Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Ann Thorac Surg ; 116(4): 834-843, 2023 10.
Article in English | MEDLINE | ID: mdl-35398036

ABSTRACT

BACKGROUND: Patients with valvular heart disease require cardiopulmonary bypass and cardiac arrest. Here, we test the hypothesis that exosomal hemoglobin formed during cardiopulmonary bypass mediates acute cardiac injury in humans and in an animal model system. METHODS: Plasma exosomes were collected from arterial blood at baseline and 30 minutes after aortic cross-clamp release in 20 patients with primary mitral regurgitation and 7 with aortic stenosis. These exosomes were injected into Sprague-Dawley rats and studied at multiple times up to 30 days. Tissue was examined by hematoxylin and eosin stain, immunohistochemistry, transmission electron microscopy, and brain natriuretic peptide. RESULTS: Troponin I levels increased from 36 ± 88 ng/L to 3622 ± 3054 ng/L and correlated with exosome hemoglobin content (Spearman r = 0.7136, < .0001, n = 24). Injection of exosomes isolated 30 minutes after cross-clamp release into Sprague-Dawley rats resulted in cardiomyocyte myofibrillar loss at 3 days. Transmission electron microscopy demonstrated accumulation of electron dense particles of ferritin within cardiomyocytes, in the interstitial space, and within exosomes. At 21 days after injection, there was myofibrillar and myosin breakdown, interstitial fibrosis, elevated brain natriuretic peptide, and left ventricle diastolic dysfunction measured by echocardiography/Doppler. Pericardial fluid exosomal hemoglobin content is fourfold higher than simultaneous plasma exosome hemoglobin, suggesting a cardiac source of exosomal hemoglobin. CONCLUSIONS: Red blood cell and cardiac-derived exosomal hemoglobin may be involved in myocardial injury during cardiopulmonary bypass in patients with valvular heart disease.


Subject(s)
Exosomes , Heart Injuries , Heart Valve Diseases , Humans , Rats , Animals , Rats, Sprague-Dawley , Natriuretic Peptide, Brain , Myocytes, Cardiac , Disease Models, Animal
2.
J Thorac Cardiovasc Surg ; 164(6): e289-e308, 2022 Dec.
Article in English | MEDLINE | ID: mdl-33451850

ABSTRACT

OBJECTIVE: Hemolysis, characterized by formation of free hemoglobin (Hb), occurs in patients undergoing cardiopulmonary bypass (CPB). However, there is no study of the dynamic changes in red blood cell (RBC)-derived exosomes (Exos) released during CPB, nor whether these particles mediate acute kidney injury (AKI). METHODS: This study is a comprehensive time-course analysis, at baseline, 30 minutes, to 24 hours post-crossclamp release (XCR) to determine (1) Exos Hb content; (2) free Hb/heme, haptoglobin, hemopexin; and (3) urinary markers of AKI over the same time period. In addition, we developed a model system in Sprague-Dawley rats to test for AKI after intravenous injection of Exos Hb released during CPB. RESULTS: In 30 patients undergoing CPB, there is a significant increase in plasma Hb-positive Exos but not microvesicles 30 minutes post-XCR versus other time points, with a simultaneous decrease in the haptoglobin/Hb ratio. These changes presage a significant increase in urine neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 at 24 hours. Intravenous injection of plasma Exos (109-10 particles obtained 30 minutes post-XCR) into rats causes AKI at 72 hours, manifested by multifocal degeneration of proximal tubular epithelium. At 21 days, there is persistent tubular injury and interstitial fibrosis. Intravenous injection of Exos from 35-day-old stored RBCs into rats results in glomerular-tubular injury, increased kidney ferritin and hemoxygenase-1 expression, and significant elevation of kidney injury molecule-1 and proteinuria at 72 hours. CONCLUSIONS: These combined studies raise the potential for RBC-derived Exos, released during CPB, to target the kidney and mediate AKI.


Subject(s)
Acute Kidney Injury , Exosomes , Rats , Animals , Cardiopulmonary Bypass/adverse effects , Haptoglobins/metabolism , Exosomes/metabolism , Rats, Sprague-Dawley , Lipocalin-2 , Biomarkers , Hemoglobins/metabolism , Disease Models, Animal , Erythrocytes/metabolism
3.
Heliyon ; 5(4): e01397, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30997426

ABSTRACT

BACKGROUND: Volume overload (VO) of isolated mitral regurgitation (MR) or aortocaval fistula (ACF) is associated with extracellular matrix degradation and cardiomyocyte myofibrillar and desmin breakdown. Left ventricular (LV) chymase activity is increased in VO and recent studies demonstrate chymase presence within cardiomyocytes. Here we test the hypothesis that chymase within the cardiomyocyte coincides with myosin and desmin breakdown in VO. METHODS AND RESULTS: Aortocaval fistula (ACF) was induced in Sprague Dawley (SD) rats and was compared to age-matched sham-operated rats at 24 hours, 4 and 12 weeks. Immunohistochemistry (IHC) and transmission electron microscopy (TEM) immunogold of LV tissue demonstrate chymase within cardiomyocytes at all ACF time points. IHC for myosin demonstrates myofibrillar disorganization starting at 24 hours. Proteolytic presence of chymase in cardiomyocytes is verified by in situ chymotryptic tissue activity that is inhibited by pretreatment with a chymase inhibitor. Real-time PCR of isolated cardiomyocytes at all ACF time points and in situ hybridization demonstrate endothelial cells and fibroblasts as a major source of chymase mRNA in addition to mast cells. Chymase added to adult rat cardiomyocytes in vitro is taken up by a dynamin-mediated process and myosin breakdown is attenuated by dynamin inhibitor, suggesting that chymase uptake is essential for myosin breakdown. In a previous study in the dog model of chronic MR, the intracellular changes were attributed to extracellular effects. However, we now demonstrate intracellular effects of chymase in both species. CONCLUSION: In response to VO, fibroblast and endothelial cells produce chymase and subsequent cardiomyocyte chymase uptake is followed by myosin degradation. The results demonstrate a novel intracellular chymase-mediated mechanism of cardiomyocyte dysfunction and adverse remodeling in a pure VO.

4.
J Mol Cell Cardiol ; 92: 1-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26807691

ABSTRACT

BACKGROUND: Previous work has identified mast cells as the major source of chymase largely associated with a profibrotic phenotype. We recently reported increased fibroblast autophagic procollagen degradation in a rat model of pure volume overload (VO). Here we demonstrate a connection between increased fibroblast chymase production and autophagic digestion of procollagen in the pure VO of aortocaval fistula (ACF) in the rat. METHODS AND RESULTS: Isolated LV fibroblasts taken from 4 and 12week ACF Sprague-Dawley rats have significant increases in chymase mRNA and chymase activity. Increased intracellular chymase protein is documented by immunocytochemistry in the ACF fibroblasts compared to cells obtained from age-matched sham rats. To implicate VO as a stimulus for chymase production, we show that isolated adult rat LV fibroblasts subjected to 24h of 20% cyclical stretch induces chymase mRNA and protein production. Exogenous chymase treatment of control isolated adult cardiac fibroblasts demonstrates that chymase is internalized through a dynamin-dependent mechanism. Chymase treatment leads to an increased formation of autophagic vacuoles, LC3-II production, autophagic flux, resulting in increased procollagen degradation. Chymase inhibitor treatment reduces cyclical stretch-induced autophagy in isolated cardiac fibroblasts, demonstrating chymase's role in autophagy induction. CONCLUSION: In a pure VO model, chymase produced in adult cardiac fibroblasts leads to autophagic degradation of newly synthesized intracellular procollagen I, suggesting a new role of chymase in extracellular matrix degradation.


Subject(s)
Aorta/metabolism , Chymases/biosynthesis , Heart Failure/metabolism , Procollagen/metabolism , Animals , Aorta/pathology , Arterio-Arterial Fistula , Autophagy/genetics , Chymases/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Heart Failure/pathology , Humans , Mast Cells/metabolism , Mast Cells/pathology , Myocardium/metabolism , Myocardium/pathology , Phagosomes/metabolism , Proteolysis , RNA, Messenger/biosynthesis , Rats
5.
J Mol Cell Cardiol ; 89(Pt B): 241-250, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26596413

ABSTRACT

In a pure volume overloaded (VO) heart, interstitial collagen loss is degraded by matrix metalloproteinases (MMPs) that leads to left ventricular (LV) dilatation and heart failure. Cardiac fibroblasts are the primary source of extracellular matrix proteins that connect cardiomyocytes. The goal of this study was to determine how VO affects intracellular procollagen in cardiac fibroblasts. Using the aortocaval fistula (ACF) model in Sprague-Dawley rats, we demonstrate that cardiac fibroblasts isolated from 4 and 12 wk ACF animals have decreased intracellular procollagen I compared to the fibroblasts from age-matched shams. The reduction of procollagen I is associated with increased autophagy as demonstrated by increased autophagic vacuoles and LC3-II expression. To test the relationship between autophagy and procollagen degradation, we treated adult cardiac fibroblasts with either an autophagy inducer, rapamycin, or an inhibitor, wortmannin, and found that procollagen I protein levels were decreased in fibroblasts treated with rapamycin and elevated in wortmannin-treated cells. In addition, we demonstrated that VO induces oxidative stresses in cardiac fibroblasts from 4 and 12 wk ACF rats. Treatment of cultured cardiac fibroblasts with an oxidative stress-inducing agent (DMNQ) induces autophagy and intracellular procollagen I and fibronectin degradation, which is reversed by wortmannin but not by the global MMP inhibitor (PD166793). Mechanical stretch of cardiac fibroblasts also induces oxidative stress and autophagic degradation of procollagen I and fibronectin. Our results suggest that in addition to the well-known effects of MMPs on extracellular collagen degradation in VO, there is a concurrent degradation of intracellular procollagen and fibronectin mediated by oxidative stress-induced autophagy in cardiac fibroblasts.


Subject(s)
Autophagy , Collagen/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Myocardium/pathology , Proteolysis , Animals , Body Weight , Cell Separation , Enzyme Activation , Fibroblasts/ultrastructure , Fibronectins/metabolism , Heart Rate , Male , Matrix Metalloproteinase 2/metabolism , Oxidative Stress , Rats, Sprague-Dawley , Stress, Mechanical , Vacuoles/metabolism , Vacuoles/ultrastructure , Vascular Fistula/pathology , Vascular Fistula/physiopathology , Ventricular Function, Left , Ventricular Remodeling
6.
PLoS One ; 9(4): e94732, 2014.
Article in English | MEDLINE | ID: mdl-24733352

ABSTRACT

Cardiac ischemia and reperfusion (I/R) injury occurs because the acute increase in oxidative/inflammatory stress during reperfusion culminates in the death of cardiomyocytes. Currently, there is no drug utilized clinically that attenuates I/R injury in patients. Previous studies have demonstrated degranulation of mast cell contents into the interstitium after I/R. Using a dog model of I/R, we tested the role of chymase, a mast cell protease, in cardiomyocyte injury using a specific oral chymase inhibitor (CI). 15 adult mongrel dogs had left anterior descending artery occlusion for 60 min and reperfusion for 100 minutes. 9 dogs received vehicle and 6 were pretreated with a specific CI. In vivo cardiac microdialysis demonstrated a 3-fold increase in interstitial fluid chymase activity in I/R region that was significantly decreased by CI. CI pretreatment significantly attenuated loss of laminin, focal adhesion complex disruption, and release of troponin I into the circulation. Microarray analysis identified an I/R induced 17-fold increase in nuclear receptor subfamily 4A1 (NR4A1) and significantly decreased by CI. NR4A1 normally resides in the nucleus but can induce cell death on migration to the cytoplasm. I/R caused significant increase in NR4A1 protein expression and cytoplasmic translocation, and mitochondrial degradation, which were decreased by CI. Immunohistochemistry also revealed a high concentration of chymase within cardiomyocytes after I/R. In vitro, chymase added to culture HL-1 cardiomyocytes entered the cytoplasm and nucleus in a dynamin-dependent fashion, and promoted cytoplasmic translocation of NR4A1 protein. shRNA knockdown of NR4A1 on pre-treatment of HL-1 cells with CI significantly decreased chymase-induced cell death and mitochondrial damage. These results suggest that the beneficial effects of an orally active CI during I/R are mediated in the cardiac interstitium as well as within the cardiomyocyte due to a heretofore-unrecognized chymase entry into cardiomyocytes.


Subject(s)
Chymases/physiology , Mitochondria/metabolism , Myocytes, Cardiac/enzymology , Wounds and Injuries/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Chymases/antagonists & inhibitors , Cytoplasm/metabolism , Dogs , Endocytosis , Ischemia/pathology , Mast Cells/enzymology , Matrix Metalloproteinase 9/metabolism , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Oligonucleotide Array Sequence Analysis , Peptide Hydrolases/chemistry , Reperfusion Injury , Transferrin/metabolism , Troponin I/blood
7.
Ther Adv Cardiovasc Dis ; 8(3): 97-118, 2014 06 01.
Article in English | MEDLINE | ID: mdl-24692245

ABSTRACT

OBJECTIVES: Heart failure is typically preceded by myocardial hypertrophy and remodeling, which can be concentric due to pressure overload (PO), or eccentric because of volume overload (VO). The molecular mechanisms that underlie these differing patterns of hypertrophy are distinct and have yet to be fully elucidated. Thus, the goal of this work is to identify novel therapeutic targets for cardiovascular conditions marked by hypertrophy that have previously been resistant to medical treatment, such as a pure VO. METHODS: Concentric or eccentric hypertrophy was induced in rats for 2 weeks with transverse aortic constriction (TAC) or aortocaval fistula (ACF), respectively. Hemodynamic and echocardiographic analysis were used to assess the development of left ventricular (LV) hypertrophy and functional differences between groups. Changes in gene expression were determined by microarray and further characterized with Ingenuity Pathway Analysis. RESULTS: Both models of hypertrophy increased LV mass. Rats with TAC demonstrated concentric LV remodeling while rats with ACF exhibited eccentric LV remodeling. Microarray analysis associated eccentric remodeling with a more extensive alteration of gene expression compared with concentric remodeling. Rats with VO had a marked activation of extracellular matrix genes, promotion of cell cycle genes, downregulation of genes associated with oxidative metabolism, and dysregulation of genes critical to cardiac contractile function. Rats with PO demonstrated similar categorical changes, but with the involvement of fewer individual genes. CONCLUSIONS: Our results indicate that eccentric remodeling is a far more complex process than concentric remodeling. This study highlights the importance of several key biological functions early in the course of VO, including regulation of matrix, metabolism, cell proliferation, and contractile function. Thus, the results of this analysis will inform the ongoing search for new treatments to prevent the progression to heart failure in VO.

8.
Circ Heart Fail ; 7(1): 194-202, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24297688

ABSTRACT

BACKGROUND: There is currently no therapy proven to attenuate left ventricular (LV) dilatation and dysfunction in volume overload induced by isolated mitral regurgitation (MR). To better understand molecular signatures underlying isolated MR, we performed LV gene expression analyses and overlaid regulated genes into ingenuity pathway analysis in patients with isolated MR. METHODS AND RESULTS: Gene arrays from LV tissue of 35 patients, taken at the time of surgical repair for isolated MR, were compared with 13 normal controls. Cine-MRI was performed in 31 patients before surgery to measure LV function and volume from serial short-axis summation. LV end-diastolic volume was 2-fold (P=0.005) higher in MR patients than in normal controls, and LV ejection fraction was 64±7% (50%-79%) in MR patients. Ingenuity pathway analysis identified significant activation of pathways involved in ß-adrenergic, cAMP, and G-protein-coupled signaling, whereas there was downregulation of pathways associated with complement activation and acute phase response. SERCA2a and phospholamban protein were unchanged in MR versus control left ventricles. However, mRNA and protein levels of the sarcoplasmic reticulum Ca2+ ATPase (SERCA) regulatory protein sarcolipin, which is predominantly expressed in normal atria, were increased 12- and 6-fold, respectively. Immunofluorescence analysis confirmed the absence of sarcolipin in normal left ventricles and its marked upregulation in MR left ventricles. CONCLUSIONS: These results demonstrate alterations in multiple pathways associated with ß-adrenergic signaling and sarcolipin in the left ventricles of patients with isolated MR and LV ejection fraction>50%, suggesting a beneficial role for ß-adrenergic blockade in isolated MR.


Subject(s)
Adrenergic Agents/metabolism , Mitral Valve Insufficiency/metabolism , Muscle Proteins/metabolism , Proteolipids/metabolism , Stroke Volume/physiology , Ventricular Dysfunction, Left/metabolism , Adult , Aged , Biopsy , Case-Control Studies , Female , Gene Expression Profiling , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Mitral Valve Insufficiency/physiopathology , Muscle Proteins/genetics , Proteolipids/genetics , Signal Transduction/physiology , Up-Regulation , Ventricular Dysfunction, Left/physiopathology
9.
Life Sci ; 92(11): 648-56, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-22728715

ABSTRACT

AIMS: Increased O-linked attachment of ß-N-acetylglucosamine (O-GlcNAc) to proteins has been implicated in the adverse effects of diabetes on the heart, although this has typically been based on models of severe hyperglycemia. Diabetes has also been associated with dysregulation of autophagy, a critical cell survival process; however, little is known regarding autophagy in the diabetic heart or whether this is influenced by O-GlcNAcylation or hemodynamic stress. MAIN METHODS: Young male rats were assigned to control (12% kcal fat/19% protein/69% carbohydrate), high fat diet (60/19/21%) and type 2 diabetic (high fat diet+low dose streptozotocin) groups for 8 weeks, followed by sham or pressure overload surgeries; animals were sacrificed 8 weeks after surgery. KEY FINDINGS: A modest increase in arterial pressure resulted in no significant effects on cardiac function in control or high fat groups, while diabetic hearts exhibited contractile dysfunction and increased apoptosis and scar formation. Immunoprecipitation studies revealed, for the first time, that Beclin-1, which plays a critical early role in autophagy, and the anti-apoptotic Bcl-2, are targets for O-GlcNAcylation. Interestingly, we also found that cardiomyocytes isolated from type 2 diabetic db/db mice exhibited a blunted autophagic response and this was at least partially reversed by inhibiting glucose entry into the hexosamine biosynthesis pathway, which regulates O-GlcNAc synthesis. We also found that acutely augmenting O-GlcNAc levels in non-diabetic cardiomyocytes mimicked the effects of diabetes by blunting autophagic signaling. SIGNIFICANCE: These data suggest that O-GlcNAc-mediated inhibition of autophagy may contribute to the abnormal response of diabetic hearts to hemodynamic stress.


Subject(s)
Acetylglucosamine/metabolism , Autophagy/physiology , Diabetic Cardiomyopathies/physiopathology , Myocytes, Cardiac/pathology , Signal Transduction , Acylation/physiology , Animals , Arterial Pressure , Body Weight , Disease Models, Animal , Hyperglycemia/physiopathology , Male , Random Allocation , Rats , Rats, Sprague-Dawley
10.
PLoS One ; 7(6): e40110, 2012.
Article in English | MEDLINE | ID: mdl-22768235

ABSTRACT

BACKGROUND: The clinical problem of a "pure volume overload" as in isolated mitral or aortic regurgitation currently has no documented medical therapy that attenuates collagen loss and the resultant left ventricular (LV) dilatation and failure. Here, we identify a potential mechanism related to upregulation of the kallikrein-kinin system in the volume overload of aortocaval fistula (ACF) in the rat. METHODOLOGY/PRINCIPAL FINDINGS: LV interstitial fluid (ISF) collection, hemodynamics, and echocardiography were performed in age-matched shams and 4 and 15 wk ACF rats. ACF rats had LV dilatation and a 2-fold increase in LV end-diastolic pressure, along with increases in LV ISF bradykinin, myocardial kallikrein and bradykinin type-2 receptor (BK(2)R) mRNA expression. Mast cell numbers were increased and interstitial collagen was decreased at 4 and 15 wk ACF, despite increases in LV ACE and chymase activities. Treatment with the kallikrein inhibitor aprotinin preserved interstitial collagen, prevented the increase in mast cells, and improved LV systolic function at 4 wk ACF. To establish a cause and effect between ISF bradykinin and mast cell-mediated collagen loss, direct LV interstitial bradykinin infusion in vivo for 24 hrs produced a 2-fold increase in mast cell numbers and a 30% decrease in interstitial collagen, which were prevented by BK(2)R antagonist. To further connect myocardial stretch with cellular kallikrein-kinin system upregulation, 24 hrs cyclic stretch of adult cardiomyocytes and fibroblasts produced increased kallikrein, BK(2)R mRNA expressions, bradykinin protein and gelatinase activity, which were all decreased by the kallikrein inhibitor-aprotinin. CONCLUSIONS/SIGNIFICANCE: A pure volume overload is associated with upregulation of the kallikrein-kinin system and ISF bradykinin, which mediates mast cell infiltration, extracellular matrix loss, and LV dysfunction-all of which are improved by kallikrein inhibition. The current investigation provides important new insights into future potential medical therapies for the volume overload of aortic and mitral regurgitation.


Subject(s)
Collagen/metabolism , Inflammation/pathology , Kallikrein-Kinin System , Myocardium/pathology , Up-Regulation , Ventricular Remodeling , Angiotensin II/blood , Angiotensin-Converting Enzyme 2 , Animals , Aprotinin/pharmacology , Bradykinin/blood , Catecholamines/blood , Cell Count , Cell Degranulation/drug effects , Chymases/metabolism , Extracellular Fluid , Gelatinases/metabolism , Heart Ventricles/diagnostic imaging , Heart Ventricles/enzymology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Immunohistochemistry , Inflammation/complications , Inflammation/genetics , Kallikrein-Kinin System/drug effects , Mast Cells/drug effects , Mast Cells/enzymology , Mast Cells/physiology , Models, Cardiovascular , Myocardium/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Bradykinin/metabolism , Ultrasonography , Up-Regulation/drug effects , Vascular Fistula/diagnostic imaging , Vascular Fistula/genetics , Vascular Fistula/pathology , Vascular Fistula/physiopathology , Ventricular Remodeling/drug effects
11.
Circulation ; 122(15): 1488-95, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20876440

ABSTRACT

BACKGROUND: The left ventricular (LV) dilatation of isolated mitral regurgitation (MR) is associated with an increase in chymase and a decrease in interstitial collagen and extracellular matrix. In addition to profibrotic effects, chymase has significant antifibrotic actions because it activates matrix metalloproteinases and kallikrein and degrades fibronectin. Thus, we hypothesize that chymase inhibitor (CI) will attenuate extracellular matrix loss and LV remodeling in MR. METHODS AND RESULTS: We studied dogs with 4 months of untreated MR (MR; n=9) or MR treated with CI (MR+CI; n=8). Cine MRI demonstrated a >40% increase in LV end-diastolic volume in both groups, consistent with a failure of CI to improve a 25% decrease in interstitial collagen in MR. However, LV cardiomyocyte fractional shortening was decreased in MR versus normal dogs (3.71±0.24% versus 4.81±0.31%; P<0.05) and normalized in MR+CI dogs (4.85±0.44%). MRI with tissue tagging demonstrated an increase in LV torsion angle in MR+CI versus MR dogs. CI normalized the significant decrease in fibronectin and FAK phosphorylation and prevented cardiomyocyte myofibrillar degeneration in MR dogs. In addition, total titin and its stiffer isoform were increased in the LV epicardium and paralleled the changes in fibronectin and FAK phosphorylation in MR+CI dogs. CONCLUSIONS: These results suggest that chymase disrupts cell surface-fibronectin connections and FAK phosphorylation that can adversely affect cardiomyocyte myofibrillar structure and function. The greater effect of CI on epicardial versus endocardial titin and noncollagen cell surface proteins may be responsible for the increase in torsion angle in chronic MR.


Subject(s)
Chymases/antagonists & inhibitors , Fibronectins/metabolism , Mitral Valve Insufficiency/physiopathology , Myocytes, Cardiac/physiology , Myofibrils/metabolism , Torsion Abnormality/physiopathology , Ventricular Remodeling/physiology , Animals , Blood Pressure/physiology , Bradykinin/metabolism , Cardiac Output/physiology , Collagen/metabolism , Dogs , Extracellular Matrix/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Heart Rate/physiology , Male , Mitral Valve Insufficiency/metabolism , Models, Animal , Myocytes, Cardiac/cytology , Torsion Abnormality/metabolism
12.
J Card Fail ; 16(9): 769-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20797601

ABSTRACT

BACKGROUND: Mast cells are increased in isolated mitral regurgitation (MR) in the dog and may mediate extracellular matrix loss and left ventricular (LV) dilatation. We tested the hypothesis that mast cell stabilization would attenuate LV remodeling and improve function in the MR dog. METHODS AND RESULTS: MR was induced in adult dogs randomized to no treatment (MR, n = 5) or to the mast cell stabilizer, ketotifen (MR + MCS, n = 4) for 4 months. LV hemodynamics were obtained at baseline and after 4 months of MR and magnetic resonance imaging (MRI) was performed at sacrifice. MRI-derived, serial, short-axis LV end-diastolic (ED) and end-systolic (ES) volumes, LVED volume/mass ratio, and LV 3-dimensional radius/wall thickness were increased in MR and MR + MCS dogs compared with normal dogs (n = 6) (P < .05). Interstitial collagen was decreased by 30% in both MR and MR + MCS versus normal dogs (P < .05). LV contractility by LV maximum time-varying elastance was significantly depressed in MR and MR + MCS dogs. Furthermore, cardiomyocyte fractional shortening was decreased in MR versus normal dogs and further depressed in MR + MCS dogs (P < .05). In vitro administration of ketotifen to normal cardiomyocytes also significantly decreased fractional shortening and calcium transients. CONCLUSIONS: Chronic mast cell stabilization did not attenuate eccentric LV remodeling or collagen loss in MR. However, MCS therapy had a detrimental effect on LV function because of a direct negative inotropic effect on cardiomyocyte function.


Subject(s)
Heart Ventricles/drug effects , Mast Cells/drug effects , Mitral Valve Insufficiency/physiopathology , Myocytes, Cardiac/drug effects , Ventricular Function, Left/drug effects , Adrenergic beta-Agonists/pharmacology , Analysis of Variance , Animals , Anti-Allergic Agents/therapeutic use , Collagen/drug effects , Dogs , Extracellular Matrix , Heart Ventricles/pathology , Hemodynamics/drug effects , Isoproterenol/pharmacology , Ketotifen/therapeutic use , Magnetic Resonance Imaging , Ventricular Remodeling
13.
J Clin Invest ; 120(4): 1229-39, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20335663

ABSTRACT

Ang I-converting enzyme (ACE) inhibitors are widely believed to suppress the deleterious cardiac effects of Ang II by inhibiting locally generated Ang II. However, the recent demonstration that chymase, an Ang II-forming enzyme stored in mast cell granules, is present in the heart has added uncertainty to this view. As discussed here, using microdialysis probes tethered to the heart of conscious mice, we have shown that chronic ACE inhibitor treatment did not suppress Ang II levels in the LV interstitial fluid (ISF) despite marked inhibition of ACE. However, chronic ACE inhibition caused a marked bradykinin/B2 receptor-mediated increase in LV ISF chymase activity that was not observed in mast cell-deficient KitW/KitW-v mice. In chronic ACE inhibitor-treated mast cell-sufficient littermates, chymase inhibition decreased LV ISF Ang II levels substantially, indicating the importance of mast cell chymase in regulating cardiac Ang II levels. Chymase-dependent processing of other regulatory peptides also promotes inflammation and tissue remodeling. We found that combined chymase and ACE inhibition, relative to ACE inhibition alone, improved LV function, decreased adverse cardiac remodeling, and improved survival after myocardial infarction in hamsters. These results suggest that chymase inhibitors could be a useful addition to ACE inhibitor therapy in the treatment of heart failure.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Chymases/physiology , Myocardial Infarction/drug therapy , Angiotensin II/antagonists & inhibitors , Angiotensin II/biosynthesis , Animals , Bradykinin/physiology , Cell Degranulation , Chymases/antagonists & inhibitors , Cricetinae , Mast Cells/enzymology , Mast Cells/physiology , Mesocricetus , Mice , Mice, Inbred C57BL , Microdialysis , Myocardial Infarction/physiopathology , Serine Endopeptidases/physiology , Ventricular Function, Left/drug effects
14.
Circulation ; 119(15): 2086-95, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19349319

ABSTRACT

BACKGROUND: The volume overload of isolated mitral regurgitation (MR) in the dog results in left ventricular (LV) dilatation and interstitial collagen loss. To better understand the mechanism of collagen loss, we performed a gene array and overlaid regulated genes into ingenuity pathway analysis. METHODS AND RESULTS: Gene arrays from LV tissue were compared in 4 dogs before and 4 months after MR. Cine-magnetic resonance-derived LV end-diastolic volume increased 2-fold (P=0.005), and LV ejection fraction increased from 41% to 53% (P<0.007). LV interstitial collagen decreased 40% (P<0.05) compared with controls, and replacement collagen was in short strands and in disarray. Ingenuity pathway analysis identified Marfan syndrome, aneurysm formation, LV dilatation, and myocardial infarction, all of which have extracellular matrix protein defects and/or degradation. Matrix metalloproteinase-1 and -9 mRNA increased 5- (P=0.01) and 10-fold (P=0.003), whereas collagen I did not change and collagen III mRNA increased 1.5-fold (P=0.02). However, noncollagen genes important in extracellular matrix structure were significantly downregulated, including decorin, fibulin 1, and fibrillin 1. In addition, connective tissue growth factor and plasminogen activator inhibitor were downregulated, along with multiple genes in the transforming growth factor-beta signaling pathway, resulting in decreased LV transforming growth factor-beta1 activity (P=0.03). CONCLUSIONS: LV collagen loss in isolated, compensated MR is chiefly due to posttranslational processing and degradation. The downregulation of multiple noncollagen genes important in global extracellular matrix structure, coupled with decreased expression of multiple profibrotic factors, explains the failure to replace interstitial collagen in the MR heart.


Subject(s)
Extracellular Matrix Proteins/biosynthesis , Gene Expression Regulation , Heart Ventricles/metabolism , Mitral Valve Insufficiency/genetics , Oligonucleotide Array Sequence Analysis , Transforming Growth Factor beta1/biosynthesis , Animals , Chronic Disease , Collagen/biosynthesis , Collagen/genetics , Decorin , Dogs , Down-Regulation , Extracellular Matrix Proteins/genetics , Female , Fibrosis , Heart Ventricles/pathology , Integrin alphaV/biosynthesis , Integrin alphaV/genetics , Magnetic Resonance Imaging , Male , Mitral Valve Insufficiency/metabolism , Mitral Valve Insufficiency/pathology , Organ Size , Phosphorylation , Protein Processing, Post-Translational , Proteoglycans/biosynthesis , Proteoglycans/genetics , Smad2 Protein/metabolism , Stroke Volume , Transforming Growth Factor beta1/genetics
15.
Circ Res ; 102(6): 677-85, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18258857

ABSTRACT

c-kit, the transmembrane tyrosine kinase receptor for stem cell factor, is required for melanocyte and mast cell development, hematopoiesis, and differentiation of spermatogonial stem cells. We show here that in the heart, c-kit is expressed not only by cardiac stem cells but also by cardiomyocytes, commencing immediately after birth and terminating a few days later, coincident with the onset of cardiomyocyte terminal differentiation. To examine the function of c-kit in cardiomyocyte terminal differentiation, we used compound heterozygous mice carrying the W (null) and W(v) (dominant negative) mutations of c-kit. In vivo, adult W/W(v) cardiomyocytes are phenotypically indistinguishable from their wild-type counterparts. After acute pressure overload adult W/W(v) cardiomyocytes reenter the cell cycle and proliferate, leading to left ventricular growth; furthermore in transgenic mice with cardiomyocyte-restricted overexpression of the dominant negative W(v) mutant, pressure overload causes cardiomyocytes to reenter the cell cycle. In contrast, in wild-type mice left ventricular growth after pressure overload results mainly from cardiomyocyte hypertrophy. Importantly, W/W(v) mice with pressure overload-induced cardiomyocyte hyperplasia had improved left ventricular function and survival. In W/W(v) mice, c-kit dysfunction also resulted in an approximately 14-fold decrease (P<0.01) in the number of c-kit(+)/GATA4(+) cardiac progenitors. These findings identify novel functions for c-kit: promotion of cardiac stem cell differentiation and regulation of cardiomyocyte terminal differentiation.


Subject(s)
Cell Differentiation , Hypertrophy, Left Ventricular/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/metabolism , Aging/metabolism , Animals , Animals, Newborn , Aorta/surgery , Blood Pressure , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Developmental , Genotype , Heart Ventricles/embryology , Heart Ventricles/growth & development , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Ligation , Male , Mice , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/pathology , Phenotype , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/metabolism , Stem Cells/pathology , Time Factors , Ventricular Function, Left
16.
Am J Physiol Heart Circ Physiol ; 293(1): H292-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17351065

ABSTRACT

Recent studies in our laboratory using the Zucker obese (ZO) and Zucker diabetic fatty (ZDF) rat models resulted in unexpectedly high mortality rates in all genotypes including healthy homozygous lean Zucker rats, possibly because of renal dysfunction. Therefore, we evaluated left ventricular (LV) and kidney morphology and function in young ZO, Zucker diabetic fatty obese (ZDFO), homozygous Zucker/ZDF lean (ZL), and Sprague-Dawley (SD) rats. Hydronephrosis was evident in ZL, ZO, and ZDFO but not SD kidneys. ZDFO rats exhibited impaired LV shortening and relaxation with increased arterial stiffness. LV wall thickness was lower and LV end-systolic wall stress was higher in ZDFO compared with SD rats. Plasma ANG II was lower in ZO and ZDFO rats, which may be a result of reduced renal parenchyma with hydronephrosis; norepinephrine was higher in ZDFO rats than SD controls. Covariate analysis indicated that LV end-systolic wall stress was associated with renal dysfunction. The presence of hydronephrosis and its association with LV dysfunction potentially limits the ZDF model for study of the effects of diabetes on renal and cardiovascular function.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Heart Ventricles/physiopathology , Hydronephrosis/physiopathology , Obesity/physiopathology , Ventricular Dysfunction, Left/physiopathology , Animals , Diabetes Mellitus, Experimental/pathology , Heart Ventricles/pathology , Hydronephrosis/pathology , Kidney/pathology , Kidney/physiopathology , Obesity/pathology , Rats , Rats, Zucker , Species Specificity , Ventricular Dysfunction, Left/complications , Ventricular Dysfunction, Left/pathology
17.
Free Radic Biol Med ; 40(6): 952-9, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540390

ABSTRACT

Xanthine oxidoreductase (XOR) activity has been previously noted to be responsive to changes in O2 tension. While prior studies have focused on the extremes (0-3% and 95-100%) of O2 tensions, we report the influence of 10% O2 on endothelial cell XOR, a concentration resembling modest arterial hypoxia commonly found in patients with chronic cardiopulmonary diseases. Exposure of bovine aortic endothelial cells to 10% O2 increased XOR mRNA and protein abundance by 50%. Concomitantly, there was a 3-fold increase in XOR activity, XOR-dependent reactive oxygen species production, and cellular export of active enzyme. Although increases in mRNA and immunoreactive protein levels were observed, inhibition of transcription, translation, or protein degradation did not significantly alter cellular XOR specific activity, suggesting only modest contributions to 10% O2-induced effects. Exposure to 10% O2 did not increase cellular HIF-1alpha protein levels and hypoxia mimics did not alter XOR activity. Treatment of control cells with adenosine resulted in increased XOR activity similar to hypoxia. Exposure to the adenosine receptor agonist NECA increased enzymatic activity 4-fold while 8SPT, an adenosine receptor antagonist, reduced hypoxic induction of XOR activity approximately 50%. Combined, these data reveal that moderate hypoxia significantly enhances endothelial XOR specific activity, release, and XOR-derived reactive oxygen species generation. These effects appear to be mediated in part via adenosine-dependent processes.


Subject(s)
Hypoxia/enzymology , Xanthine Oxidase/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Aorta/cytology , Cattle , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Induction , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Oxygen/pharmacology , Purinergic P1 Receptor Antagonists , Reactive Oxygen Species/metabolism
18.
Am J Physiol Heart Circ Physiol ; 285(2): H784-92, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12663259

ABSTRACT

In the current study, interstitial fluid (ISF), bradykinin (BK), and angiotensin II (ANG II) levels were measured using cardiac microdialysis in conscious, nonsedated rats at baseline and at 48 h and 5 days after each of the following: sham surgery (sham, n = 6), sham + administration of ANG-converting enzyme inhibitor ramipril (R, n = 6), creation of aortocaval fistula (ACF, n = 6), ACF + R (n = 6), and ACF + R + BK2 receptor antagonist (HOE-140) administration (n = 6). At 5 days, both ISF ANG II and BK increased in ACF rats (P < 0.05); however, in ACF + R rats, ISF ANG II did not differ from basal levels and ISF BK increased greater than threefold above baseline at 2 and 5 days (P < 0.05). Five days after ACF, the left ventricular (LV) weight-to-body weight ratio increased 30% (P < 0.05) in ACF but did not differ from sham in ACF + R and ACF + R + HOE-140 rats despite similar systemic arterial pressures across all ACF groups. However, ACF + R + HOE-140 rats had greater postmortem wall thickness-to-diameter ratio and smaller cross-sectional diameter compared with ACF + R rats. There was a significant increase in mast cell density in ACF and ACF + R rats that decreased below sham in ACF + R + HOE-140 rats. These results suggest a potentially important interaction of mast cells and BK in the cardiac interstitium that modulates the pattern of LV remodeling in the acute phase of volume overload.


Subject(s)
Bradykinin/metabolism , Mast Cells/metabolism , Myocardium/metabolism , Myocardium/pathology , Ventricular Remodeling/physiology , Acute Disease , Angiotensin II/blood , Angiotensin II/metabolism , Animals , Aorta , Arteriovenous Fistula , Bradykinin/blood , Cardiac Volume/physiology , Cell Count , Chymases , Consciousness , Disease Models, Animal , Extracellular Space/metabolism , Mast Cells/cytology , Microdialysis , Peptidyl-Dipeptidase A/metabolism , Rats , Rats, Sprague-Dawley , Serine Endopeptidases/metabolism , Vena Cava, Inferior
SELECTION OF CITATIONS
SEARCH DETAIL
...