Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(12)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207723

ABSTRACT

A hypofibrotic phenotype has been observed in cardiac fibroblasts (CFs) isolated from a volume overload heart failure model, aortocaval fistula (ACF). This paradoxical phenotype results in decreased ECM synthesis despite increased TGF-ß presence. Since ACF results in decreased tissue stiffness relative to control (sham) hearts, this study investigates whether the effects of substrate stiffness could account for the observed hypofibrotic phenotype in CFs isolated from ACF. CFs isolated from ACF and sham hearts were plated on polyacrylamide gels of a range of stiffness (2 kPa to 50 kPa). Markers related to cytoskeletal and fibrotic proteins were measured. Aspects of the hypofibrotic phenotype observed in ACF CFs were recapitulated by sham CFs on soft substrates. For instance, sham CFs on the softest gels compared to ACF CFs on the stiffest gels results in similar CTGF (0.80 vs. 0.76) and transgelin (0.44 vs. 0.57) mRNA expression. The changes due to stiffness may be explained by the observed decreased nuclear translocation of transcriptional regulators, MRTF-A and YAP. ACF CFs appear to have a mechanical memory of a softer environment, supported by a hypofibrotic phenotype overall compared to sham with less YAP detected in the nucleus, and less CTGF and transgelin on all stiffnesses.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation , Heart Failure/metabolism , Myocardium/metabolism , Stress, Mechanical , Animals , Fibroblasts/pathology , Fibrosis , Heart Failure/pathology , Male , Myocardium/pathology , Rats , Rats, Sprague-Dawley
2.
J Cardiovasc Pharmacol ; 77(6): 707-717, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34016841

ABSTRACT

ABSTRACT: Arachidonic acid-derived lipid mediators play crucial roles in the development and progression of cardiovascular diseases. Eicosanoid metabolites generated by lipoxygenases and cytochrome P450 enzymes produce several classes of molecules, including the epoxyeicosatrienoic acid (EET) and hydroxyeicosatetraenoic acids (HETE) family of bioactive lipids. In general, the cardioprotective effects of EETs have been documented across a number of cardiac diseases. In contrast, members of the HETE family have been shown to contribute to the pathogenesis of ischemic cardiac disease, maladaptive cardiac hypertrophy, and heart failure. The net effect of 12(S)- and 20-HETE depends upon the relative amounts generated, ratio of HETEs:EETs produced, timing of synthesis, as well as cellular and subcellular mechanisms activated by each respective metabolite. HETEs are synthesized by and affect multiple cell types within the myocardium. Moreover, cytochrome P450-derived and lipoxygenase- derived metabolites have been shown to directly influence cardiac myocyte growth and the regulation of cardiac fibroblasts. The mechanistic data uncovered thus far have employed the use of enzyme inhibitors, HETE antagonists, and the genetic manipulation of lipid-producing enzymes and their respective receptors, all of which influence a complex network of outcomes that complicate data interpretation. This review will summarize and integrate recent findings on the role of 12(S)-/20-HETE in cardiac diseases.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Cardiovascular Diseases/physiopathology , Hydroxyeicosatetraenoic Acids/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/antagonists & inhibitors , Animals , Animals, Genetically Modified , Cardiomegaly/physiopathology , Cytochrome P-450 Enzyme System/metabolism , Heart Failure/physiopathology , Humans , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Myocardial Ischemia/physiopathology , Receptors, G-Protein-Coupled/metabolism
3.
J Biomech Eng ; 143(8)2021 08 01.
Article in English | MEDLINE | ID: mdl-33729495

ABSTRACT

Pressure overload (PO) and volume overload (VO) of the heart result in distinctive changes to geometry, due to compensatory structural remodeling. This remodeling potentially leads to changes in tissue mechanical properties. Understanding such changes is important, as tissue modulus has an impact on cardiac performance, disease progression, and influences on cell phenotype. Pressure-volume (PV) loop analysis, a clinically relevant method for measuring left ventricular (LV) chamber stiffness, was performed in vivo on control rat hearts and rats subjected to either chronic PO through Angiotensin-II infusion (4-weeks) or VO (8-weeks). Immediately following PV loops, biaxial testing was performed on LV free wall tissue to directly measure tissue mechanical properties. The ß coefficient, an index of chamber stiffness calculated from the PV loop analysis, increased 98% in PO (n = 4) and decreased 38% in VO (n = 5) compared to control (n = 6). Material constants of LV walls obtained from ex vivo biaxial testing (n = 9-10) were not changed in Angiotensin-II induced PO and decreased by about half in VO compared to control (47% in the circumferential and 57% the longitudinal direction). PV loop analysis showed the expected increase in chamber stiffness of PO and expected decrease in chamber stiffness of VO. Biaxial testing showed a decreased modulus of the myocardium of the VO model, but no changes in the PO model, this suggests the increased chamber stiffness in PO, as shown in the PV loop analysis, may be secondary to changes in tissue mass and/or geometry but not an increase in passive tissue mechanical properties.


Subject(s)
Angiotensins
4.
Pharmacy (Basel) ; 8(1)2019 Dec 20.
Article in English | MEDLINE | ID: mdl-31861770

ABSTRACT

Goodman and Gilman's The Pharmacological Basis of Therapeutics (GGPBT) has been a cornerstone in the education of pharmacists, physicians, and pharmacologists for decades. The objectives of this study were to describe and evaluate the 13th edition of GGPBT on bases including: (1) author characteristics; (2) recency of citations; (3) conflict of interest (CoI) disclosure; (4) expert evaluation of chapters. Contributors' (N = 115) sex, professional degrees, and presence of undisclosed potential CoI-as reported by the Center for Medicare and Medicaid's Open Payments (2013-2017)-were examined. The year of publication of citations was extracted relative to Katzung's Basic and Clinical Pharmacology (KatBCP), and DiPiro's Pharmacotherapy: A Pathophysiologic Approach (DiPPAPA). Content experts provided thorough chapter reviews. The percent of GGPBT contributors that were female (20.9%) was equivalent to those in KatBCP (17.0%). Citations in GGPBT (11.5 ± 0.2 years) were significantly older than those in KatBCP (10.4 ± 0.2) and DiPPAPA (9.1 ± 0.1, p < 0.0001). Contributors to GGPBT received USD 3 million in undisclosed remuneration (Maximum author = USD 743,718). In contrast, DiPPAPA made CoI information available. Reviewers noted several strengths but also some areas for improvement. GGPBT will continue to be an important component of the biomedical curriculum. Areas of improvement include a more diverse authorship, improved conflict of interest transparency, and a greater inclusion of more recent citations.

5.
Am J Physiol Heart Circ Physiol ; 316(3): H596-H608, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30575422

ABSTRACT

Hemodynamic load regulates cardiac remodeling. In contrast to pressure overload (increased afterload), hearts subjected to volume overload (VO; preload) undergo a distinct pattern of eccentric remodeling, chamber dilation, and decreased extracellular matrix content. Critical profibrotic roles of cardiac fibroblasts (CFs) in postinfarct remodeling and in response to pressure overload have been well established. Little is known about the CF phenotype in response to VO. The present study characterized the phenotype of primary cultures of CFs isolated from hearts subjected to 4 wk of VO induced by an aortocaval fistula. Compared with CFs isolated from sham hearts, VO CFs displayed a "hypofibrotic" phenotype, characterized by a ~50% decrease in the profibrotic phenotypic markers α-smooth muscle actin, connective tissue growth factor, and collagen type I, despite increased levels of profibrotic transforming growth factor-ß1 and an intact canonical transforming growth factor-ß signaling pathway. Actin filament dynamics were characterized, which regulate the CF phenotype in response to biomechanical signals. Actin polymerization was determined by the relative amounts of G-actin monomers versus F-actin. Compared with sham CFs, VO CFs displayed ~78% less F-actin and an increased G-actin-to-F-actin ratio (G/F ratio). In sham CFs, treatment with the Rho kinase inhibitor Y-27632 to increase the G/F ratio resulted in recapitulation of the hypofibrotic CF phenotype observed in VO CFs. Conversely, treatment of VO CFs with jasplakinolide to decrease the G/F ratio restored a more profibrotic response (>2.5-fold increase in α-smooth muscle actin, connective tissue growth factor, and collagen type I). NEW & NOTEWORTHY The present study is the first to describe a "hypofibrotic" phenotype of cardiac fibroblasts isolated from a volume overload model. Our results suggest that biomechanical regulation of actin microfilament stability and assembly is a critical mediator of cardiac fibroblast phenotypic modulation.


Subject(s)
Cytoskeleton/metabolism , Fibroblasts/metabolism , Heart Failure/metabolism , Actins/metabolism , Animals , Cells, Cultured , Collagen/metabolism , Fibroblasts/pathology , Heart Failure/pathology , Male , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Phenotype , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/metabolism
6.
J Vis Exp ; (111)2016 05 30.
Article in English | MEDLINE | ID: mdl-27285607

ABSTRACT

While the isolation and culture of vascular smooth muscle cells (VSMCs) from large vessels is well established, we sought to isolate and culture VSMCs from the coronary circulation. Hearts with intact aortic arches were removed and perfused via retrograde Langendorff with digestion solution containing 300 Units/ml of collagenase type II, 0.1 mg/ml soybean trypsin inhibitor and 1 M CaCl2. The perfusates were collected at 15 min intervals for 90 min, pelleted by centrifugation, resuspended in plating media, and plated on tissue culture dishes. VSMCs were characterized by presence of SM22α, α-SMA, and vimentin. One of the main advantages of using this technique is the ability to isolate VSMCs from the coronary circulation of mice. Although the small number of cells obtained can limit some of the applications for which the cells can be utilized, isolated coronary VSMCs can be used in a variety of well-established cell culture techniques and assays. Studies investigating VSMCs from genetically modified mice can provide further information about structure-function and signaling processes associated with vascular pathologies.


Subject(s)
Cell Culture Techniques , Cell Separation , Coronary Vessels/cytology , Myocytes, Smooth Muscle , Animals , Cells, Cultured , Mice , Muscle, Smooth, Vascular/cytology
7.
Gene Expr ; 17(1): 47-59, 2016.
Article in English | MEDLINE | ID: mdl-27342733

ABSTRACT

Early life exposures can increase the risk of developing chronic diseases including nonalcoholic fatty liver disease. Maternal high-fat diet increases susceptibility to development of steatosis in the offspring. We determined the effect of maternal high-fat diet exposure in utero and during lactation on offspring liver histopathology, particularly fibrosis. Female C57Bl/6J mice were fed a control or high-fat diet (HFD) for 8 weeks and bred with lean males. Nursing dams were continued on the same diet with offspring sacrificed during the perinatal period or maintained on either control or high-fat diet for 12 weeks. Increased hepatocyte proliferation and stellate cell activation were observed in the liver of HFD-exposed pups. Offspring exposed to perinatal high-fat diet and high-fat diet postweaning showed extensive hepatosteatosis compared to offspring on high-fat diet after perinatal control diet. Offspring exposed to perinatal high-fat diet and then placed on control diet for 12 weeks developed steatosis and pericellular fibrosis. Importantly, we found that exposure to perinatal high-fat diet unexpectedly promotes more rapid disease progression of nonalcoholic fatty liver disease, with a sustained fibrotic phenotype, only in adult offspring fed a postweaning control diet.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/etiology , Fibrosis/etiology , Liver/pathology , Prenatal Exposure Delayed Effects/etiology , Animals , Cell Proliferation/physiology , Disease Progression , Fatty Liver/pathology , Female , Fibrosis/pathology , Hepatocytes/pathology , Lactation/physiology , Male , Maternal Exposure , Maternal Nutritional Physiological Phenomena/physiology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology
8.
Vascul Pharmacol ; 76: 28-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26133668

ABSTRACT

Cardiovascular complications are a leading cause of morbidity and mortality in type 2 diabetes mellitus (T2DM) and are associated with alterations of blood vessel structure and function. Although endothelial dysfunction and aortic stiffness have been documented, little is known about the effects of T2DM on coronary microvascular structural remodeling. The renin-angiotensin-aldosterone system plays an important role in large artery stiffness and mesenteric vessel remodeling in hypertension and T2DM. The goal of this study was to determine whether the blockade of AT1R signaling dictates vascular smooth muscle growth that partially underlies coronary arteriole remodeling in T2DM. Control and db/db mice were given AT1R blocker losartan via drinking water for 4 weeks. Using pressure myography, we found that coronary arterioles from 16-week db/db mice undergo inward hypertrophic remodeling due to increased wall thickness and wall-to-lumen ratio with a decreased lumen diameter. This remodeling was accompanied by decreased elastic modulus (decreased stiffness). Losartan treatment decreased wall thickness, wall-to-lumen ratio, and coronary arteriole cell number in db/db mice. Losartan treatment did not affect incremental elastic modulus. However, losartan improved coronary flow reserve. Our data suggest that Ang II-AT1R signaling mediates, at least in part, coronary arteriole inward hypertrophic remodeling in T2DM without affecting vascular mechanics, further suggesting that targeting the coronary microvasculature in T2DM may help reduce cardiac ischemic events.


Subject(s)
Angiotensin Receptor Antagonists/pharmacology , Arterioles/drug effects , Coronary Vessels/drug effects , Diabetes Mellitus, Experimental/diet therapy , Diabetes Mellitus, Type 2/drug therapy , Losartan/pharmacology , Angiotensin II/pharmacology , Animals , Arterioles/metabolism , Blood Pressure/drug effects , Coronary Vessels/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypertension/drug therapy , Hypertension/metabolism , Male , Mice , Microvessels/drug effects , Microvessels/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Pilot Projects , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/drug effects
9.
Ann Thorac Surg ; 100(3): 1013-9; discussion 1019-20, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26163359

ABSTRACT

BACKGROUND: The hybrid palliation for hypoplastic left heart syndrome has emerged as an alternative approach to the Norwood procedure. The development of patent ductus arteriosus (PDA) in-stent stenosis can cause retrograde aortic arch stenosis (RAAS), leading to significant morbidity. This study aimed to identify potential mechanisms of PDA in-stent stenosis contributing to RAAS. METHODS: Tissues from stented PDAs were collected from 17 patients undergoing comprehensive stage II repair between 2009 and 2014. Patients requiring RAAS intervention based on cardiology-surgery consensus were defined as RAAS(+) (n = 10), whereas patients without any RAAS intervention were defined as RAAS(-) (n = 7). Tissues were examined by quantitative polymerase chain reaction analysis for vascular smooth muscle cell (VSMC) differentiation and proliferation markers. RESULTS: Patient characteristics were hypoplastic left heart syndrome with aortic atresia in 6 and with aortic stenosis in 3; unbalanced atrioventricular canal in 3; double-inlet left ventricle/transposition of the great arteries in 3; and double-outlet right ventricle in 2. VSMC differentiation markers (ß-actin, SM22, and calponin) and signaling pathways for VSMC modulation (transforming growth factor-ß1, Notch, and platelet derived growth factor-BB) were significantly higher in the RAAS(+) than in RAAS(-) patients. The proliferation marker Ki67 was increased in RAAS(+) patients. Cell cycle markers were comparable in both groups. CONCLUSIONS: Increased VSMC differentiation and proliferation markers suggest a mechanism for inward neointima formation of the PDA in RAAS. The apparent lack of change in cell cycle markers is contrary to coronary artery in-stent stenosis, suggesting further targets should be examined. Combined primary in vitro PDA cell culture and proteomics can be strong tools to elucidate targets to reduce PDA in-stent stenosis for RAAS in the future.


Subject(s)
Aortic Valve Stenosis/etiology , Ductus Arteriosus, Patent/etiology , Hypoplastic Left Heart Syndrome/surgery , Muscle, Smooth, Vascular/pathology , Postoperative Complications/etiology , Stents , Aortic Valve Stenosis/genetics , Cardiac Surgical Procedures , Cell Differentiation/genetics , Cell Proliferation/genetics , Ductus Arteriosus, Patent/genetics , Humans , Infant, Newborn , Postoperative Complications/genetics
10.
Am J Physiol Heart Circ Physiol ; 307(11): H1605-17, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25260618

ABSTRACT

Aortocaval fistula (ACF)-induced volume overload (VO) heart failure (HF) results in progressive left ventricular (LV) dysfunction. Hemodynamic load reversal during pre-HF (4 wk post-ACF; REV) results in rapid structural but delayed functional recovery. This study investigated myocyte and myofilament function in ACF and REV and tested the hypothesis that a myofilament Ca(2+) sensitizer would improve VO-induced myofilament dysfunction in ACF and REV. Following the initial sham or ACF surgery in male Sprague-Dawley rats (200-240 g) at week 0, REV surgery and experiments were performed at weeks 4 and 8, respectively. In ACF, decreased LV function is accompanied by impaired sarcomeric shortening and force generation and decreased Ca(2+) sensitivity, whereas, in REV, impaired LV function is accompanied by decreased Ca(2+) sensitivity. Intravenous levosimendan (Levo) elicited the best inotropic and lusitropic responses and was selected for chronic oral studies. Subsets of ACF and REV rats were given vehicle (water) or Levo (1 mg/kg) in drinking water from weeks 4-8. Levo improved systolic (% fractional shortening, end-systolic elastance, and preload-recruitable stroke work) and diastolic (τ, dP/dtmin) function in ACF and REV. Levo improved Ca(2+) sensitivity without altering the amplitude and kinetics of the intracellular Ca(2+) transient. In ACF-Levo, increased cMyBP-C Ser-273 and Ser-302 and cardiac troponin I Ser-23/24 phosphorylation correlated with improved diastolic relaxation, whereas, in REV-Levo, increased cMyBP-C Ser-273 phosphorylation and increased α-to-ß-myosin heavy chain correlated with improved diastolic relaxation. We concluded that Levo improves LV function, and myofilament composition and regulatory protein phosphorylation likely play a key role in improving function.


Subject(s)
Calcium Signaling/drug effects , Cardiotonic Agents/pharmacology , Heart Failure/drug therapy , Hydrazones/pharmacology , Myofibrils/drug effects , Pyridazines/pharmacology , Ventricular Dysfunction, Left/drug therapy , Ventricular Function, Left/drug effects , Animals , Arterio-Arterial Fistula/pathology , Cardiotonic Agents/therapeutic use , Heart Failure/diagnostic imaging , Heart Failure/physiopathology , Hydrazones/therapeutic use , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Pyridazines/therapeutic use , Rats , Rats, Sprague-Dawley , Sarcomeres/pathology , Simendan , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology
12.
Pflugers Arch ; 466(6): 1065-77, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24488008

ABSTRACT

Two main hemodynamic overload mechanisms [i.e., volume and pressure overload (VO and PO, respectively] result in heart failure (HF), and these two mechanisms have divergent pathologic alterations and different pathophysiological mechanisms. Extensive evidence from animal models and human studies of PO demonstrate a clear association with alterations in Ca(2+) homeostasis. By contrast, emerging evidence from animal models and patients with regurgitant valve disease and dilated cardiomyopathy point toward a more prominent role of myofilament dysfunction. With respect to VO HF, key features of excitation-contraction coupling defects, myofilament dysfunction, and extracellular matrix composition will be discussed.


Subject(s)
Excitation Contraction Coupling , Heart Failure/metabolism , Hemodynamics , Myofibrils/metabolism , Animals , Heart Failure/physiopathology , Humans , Myofibrils/physiology
13.
J Mol Cell Cardiol ; 60: 27-35, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23583836

ABSTRACT

The mature aortic valve is composed of a structured trilaminar extracellular matrix that is interspersed with aortic valve interstitial cells (AVICs) and covered by endothelium. Dysfunction of the valvular endothelium initiates calcification of neighboring AVICs leading to calcific aortic valve disease (CAVD). The molecular mechanism by which endothelial cells communicate with AVICs and cause disease is not well understood. Using a co-culture assay, we show that endothelial cells secrete a signal to inhibit calcification of AVICs. Gain or loss of nitric oxide (NO) prevents or accelerates calcification of AVICs, respectively, suggesting that the endothelial cell-derived signal is NO. Overexpression of Notch1, which is genetically linked to human CAVD, retards the calcification of AVICs that occurs with NO inhibition. In AVICs, NO regulates the expression of Hey1, a downstream target of Notch1, and alters nuclear localization of Notch1 intracellular domain. Finally, Notch1 and NOS3 (endothelial NO synthase) display an in vivo genetic interaction critical for proper valve morphogenesis and the development of aortic valve disease. Our data suggests that endothelial cell-derived NO is a regulator of Notch1 signaling in AVICs in the development of the aortic valve and adult aortic valve disease.


Subject(s)
Aortic Valve/metabolism , Heart Defects, Congenital/metabolism , Heart Valve Diseases/metabolism , Nitric Oxide/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Active Transport, Cell Nucleus/genetics , Animals , Aortic Valve/pathology , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Bicuspid Aortic Valve Disease , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/pathology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Heart Valve Diseases/genetics , Heart Valve Diseases/pathology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Nitric Oxide/genetics , Receptor, Notch1/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Swine
14.
J Mol Cell Cardiol ; 57: 47-58, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23220155

ABSTRACT

Hearts in volume overload (VO) undergo progressive ventricular hypertrophy resulting in chronic heart failure that is unresponsive to ß-adrenergic agonists. This study compared left ventricular (LV) and isolated cardiomyocyte contractility and ß-adrenergic responsiveness in rats with end-stage VO heart failure (HF). Adult male Sprague-Dawley rats were studied 21 weeks after aortocaval fistula (ACF) or sham surgery. Echocardiography revealed decreased fractional shortening accompanied by increased LV chamber diameter and decreased eccentric dilatation index at end-stage ACF compared to sham. Hemodynamic measurements showed a decrease in the slope of end-systolic pressure-volume relationship, indicating systolic dysfunction. Isolated LV myocytes from ACF exhibited decreased peak sarcomere shortening and kinetics. Both Ca2+ transient amplitude and kinetics were increased in ACF myocytes, with no change under the integrated Ca2+ curves relating to contraction and relaxation phases. Increases in ryanodine receptor and phospholamban phosphorylation, along with a decrease in SERCA2 levels, were observed in ACF. These changes were associated with decreased expression of ß-myosin heavy chain, cardiac troponin I and cardiac myosin binding protein-C. In vivo inotropic responses to ß-adrenergic stimulation were attenuated in ACF. Interestingly, ACF myocytes exhibited a similar peak shortening to those of sham in response to a ß-adrenergic agonist. The protein expression of the gap junction protein connexin-43 was decreased, although its phosphorylation at Ser-368 increased. These changes were associated with alterations in Src and ZO-1. In summary, these data suggest that the disconnect in ß-adrenergic responsiveness between in vivo and in vitro conditions may be associated with altered myofilament Ca2+ sensitivity and connexin-43 degradation.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Heart Failure/physiopathology , Isoproterenol/pharmacology , Animals , Calcium Signaling , Cells, Cultured , Connexin 43/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Heart Failure/metabolism , Heart Failure/pathology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Kinetics , Male , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Rats , Rats, Sprague-Dawley , Troponin I/metabolism , Ventricular Myosins/genetics , Ventricular Myosins/metabolism , Ventricular Remodeling
16.
Vascul Pharmacol ; 57(5-6): 187-93, 2012.
Article in English | MEDLINE | ID: mdl-22885305

ABSTRACT

The goals of the present study were to compare coronary resistance microvessel (CRM) remodeling between type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) mice, and to determine the impact of aerobic exercise training on CRM remodeling in diabetes. Eight week old male mice were divided into T1DM: control sedentary (Control-SD), T1DM sedentary (T1DM-SD) induced by streptozotocin, and T1DM exercise trained (T1DM-TR); T2DM: control sedentary (Db/db-SD), T2DM sedentary (db/db-SD), and T2DM trained (db/db-TR). Aerobic exercise training (TR) was performed on a mouse treadmill for 8weeks. CRMs were isolated and mounted on a pressure myograph to measure and record vascular remodeling and mechanics. CRM diameters, wall thickness, stress-strain, incremental modulus remained unchanged in T1DM-SD mice compared to control, and exercise training showed no effect. In contrast, CRMs isolated from db/db-SD mice exhibited decreased luminal diameter with thicker microvascular walls, which significantly increased the wall:lumen ratio (Db/db-SD: 5.8±0.3 vs. db/db-SD: 8.9±0.7, p<0.001). Compared to db/db-SD mice, coronary arterioles isolated from db/db-TR mice had similar internal diameter and wall thickness, while wall:lumen ratio (6.8±0.2, p<0.05) and growth index (db/db-SD: 16.2 vs. db/db-TR: 4.3, % over Db/db) were reduced. These data show that CRMs undergo adverse inward hypertrophic remodeling only in T2DM, but not T1DM, and that aerobic exercise training can partially mitigate this process.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/therapy , Exercise Therapy/methods , Animals , Coronary Vessels/metabolism , Coronary Vessels/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Microvessels/pathology , Physical Conditioning, Animal/physiology , Streptozocin
17.
J Mol Cell Cardiol ; 53(5): 626-38, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22906538

ABSTRACT

Collagen XIV is a fibril-associated collagen with an interrupted triple helix (FACIT). Previous studies have shown that this collagen type regulates early stages of fibrillogenesis in connective tissues of high mechanical demand. Mice null for Collagen XIV are viable, however formation of the interstitial collagen network is defective in tendons and skin leading to reduced biomechanical function. The assembly of a tightly regulated collagen network is also required in the heart, not only for structural support but also for controlling cellular processes. Collagen XIV is highly expressed in the embryonic heart, notably within the cardiac interstitium of the developing myocardium, however its role has not been elucidated. To test this, we examined cardiac phenotypes in embryonic and adult mice devoid of Collagen XIV. From as early as E11.5, Col14a1(-/-) mice exhibit significant perturbations in mRNA levels of many other collagen types and remodeling enzymes (MMPs, TIMPs) within the ventricular myocardium. By post natal stages, collagen fibril organization is in disarray and the adult heart displays defects in ventricular morphogenesis. In addition to the extracellular matrix, Col14a1(-/-) mice exhibit increased cardiomyocyte proliferation at post natal, but not E11.5 stages, leading to increased cell number, yet cell size is decreased by 3 months of age. In contrast to myocytes, the number of cardiac fibroblasts is reduced after birth associated with increased apoptosis. As a result of these molecular and cellular changes during embryonic development and post natal maturation, cardiac function is diminished in Col14a1(-/-) mice from 3 months of age; associated with dilation in the absence of hypertrophy, and reduced ejection fraction. Further, Col14a1 deficiency leads to a greater increase in left ventricular wall thickening in response to pathological pressure overload compared to wild type animals. Collectively, these studies identify a new role for type XIV collagen in the formation of the cardiac interstitium during embryonic development, and highlight the importance of the collagen network for myocardial cell survival, and function of the working myocardium after birth.


Subject(s)
Collagen/deficiency , Glycoproteins/deficiency , Heart/growth & development , Myocardium/metabolism , Animals , Cell Proliferation , Collagen/genetics , Collagen/physiology , Glycoproteins/genetics , Glycoproteins/physiology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , In Vitro Techniques , Male , Mice , Mice, Transgenic , Myocardial Contraction , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Stroke Volume , Transcription, Genetic , Ventricular Function, Left , Ventricular Pressure , Ventricular Remodeling
18.
J Appl Physiol (1985) ; 113(7): 1128-40, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837170

ABSTRACT

Previous studies from our laboratory showed that coronary arterioles from type 2 diabetic mice undergo inward hypertrophic remodeling and reduced stiffness. The aim of the current study was to determine if coronary resistance microvessels (CRMs) in Ossabaw swine with metabolic syndrome (MetS) undergo remodeling distinct from coronary conduit arteries. Male Ossabaw swine were fed normal (n = 7, Lean) or hypercaloric high-fat (n = 7, MetS) diets for 6 mo, and then CRMs were isolated and mounted on a pressure myograph. CRMs isolated from MetS swine exhibited decreased luminal diameters (126 ± 5 and 105 ± 9 µm in Lean and MetS, respectively, P < 0.05) with thicker walls (18 ± 3 and 31 ± 3 µm in Lean and MetS, respectively, P < 0.05), which doubled the wall-to-lumen ratio (14 ± 2 and 30 ± 2 in Lean and MetS, respectively, P < 0.01). Incremental modulus of elasticity (IME) and beta stiffness index (BSI) were reduced in CRMs isolated from MetS pigs (IME: 3.6 × 10(6) ± 0.7 × 10(6) and 1.1 × 10(6) ± 0.2 × 10(6) dyn/cm(2) in Lean and MetS, respectively, P < 0.001; BSI: 10.3 ± 0.4 and 7.3 ± 1.8 in Lean and MetS, respectively, P < 0.001). BSI in the left anterior descending coronary artery was augmented in pigs with MetS. Structural changes were associated with capillary rarefaction, decreased hyperemic-to-basal coronary flow velocity ratio, and augmented myogenic tone. MetS CRMs showed a reduced collagen-to-elastin ratio, while immunostaining for the receptor for advanced glycation end products was selectively increased in the left anterior descending coronary artery. These data suggest that MetS causes hypertrophic inward remodeling of CRMs and capillary rarefaction, which contribute to decreased coronary flow and myocardial ischemia. Moreover, our data demonstrate novel differential remodeling between coronary micro- and macrovessels in a clinically relevant model of MetS.


Subject(s)
Coronary Circulation/physiology , Coronary Vessels/physiopathology , Metabolic Syndrome/physiopathology , Microvessels/physiopathology , Obesity/physiopathology , Animals , Blood Flow Velocity/physiology , Collagen/metabolism , Coronary Vessels/metabolism , Elastin/metabolism , Male , Metabolic Syndrome/metabolism , Microvessels/metabolism , Obesity/metabolism , Swine
19.
Circ Heart Fail ; 5(4): 452-61, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22661498

ABSTRACT

BACKGROUND: Air pollution is a pervasive environmental health hazard that occurs over a lifetime of exposure in individuals from many industrialized societies. However, studies have focused primarily on exposure durations that correspond to only a portion of the lifespan. We therefore tested the hypothesis that exposure over a considerable portion of the lifespan would induce maladaptive cardiovascular responses. METHODS AND RESULTS: C57BL/6 male mice were exposed to concentrated ambient particles <2.5 µm (particulate matter, PM or PM(2.5)) or filtered air (FA), 6 h/d, 5 d/wk, for 9 months. Assessment of cardiac contractile function, coronary arterial flow reserve, isolated cardiomyocyte function, expression of hypertrophic markers, calcium handling proteins, and cardiac fibrosis were then performed. Mean daily concentrations of PM(2.5) in the exposure chamber versus ambient daily PM(2.5) concentration at the study site were 85.3 versus 10.6 µg/m(3) (7.8-fold concentration), respectively. PM(2.5) exposure resulted in increased hypertrophic markers leading to adverse ventricular remodeling characterized by myosin heavy chain (MHC) isoform switch and fibrosis, decreased fractional shortening (39.8 ± 1.4 FA versus 27.9 ± 1.3 PM, FS%), and mitral inflow patterns consistent with diastolic dysfunction (1.95 ± 0.05 FA versus 1.52 ± 0.07 PM, E/A ratio). Contractile reserve to dobutamine was depressed (62.3 ± 0.9 FA versus 49.2 ± 1.5 PM, FS%) in response to PM(2.5) without significant alterations in maximal vasodilator flow reserve. In vitro cardiomyocyte function revealed depressed peak shortening (8.7 ± 0.6 FA versus 7.0 ± 0.4 PM, %PS) and increased time-to-90% shortening (72.5 ± 3.2 FA versus 82.8 ± 3.2 PM, ms) and re-lengthening (253.1 ± 7.9 FA versus 282.8 ± 9.3 PM, ms), which were associated with upregulation of profibrotic markers and decreased total antioxidant capacity. Whole-heart SERCA2a levels and the ratio of α/ß-MHC were both significantly decreased (P<0.05) in PM(2.5)-exposed animals, suggesting a switch to fetal programming. CONCLUSIONS: Long-term exposure to environmentally relevant concentrations of PM(2.5) resulted in a cardiac phenotype consistent with incipient heart failure.


Subject(s)
Cardiovascular Diseases/etiology , Particulate Matter/toxicity , Ventricular Function, Left , Ventricular Remodeling , Animals , Biomarkers/metabolism , Blood Pressure , Calcium/metabolism , Cardiomegaly/etiology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Collagen/metabolism , Fibrosis , Fractional Flow Reserve, Myocardial , Heart Failure/etiology , Heart Failure/pathology , Heart Failure/physiopathology , Inhalation Exposure , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myosin Heavy Chains/metabolism , Phenotype , Protein Isoforms , Risk Assessment , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Time Factors
20.
PLoS Genet ; 8(5): e1002690, 2012.
Article in English | MEDLINE | ID: mdl-22589735

ABSTRACT

Defects of atrial and ventricular septation are the most frequent form of congenital heart disease, accounting for almost 50% of all cases. We previously reported that a heterozygous G296S missense mutation of GATA4 caused atrial and ventricular septal defects and pulmonary valve stenosis in humans. GATA4 encodes a cardiac transcription factor, and when deleted in mice it results in cardiac bifida and lethality by embryonic day (E)9.5. In vitro, the mutant GATA4 protein has a reduced DNA binding affinity and transcriptional activity and abolishes a physical interaction with TBX5, a transcription factor critical for normal heart formation. To characterize the mutation in vivo, we generated mice harboring the same mutation, Gata4 G295S. Mice homozygous for the Gata4 G295S mutant allele have normal ventral body patterning and heart looping, but have a thin ventricular myocardium, single ventricular chamber, and lethality by E11.5. While heterozygous Gata4 G295S mutant mice are viable, a subset of these mice have semilunar valve stenosis and small defects of the atrial septum. Gene expression studies of homozygous mutant mice suggest the G295S protein can sufficiently activate downstream targets of Gata4 in the endoderm but not in the developing heart. Cardiomyocyte proliferation deficits and decreased cardiac expression of CCND2, a member of the cyclin family and a direct target of Gata4, were found in embryos both homozygous and heterozygous for the Gata4 G295S allele. To further define functions of the Gata4 G295S mutation in vivo, compound mutant mice were generated in which specific cell lineages harbored both the Gata4 G295S mutant and Gata4 null alleles. Examination of these mice demonstrated that the Gata4 G295S protein has functional deficits in early myocardial development. In summary, the Gata4 G295S mutation functions as a hypomorph in vivo and leads to defects in cardiomyocyte proliferation during embryogenesis, which may contribute to the development of congenital heart defects in humans.


Subject(s)
Atrial Septum , Cell Proliferation , GATA4 Transcription Factor , Heart Defects, Congenital , Heart Ventricles , Myocytes, Cardiac , Animals , Atrial Septum/metabolism , Atrial Septum/physiopathology , Cyclin D2/metabolism , Embryonic Development/genetics , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/physiopathology , Heart Ventricles/growth & development , Heart Ventricles/metabolism , Humans , Mice , Mice, Mutant Strains , Mutation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...