Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nature ; 587(7834): 460-465, 2020 11.
Article in English | MEDLINE | ID: mdl-33149301

ABSTRACT

Atrial fibrillation, the most common cardiac arrhythmia, is an important contributor to mortality and morbidity, and particularly to the risk of stroke in humans1. Atrial-tissue fibrosis is a central pathophysiological feature of atrial fibrillation that also hampers its treatment; the underlying molecular mechanisms are poorly understood and warrant investigation given the inadequacy of present therapies2. Here we show that calcitonin, a hormone product of the thyroid gland involved in bone metabolism3, is also produced by atrial cardiomyocytes in substantial quantities and acts as a paracrine signal that affects neighbouring collagen-producing fibroblasts to control their proliferation and secretion of extracellular matrix proteins. Global disruption of calcitonin receptor signalling in mice causes atrial fibrosis and increases susceptibility to atrial fibrillation. In mice in which liver kinase B1 is knocked down specifically in the atria, atrial-specific knockdown of calcitonin promotes atrial fibrosis and increases and prolongs spontaneous episodes of atrial fibrillation, whereas atrial-specific overexpression of calcitonin prevents both atrial fibrosis and fibrillation. Human patients with persistent atrial fibrillation show sixfold lower levels of myocardial calcitonin compared to control individuals with normal heart rhythm, with loss of calcitonin receptors in the fibroblast membrane. Although transcriptome analysis of human atrial fibroblasts reveals little change after exposure to calcitonin, proteomic analysis shows extensive alterations in extracellular matrix proteins and pathways related to fibrogenesis, infection and immune responses, and transcriptional regulation. Strategies to restore disrupted myocardial calcitonin signalling thus may offer therapeutic avenues for patients with atrial fibrillation.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcitonin/metabolism , Fibrinogen/biosynthesis , Heart Atria/metabolism , Myocardium/metabolism , Paracrine Communication , Animals , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Atrial Fibrillation , Collagen Type I/metabolism , Female , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Heart Atria/cytology , Heart Atria/pathology , Heart Atria/physiopathology , Humans , Male , Mice , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Receptors, Calcitonin/metabolism
2.
Int J Mol Sci ; 24(20)2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37894719

ABSTRACT

NTPDase1/CD39, the major vascular ectonucleotidase, exerts thrombo-immunoregulatory function by controlling endothelial P2 receptor activation. Despite the well-described release of ATP from endothelial cells, few data are available regarding the potential role of CD39 as a regulator of arterial diameter. We thus investigated the contribution of CD39 in short-term diameter adaptation and long-term arterial remodeling in response to flow using Entpd1-/- male mice. Compared to wild-type littermates, endothelial-dependent relaxation was modified in Entpd1-/- mice. Specifically, the vasorelaxation in response to ATP was potentiated in both conductance (aorta) and small resistance (mesenteric and coronary) arteries. By contrast, the relaxing responses to acetylcholine were supra-normalized in thoracic aortas while decreased in resistance arteries from Entpd1-/- mice. Acute flow-mediated dilation, measured via pressure myography, was dramatically diminished and outward remodeling induced by in vivo chronic increased shear stress was altered in the mesenteric resistance arteries isolated from Entpd1-/- mice compared to wild-types. Finally, changes in vascular reactivity in Entpd1-/- mice were also evidenced by a decrease in the coronary output measured in isolated perfused hearts compared to the wild-type mice. Our results highlight a key regulatory role for purinergic signaling and CD39 in endothelium-dependent short- and long-term arterial diameter adaptation to increased flow.


Subject(s)
Adenosine Triphosphate , Endothelial Cells , Male , Animals , Mice , Antigens, CD/genetics , Apyrase/physiology , Vasodilation , Endothelium, Vascular
3.
Circulation ; 138(16): 1677-1692, 2018 10 16.
Article in English | MEDLINE | ID: mdl-29674325

ABSTRACT

BACKGROUND: Pharmacogenomic studies have shown that ADCY9 genotype determines the effects of the CETP (cholesteryl ester transfer protein) inhibitor dalcetrapib on cardiovascular events and atherosclerosis imaging. The underlying mechanisms responsible for the interactions between ADCY9 and CETP activity have not yet been determined. METHODS: Adcy9-inactivated ( Adcy9Gt/Gt) and wild-type (WT) mice, that were or not transgenic for the CETP gene (CETPtg Adcy9Gt/Gt and CETPtg Adcy9WT), were submitted to an atherogenic protocol (injection of an AAV8 [adeno-associated virus serotype 8] expressing a PCSK9 [proprotein convertase subtilisin/kexin type 9] gain-of-function variant and 0.75% cholesterol diet for 16 weeks). Atherosclerosis, vasorelaxation, telemetry, and adipose tissue magnetic resonance imaging were evaluated. RESULTS: Adcy9Gt/Gt mice had a 65% reduction in aortic atherosclerosis compared to WT ( P<0.01). CD68 (cluster of differentiation 68)-positive macrophage accumulation and proliferation in plaques were reduced in Adcy9Gt/Gt mice compared to WT animals ( P<0.05 for both). Femoral artery endothelial-dependent vasorelaxation was improved in Adcy9Gt/Gt mice (versus WT, P<0.01). Selective pharmacological blockade showed that the nitric oxide, cyclooxygenase, and endothelial-dependent hyperpolarization pathways were all responsible for the improvement of vasodilatation in Adcy9Gt/Gt ( P<0.01 for all). Aortic endothelium from Adcy9Gt/Gt mice allowed significantly less adhesion of splenocytes compared to WT ( P<0.05). Adcy9Gt/Gt mice gained more weight than WT with the atherogenic diet; this was associated with an increase in whole body adipose tissue volume ( P<0.01 for both). Feed efficiency was increased in Adcy9Gt/Gt compared to WT mice ( P<0.01), which was accompanied by prolonged cardiac RR interval ( P<0.05) and improved nocturnal heart rate variability ( P=0.0572). Adcy9 inactivation-induced effects on atherosclerosis, endothelial function, weight gain, adipose tissue volume, and feed efficiency were lost in CETPtg Adcy9Gt/Gt mice ( P>0.05 versus CETPtg Adcy9WT). CONCLUSIONS: Adcy9 inactivation protects against atherosclerosis, but only in the absence of CETP activity. This atheroprotection may be explained by decreased macrophage accumulation and proliferation in the arterial wall, and improved endothelial function and autonomic tone.


Subject(s)
Adenylyl Cyclases/deficiency , Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Cholesterol Ester Transfer Proteins/deficiency , Plaque, Atherosclerotic , Adenylyl Cyclases/genetics , Adiposity , Animals , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Autonomic Nervous System/physiopathology , Biological Factors/metabolism , Cell Proliferation , Cholesterol Ester Transfer Proteins/genetics , Diet, High-Fat , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/pathology , Lipids/blood , Lipolysis , Macrophages/enzymology , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Proprotein Convertase 9/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Signal Transduction , Vasodilation , Weight Gain
4.
Am J Physiol Heart Circ Physiol ; 316(6): H1281-H1296, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30901279

ABSTRACT

MK5 is a protein serine/threonine kinase activated by p38, ERK3, and ERK4 MAPKs. MK5 mRNA and immunoreactivity are detected in mouse cardiac fibroblasts, and MK5 haplodeficiency attenuates the increase in collagen 1-α1 mRNA evoked by pressure overload. The present study examined the effect of MK5 haplodeficiency on reparative fibrosis following myocardial infarction (MI). Twelve-week-old MK5+/- and wild-type littermate (MK5+/+) mice underwent ligation of the left anterior descending coronary artery (LADL). Surviving mice were euthanized 8 or 21 days post-MI. Survival rates did not differ significantly between MK5+/+ and MK5+/- mice, with rupture of the LV wall being the primary cause of death. Echocardiographic imaging revealed similar increases in LV end-diastolic diameter, myocardial performance index, and wall motion score index in LADL-MK5+/+ and LADL-MK5+/- mice. Area at risk did not differ between LADL-MK5+/+ and LADL-MK5+/- hearts. In contrast, infarct size, scar area, and scar collagen content were reduced in LADL-MK5+/- hearts. Immunohistochemical analysis of mice experiencing heart rupture revealed increased MMP-9 immunoreactivity in the infarct border zone of LADL-MK5+/- hearts compared with LADL-MK5+/+. Although inflammatory cell infiltration was similar in LADL-MK5+/+ and LADL-MK5+/- hearts, angiogenesis was more pronounced in the infarct border zone of LADL-MK5+/- mice. Characterization of ventricular fibroblasts revealed reduced motility and proliferation in fibroblasts isolated from MK5-/- mice compared with those from both wild-type and haplodeficient mice. siRNA-mediated knockdown of MK5 in fibroblasts from wild-type mice also impaired motility. Hence, reduced MK5 expression alters fibroblast function and scar morphology but not mortality post-MI. NEW & NOTEWORTHY MK5/PRAK is a protein serine/threonine kinase activated by p38 MAPK and/or atypical MAPKs ERK3/4. MK5 haplodeficiency reduced infarct size, scar area, and scar collagen content post-myocardial infarction. Motility and proliferation were reduced in cultured MK5-null cardiac myofibroblasts.


Subject(s)
Cicatrix/enzymology , Collagen/metabolism , Haploinsufficiency , Intracellular Signaling Peptides and Proteins/deficiency , Myocardial Infarction/enzymology , Myocardium/enzymology , Myofibroblasts/enzymology , Protein Serine-Threonine Kinases/deficiency , Wound Healing , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Cicatrix/pathology , Cicatrix/physiopathology , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/genetics , Male , Matrix Metalloproteinase 9/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myofibroblasts/pathology , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Ventricular Function, Left , Ventricular Remodeling
5.
Am J Physiol Heart Circ Physiol ; 313(1): H46-H58, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28432058

ABSTRACT

MAPK-activated protein kinase-5 (MK5) is a protein serine/threonine kinase that is activated by p38 MAPK and the atypical MAPKs ERK3 and ERK4. The physiological function(s) of MK5 remains unknown. Here, we examined the effect of MK5 haplodeficiency on cardiac function and myocardial remodeling. At 12 wk of age, MK5 haplodeficient mice (MK5+/-) were smaller than age-matched wild-type littermates (MK5+/+), with similar diastolic function but reduced systolic function. Transverse aortic constriction (TAC) was used to induce chronic pressure overload in 12-wk-old male MK5+/- and MK5+/+ mice. Two weeks post-TAC, heart weight-to-tibia length ratios were similarly increased in MK5+/- and MK5+/+ hearts, as was the abundance of B-type natriuretic peptide and ß-myosin heavy chain mRNA. Left ventricular ejection fraction was reduced in both MK5+/+ and MK5+/- mice, whereas regional peak systolic tissue velocities were reduced and isovolumetric relaxation time was prolonged in MK5+/+ hearts but not in MK5+/- hearts. The TAC-induced increase in collagen type 1-α1 mRNA observed in MK5+/+ hearts was markedly attenuated in MK5+/- hearts. Eight weeks post-TAC, systolic function was equally impaired in MK5+/+ and MK5+/- mice. In contrast, the increase in E wave deceleration rate and progression of hypertrophy observed in TAC MK5+/+ mice were attenuated in TAC MK5+/- mice. MK5 immunoreactivity was detected in adult fibroblasts but not in myocytes. MK5+/+, MK5+/-, and MK5-/- fibroblasts all expressed α-smooth muscle actin in culture. Hence, reduced MK5 expression in cardiac fibroblasts was associated with the attenuation of both hypertrophy and development of a restrictive filling pattern during myocardial remodeling in response to chronic pressure overload.NEW & NOTEWORTHY MAPK-activated protein kinase-5 (MK5)/p38-regulated/activated protein kinase is a protein serine/threonine kinase activated by p38 MAPK and/or the atypical MAPKs ERK3 and ERK4. MK5 immunoreactivity was detected in adult ventricular fibroblasts but not in myocytes. MK5 haplodeficiency attenuated the progression of hypertrophy, reduced collagen type 1 mRNA, and protected diastolic function in response to chronic pressure overload.


Subject(s)
Hypertrophy, Left Ventricular/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling/physiology , Animals , Haplotypes/genetics , Hypertrophy, Left Ventricular/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Stroke Volume , Ventricular Dysfunction, Left/complications
6.
J Transl Med ; 13: 142, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25948279

ABSTRACT

BACKGROUND: The role of endothelial progenitor cells (EPCs) in vascular repair is related to their recruitment at the sites of injury and their interaction with different components of the circulatory system. We have previously shown that EPCs bind and inhibit platelet function and impair thrombus formation via prostacyclin secretion, but the role of EPC binding to platelet P-selectin in this process has not been fully characterized. In the present study, we assessed the impact of EPCs on thrombus formation and we addressed the implication of P-selectin in this process. METHODS: EPCs were generated from human peripheral blood mononuclear cells cultured on fibronectin in conditioned media. The impact of EPCs on platelet aggregation and thrombus formation was investigated in P-selectin deficient (P-sel(-/-)) mice and their wild-type (WT) counterparts. RESULTS: EPCs significantly and dose-dependently impaired collagen-induced whole blood platelet aggregation in WT mice, whereas no effects were observed in P-sel(-/-) mice. Moreover, in a ferric chloride-induced arterial thrombosis model, infusion of EPCs significantly reduced thrombus formation in WT, but not in P-sel(-/-) mice. Furthermore, the relative mass of thrombi generated in EPC-treated P-sel(-/-) mice were significantly larger than those in EPC-treated WT mice, and the number of EPCs recruited within the thrombi and along the arterial wall was reduced in P-sel(-/-) mice as compared to WT mice. CONCLUSION: This study shows that EPCs impair platelet aggregation and reduce thrombus formation via a cellular mechanism involving binding to platelet P-selectin. These findings add new insights into the role of EPC-platelet interactions in the regulation of thrombotic events during vascular repair.


Subject(s)
Blood Platelets/metabolism , Endothelial Progenitor Cells/cytology , Gene Expression Regulation , P-Selectin/genetics , Adult , Animals , Carotid Arteries/pathology , Female , Humans , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Middle Aged , P-Selectin/metabolism , Phenotype , Platelet Aggregation , Protein Binding , Thrombosis/metabolism , Thrombosis/pathology , Young Adult
7.
Front Physiol ; 15: 1320065, 2024.
Article in English | MEDLINE | ID: mdl-38426206

ABSTRACT

Background: Angiopoietin-like 2 (ANGPTL2) is a pro-inflammatory and pro-oxidant circulating protein that predicts and promotes chronic inflammatory diseases such as atherosclerosis in humans. Transgenic murine models demonstrated the deleterious role of ANGPTL2 in vascular diseases, while deletion of ANGPTL2 was protective. The nature of its role in cardiac tissues is, however, less clear. Indeed, in adult mice knocked down (KD) for ANGPTL2, we recently reported a mild left ventricular (LV) dysfunction originating from a congenital aortic valve stenosis, demonstrating that ANGPTL2 is essential to cardiac development and function. Hypothesis: Because we originally demonstrated that the KD of ANGPTL2 protected vascular endothelial function via an upregulation of arterial NOX4, promoting the beneficial production of dilatory H2O2, we tested the hypothesis that increased cardiac NOX4 could negatively affect cardiac redox and remodeling and contribute to LV dysfunction observed in adult Angptl2-KD mice. Methods and results: Cardiac expression and activity of NOX4 were higher in KD mice, promoting higher levels of cardiac H2O2 when compared to wild-type (WT) mice. Immunofluorescence showed that ANGPTL2 and NOX4 were co-expressed in cardiac cells from WT mice and both proteins co-immunoprecipitated in HEK293 cells, suggesting that ANGPTL2 and NOX4 physically interact. Pressure overload induced by transverse aortic constriction surgery (TAC) promoted LV systolic dysfunction in WT mice but did not further exacerbate the dysfunction in KD mice. Importantly, the severity of LV systolic dysfunction in KD mice (TAC and control SHAM) correlated with cardiac Nox4 expression. Injection of an adeno-associated virus (AAV9) delivering shRNA targeting cardiac Nox4 expression fully reversed LV systolic dysfunction in KD-SHAM mice, demonstrating the causal role of NOX4 in cardiac dysfunction in KD mice. Targeting cardiac Nox4 expression in KD mice also induced an antioxidant response characterized by increased expression of NRF2/KEAP1 and catalase. Conclusion: Together, these data reveal that the absence of ANGPTL2 induces an upregulation of cardiac NOX4 that contributes to oxidative stress and LV dysfunction. By interacting and repressing cardiac NOX4, ANGPTL2 could play a new beneficial role in the maintenance of cardiac redox homeostasis and function.

8.
Physiol Rep ; 12(11): e16108, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872461

ABSTRACT

ERK3/MAPK6 activates MAP kinase-activated protein kinase (MK)-5 in selected cell types. Male MK5 haplodeficient mice show reduced hypertrophy and attenuated increase in Col1a1 mRNA in response to increased cardiac afterload. In addition, MK5 deficiency impairs cardiac fibroblast function. This study determined the effect of reduced ERK3 on cardiac hypertrophy following transverse aortic constriction (TAC) and fibroblast biology in male mice. Three weeks post-surgery, ERK3, but not ERK4 or p38α, co-immunoprecipitated with MK5 from both sham and TAC heart lysates. The increase in left ventricular mass and myocyte diameter was lower in TAC-ERK3+/- than TAC-ERK3+/+ hearts, whereas ERK3 haploinsufficiency did not alter systolic or diastolic function. Furthermore, the TAC-induced increase in Col1a1 mRNA abundance was diminished in ERK3+/- hearts. ERK3 immunoreactivity was detected in atrial and ventricular fibroblasts but not myocytes. In both quiescent fibroblasts and "activated" myofibroblasts isolated from adult mouse heart, siRNA-mediated knockdown of ERK3 reduced the TGF-ß-induced increase in Col1a1 mRNA. In addition, intracellular type 1 collagen immunoreactivity was reduced following ERK3 depletion in quiescent fibroblasts but not myofibroblasts. Finally, knocking down ERK3 impaired motility in both atrial and ventricular myofibroblasts. These results suggest that ERK3 plays an important role in multiple aspects of cardiac fibroblast biology.


Subject(s)
Fibroblasts , Animals , Male , Mice , Fibroblasts/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain/metabolism , Myocardium/metabolism , Myocardium/cytology , Mitogen-Activated Protein Kinase 6/metabolism , Mitogen-Activated Protein Kinase 6/genetics , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Cells, Cultured , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/genetics , Myocytes, Cardiac/metabolism
9.
J Mol Cell Cardiol ; 62: 189-202, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23756157

ABSTRACT

Endothelin receptors are present on the nuclear membranes in adult cardiac ventricular myocytes. The objectives of the present study were to determine 1) which endothelin receptor subtype is in cardiac nuclear membranes, 2) if the receptor and ligand traffic from the cell surface to the nucleus, and 3) the effect of increased intracellular ET-1 on nuclear Ca(2+) signaling. Confocal microscopy using fluorescently-labeled endothelin analogs confirmed the presence of ETB at the nuclear membrane of rat cardiomyocytes in skinned-cells and isolated nuclei. Furthermore, in both cardiac myocytes and aortic endothelial cells, endocytosed ET:ETB complexes translocated to lysosomes and not the nuclear envelope. Although ETA and ETB can form heterodimers, the presence or absence of ETA did not alter ETB trafficking. Treatment of isolated nuclei with peptide: N-glycosidase F did not alter the electrophoretic mobility of ETB. The absence of N-glycosylation further indicates that these receptors did not originate at the cell surface. Intracellular photolysis of a caged ET-1 analog ([Trp-ODMNB(21)]ET-1) evoked an increase in nucleoplasmic Ca(2+) ([Ca(2+)]n) that was attenuated by inositol 1,4,5-trisphosphate receptor inhibitor 2-aminoethoxydiphenyl borate and prevented by pre-treatment with ryanodine. A caged cell-permeable analog of the ETB-selective antagonist IRL-2500 blocked the ability of intracellular cET-1 to increase [Ca(2+)]n whereas extracellular application of ETA and ETB receptor antagonists did not. These data suggest that 1) the endothelin receptor in the cardiac nuclear membranes is ETB, 2) ETB traffics directly to the nuclear membrane after biosynthesis, 3) exogenous endothelins are not ligands for ETB on nuclear membranes, and 4) ETB associated with the nuclear membranes regulates nuclear Ca(2+) signaling.


Subject(s)
Calcium/metabolism , Endothelins/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Cardiac/metabolism , Animals , Aorta/cytology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Immunoblotting , Immunoprecipitation , Microscopy, Confocal , Myocytes, Cardiac/drug effects , Nuclear Envelope/metabolism , Rats , Receptors, Endothelin/metabolism , Ryanodine/pharmacology
10.
Can J Cardiol ; 39(7): 952-962, 2023 07.
Article in English | MEDLINE | ID: mdl-37054880

ABSTRACT

BACKGROUND: Polymorphisms in the adenylate cyclase 9 (ADCY9) gene influence the benefits of the cholesteryl ester transfer protein (CETP) modulator dalcetrapib on cardiovascular events after acute coronary syndrome. We hypothesized that Adcy9 inactivation could improve cardiac function and remodelling following myocardial infarction (MI) in absence of CETP activity. METHODS: Wild-type (WT) and Adcy9-inactivated (Adcy9Gt/Gt) male mice, transgenic or not for human CETP (tgCETP+/-), were subjected to MI by permanent left anterior descending coronary artery ligation and studied for 4 weeks. Left ventricular (LV) function was assessed by echocardiography at baseline, 1, and 4 weeks after MI. At sacrifice, blood, spleen and bone marrow cells were collected for flow cytometry analysis, and hearts were harvested for histologic analyses. RESULTS: All mice developed LV hypertrophy, dilation, and systolic dysfunction, but Adcy9Gt/Gt mice exhibited reduced pathologic LV remodelling and better LV function compared with WT mice. There were no differences between tgCETP+/- and Adcy9Gt/Gt tgCETP+/- mice, which both exhibited intermediate responses. Histologic analyses showed smaller cardiomyocyte size, reduced infarct size, and preserved myocardial capillary density in the infarct border zone in Adcy9Gt/Gt vs WT mice. Count of bone marrow T cells and B cells were significantly increased in Adcy9Gt/Gt mice compared with the other genotypes. CONCLUSIONS: Adcy9 inactivation reduced infarct size, pathologic remodelling, and cardiac dysfunction. These changes were accompanied by preserved myocardial capillary density and increased adaptive immune response. Most of the benefits of Adcy9 inactivation were only observed in the absence of CETP.


Subject(s)
Myocardial Infarction , Animals , Humans , Male , Mice , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Myocardial Infarction/complications , Myocardium/pathology , Myocytes, Cardiac/metabolism , Ventricular Remodeling/physiology
11.
J Cell Physiol ; 226(5): 1157-65, 2011 May.
Article in English | MEDLINE | ID: mdl-20945344

ABSTRACT

Recent studies have revealed the existence of multipotent nestin-immunoreactive cells in the adult mammalian heart. These cells were recruited to infarct site following ischemic injury and differentiated to a vascular lineage leading to de novo blood vessel formation. Here, we show that a sub-population of cardiac resident nestin((+)) cells can further differentiate to a neuronal-like fate in vivo following myocardial infarction. In the ischemically damaged rat heart, neurofilament-M((+)) fibres were detected innervating the peri-infarct/infarct region and the preponderance of these fibres were physically associated with processes emanating from nestin((+)) cells. One week after isogenic heterotopic cardiac transplantation, the beating transplanted rat heart was devoid of neurofilament-M((+)) fibre staining. The superimposition of an ischemic insult to the transplanted heart led to the de novo synthesis of neurofilament-M((+)) fibres by cardiac resident nestin((+)) cells. Nerve growth factor infusion and the exposure of normal rats to intermittent hypoxia significantly increased the density of neurofilament-M((+)) fibres in the heart. However, these newly formed neurofilament-M((+)) fibres were not physically associated with nestin((+)) processes. These data highlight a novel paradigm of reparative fibrosis as a subpopulation of cardiac resident nestin((+)) cells directly contributed to neural remodelling of the peri-infarct/infarct region of the ischemically damaged rat heart via the de novo synthesis of neurofilament-M fibres.


Subject(s)
Adrenergic Fibers/pathology , Adult Stem Cells/pathology , Cell Differentiation , Heart/innervation , Intermediate Filament Proteins/metabolism , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Nerve Tissue Proteins/metabolism , Neurons/pathology , Adrenergic Fibers/drug effects , Adrenergic Fibers/metabolism , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Cell Lineage , Disease Models, Animal , Fibrosis , Heart Transplantation , Hypoxia/metabolism , Hypoxia/pathology , Intermediate Filament Proteins/genetics , Male , Myocardial Infarction/metabolism , Myocardial Ischemia/metabolism , Nerve Growth Factor/administration & dosage , Nerve Tissue Proteins/genetics , Nestin , Neurofilament Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidopamine/administration & dosage , Phenotype , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
12.
Pflugers Arch ; 462(3): 371-83, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21617900

ABSTRACT

We previously reported that in healthy mouse cerebral arteries, endothelial nitric oxide synthase (eNOS) produces H2O2, leading to endothelium-dependent dilation. In contrast, thromboxane A2 (TXA2), a potent pro-oxidant and pro-inflammatory endogenous vasoconstrictor, is associated with eNOS dysfunction. Our objectives were to elucidate whether (1) the cerebrovascular eNOS-H2O2 pathway was sensitive to oxidative stress associated with aging and dyslipidemia and (2) TXA2 contributed to cerebral eNOS dysfunction. Atherosclerotic (ATX = LDLR(-/-); hApoB(+/+)) and wild-type (WT) control mice were used at 3 and 12 months old (m/o). Three-m/o ATX mice were treated with the cardio-protective polyphenol catechin for 9 months. Dilations to ACh and the simultaneous eNOS-derived H2O2 production were recorded in isolated pressurized cerebral arteries. The age-associated decrease in cerebral eNOS-H2O2 pathway observed in WT was premature in ATX mice, decreasing at 3 m/o and abolished at 12 m/o. Thromboxane synthase inhibition by furegrelate increased dilations at 12 months in WT and at 3 and 12 months in ATX mice, suggesting an anti-dilatory role of TXA2 with age hastened by dyslipidemia. In addition, the non-selective NADP(H) oxidase inhibitor apocynin improved the eNOS-H2O2 pathway only in 12-m/o ATX mice. Catechin normalized the function of this pathway, which became sensitive to L-NNA and insensitive to furegrelate or apocynin; catechin also prevented the rise in TXA2 synthase expression. In conclusion, the age-dependent cerebral endothelial dysfunction is precocious in dyslipidemia and involves TXA2 production that limits eNOS activity. Preventive catechin treatment reduced the impact of endogenous TXA2 on the control of cerebral tone and maintained eNOS function.


Subject(s)
Aging/physiology , Atherosclerosis/enzymology , Cerebral Arteries/enzymology , Nitric Oxide Synthase Type III/metabolism , Thromboxane A2/metabolism , Up-Regulation , Acetophenones/metabolism , Animals , Antioxidants/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Benzofurans/metabolism , Catechin/therapeutic use , Cerebral Arteries/pathology , Enzyme Inhibitors/metabolism , Hemodynamics , Humans , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidants/metabolism , Oxidative Stress , Receptors, LDL/genetics , Receptors, LDL/metabolism , Thromboxane A2/genetics , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxane-A Synthase/metabolism
13.
Am J Physiol Heart Circ Physiol ; 301(4): H1706-15, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21803942

ABSTRACT

Calcium flux into and out of the sarco(endo)plasmic reticulum is vitally important to cardiac function because the cycle of calcium entry and exit controls contraction and relaxation. Putative estrogen and androgen consensus binding sites near to a CpG island are present in the cardiac calsequestrin 2 (CSQ2) promoter. Cardiomyocytes express sex hormone receptors and respond to sex hormones. We hypothesized that sex hormones control CSQ2 expression in cardiomyocytes and so affect cardiac structure/function. Echocardiographic analysis of male and female C57bl6n mice identified thinner walled and lighter hearts in females and significant concentric remodeling after long-term gonadectomy. CSQ2 and sodium-calcium exchanger-1 (NCX1) expression was significantly increased in female compared with male hearts and decreased postovariectomy. NCX1, but not CSQ2, expression was increased postcastration. CSQ2 expression was reduced when H9c2 cells were cultured in hormone-deficient media; increased when estrogen receptor-α (ERα), estrogen receptor-ß (ERß), or androgen agonists were added; and increased in hearts from ERß-deficient mice. CSQ2 expression was reduced in mice fed a diet low in the methyl donor folic acid and in cells treated with 5-azadeoxycytidine suggesting an involvement of DNA methylation. DNA methylation in CpG in the CSQ2 CpG island was significantly different in males and females and was additionally changed postgonadectomy. Expression of DNA methyltransferases 1, 3a, and 3b was unchanged. These studies strongly link sex hormone-directed changes in CSQ2 expression to DNA methylation with changed expression correlated with altered left ventricular structure and function.


Subject(s)
Gonadal Steroid Hormones/physiology , Ventricular Function, Left/physiology , Animals , Base Sequence , Calcium/physiology , Calsequestrin/genetics , Calsequestrin/physiology , Cell Line , DNA Methylation , Echocardiography , Estrogen Receptor beta/genetics , Estrogen Receptor beta/physiology , Female , Gene Expression Regulation , Gonadal Steroid Hormones/biosynthesis , Gonadal Steroid Hormones/genetics , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myocardium/cytology , Orchiectomy , Ovariectomy , Sodium-Calcium Exchanger/genetics
14.
Am J Physiol Heart Circ Physiol ; 301(3): H813-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21685264

ABSTRACT

Patients with very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency frequently present cardiomyopathy and heartbeat disorders. However, the underlying factors, which may be of cardiac or extra cardiac origins, remain to be elucidated. In this study, we tested for metabolic and functional alterations in the heart from 3- and 7-mo-old VLCAD null mice and their littermate counterparts, using validated experimental paradigms, namely, 1) ex vivo perfusion in working mode, with concomitant evaluation of myocardial contractility and metabolic fluxes using (13)C-labeled substrates under various conditions; as well as 2) in vivo targeted lipidomics, gene expression analysis as well as electrocardiogram monitoring by telemetry in mice fed various diets. Unexpectedly, when perfused ex vivo, working VLCAD null mouse hearts maintained values similar to those of the controls for functional parameters and for the contribution of exogenous palmitate to ß-oxidation (energy production), even at high palmitate concentration (1 mM) and increased energy demand (with 1 µM epinephrine) or after fasting. However, in vivo, these hearts displayed a prolonged rate-corrected QT (QTc) interval under all conditions examined, as well as the following lipid alterations: 1) age- and condition-dependent accumulation of triglycerides, and 2) 20% lower docosahexaenoic acid (an omega-3 polyunsaturated fatty acid) in membrane phospholipids. The latter was independent of liver but affected by feeding a diet enriched in saturated fat (exacerbated) or fish oil (attenuated). Our finding of a longer QTc interval in VLCAD null mice appears to be most relevant given that such condition increases the risk of sudden cardiac death.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Heart Conduction System/physiopathology , Lipid Metabolism/genetics , Long QT Syndrome/enzymology , Metabolism, Inborn Errors/enzymology , Mitochondrial Diseases/enzymology , Muscular Diseases/enzymology , Myocardium/enzymology , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Age Factors , Aging , Analysis of Variance , Animals , Congenital Bone Marrow Failure Syndromes , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Electrocardiography, Ambulatory , Fish Oils/administration & dosage , Fish Oils/metabolism , Lipid Metabolism, Inborn Errors , Liver/metabolism , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Long QT Syndrome/prevention & control , Male , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases/complications , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Muscular Diseases/complications , Muscular Diseases/genetics , Muscular Diseases/physiopathology , Myocardial Contraction , Oxidation-Reduction , Palmitic Acid/metabolism , Perfusion , Telemetry , Triglycerides/metabolism
15.
Am J Physiol Heart Circ Physiol ; 300(3): H1032-43, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21186270

ABSTRACT

Severe dyslipidemia and the associated oxidative stress could accelerate the age-related decline in cerebrovascular endothelial function and cerebral blood flow (CBF), leading to neuronal loss and impaired learning abilities. We hypothesized that a chronic treatment with the polyphenol catechin would prevent endothelial dysfunction, maintain CBF responses, and protect learning abilities in atherosclerotic (ATX) mice. We treated ATX (C57Bl/6-LDLR(-/-)hApoB(+/+); 3 mo old) mice with catechin (30 mg · kg(-1) · day(-1)) for 3 mo, and C57Bl/6 [wild type (WT), 3 and 6 mo old] mice were used as controls. ACh- and flow-mediated dilations (FMD) were recorded in pressurized cerebral arteries. Basal CBF and increases in CBF induced by whisker stimulation were measured by optical coherence tomography and Doppler, respectively. Learning capacities were evaluated with the Morris water maze test. Compared with 6-mo-old WT mice, cerebral arteries from 6-mo-old ATX mice displayed a higher myogenic tone, lower responses to ACh and FMD, and were insensitive to NOS inhibition (P < 0.05), suggesting endothelial dysfunction. Basal and increases in CBF were lower in 6-mo-old ATX than WT mice (P < 0.05). A decline in the learning capabilities was also observed in ATX mice (P < 0.05). Catechin 1) reduced cerebral superoxide staining (P < 0.05) in ATX mice, 2) restored endothelial function by reducing myogenic tone, improving ACh- and FMD and restoring the sensitivity to nitric oxide synthase inhibition (P < 0.05), 3) increased the changes in CBF during stimulation but not basal CBF, and 4) prevented the decline in learning abilities (P < 0.05). In conclusion, catechin treatment of ATX mice prevents cerebrovascular dysfunctions and the associated decline in learning capacities.


Subject(s)
Catechin/pharmacology , Cerebrovascular Circulation/drug effects , Intracranial Arteriosclerosis/drug therapy , Learning/drug effects , Vasodilation/drug effects , Vasodilator Agents/therapeutic use , Animals , Apolipoprotein B-100/deficiency , Apolipoprotein B-100/genetics , Blood Pressure/drug effects , Blood Pressure/genetics , Brain/blood supply , Brain/drug effects , Cerebral Arteries/drug effects , Cholesterol/blood , Heart Rate/drug effects , Heart Rate/genetics , Lipoproteins, LDL/blood , Lipoproteins, LDL/deficiency , Lipoproteins, LDL/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Triglycerides/blood
16.
Am J Physiol Heart Circ Physiol ; 301(5): H2081-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21926346

ABSTRACT

The cardiac cycle imposes a mechanical stress that dilates elastic carotid arteries, while shear stress largely contributes to the endothelium-dependent dilation of downstream cerebral arteries. In the presence of dyslipidemia, carotid arteries stiffen while the endothelial function declines. We reasoned that stiffening of carotid arteries would be prevented by reducing resting heart rate (HR), while improving the endothelial function would regulate cerebral artery compliance and function. Thus we treated or not 3-mo-old male atherosclerotic mice (ATX; LDLr(-/-):hApoB(+/+)) for 3 mo with the sinoatrial pacemaker current inhibitor ivabradine (IVA), the ß-blocker metoprolol (METO), or subjected mice to voluntary physical training (PT). Arterial (carotid and cerebral artery) compliance and endothelium-dependent flow-mediated cerebral dilation were measured in isolated pressurized arteries. IVA and METO similarly reduced (P < 0.05) 24-h HR by ≈15%, while PT had no impact. As expected, carotid artery stiffness increased (P < 0.05) in ATX mice compared with wild-type mice, while cerebral artery stiffness decreased (P < 0.05); this paradoxical increase in cerebrovascular compliance was associated with endothelial dysfunction and an augmented metalloproteinase-9 (MMP-9) activity (P < 0.05), without changing the lipid composition of the wall. Reducing HR (IVA and METO) limited carotid artery stiffening, but plaque progression was prevented by IVA only. In contrast, IVA maintained and PT improved cerebral endothelial nitric oxide synthase-dependent flow-mediated dilation and wall compliance, and both interventions reduced MMP-9 activity (P < 0.05); METO worsened endothelial dysfunction and compliance and did not reduce MMP-9 activity. In conclusion, HR-dependent mechanical stress contributes to carotid artery wall stiffening in severely dyslipidemic mice while cerebrovascular compliance is mostly regulated by the endothelium.


Subject(s)
Atherosclerosis/etiology , Carotid Arteries/physiopathology , Carotid Artery Diseases/etiology , Cerebral Arteries/physiopathology , Cerebrovascular Disorders/etiology , Dyslipidemias/complications , Endothelium, Vascular/physiopathology , Heart Rate , Animals , Anti-Arrhythmia Agents/pharmacology , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Atherosclerosis/therapy , Benzazepines/pharmacology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/physiopathology , Carotid Artery Diseases/prevention & control , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/metabolism , Cerebrovascular Disorders/physiopathology , Cerebrovascular Disorders/prevention & control , Compliance , Disease Models, Animal , Disease Progression , Dyslipidemias/genetics , Dyslipidemias/metabolism , Dyslipidemias/physiopathology , Dyslipidemias/therapy , Endothelium, Vascular/drug effects , Exercise Therapy , Genotype , Heart Rate/drug effects , Humans , Ivabradine , Male , Matrix Metalloproteinase 9/metabolism , Metoprolol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , Severity of Illness Index , Stress, Mechanical , Time Factors , Vasodilation
17.
Arterioscler Thromb Vasc Biol ; 30(12): 2424-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21071692

ABSTRACT

OBJECTIVE: CD40 ligand is a thromboinflammatory molecule that predicts cardiovascular events. Platelets constitute the major source of soluble CD40 ligand (sCD40L), which has been shown to influence platelet activation, although its exact functional impact on platelets and the underlying mechanisms remain undefined. We aimed to determine the impact and the signaling mechanisms of sCD40L on platelets. METHODS AND RESULTS: sCD40L strongly enhances platelet activation and aggregation. Human platelets treated with a mutated form of sCD40L that does not bind CD40, and CD40(-/-) mouse platelets failed to elicit such responses. Furthermore, sCD40L stimulation induces the association of the tumor necrosis factor receptor-associated factor-2 with platelet CD40. Notably, sCD40L primes platelets through activation of the small GTPase Rac1 and its downstream target p38 mitogen-activated protein kinase, which leads to platelet shape change and actin polymerization. Moreover, sCD40L exacerbates thrombus formation and leukocyte infiltration in wild-type mice but not in CD40(-/-) mice. CONCLUSIONS: sCD40L enhances agonist-induced platelet activation and aggregation through a CD40-dependent tumor necrosis factor receptor-associated factor-2/Rac1/p38 mitogen-activated protein kinase signaling pathway. Thus, sCD40L is an important platelet primer predisposing platelets to enhanced thrombus formation in response to vascular injury. This may explain the link between circulating levels of sCD40L and cardiovascular diseases.


Subject(s)
Blood Platelets/enzymology , CD40 Antigens/blood , CD40 Ligand/blood , Platelet Aggregation , Signal Transduction , TNF Receptor-Associated Factor 2/blood , Thrombosis/blood , p38 Mitogen-Activated Protein Kinases/blood , rac1 GTP-Binding Protein/blood , Actins/blood , Animals , Blood Platelets/immunology , CD40 Antigens/genetics , CD40 Ligand/genetics , Cell Shape , Disease Models, Animal , Female , Humans , Leukocytes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neuropeptides/blood , Recombinant Proteins/blood , Thrombosis/enzymology , Thrombosis/immunology , Time Factors , rac GTP-Binding Proteins/blood
18.
J Am Heart Assoc ; 10(4): e017791, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33533257

ABSTRACT

Background Mitogen-activated protein kinase-activated protein kinase-2 (MK2) is a protein serine/threonine kinase activated by p38α/ß. Herein, we examine the cardiac phenotype of pan MK2-null (MK2-/-) mice. Methods and Results Survival curves for male MK2+/+ and MK2-/- mice did not differ (Mantel-Cox test, P=0.580). At 12 weeks of age, MK2-/- mice exhibited normal systolic function along with signs of possible early diastolic dysfunction; however, aging was not associated with an abnormal reduction in diastolic function. Both R-R interval and P-R segment durations were prolonged in MK2-deficient mice. However, heart rates normalized when isolated hearts were perfused ex vivo in working mode. Ca2+ transients evoked by field stimulation or caffeine were similar in ventricular myocytes from MK2+/+ and MK2-/- mice. MK2-/- mice had lower body temperature and an age-dependent reduction in body weight. mRNA levels of key metabolic genes, including Ppargc1a, Acadm, Lipe, and Ucp3, were increased in hearts from MK2-/- mice. For equivalent respiration rates, mitochondria from MK2-/- hearts showed a significant decrease in Ca2+ sensitivity to mitochondrial permeability transition pore opening. Eight weeks of pressure overload increased left ventricular mass in MK2+/+ and MK2-/- mice; however, after 2 weeks the increase was significant in MK2+/+ but not MK2-/- mice. Finally, the pressure overload-induced decrease in systolic function was attenuated in MK2-/- mice 2 weeks, but not 8 weeks, after constriction of the transverse aorta. Conclusions Collectively, these results implicate MK2 in (1) autonomic regulation of heart rate, (2) cardiac mitochondrial function, and (3) the early stages of myocardial remodeling in response to chronic pressure overload.


Subject(s)
Blood Pressure/physiology , Bradycardia/physiopathology , Cardiomyopathy, Hypertrophic/physiopathology , Heart Rate/physiology , Mitochondria, Heart/metabolism , Ventricular Function, Left/physiology , Ventricular Remodeling , Animals , Bradycardia/diagnosis , Bradycardia/metabolism , Cardiomyopathy, Hypertrophic/diagnosis , Cardiomyopathy, Hypertrophic/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/deficiency
19.
Circulation ; 120(22): 2230-9, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19917882

ABSTRACT

BACKGROUND: Interactions of endothelial progenitor cells (EPCs) with vascular and blood cells contribute to vascular homeostasis. Although platelets promote the homing of EPCs to sites of vascular injury and their differentiation into endothelial cells, the functional consequences of such interactions on platelets remain unknown. Herein, we addressed the interactions between EPCs and platelets and their impact on platelet function and thrombus formation. METHODS AND RESULTS: Cultured on fibronectin in conditioned media, human peripheral blood mononuclear cells differentiated, within 10 days of culture, into EPCs, which uptake acetylated low-density lipoprotein, bind ulex-lectin, lack monocyte/leukocyte markers (CD14, P-selectin glycoprotein ligand-1, L-selectin), express progenitor/endothelial markers (CD34, vascular endothelial growth factor receptor-2, von Willebrand factor, and vascular endothelial cadherin), and proliferate in culture. These EPCs bound activated platelets via CD62P and inhibited its translocation, glycoprotein IIb/IIIa activation, aggregation, and adhesion to collagen, mainly via prostacyclin secretion. Indeed, this was associated with upregulation of cyclooxygenase-2 and inducible nitric oxide synthase. However, the effects on platelets in vitro were reversed by cyclooxygenase and cyclooxygenase-2 inhibition but not by nitric oxide or inducible nitric oxide synthase inhibition. Moreover, in a ferric chloride-induced murine arterial thrombosis model, injection of EPCs led to their incorporation into sites of injury and impaired thrombus formation, leading to an incomplete occlusion with 50% residual flow. CONCLUSIONS: Peripheral blood mononuclear cell-derived EPCs bind platelets via CD62P and inhibit platelet activation, aggregation, adhesion to collagen, and thrombus formation, predominantly via upregulation of cyclooxygenase-2 and secretion of prostacyclin. These findings add new insights into the biology of EPCs and define their potential roles in regulating platelet function and thrombosis.


Subject(s)
Blood Platelets/cytology , Blood Platelets/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Thrombosis/physiopathology , Carotid Artery Injuries/physiopathology , Cells, Cultured , Epoprostenol/metabolism , Hematopoietic Stem Cells/metabolism , Hemostasis/physiology , Humans , Leukocytes, Mononuclear/cytology , Nitric Oxide/metabolism , Platelet Adhesiveness/physiology , Platelet Aggregation/physiology , Regional Blood Flow
20.
Cardiology ; 117(3): 234-42, 2010.
Article in English | MEDLINE | ID: mdl-21212673

ABSTRACT

OBJECTIVES: To determine if heart rate (HR) reduction with ivabradine (IVA), a selective inhibitor of the pacemaker I(f) current, prevents cardiac dysfunction associated with dyslipidemia. METHODS: New Zealand White rabbits received either a standard diet, a 0.5% cholesterol-enriched diet only (CD), or a 0.5% CD with IVA (17 mg/kg/day) for 12 weeks. HR, left ventricular (LV) systolic function, diastolic function and LV regional myocardial performance index (MPI) were studied using echocardiography. Histological analysis included cardiac interstitial fibrosis and collagen type I fibers. Plasma levels of angiotensin II and aldosterone were quantified by immunoassays. RESULTS: IVA reduced HR by approximately 11%. IVA improved MPI and attenuated LV diastolic dysfunction (DD) (92% mild and 8% moderate DD with IVA vs. 54% mild and 46% moderate DD in CD group). IVA also reduced atrial fibrosis (p = 0.027), ventricular fibrosis (p = 0.0002) and ventricular collagen type I (p = 0.0042). IVA decreased plasma angiotensin II levels (p = 0.042), and both angiotensin II and aldosterone levels were correlated with HR (p = 0.038 and 0.008). CONCLUSION: Selective HR reduction with IVA reduces DD and cardiac fibrosis in hypercholesterolemic rabbits. These beneficial effects of IVA support testing pure HR reduction in patients with diastolic heart failure.


Subject(s)
Benzazepines/pharmacology , Cyclic Nucleotide-Gated Cation Channels/pharmacology , Heart Rate/drug effects , Hypercholesterolemia/physiopathology , Ventricular Dysfunction, Left/physiopathology , Aldosterone/blood , Angiotensin II/blood , Animals , Diastole/drug effects , Echocardiography, Doppler, Pulsed , Fibrosis , Heart Atria/pathology , Heart Ventricles/pathology , Hemodynamics , Ivabradine , Oxidative Stress , Rabbits , Sinoatrial Node/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL