Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(1): e51632, 2013.
Article in English | MEDLINE | ID: mdl-23308102

ABSTRACT

Autophagy is a bulk degradation mechanism for cytosolic proteins and organelles. The heart undergoes hypertrophy in response to mechanical load but hypertrophy can regress upon unloading. We hypothesize that autophagy plays an important role in mediating regression of cardiac hypertrophy during unloading. Mice were subjected to transverse aortic constriction (TAC) for 1 week, after which the constriction was removed (DeTAC). Regression of cardiac hypertrophy was observed after DeTAC, as indicated by reduction of LVW/BW and cardiomyocyte cross-sectional area. Indicators of autophagy, including LC3-II expression, p62 degradation and GFP-LC3 dots/cell, were significantly increased after DeTAC, suggesting that autophagy is induced. Stimulation of autophagy during DeTAC was accompanied by upregulation of FoxO1. Upregulation of FoxO1 and autophagy was also observed in vitro when cultured cardiomyocytes were subjected to mechanical stretch followed by incubation without stretch (de-stretch). Transgenic mice with cardiac-specific overexpression of FoxO1 exhibited smaller hearts and upregulation of autophagy. Overexpression of FoxO1 in cultured cardiomyocytes significantly reduced cell size, an effect which was attenuated when autophagy was inhibited. To further examine the role of autophagy and FoxO1 in mediating the regression of cardiac hypertrophy, beclin1+/- mice and cultured cardiomyocytes transduced with adenoviruses harboring shRNA-beclin1 or shRNA-FoxO1 were subjected to TAC/stretch followed by DeTAC/de-stretch. Regression of cardiac hypertrophy achieved after DeTAC/de-stretch was significantly attenuated when autophagy was suppressed through downregulation of beclin1 or FoxO1. These results suggest that autophagy and FoxO1 play an essential role in mediating regression of cardiac hypertrophy during mechanical unloading.


Subject(s)
Autophagy , Cardiomegaly/physiopathology , Heart/physiopathology , Animals , Autophagy/drug effects , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Size , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Up-Regulation , Weight-Bearing
2.
Curr Protoc Mouse Biol ; 1: 123-139, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21743842

ABSTRACT

The majority of current cardiovascular research involves studies in genetically engineered mouse models. The measurement of heart rate is central to understanding cardiovascular control under normal conditions, with altered autonomic tone, superimposed stress or disease states, both in wild type mice as well as those with altered genes. Electrocardiography (ECG) is the "gold standard" using either hard wire or telemetry transmission. In addition, heart rate is measured or monitored from the frequency of the arterial pressure pulse or cardiac contraction, or by pulse oximetry. For each of these techniques, discussions of materials and methods, as well as advantages and limitations are covered. However, only the direct ECG monitoring will determine not only the precise heart rates but also whether the cardiac rhythm is normal or not.

3.
Circulation ; 124(4): 406-15, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21747053

ABSTRACT

BACKGROUND: Cardiac overload, a major cause of heart failure, induces the expression of the heat shock protein H11 kinase/Hsp22 (Hsp22). METHODS AND RESULTS: To determine the specific function of Hsp22 in that context, a knockout mouse model of Hsp22 deletion was generated. Although comparable to wild-type mice in basal conditions, knockout mice exposed to pressure overload developed less hypertrophy and showed ventricular dilation, impaired contractile function, increased myocyte length and accumulation of interstitial collagen, faster transition into heart failure, and increased mortality. Microarrays revealed that hearts from knockout mice failed to transactivate genes regulated by the transcription factor STAT3. Accordingly, nuclear STAT3 tyrosine phosphorylation was decreased in knockout mice. Silencing and overexpression experiments in isolated neonatal rat cardiomyocytes showed that Hsp22 activates STAT3 via production of interleukin-6 by the transcription factor nuclear factor-κB. In addition to its transcriptional function, STAT3 translocates to the mitochondria where it increases oxidative phosphorylation. Both mitochondrial STAT3 translocation and respiration were also significantly decreased in knockout mice. CONCLUSIONS: This study found that Hsp22 represents a previously undescribed activator of both nuclear and mitochondrial functions of STAT3, and its deletion in the context of pressure overload in vivo accelerates the transition into heart failure and increases mortality.


Subject(s)
Gene Deletion , HSP20 Heat-Shock Proteins/genetics , Heart Failure/genetics , Mitochondria, Heart/genetics , Muscle Proteins/genetics , STAT3 Transcription Factor/genetics , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cell Nucleus/enzymology , Cell Nucleus/genetics , Cells, Cultured , Collagen/metabolism , Gene Expression Profiling , Heart Failure/enzymology , Heart Failure/mortality , Heat-Shock Proteins , Interleukin-6/biosynthesis , Male , Mice , Mice, Knockout , Mitochondria, Heart/enzymology , Molecular Chaperones , Myocytes, Cardiac/enzymology , NF-kappa B/metabolism , Oxidative Phosphorylation , Rats
4.
Curr Protoc Mouse Biol ; 1: 105-122, 2011.
Article in English | MEDLINE | ID: mdl-21686061

ABSTRACT

The use of mice for the evaluation and study of cardiovascular pathophysiology is growing rapidly, primarily due to the relative ease for developing genetically engineered mouse models. Arterial pressure monitoring is central to the evaluation of the phenotypic changes associated with cardiovascular pathology and interventions in these transgenic and knockout models. There are four major techniques for measuring arterial pressure in the mouse: tail cuff system, implanted fluid filled catheters, Millar catheters and implanted telemetry systems. Here we provide protocols for their use and discuss the advantages and limitations for each of these techniques .

5.
Circ Res ; 109(2): 161-71, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21617130

ABSTRACT

RATIONALE: Overexpression of muscle atrophy F-box (MAFbx/atrogin-1), an E3 ubiquitin ligase, induces proteasomal degradation in cardiomyocytes. The role of endogenous MAFbx in regulating cardiac hypertrophy and failure remains unclear. OBJECTIVE: We investigated the role of MAFbx in regulating cardiac hypertrophy and function in response to pressure overload. Transverse aortic constriction (TAC) was applied to MAFbx knockout (KO) and wild-type (WT) mice. METHODS AND RESULTS: Expression of MAFbx in WT mice was significantly increased by TAC. TAC-induced increases in cardiac hypertrophy were significantly smaller in MAFbx KO than in WT mice. There was significantly less lung congestion and interstitial fibrosis in MAFbx KO than in WT mice. MAFbx KO also inhibited ß-adrenergic cardiac hypertrophy. DNA microarray analysis revealed that activation of genes associated with the transcription factor binding site for the nuclear factor-κB family were inhibited in MAFbx KO mice compared with WT mice after TAC. Although the levels of IκB-α were significantly decreased after TAC in WT mice, they were increased in MAFbx KO mice. MAFbx regulates ubiquitination and proteasomal degradation of IκB-α in cardiomyocytes. In primary cultured rat cardiomyocytes, phenylephrine-induced activation of nuclear factor-κB and hypertrophy were significantly suppressed by MAFbx knockdown but were partially rescued by overexpression of nuclear factor-κB p65. CONCLUSIONS: MAFbx plays an essential role in mediating cardiac hypertrophy in response to pressure overload. Downregulation of MAFbx inhibits cardiac hypertrophy in part through stabilization of IκB-α and inactivation of nuclear factor-κB. Taken together, inhibition of MAFbx attenuates pathological hypertrophy, thereby protecting the heart from progression into heart failure.


Subject(s)
Cardiomegaly/metabolism , Muscle Proteins/physiology , NF-kappa B/metabolism , SKP Cullin F-Box Protein Ligases/physiology , Animals , Cardiomegaly/etiology , Cells, Cultured , Constriction, Pathologic , Gene Expression , Gene Expression Regulation/physiology , I-kappa B Proteins/metabolism , Mice , Mice, Knockout , Muscle Proteins/deficiency , Muscle Proteins/metabolism , NF-KappaB Inhibitor alpha , Protective Agents , Rats , SKP Cullin F-Box Protein Ligases/deficiency , SKP Cullin F-Box Protein Ligases/metabolism
6.
Cardiovasc Res ; 89(2): 353-61, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20833651

ABSTRACT

AIMS: Improving the sarco(endo)plasmic reticulum (SR) Ca(2+)-ATPase (SERCA) function has clinical implications in treating heart failure. The present study aimed to determine the effect of constitutive activation of the SERCA pump on cardiac contractility in normal mice and during pressure-overload-induced cardiac hypertrophy. METHODS AND RESULTS: The SERCA pump was constitutively activated in both atrial and ventricular chambers of the mouse heart by ablating its key regulators, phospholamban (PLN) and sarcolipin (SLN). The double-knockout (dKO) mice for PLN and SLN showed increased SERCA pump activity, Ca(2+) transients and SR Ca(2+) load, and developed cardiac hypertrophy. Echocardiographic measurements showed that the basal cardiac function was not affected in the young dKO mice. However, the cardiac function worsened upon ageing and when subjected to pressure overload. CONCLUSION: Our studies suggest that the constitutive activation of the SERCA pump is detrimental to cardiac function. Our findings also emphasize the need for dynamic regulation of the SERCA pump by PLN and/or SLN to maintain cardiac contractility in normal conditions and during pathophysiological states.


Subject(s)
Calcium-Binding Proteins/deficiency , Cardiomegaly/metabolism , Muscle Proteins/deficiency , Myocardial Contraction , Myocardium/metabolism , Proteolipids/deficiency , Age Factors , Aging , Animals , Aorta/surgery , Calcium/metabolism , Calcium Signaling , Calcium-Binding Proteins/genetics , Cardiomegaly/diagnostic imaging , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Disease Models, Animal , Enzyme Activation , Female , Gene Expression Regulation , Ligation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Myocardial Contraction/genetics , Proteolipids/genetics , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Stroke Volume , Ultrasonography , Ventricular Function, Left
7.
Am J Physiol Heart Circ Physiol ; 299(3): H707-12, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20562336

ABSTRACT

Adenylyl cyclase (AC) type 5 (AC5) and AC type 6 (AC6) are the two major AC isoforms in the heart. Cardiac overexpression of AC6 has been shown to be protective in response to several interventions. In this investigation, we examined the effects of chronic pressure overload in AC6 transgenic (TG) mice. In the absence of any stress, AC6 TG mice exhibited enhanced contractile function compared with their wild-type (WT) littermates, i.e., increased (P < 0.05) left ventricular (LV) ejection fraction (EF) (75 +/- 0.9 vs. 71 +/- 0.5%) and LV dP/dt (7,850 +/- 526 vs. 6,374 +/- 315 mmHg/s). Forskolin (25 microg x kg(-1) x min(-1) for 5 min) increased LVEF more (P < 0.05) in AC6 TG mice (14.8 +/- 1.0%) than in WT mice (7.7 +/- 1.0%). Also, isoproterenol (0.04 microg x kg(-1) x min(-1) for 5 min) increased LVEF more (P < 0.05) in AC6 TG mice (18.0 +/- 1.2%) than in WT mice (11.6 +/- 2.1%). Pressure overload, induced by 4 wk of transverse aortic constriction (TAC), increased the LV weight-to-body weight ratio and myocyte cross-sectional area similarly in both groups, but reduced LVEF more in AC6 TG mice (22%) compared with WT mice (9%), despite the higher starting level of LVEF in AC6 TG mice. LV systolic wall stress increased more in AC6 TG mice than in WT mice, which could be responsible for the reduced LVEF in AC6 TG mice with chronic pressure overload. In addition, LV dP/dt was no longer elevated in AC6 TG mice after TAC compared with WT mice. LV end-diastolic diameter was also greater (P < 0.05) in AC6 TG mice (3.8 +/- 0.07 mm) than in WT mice (3.6 +/- 0.05 mm) after TAC. Thus, in contrast to other interventions previously reported to be salutary with cardiac AC6 overpression, the response to chronic pressure overload was not; actually, AC6 TG mice fared worse than WT mice. The mechanism may be due to the increased LV systolic wall stress in AC6 TG mice with chronic pressure overload.


Subject(s)
Adenylyl Cyclases/metabolism , Heart Ventricles/metabolism , Heart/physiopathology , Hypertrophy, Left Ventricular/metabolism , Myocardium/metabolism , Ventricular Dysfunction, Left/metabolism , Adenylyl Cyclases/genetics , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/physiology , Colforsin/pharmacology , Echocardiography , Heart/drug effects , Heart Rate/drug effects , Heart Rate/physiology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics , Hypertrophy, Left Ventricular/physiopathology , Isoproterenol , Mice , Mice, Transgenic , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Stress, Physiological/physiology , Ventricular Dysfunction, Left/physiopathology
8.
J Biol Chem ; 285(26): 20281-90, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20404348

ABSTRACT

MicroRNA-21 (miR-21) is highly up-regulated during hypertrophic and cancerous cell growth. In contrast, we found that it declines in cardiac myocytes upon exposure to hypoxia. Thus, the objective was to explore its role during hypoxia. We show that miR-21 not only regulates phosphatase and tensin homologue deleted on chromosome 10 (PTEN), but also targets Fas ligand (FasL). During prolonged hypoxia, down-regulation of miR-21 proved necessary and sufficient for enhancing expression of both proteins. We demonstrate here for the first time that miR-21 is positively regulated via an AKT-dependent pathway, which is depressed during prolonged hypoxia. Accordingly, hypoxia-induced down-regulation of miR-21 and up-regulation of FasL and PTEN were reversed by activated AKT and reproduced by a dominant negative mutant, wortmannin, or PTEN. Moreover, the antiapoptotic function of AKT partly required miR-21, which was sufficient for inhibition of caspase-8 activity and mitochondrial damage. In consensus, overexpression of miR-21 in a transgenic mouse heart resulted in suppression of ischemia-induced up-regulation of PTEN and FasL expression, an increase in phospho-AKT, a smaller infarct size, and ameliorated heart failure. Thus, we have identified a unique aspect of the function of AKT by which it inhibits apoptosis through miR-21-dependent suppression of FasL.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 3' Untranslated Regions/genetics , Animals , Animals, Newborn , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Fas Ligand Protein/genetics , Humans , Immunohistochemistry , Mice , Mice, Transgenic , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Rats , Rats, Sprague-Dawley
9.
Am J Physiol Heart Circ Physiol ; 297(5): H1814-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19717730

ABSTRACT

Calcineurin is a Ca(2+)/calmodulin-dependent protein phosphatase that induces myocardial growth in response to several physiological and pathological stimuli. Calcineurin inhibition, induced either via cyclosporine or genetically, can decrease myocardial hypertrophy secondary to pressure overload without affecting left ventricular (LV) systolic function. Since hypertrophy can also affect LV diastolic function, the goal of this study was to examine the effects of chronic pressure overload (2 wk aortic banding) in transgenic (Tg) mice overexpressing Zaki-4beta (TgZ), a specific endogenous inhibitor of calcineurin, on LV diastolic function. As expected, in the TgZ mice with calcineurin inhibitor overexpression, aortic banding reduced the degree of LV hypertrophy, as assessed by LV weight-to-body weight ratio (3.5 + or - 0.1) compared with that in non-Tg mice (4.6 + or - 0.2). LV systolic function remained compensated in both groups with pressure overload. However, the LV end-diastolic stress-to-LV end-diastolic dimension ratio, an index of diastolic stiffness and LV pressure half-time and isovolumic relaxation time, two indexes of isovolumic relaxation, increased significantly more in TgZ mice with aortic banding. Protein levels of phosphorylated phospholamban (PS16), sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a, phosphorylated ryanodine receptor, and the Na(+)/Ca(2+) exchanger were also reduced significantly (P < 0.05) in the banded TgZ mice. As expected, genetic calcineurin inhibition inhibited the development of LV hypertrophy with chronic pressure overload but also induced LV diastolic dysfunction, as reflected by both impaired isovolumic relaxation and increased myocardial stiffness. Thus genetic calcineurin inhibition reveals a new mechanism regulating LV diastolic function.


Subject(s)
Calcineurin Inhibitors , Diastole , Hypertrophy, Left Ventricular/prevention & control , Myocardium/enzymology , Proteins/metabolism , Ventricular Dysfunction, Left/enzymology , Animals , Aorta/surgery , Calcineurin/metabolism , Calcium-Binding Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Diastole/genetics , Disease Models, Animal , Elasticity , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Intracellular Signaling Peptides and Proteins , Ligation , Mice , Mice, Transgenic , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phosphorylation , Proteins/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism , Stroke Volume , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure
10.
Proc Natl Acad Sci U S A ; 105(52): 20900-5, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19106302

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) is a master regulator of growth and death in cardiac myocytes. GSK-3 is inactivated by hypertrophic stimuli through phosphorylation-dependent and -independent mechanisms. Inactivation of GSK-3 removes the negative constraint of GSK-3 on hypertrophy, thereby stimulating cardiac hypertrophy. N-terminal phosphorylation of the GSK-3 isoforms GSK-3alpha and GSK-3beta by upstream kinases (e.g., Akt) is a major mechanism of GSK-3 inhibition. Nonetheless, its role in mediating cardiac hypertrophy and failure remains to be established. Here we evaluated the role of Serine(S)21 and S9 phosphorylation of GSK-3alpha and GSK-3beta in the regulation of cardiac hypertrophy and function during pressure overload (PO), using GSK-3alpha S21A knock-in (alphaKI) and GSK-3beta S9A knock-in (betaKI) mice. Although inhibition of S9 phosphorylation during PO in the betaKI mice attenuated hypertrophy and heart failure (HF), inhibition of S21 phosphorylation in the alphaKI mice unexpectedly promoted hypertrophy and HF. Inhibition of S21 phosphorylation in GSK-3alpha, but not of S9 phosphorylation in GSK-3beta, caused phosphorylation and down-regulation of G1-cyclins, due to preferential localization of GSK-3alpha in the nucleus, and suppressed E2F and markers of cell proliferation, including phosphorylated histone H3, under PO, thereby contributing to decreases in the total number of myocytes in the heart. Restoration of the E2F activity by injection of adenovirus harboring cyclin D1 with a nuclear localization signal attenuated HF under PO in the alphaKI mice. Collectively, our results reveal that whereas S9 phosphorylation of GSK-3beta mediates pathological hypertrophy, S21 phosphorylation of GSK-3alpha plays a compensatory role during PO, in part by alleviating the negative constraint on the cell cycle machinery in cardiac myocytes.


Subject(s)
Blood Pressure , Glycogen Synthase Kinase 3/metabolism , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Animals , Blood Pressure/genetics , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin G , Cyclin G1 , Cyclins/genetics , Cyclins/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Gene Knock-In Techniques , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Heart Failure/enzymology , Heart Failure/genetics , Histones/genetics , Histones/metabolism , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/pathology , Phosphorylation/genetics
11.
Circ Res ; 103(11): 1309-18, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18927464

ABSTRACT

Mammalian sterile 20-like kinase (Mst)1 plays an important role in mediating apoptosis and inhibiting hypertrophy in the heart. Because Hippo, a Drosophila homolog of Mst1, forms a signaling complex with Warts, a serine/threonine kinase, which in turn stimulates cell death and inhibits cell proliferation, mammalian homologs of Warts, termed Lats1 and Lats2, may mediate the function of Mst1. We here show that Lats2, but not Lats1, dose-dependently increased apoptosis in cultured cardiac myocytes. Lats2 also dose-dependently reduced [(3)H]phenylalanine incorporation and cardiac myocyte size, whereas dominant negative Lats2 (DN-Lats2) increased them, suggesting that endogenous Lats2 negatively regulates myocyte growth. DN-Lats2 significantly attenuated induction of apoptosis and inhibition of hypertrophy by Mst1, indicating that Lats2 mediates the function of Mst1 in cardiac myocytes. Cardiac specific overexpression of Lats2 in transgenic mice significantly reduced the size of left and right ventricles, whereas that of DN-Lats2 caused hypertrophy in both ventricles. Overexpression of Lats2 reduced left ventricular systolic and diastolic function without affecting baseline levels of myocardial apoptosis. Expression of endogenous Lats2 was significantly upregulated in response to transverse aortic constriction. Overexpression of DN-Lats2 significantly enhanced cardiac hypertrophy and inhibited cardiac myocyte apoptosis induced by transverse aortic constriction. These results suggest that Lats2 is necessary and sufficient for negatively regulating ventricular mass in the heart. Although Lats2 is required for cardiac myocyte apoptosis in response to pressure overload, it was not sufficient to induce apoptosis at baseline. In conclusion, Lats2 affects both growth and death of cardiac myocytes, but it primarily regulates the size of the heart and acts as an endogenous negative regulator of cardiac hypertrophy.


Subject(s)
Cardiomegaly/prevention & control , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Adenoviridae/genetics , Analysis of Variance , Animals , Apoptosis , Cell Size , Cells, Cultured , DNA Fragmentation , Genes, Dominant , Humans , Mice , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/deficiency , RNA, Messenger/genetics , Tumor Suppressor Proteins/deficiency
12.
Am J Physiol Heart Circ Physiol ; 295(4): H1385-93, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18676687

ABSTRACT

We tested the possibility that proteasome inhibition may reverse preexisting cardiac hypertrophy and improve remodeling upon pressure overload. Mice were submitted to aortic banding and followed up for 3 wk. The proteasome inhibitor epoxomicin (0.5 mg/kg) or the vehicle was injected daily, starting 2 wk after banding. At the end of the third week, vehicle-treated banded animals showed significant (P<0.05) increase in proteasome activity (PA), left ventricle-to-tibial length ratio (LV/TL), myocyte cross-sectional area (MCA), and myocyte apoptosis compared with sham-operated animals and developed signs of heart failure, including increased lung weight-to-TL ratio and decreased ejection fraction. When compared with that group, banded mice treated with epoxomicin showed no increase in PA, a lower LV/TL and MCA, reduced apoptosis, stabilized ejection fraction, and no signs of heart failure. Because overload-mediated cardiac remodeling largely depends on the activation of the proteasome-regulated transcription factor NF-kappaB, we tested whether epoxomicin would prevent this activation. NF-kappaB activity increased significantly upon overload, which was suppressed by epoxomicin. The expression of NF-kappaB-dependent transcripts, encoding collagen types I and III and the matrix metalloprotease-2, increased (P<0.05) after banding, which was abolished by epoxomicin. The accumulation of collagen after overload, as measured by histology, was 75% lower (P<0.05) with epoxomicin compared with vehicle. Myocyte apoptosis increased by fourfold in hearts submitted to aortic banding compared with sham-operated hearts, which was reduced by half upon epoxomicin treatment. Therefore, we propose that proteasome inhibition after the onset of pressure overload rescues ventricular remodeling by stabilizing cardiac function, suppressing further progression of hypertrophy, repressing collagen accumulation, and reducing myocyte apoptosis.


Subject(s)
Cardiomegaly/drug therapy , Heart Failure/prevention & control , Myocardium/enzymology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Aorta/surgery , Apoptosis/drug effects , Blood Pressure , Cardiomegaly/complications , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Disease Models, Animal , Heart Failure/enzymology , Heart Failure/etiology , Heart Failure/physiopathology , Ligation , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Myocardial Contraction/drug effects , Myocardium/pathology , NF-kappa B/metabolism , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/metabolism , Stroke Volume/drug effects , Time Factors
13.
Am J Physiol Heart Circ Physiol ; 295(2): H860-6, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18567712

ABSTRACT

The objective of this study was to test the hypothesis that the mechanism mediating left ventricular (LV) dysfunction in the aging rat heart involves, in part, changes in cardiac cytoskeletal components. Our results show that there were no significant differences in heart rate, LV pressure, or LV diameter between conscious, instrumented young [5.9 +/- 0.3 mo (n = 9)] and old rats [30.6 +/- 0.1 mo (n = 10)]. However, the first derivative of LV pressure (LV dP/dt) was reduced (8,309 +/- 790 vs. 11,106 +/- 555 mmHg/s, P < 0.05) and isovolumic relaxation time (tau) was increased (8.7 +/- 0.7 vs. 6.3 +/- 0.6 ms, P < 0.05) in old vs. young rats, respectively. The differences in baseline LV function in young and old rats, which were modest, were accentuated after beta-adrenergic receptor stimulation with dobutamine (20 mug/kg), which increased LV dP/dt by 170 +/- 9% in young rats, significantly more (P < 0.05) than observed in old rats (115 +/- 5%). Volume loading in anesthetized rats demonstrated significantly impaired LV compliance in old rats, as measured by the LV end-diastolic pressure and dimension relationship. In old rat hearts, there was a significant (P < 0.05) increase in the percentage of LV collagen (2.4 +/- 0.2 vs. 1.3 +/- 0.2%), alpha-tubulin (92%), and beta-tubulin (2.3-fold), whereas intact desmin decreased by 51%. Thus the cardiomyopathy of aging in old, conscious rats may be due not only to increases in collagen but also to alterations in cytoskeletal proteins.


Subject(s)
Aging/metabolism , Cardiomyopathies/metabolism , Cytoskeletal Proteins/metabolism , Myocardium/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Adrenergic beta-Agonists/administration & dosage , Age Factors , Animals , Cardiomyopathies/physiopathology , Collagen/metabolism , Compliance , Consciousness , Crosses, Genetic , Desmin/metabolism , Dobutamine/administration & dosage , Dose-Response Relationship, Drug , Heart Rate , Myocardial Contraction , Rats , Rats, Inbred BN , Rats, Inbred F344 , Time Factors , Tubulin/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/drug effects , Ventricular Pressure
14.
J Biol Chem ; 283(32): 22157-65, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18508765

ABSTRACT

Myocardial infarction (MI) is often followed by heart failure (HF), but the mechanisms precipitating the transition to HF remain largely unknown. A genomic profile was performed in a monkey model of MI, from the myocardium adjacent to chronic (2-month) MI followed by 3 weeks of pacing to develop HF. The transcript of the gene encoding the cell cycle-related kinase (CCRK) was down-regulated by 50% in HF heart compared with control (p<0.05), which was confirmed by quantitative PCR. The CCRK sequence cloned from a heart library showed a conservation of the N-terminal kinase domain when compared with the "generic" isoform cloned previously but a different C-terminal half due to alternative splicing with frameshift. The homology of the cardiac sequence was 100% between mice and humans. Expression of the corresponding protein, measured upon generation of a monoclonal antibody, was limited to heart, liver, and kidney. Upon overexpression in cardiac myocytes, both isoforms promote cell growth and reduce apoptosis by chelerythrine (p<0.05 versus control). Using a yeast two-hybrid screening, we found an interaction of the generic but not the cardiac CCRK with cyclin H and casein kinase 2. In addition, only the generic CCRK phosphorylates the cyclin-dependent kinase 2, which was accompanied by a doubling of myocytes in the S and G(2) phases of the cell cycle (p < 0.05 versus control). Therefore, the heart expresses a splice variant of CCRK, which promotes cardiac cell growth and survival; differs from the generic isoform in terms of protein-protein interactions, substrate specificity and regulation of the cell cycle; and is down-regulated significantly in HF.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Heart Failure/enzymology , Myocardium/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cell Proliferation , Cell Survival , Cells, Cultured , Cloning, Molecular , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/genetics , Disease Models, Animal , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Macaca fascicularis , Male , Mice , Molecular Sequence Data , Protein Binding , Rats , Rats, Wistar , Sequence Alignment , Substrate Specificity
15.
Circ Res ; 101(11): 1164-74, 2007 Nov 26.
Article in English | MEDLINE | ID: mdl-17901358

ABSTRACT

Glycogen synthase kinase (GSK)-3, a negative regulator of cardiac hypertrophy, is inactivated in failing hearts. To examine the histopathological and functional consequence of the persistent inhibition of GSK-3beta in the heart in vivo, we generated transgenic mice with cardiac-specific overexpression of dominant negative GSK-3beta (Tg-GSK-3beta-DN) and tetracycline-regulatable wild-type GSK-3beta. GSK-3beta-DN significantly reduced the kinase activity of endogenous GSK-3beta, inhibited phosphorylation of eukaryotic translation initiation factor 2B epsilon, and induced accumulation of beta-catenin and myeloid cell leukemia-1, confirming that GSK-3beta-DN acts as a dominant negative in vivo. Tg-GSK-3beta-DN exhibited concentric hypertrophy at baseline, accompanied by upregulation of the alpha-myosin heavy chain gene and increases in cardiac function, as evidenced by a significantly greater Emax after dobutamine infusion and percentage of contraction in isolated cardiac myocytes, indicating that inhibition of GSK-3beta induces well-compensated hypertrophy. Although transverse aortic constriction induced a similar increase in hypertrophy in both Tg-GSK-3beta-DN and nontransgenic mice, Tg-GSK-3beta-DN exhibited better left ventricular function and less fibrosis and apoptosis than nontransgenic mice. Induction of the GSK-3beta transgene in tetracycline-regulatable wild-type GSK-3beta mice induced left ventricular dysfunction and premature death, accompanied by increases in apoptosis and fibrosis. Overexpression of GSK-3beta-DN in cardiac myocytes inhibited tumor necrosis factor-alpha-induced apoptosis, and the antiapoptotic effect of GSK-3beta-DN was abrogated in the absence of myeloid cell leukemia-1. These results suggest that persistent inhibition of GSK-3beta induces compensatory hypertrophy, inhibits apoptosis and fibrosis, and increases cardiac contractility and that the antiapoptotic effect of GSK-3beta inhibition is mediated by myeloid cell leukemia-1. Thus, downregulation of GSK-3beta during heart failure could be compensatory.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Heart Failure/drug therapy , Animals , Apoptosis , Cardiomegaly/etiology , Cardiotonic Agents , Down-Regulation , Enzyme Inhibitors/pharmacology , Fibrosis/etiology , Glycogen Synthase Kinase 3 beta , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein , Myocardial Contraction , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology
16.
Circ Res ; 100(9): 1344-52, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17395874

ABSTRACT

Mammalian sterile 20-like kinase-1 (Mst1) plays an important role in mediating cardiac myocyte apoptosis in response to ischemia/reperfusion. Whether or not Mst1 is also involved in the long-term development of heart failure after myocardial infarction (MI) is unknown. We addressed this issue using transgenic mice with cardiac specific overexpression of dominant negative Mst1 (Tg-DN-Mst1). The left coronary artery was permanently ligated, and the size of MI was similar between Tg-DN-Mst1 and nontransgenic controls (NTg). After 4 weeks, Mst1 was significantly activated in the remodeling area in NTg, but not in Tg-DN-Mst1. Although left ventricular (LV) enlargement was significantly attenuated in Tg-DN-Mst1 compared with NTg, neither LV weight/body weight nor myocyte cross sectional area was statistically different between Tg-DN-Mst1 and NTg. LV ejection fraction was significantly greater in Tg-DN-Mst1 than in NTg (53 versus 38%, P<0.01), whereas LV end-diastolic pressure (6 versus 12 mm Hg, P<0.05) and lung weight/body weight (9.8 versus 12.2 P<0.05) were significantly smaller in Tg-DN-Mst1 than in NTg. The number of TUNEL-positive myocytes (0.17 versus 0.28%, P<0.05) and amount of interstitial fibrosis (5.0 versus 7.1%, P<0.05) in the remodeling area were significantly less in Tg-DN-Mst1 than in NTg. Upregulation of matrix metalloproteinase 2 and proinflammatory cytokines was significantly attenuated in Tg-DN-Mst1. These results indicate that endogenous Mst1 plays an important role in mediating cardiac dilation, apoptosis, fibrosis, and cardiac dysfunction, but not cardiac hypertrophy, after MI. Inhibition of Mst1 improves cardiac function without attenuating cardiac hypertrophy. Thus, Mst1 may be an important target of heart failure treatment.


Subject(s)
Apoptosis , Cardiomegaly/etiology , Hepatocyte Growth Factor/physiology , Myocardial Infarction/complications , Myocardium/pathology , Proto-Oncogene Proteins/physiology , Ventricular Dysfunction, Left/prevention & control , Animals , Cytokines/physiology , Fibrosis , Hepatocyte Growth Factor/antagonists & inhibitors , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Ventricular Remodeling
17.
Am J Physiol Heart Circ Physiol ; 292(2): H971-5, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17287453

ABSTRACT

Both enhanced sympathetic drive and altered autonomic control are involved in the pathogenesis of heart failure. The goal of the present study was to determine the extent to which chronically enhanced sympathetic drive, in the absence of heart failure, alters reflex autonomic control in conscious, transgenic (TG) rabbits with overexpressed cardiac Gsalpha. Nine TG rabbits and seven wild-type (WT) littermates were instrumented with a left ventricular (LV) pressure micromanometer and arterial catheters and studied in the conscious state. Compared with WT rabbits, LV function was enhanced in TG rabbits, as reflected by increased levels of LV dP/dt (5,600 +/- 413 vs. 3,933 +/- 161 mmHg/s). Baseline heart rate was also higher (P < 0.05) in conscious TG (247 +/- 10 beats/min) than in WT (207 +/- 10 beats/min) rabbits and was higher in TG after muscarinic blockade (281 +/- 9 vs. 259 +/- 8 beats/min) or combined beta-adrenergic receptor and muscarinic blockade (251 +/- 6 vs. 225 +/- 9 beats/min). Bradycardia was blunted (P < 0.05), whether induced by intravenous phenylephrine (arterial baroreflex), by cigarette smoke inhalation (nasopharyngeal reflex), or by veratrine administration (Bezold-Jarisch reflex). With veratrine administration, the bradycardia was enhanced in TG for any given decrease in arterial pressure. Thus the chronically enhanced sympathetic drive in TG rabbits with overexpressed cardiac Gsalpha resulted in enhanced LV function and heart rate and impaired reflex autonomic control. The impaired reflex control was generalized, not only affecting the high-pressure arterial baroreflex but also the low-pressure Bezold-Jarisch reflex and the nasopharyngeal reflex.


Subject(s)
Bradycardia/physiopathology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Myocardium/metabolism , Reflex , Sympathetic Nervous System/physiopathology , Adrenergic beta-Antagonists/pharmacology , Animals , Animals, Genetically Modified , Atropine/pharmacology , Autonomic Nervous System/physiopathology , Baroreflex , Blood Pressure , Bradycardia/chemically induced , Bradycardia/metabolism , Electric Stimulation , GTP-Binding Protein alpha Subunits, Gs/genetics , Heart Rate , Muscarinic Antagonists/pharmacology , Phenylephrine , Propranolol/pharmacology , Rabbits/genetics , Smoke/adverse effects , Sympathetic Nervous System/drug effects , Nicotiana , Vagus Nerve/physiopathology , Ventricular Function, Left , Veratrine
18.
Circ Res ; 100(3): 416-24, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17234972

ABSTRACT

MicroRNAs are naturally existing, small, noncoding RNA molecules that downregulate posttranscriptional gene expression. Their expression pattern and function in the heart remain unknown. Here we report an array of microRNAs that are differentially and temporally regulated during cardiac hypertrophy. Significantly, the muscle-specific microRNA-1 (miR-1) was singularly downregulated as early as day 1 (0.56+/-0.036), persisting through day 7 (0.29+/-0.14), after aortic constriction-induced hypertrophy in a mouse model. Overexpression experiments showed that miR-1 inhibited its in silico-predicted, growth-related targets, including Ras GTPase-activating protein (RasGAP), cyclin-dependent kinase 9 (Cdk9), fibronectin, and Ras homolog enriched in brain (Rheb), in addition to protein synthesis and cell size. Thus, we propose that microRNAs play an essential regulatory role in the development of cardiac hypertrophy, wherein downregulation of miR-1 is necessary for the relief of growth-related target genes from its repressive influence and induction of hypertrophy.


Subject(s)
Cardiomyopathy, Hypertrophic/etiology , Gene Expression Regulation , MicroRNAs/physiology , Animals , Aortic Valve Stenosis/complications , Blotting, Northern , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Division/drug effects , Cell Size , Cells, Cultured/metabolism , Constriction , Culture Media/pharmacology , Culture Media, Serum-Free/pharmacology , Cytomegalovirus/genetics , Disease Progression , Down-Regulation , Gene Expression Profiling , Genetic Vectors/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , MicroRNAs/classification , MicroRNAs/genetics , MicroRNAs/isolation & purification , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA Polymerase III/physiology , RNA, Small Nuclear/physiology , Rats , Rats, Sprague-Dawley
19.
Circulation ; 114(17): 1821-8, 2006 Oct 24.
Article in English | MEDLINE | ID: mdl-17043166

ABSTRACT

BACKGROUND: The adaptation of cardiac mass to hemodynamic overload requires an adaptation of protein turnover, ie, the balance between protein synthesis and degradation. We tested 2 hypotheses: (1) chronic left ventricular hypertrophy (LVH) activates the proteasome system of protein degradation, especially in the myocardium submitted to the highest wall stress, ie, the subendocardium, and (2) the proteasome system is required for the development of LVH. METHODS AND RESULTS: Gene and protein expression of proteasome subunits and proteasome activity were measured separately from left ventricular subendocardium and subepicardium, right ventricle, and peripheral tissues in a canine model of severe, chronic (2 years) LVH induced by aortic banding and then were compared with controls. Both gene and protein expressions of proteasome subunits were increased in LVH versus control (P<0.05), which was accompanied by a significant (P<0.05) increase in proteasome activity. Posttranslational modification of the proteasome was also detected by 2-dimensional gel electrophoresis. These changes were found specifically in left ventricular subendocardium but not in left ventricular subepicardium, right ventricle, or noncardiac tissues from the same animals. In a mouse model of chronic pressure overload, a 50% increase in heart mass and a 2-fold increase in proteasome activity (both P<0.05 versus sham) were induced. In that model, the proteasome inhibitor epoxomicin completely prevented LVH while blocking proteasome activation. CONCLUSIONS: The increase in proteasome expression and activity found during chronic pressure overload in myocardium submitted to higher stress is also required for the establishment of LVH.


Subject(s)
Hypertrophy, Left Ventricular/metabolism , Muscle Proteins/metabolism , Proteasome Endopeptidase Complex/physiology , Ventricular Remodeling/physiology , Adaptation, Physiological , Animals , Aorta, Thoracic , Disease Models, Animal , Dogs , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Profiling , Ligation , Male , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oligopeptides/pharmacology , Polyubiquitin/metabolism , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Protein Subunits , Stress, Physiological/metabolism
20.
J Mol Cell Cardiol ; 41(4): 680-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16928383

ABSTRACT

We examined pressure overload left ventricular (LV) hypertrophy (H) induced by aortic banding in transgenic mice with cardiac-specific expression of a dominant negative (DN) p38alpha (TG) and wild type controls (WT). In response to chronic pressure overload, induced by aortic constriction, LV/BW increased more, p<0.05, in female TG (6.4+/-0.2, n=7) than in WT female (5.1+/-0.2, n=10), or male TG or WT (5.0+/-0.2, n=10 vs. 5.5+/-0.2, n=8). Lung/BW, an index of LV decompensation, was significantly higher, p<0.05, in banded female TG (14+/-1.2 mg/g) than in WT females (9.0+/-0.8), or male TG or WT (8.2+/-0.7 vs. 9.3+/-1.3). This was associated with higher premature mortality, p<0.05, in banded female TG mice (42%) compared with banded WT females (10%), TG males (13%), or WT males (17%). In male, but not female, TG mice, the number of TUNEL-positive cells was smaller, p<0.05, compared with WT. Phospho-Akt kinase activity increased (p<0.05) in female TG after banding, but not in males. After ovariectomy, chronic pressure overload no longer induced greater mortality, greater LVH, or p-Akt levels in female TG mice, and like male TG mice, apoptosis was protected. DN-p38alpha enhanced estrogen-induced activation of Akt in cultured cardiac myocytes. Thus, inhibition of p38alpha MAPK paradoxically augments LVH resulting in cardiac decompensation and increased mortality in response to pressure overload more in female mice than male mice, which could be due to increased Akt activation and/or through cross-talk between p38alpha MAPK and Akt.


Subject(s)
Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Ventricular Pressure , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Apoptosis , Cells, Cultured , Female , Male , Mice , Mice, Transgenic , Ovariectomy/adverse effects , Phosphorylation , Rats , Rats, Wistar , Sex Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...