found that chemokines are upregulated in the pressure-overloaded RV and that CXCL16, CX3CL1, and CCL5 regulate expression and posttranslational modifications of SLRPs in cardiac fibroblasts. In the pressure-overloaded RV, protein levels of lumican were increased, and a glycosylated form of decorin with a high molecular weight appeared.


Subject(s)
Chemokines/metabolism , Extracellular Matrix/metabolism , Hypertrophy, Right Ventricular/metabolism , Leucine/metabolism , Proteoglycans/metabolism , Ventricular Dysfunction, Right/metabolism , Adolescent , Animals , Case-Control Studies , Chemokine CCL5/metabolism , Chemokine CX3CL1/metabolism , Chemokine CXCL16 , Chemokine CXCL6/metabolism , Chemokines, CXC/metabolism , Child , Child, Preschool , Female , Fibroblasts/metabolism , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Models, Animal , Pulmonary Valve Stenosis/metabolism , Receptors, Scavenger/metabolism
8.
Lipids Health Dis ; 10: 245, 2011 Dec 29.
Article in English | MEDLINE | ID: mdl-22206454

ABSTRACT

BACKGROUND: In the western world, heart failure (HF) is one of the most important causes of cardiovascular mortality. Supplement with n-3 polyunsaturated fatty acids (PUFA) has been shown to improve cardiac function in HF and to decrease mortality after myocardial infarction (MI). The molecular structure and composition of n-3 PUFA varies between different marine sources and this may be of importance for their biological effects. Krill oil, unlike fish oil supplements, contains the major part of the n-3 PUFA in the form of phospholipids. This study investigated effects of krill oil on cardiac remodeling after experimental MI. Rats were randomised to pre-treatment with krill oil or control feed 14 days before induction of MI. Seven days post-MI, the rats were examined with echocardiography and rats in the control group were further randomised to continued control feed or krill oil feed for 7 weeks before re-examination with echocardiography and euthanization. RESULTS: The echocardiographic evaluation showed significant attenuation of LV dilatation in the group pretreated with krill oil compared to controls. Attenuated heart weight, lung weight, and levels of mRNA encoding classical markers of LV stress, matrix remodeling and inflammation reflected these findings. The total composition of fatty acids were examined in the left ventricular (LV) tissue and all rats treated with krill oil showed a significantly higher proportion of n-3 PUFA in the LV tissue, although no difference was seen between the two krill oil groups. CONCLUSIONS: Supplement with krill oil leads to a proportional increase of n-3 PUFA in myocardial tissue and supplement given before induction of MI attenuates LV remodeling.


Subject(s)
Cardiotonic Agents/pharmacology , Dilatation, Pathologic/prevention & control , Euphausiacea/chemistry , Fatty Acids, Omega-3/pharmacology , Myocardial Infarction/pathology , Oils/pharmacology , Ventricular Remodeling/drug effects , Animals , Cardiotonic Agents/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Gene Expression/drug effects , Heart/drug effects , Heart/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Lipids/blood , Male , Myocardium/enzymology , Myocardium/metabolism , Myocardium/pathology , Oils/therapeutic use , Organ Size/drug effects , Random Allocation , Rats , Rats, Wistar
9.
PLoS One ; 6(4): e18668, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21533157

ABSTRACT

RATIONALE: Inflammatory mechanisms have been suggested to play a role in the development of heart failure (HF), but a role for chemokines is largely unknown. Based on their role in inflammation and matrix remodeling in other tissues, we hypothesized that CXCL13 and CXCR5 could be involved in cardiac remodeling during HF. OBJECTIVE: We sought to analyze the role of the chemokine CXCL13 and its receptor CXCR5 in cardiac pathophysiology leading to HF. METHODS AND RESULTS: Mice harboring a systemic knockout of the CXCR5 (CXCR5(-/-)) displayed increased mortality during a follow-up of 80 days after aortic banding (AB). Following three weeks of AB, CXCR5(-/-) developed significant left ventricular (LV) dilatation compared to wild type (WT) mice. Microarray analysis revealed altered expression of several small leucine-rich proteoglycans (SLRPs) that bind to collagen and modulate fibril assembly. Protein levels of fibromodulin, decorin and lumican (all SLRPs) were significantly reduced in AB CXCR5(-/-) compared to AB WT mice. Electron microscopy revealed loosely packed extracellular matrix with individual collagen fibers and small networks of proteoglycans in AB CXCR5(-/-) mice. Addition of CXCL13 to cultured cardiac fibroblasts enhanced the expression of SLRPs. In patients with HF, we observed increased myocardial levels of CXCR5 and SLRPs, which was reversed following LV assist device treatment. CONCLUSIONS: Lack of CXCR5 leads to LV dilatation and increased mortality during pressure overload, possibly via lack of an increase in SLRPs. This study demonstrates a critical role of the chemokine CXCL13 and CXCR5 in survival and maintaining of cardiac structure upon pressure overload, by regulating proteoglycans essential for correct collagen assembly.


Subject(s)
Chemokines/metabolism , Heart Ventricles/metabolism , Signal Transduction , Animals , Mice , Mice, Knockout , Microscopy, Electron , Oligonucleotide Array Sequence Analysis , Pressure
10.
Ann Med ; 43(2): 90-103, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21039303

ABSTRACT

Accumulating evidence suggests that inflammatory pathways play an essential role in all stages of atherogenesis. Inflammatory processes are not only involved in plaque progression, but seem also to play a critical role in plaque rupture. Members of the tumor necrosis factor (TNF) superfamiliy are potent regulators of inflammation and cell survival and consist of 20 ligands that signal through 29 different receptors. Several lines of evidence suggest that TNF-related molecules are involved in the development of acute coronary syndromes (ACS). Most, convincing evidence exists for CD40 ligand-CD40 interaction, but several other members of the TNF superfamily seem also to be involved in this immune-mediated promotion of plaque instability, including LIGHT, receptor activator of nuclear factor κB ligand, and TNF-α. These plaque destabilization pathways involve the bidirectional interaction between platelets and endothelial cells/monocytes, activation of vascular smooth muscle cells, and co-stimulatory effects on T cells, promoting inflammation, thrombus formation, matrix degradation, and apoptosis. TNF-related pathways could contribute to the non-resolving inflammation that characterizes atherosclerosis, representing pathogenic loops that are operating during plaque rupture and the development of ACS. These TNF-related molecules could also represent attractive new targets for therapy in this disorder.


Subject(s)
Acute Coronary Syndrome/physiopathology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factors/metabolism , Acute Coronary Syndrome/drug therapy , Animals , Atherosclerosis/drug therapy , Atherosclerosis/physiopathology , Drug Delivery Systems , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Ligands , Signal Transduction
11.
Am J Physiol Heart Circ Physiol ; 300(4): H1291-302, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21186275

ABSTRACT

CCN2/connective tissue growth factor (CTGF), a CCN family matricellular protein repressed in healthy hearts after birth, is induced in heart failure of various etiologies. Multiple cellular and biological functions have been assigned to CCN2/CTGF depending on cellular context. However, the functions and mechanisms of action of CCN2/CTGF in the heart as well as its roles in cardiac physiology and pathophysiology remain unknown. Transgenic mice with cardiac-restricted overexpression of CTGF (Tg-CTGF) were generated and compared with nontransgenic littermate control (NLC) mice. Tg-CTGF mice displayed slightly lower cardiac mass and inconspicuous increase of myocardial collagen compared with NLC mice but no evidence of contractile dysfunction. Analysis of the myocardial transcriptome by DNA microarray revealed activation of several distinct gene programs in Tg-CTGF hearts involved in cardioprotection and growth inhibition. Indeed, Tg-CTGF mice subjected to ischemia-reperfusion injury by in situ transient occlusion of the left anterior descending coronary artery in vivo displayed reduced vulnerability with markedly diminished infarct size. These findings were recapitulated in isolated hearts perfused with recombinant human (h)CTGF before the ischemia-reperfusion procedure. Consistently, Tg-CTGF hearts, as well as isolated adult cardiac myocytes exposed to recombinant hCTGF, displayed enhanced phosphorylation and activity of the Akt/p70S6 kinase/GSK-3ß salvage kinase pathway and induction of several genes with reported cardioprotective functions. Inhibition of Akt activities also prevented the cardioprotective phenotype of hearts from Tg-CTGF mice. This report provides novel evidence that CTGF confers cardioprotection by salvage phosphokinase signaling leading to inhibition of GSK-3ß activities, activation of phospho-SMAD2, and reprogramming of gene expression.


Subject(s)
Connective Tissue Growth Factor/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Animals , Cardiotonic Agents/pharmacology , Cells, Cultured , Connective Tissue Growth Factor/genetics , Gene Expression Profiling , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Mice , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Smad2 Protein/metabolism
12.
Mutat Res ; 666(1-2): 32-8, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19481677

ABSTRACT

Base excision repair (BER) is the major pathway to counteract the genotoxic effect of endogenous DNA damaging agents. The present study investigated the enzymatic activities and gene transcription of DNA glycosylases initiating BER in an experimental heart failure (HF) model. Rats were subjected to myocardial infarction or sham-operated. Twenty-eight days after surgical intervention cell-free protein extracts, total RNA and genomic DNA were isolated to analyze DNA glycosylase and AP-endonuclease activities, transcript levels of DNA glycosylases and accumulation of oxidative DNA damage. The capacity to remove major oxidation products (e.g., formamidopyrimidine and 5-hydroxycytosine) was significantly increased in the border zone of infarcted area, while the capacity to remove the highly mutagenic 8-oxoguanine residue was enhanced both in non-infarcted and infarcted areas of left ventricle (LV). DNA glycosylase activities towards 3-methyladenine and uracil were up-regulated in infarcted and non-infarcted areas of LV, indicating that generation of alkylated and deaminated base lesions on DNA increase during HF. Finally, we found no difference in accumulation of oxidative DNA damage in myocardial tissue between rats with post-myocardial infarction and sham-operated rats. This up-regulation of activities, initiating the BER pathway, could play an important role during HF by counteracting the effect of genotoxic stress, structural damage of tissue and myocardial remodeling.


Subject(s)
DNA Repair , DNA, Mitochondrial/metabolism , Guanine/analogs & derivatives , Heart Failure/genetics , Myocardium/metabolism , Animals , DNA Damage , DNA Glycosylases/metabolism , Gene Expression Regulation, Enzymologic , Guanine/metabolism , Male , Oxidative Stress , Rats , Rats, Wistar , Up-Regulation
13.
J Mol Cell Cardiol ; 46(1): 100-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18930063

ABSTRACT

Chronic ventricular pressure overload states, such as hypertension, and elevated levels of neurohormones (norepinephrine, angiotensin II, endothelin-1) initiate cardiac hypertrophy and dysfunction and share the property of being able to bind to Gq-coupled 7-transmembrane receptors. The goal of the current study was to determine the role of endogenous cardiac myocyte Gq signaling and its role in cardiac hypertrophy and dysfunction during high blood pressure (BP). We induced renal artery stenosis for 8 weeks in control mice and mice expressing a peptide inhibitor of Gq signaling (GqI) using a 2 kidney, 1 clip renal artery stenosis model. 8 weeks following chronic high BP, control mice had cardiac hypertrophy and depressed function. Inhibition of cardiomyocyte Gq signaling did not reverse cardiac hypertrophy but attenuated increases in a profile of cardiac profibrotic genes and genes associated with remodeling. Inhibition of Gq signaling also attenuated the loss of cardiac function. We determined that Gq signaling downstream of angiotensin II receptor stimulation negatively impacted beta-adrenergic receptor (AR) responses and inhibition of Gq signaling was sufficient to restore betaAR-mediated responses. Therefore, in this study we found that Gq signaling negatively impacts cardiac function during high BP. Specifically, we found that inhibition of AT1-Gq signaling augmented betaAR mediated effects in a renal artery stenosis model of hypertension. These observations may underlie additional, beneficial effects of angiotensinogen converting enzyme (ACE) inhibitors and angiotensin receptor antagonists observed during times of hemodynamic stress.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin II/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Renal Artery Obstruction/pathology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Echocardiography/methods , Female , Hemodynamics , Hypertension , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 105(34): 12457-62, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18711143

ABSTRACT

G protein-coupled receptor (GPCR) kinases (GRKs) are critical regulators of cellular signaling and function. In cardiomyocytes, GRK2 and GRK5 are two GRKs important for myocardial regulation, and both have been shown to be up-regulated in the dysfunctional heart. We report that increased levels and activity of GRK5 in failing myocardium may have unique significance due to its nuclear localization, a property not shared by GRK2. We find that transgenic mice with elevated cardiac GRK5 levels have exaggerated hypertrophy and early heart failure compared with control mice after pressure overload. This pathology is not present in cardiac GRK2-overexpressing mice or in mice with overexpression of a mutant GRK5 that is excluded from the nucleus. Nuclear accumulation of GRK5 is enhanced in myocytes after aortic banding in vivo and in vitro in myocytes after increased G alpha q activity, the trigger for pressure-overload hypertrophy. GRK5 enhances activation of MEF2 in concert with Gq signals, demonstrating that nuclear localized GRK5 regulates gene transcription via a pathway critically linked to myocardial hypertrophy. Mechanistically, we show that this is due to GRK5 acting, in a non-GPCR manner, as a class II histone deacetylase (HDAC) kinase because it can associate with and phosphorylate the myocyte enhancer factor-2 repressor, HDAC5. Moreover, significant HDAC activity can be found with GRK5 in the heart. Our data show that GRK5 is a nuclear HDAC kinase that plays a key role in maladaptive cardiac hypertrophy apparently independent of any action directly on GPCRs.


Subject(s)
Cell Nucleus/enzymology , G-Protein-Coupled Receptor Kinase 5/physiology , Myocytes, Cardiac/enzymology , Animals , G-Protein-Coupled Receptor Kinase 5/analysis , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Heart Failure/enzymology , Heart Failure/etiology , Histone Deacetylases/metabolism , Hypertrophy/enzymology , Hypertrophy/etiology , MEF2 Transcription Factors , Mice , Mice, Transgenic , Myocytes, Cardiac/ultrastructure , Myogenic Regulatory Factors/metabolism , Up-Regulation
15.
Circ Res ; 103(4): 413-22, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18635825

ABSTRACT

Myocardial G protein-coupled receptor kinase (GRK)2 is a critical regulator of cardiac beta-adrenergic receptor (betaAR) signaling and cardiac function. Its upregulation in heart failure may further depress cardiac function and contribute to mortality in this syndrome. Preventing GRK2 translocation to activated betaAR with a GRK2-derived peptide that binds G(beta)gamma (betaARKct) has benefited some models of heart failure, but the precise mechanism is uncertain, because GRK2 is still present and betaARKct has other potential effects. We generated mice in which cardiac myocyte GRK2 expression was normal during embryonic development but was ablated after birth (alphaMHC-Cre x GRK2 fl/fl) or only after administration of tamoxifen (alphaMHC-MerCreMer x GRK2 fl/fl) and examined the consequences of GRK2 ablation before and after surgical coronary artery ligation on cardiac adaptation after myocardial infarction. Absence of GRK2 before coronary artery ligation prevented maladaptive postinfarction remodeling and preserved betaAR responsiveness. Strikingly, GRK2 ablation initiated 10 days after infarction increased survival, enhanced cardiac contractile performance, and halted ventricular remodeling. These results demonstrate a specific causal role for GRK2 in postinfarction cardiac remodeling and heart failure and support therapeutic approaches of targeting GRK2 or restoring betaAR signaling by other means to improve outcomes in heart failure.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Heart Failure/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Estrogen Antagonists/pharmacology , G-Protein-Coupled Receptor Kinase 2/genetics , Gene Expression Regulation , Heart Failure/pathology , Heart Failure/prevention & control , Ligation , Mice , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Receptors, Adrenergic, beta/metabolism , Tamoxifen/pharmacology
16.
Circulation ; 117(11): 1378-87, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18316484

ABSTRACT

BACKGROUND: A salient characteristic of dysfunctional myocardium progressing to heart failure is an upregulation of the adenylyl cyclase inhibitory guanine nucleotide (G) protein alpha subunit, G alpha(i2). It has not been determined conclusively whether increased Gi activity in the heart is beneficial or deleterious in vivo. Gi signaling has been implicated in the mechanism of cardioprotective agents; however, no in vivo evidence exists that any of the G alpha subunits are cardioprotective. We have created a novel molecular tool to specifically address the role of Gi proteins in normal and dysfunctional myocardium. METHODS AND RESULTS: We have developed a class-specific Gi inhibitor peptide, GiCT, composed of the region of G alpha(i2) that interacts specifically with G protein-coupled receptors. GiCT inhibits Gi signals specifically in vitro and in vivo, whereas Gs and Gq signals are not affected. In vivo expression of GiCT in transgenic mice effectively causes a "functional knockout" of cardiac G alpha(i2) signaling. Inducible, cardiac-specific GiCT transgenic mice display a baseline phenotype consistent with nontransgenic mice. However, when subjected to ischemia/reperfusion injury, GiCT transgenic mice demonstrate a significant increase in infarct size compared with nontransgenic mice (from 36.9+/-2.5% to 50.9+/-4.3%). Mechanistically, this post-ischemia/reperfusion phenotype includes increased myocardial apoptosis and resultant decreased contractile performance. CONCLUSIONS: Overall, our results demonstrate the in vivo utility of GiCT to dissect specific mechanisms attributed to Gi signaling in stressed myocardium. Our results with GiCT indicate that upregulation of G alpha(i2) is an adaptive protective response after ischemia to shield myocytes from apoptosis.


Subject(s)
Apoptosis/physiology , GTP-Binding Protein alpha Subunit, Gi2/physiology , Mitochondria, Heart/physiology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Cells, Cultured/metabolism , GTP-Binding Protein alpha Subunit, Gi2/antagonists & inhibitors , GTP-Binding Protein alpha Subunit, Gi2/chemistry , GTP-Binding Protein alpha Subunit, Gi2/genetics , Heart Failure/etiology , Heart Failure/physiopathology , Heart Failure/prevention & control , Humans , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Oxidative Stress , Peptide Fragments/genetics , Rats , Receptors, G-Protein-Coupled/physiology , Recombinant Fusion Proteins/physiology , Signal Transduction/physiology , Transduction, Genetic
17.
Circ Res ; 102(7): 786-94, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18292599

ABSTRACT

S100A1, a Ca(2+)-binding protein of the EF-hand type, is known to modulate sarcoplasmic reticulum Ca(2+) handling in skeletal muscle and cardiomyocytes. Recently, S100A1 has been shown to be expressed in endothelial cells (ECs). Because intracellular Ca(2+) ([Ca(2+)](i)) transients can be involved in important EC functions and endothelial NO synthase activity, we sought to investigate the impact of endothelial S100A1 on the regulation of endothelial and vascular function. Thoracic aortas from S100A1 knockout mice (SKO) showed significantly reduced relaxation in response to acetylcholine compared with wild-type vessels, whereas direct vessel relaxation using sodium nitroprusside was unaltered. Endothelial dysfunction attributable to the lack of S100A1 expression could also be demonstrated in vivo and translated into hypertension of SKO. Mechanistically, both basal and acetylcholine-induced endothelial NO release of SKO aortas was significantly reduced compared with wild type. Impaired endothelial NO production in SKO could be attributed, at least in part, to diminished agonist-induced [Ca(2+)](i) transients in ECs. Consistently, silencing endothelial S100A1 expression in wild type also reduced [Ca(2+)](i) and NO generation. Moreover, S100A1 overexpression in ECs further increased NO generation that was blocked by the inositol-1,4,5-triphosphate receptor blocker 2-aminoethoxydiphenylborate. Finally, cardiac endothelial S100A1 expression was shown to be downregulated in heart failure in vivo. Collectively, endothelial S100A1 critically modulates vascular function because lack of S100A1 expression leads to decreased [Ca(2+)](i) and endothelial NO release, which contributes, at least partially, to impaired endothelium-dependent vascular relaxation and hypertension in SKO mice. Targeting endothelial S100A1 expression may, therefore, be a novel therapeutic means to improve endothelial function in vascular disease or heart failure.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , S100 Proteins/metabolism , Vasoconstriction/physiology , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Calcium/metabolism , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertension/metabolism , Hypertension/pathology , Hypertension/physiopathology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitroprusside/pharmacology , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL