Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Rev Sci Instrum ; 87(8): 084301, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27587132

ABSTRACT

Mechanical stimulation appears to be a critical modulator for many aspects of biology, both of living tissue and cells. The cell-stretcher, a novel device for the mechanical uniaxial stimulation of populations of cells, is described. The system is based on a variable stroke cam-lever-tappet mechanism which allows the delivery of cyclic stimuli with frequencies of up to 10 Hz and deformation between 1% and 20%. The kinematics is presented and a simulation of the dynamics of the system is shown, in order to compute the contact forces in the mechanism. The cells, following cultivation and preparation, are plated on an ad hoc polydimethylsiloxane membrane which is then loaded on the clamps of the cell-stretcher via force-adjustable magnetic couplings. In order to show the viability of the experimentation and biocompatibility of the cell-stretcher, a set of two in vitro tests were performed. Human epithelial carcinoma cell line A431 and Adult Mouse Ventricular Fibroblasts (AMVFs) from a dual reporter mouse were subject to 0.5 Hz, 24 h cyclic stretching at 15% strain, and to 48 h stimulation at 0.5 Hz and 15% strain, respectively. Visual analysis was performed on A431, showing definite morphological changes in the form of cellular extroflections in the direction of stimulation compared to an unstimulated control. A cytometric analysis was performed on the AMVF population. Results show a post-stimulation live-dead ratio deviance of less than 6% compared to control, which proves that the environment created by the cell-stretcher is suitable for in vitro experimentation.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Shear Strength , Animals , Cell Line, Tumor , Humans , Mice
2.
Mater Sci Eng C Mater Biol Appl ; 49: 217-224, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25686942

ABSTRACT

Atomic force microscopy (AFM) for single-cell force spectroscopy (SCFS) and Poisson statistic were used to analyze the detachment work recorded during the removal of gold-covered microspheres from cardiac fibroblasts. The effect of Cytochalasin D, a disruptor of the actin cytoskeleton, on cell adhesion was also tested. The adhesion work was assessed using a Poisson analysis also derived from single-cell force spectroscopy retracting curves. The use of Poisson analysis to get adhesion work from AFM curves is quite a novel method, and in this case, proved to be effective to study the short-range and long-range contributions to the adhesion work. This method avoids the difficult identification of minor peaks in the AFM retracting curves by creating what can be considered an average adhesion work. Even though the effect of actin depolymerisation is well documented, its use revealed that control cardiac fibroblasts (CT) exhibit a work of adhesion at least 5 times higher than that of the Cytochalasin treated cells. However, our results indicate that in both cells short-range and long-range contributions to the adhesion work are nearly equal and the same heterogeneity index describes both cells. Therefore, we infer that the different adhesion behaviors might be explained by the presence of fewer membrane adhesion molecules available at the AFM tip-cell interface under circumstances where the actin cytoskeleton has been disrupted.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Fibroblasts/physiology , Heart/physiology , Actin Cytoskeleton/metabolism , Animals , Cytochalasin D/metabolism , Fibroblasts/metabolism , Gold/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Atomic Force/methods
3.
Mater Sci Eng C Mater Biol Appl ; 40: 427-34, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24857511

ABSTRACT

AFM was used to collect the whole force-deformation cell curves. They provide both the elasticity and adhesion behavior of mouse primary cardiac fibroblasts. To confirm the hypothesis that a link exists between the membrane receptors and the cytoskeletal filaments causing therefore changing in both elasticity and adhesion behavior, actin-destabilizing Cytochalsin D was administrated to the fibroblasts. From immunofluorescence observation and AFM loading/unloading curves, cytoskeletal reorganization as well as a change in the elasticity and adhesion was indeed observed. Elasticity of control fibroblasts is three times higher than that for fibroblasts treated with 0.5 µM Cytochalasin. Moreover, AFM loading-unloading curves clearly show the different mechanical behavior of the two different cells analyzed: (i) for control cells the AFM cantilever rises during the dwell time while cells with Cytochalasin fail to show such an active resistance; (ii) the maximum force to deform control cells is quite higher and as far as adhesion is concern (iii) the maximum separation force, detachment area and the detachment process time are much larger for control compared to the Cytochalasin treated cells. Therefore, alterations in the cytoskeleton suggest that a link must exist between the membrane receptors and the cytoskeletal filaments beneath the cellular surface and inhibition of actin polymerization has effects on the whole cell mechanical behavior as well as adhesion.


Subject(s)
Fibroblasts/cytology , Microscopy, Atomic Force , Myocytes, Cardiac/cytology , Animals , Cell Adhesion/drug effects , Cells, Cultured , Cytochalasins/pharmacology , Cytoskeleton/drug effects , Elasticity , Fibroblasts/drug effects , Mice
5.
Circ Res ; 89(7): 591-8, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11577024

ABSTRACT

Physiological and pathological cardiac hypertrophy have directionally opposite changes in transcription of thyroid hormone (TH)-responsive genes, including alpha- and beta-myosin heavy chain (MyHC) and sarcoplasmic reticulum Ca(2+)-ATPase (SERCA), and TH treatment can reverse molecular and functional abnormalities in pathological hypertrophy, such as pressure overload. These findings suggest relative hypothyroidism in pathological hypertrophy, but serum levels of TH are usually normal. We studied the regulation of TH receptors (TRs) beta1, alpha1, and alpha2 in pathological and physiological rat cardiac hypertrophy models with hypothyroid- and hyperthyroid-like changes in the TH target genes, alpha- and beta-MyHC and SERCA. All 3 TR subtypes in myocytes were downregulated in 2 hypertrophy models with a hypothyroid-like mRNA phenotype, phenylephrine in culture and pressure overload in vivo. Myocyte TRbeta1 was upregulated in models with a hyperthyroid-like phenotype, TH (triiodothyronine, T3), in culture and exercise in vivo. In myocyte culture, TR overexpression, or excess T3, reversed the effects of phenylephrine on TH-responsive mRNAs and promoters. In addition, TR cotransfection and treatment with the TRbeta1-selective agonist GC-1 suggested different functional coupling of the TR isoforms, TRbeta1 to transcription of beta-MyHC, SERCA, and TRbeta1, and TRalpha1 to alpha-MyHC transcription and increased myocyte size. We conclude that TR isoforms have distinct regulation and function in rat cardiac myocytes. Changes in myocyte TR levels can explain in part the characteristic molecular phenotypes in physiological and pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly/physiopathology , Gene Expression Regulation , Myocardium/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Animals , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Male , Motor Activity , Myocardium/cytology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phenotype , Phenylephrine/pharmacology , Physical Conditioning, Animal , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Thyroid Hormone/agonists , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Transfection , Triiodothyronine/pharmacology
6.
J Biol Chem ; 276(31): 29490-8, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11382751

ABSTRACT

We have demonstrated that two hypertrophic agents, interleukin-1 beta (IL-1 beta) and leukemic inhibitory factor (LIF), altered cardiac myocyte morphology with striking similarity and prompted us to investigate the common actions of these cytokines. We compared the phosphorylation/activation of signal tranducer and activator of transcription 3 (STAT3), extracellular signal-regulated kinase (ERK), p38(MAPK), and c-Jun N-terminal kinase mitogen-activated protein kinases (MAPKs). The phosphorylation of STAT3 by IL-1 beta was delayed (>60 min), whereas the response to LIF was rapid (<10 min) and transient. We confirmed that IL-1 beta potently stimulated all three MAPK subfamilies. In contrast, LIF promoted strong activation of ERKs, marginal activation of p38(MAPK), and no c-Jun N-terminal kinase activation. To test the roles of ERKs and p38(MAPK), myocytes were pretreated with PD98059 and SB203580. Either inhibitor alone prevented STAT3 phosphorylation, implicating ERKs and p38(MAPK) in the delayed STAT3 response to IL-1 beta. The interplay of MAPKs and STAT3 phosphorylation in regulating IL-1 beta-stimulated hypertrophy was investigated by evaluating the effect of MAPK inhibitors on atrial natriuretic factor (ANF) expression and myocyte morphology. The specific inhibition of either ERK or p38(MAPK) attenuated the IL-1 beta- or LIF-stimulated ANF expression by up to 70%. Inhibition was not further increased in the presence of both inhibitors. Furthermore, although individual inhibition of ERK or p38(MAPK) did not affect morphology, co-treatment with both inhibitors abrogated the hypertrophic morphology stimulated by IL-1 beta but not by LIF. Taken together, our data indicate that the activation of ERK and p38(MAPK) is essential in regulating a delayed STAT3 phosphorylation as well as changes in ANF expression and morphology that follow IL-1 beta treatment. Thus, the role of MAPKs in the hypertrophic response can be dictated at least partly by the nature of the hypertrophic agent employed.


Subject(s)
Atrial Natriuretic Factor/genetics , DNA-Binding Proteins/metabolism , Interleukin-1/pharmacology , Interleukin-6 , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Myocardium/metabolism , Trans-Activators/metabolism , Acute-Phase Proteins/metabolism , Animals , Animals, Newborn , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Culture Media, Serum-Free , Cycloheximide/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Growth Inhibitors/pharmacology , Heart/drug effects , Heart/physiology , Heart Ventricles , Hypertrophy , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Kinetics , Leukemia Inhibitory Factor , Lymphokines/pharmacology , MAP Kinase Signaling System/drug effects , Phenylephrine/pharmacology , Phosphorylation , Protein Transport , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor , p38 Mitogen-Activated Protein Kinases
7.
Heart Fail Rev ; 6(2): 81-94, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11309527

ABSTRACT

The prevalence of congestive heart failure and its continued poor prognosis despite presently available therapeutic options emphasize the importance of pursuing the observations suggesting an important role for an immunomodulatory approach to decompensated cardiac failure. Furthermore, there are several pieces of background information that suggest that cytokines like IL-1 may play a significant role in the pathogenesis of several forms of myocardial dysfunction. Although it seems clear that IL-1 is not acting alone under circumstances of myocardial injury, but in concert with other pro-inflammatory molecules and their effectors, we believe that continued investigations into the cytokine hypothesis will ultimately increase the understanding of how pro-inflammatory molecules influence myocardial function and how the modulation of such factors may improve the myocardial response to injury. The specific observations that emphasize the importance of pursuing a substantive role for IL-1 in this process are: (1) IL-1 is elevated in several cardiac disease states, (2) IL-1 is produced by myocardial cells themselves in response to injury, (3)The alterations in gene expression seen in response IL-1 resembles in many ways the phenotype of the failing heart, and (4) The co-localization of the IL-1 response with that of several previously described negative transcriptional regulators (making them potential targets for therapeutic manipulation).


Subject(s)
Heart Failure/metabolism , Interleukin-1/metabolism , Animals , Disease Progression , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Humans , Interleukin-1/genetics , Myosins/metabolism , Signal Transduction , Transcription Factors/metabolism
8.
Circulation ; 103(8): 1089-94, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11222471

ABSTRACT

BACKGROUND: We have previously demonstrated that changes in myosin heavy chain (MHC) isoforms that occur in failing human hearts resemble the pattern produced in rodent myocardium in response to hypothyroidism. Because thyroid hormone status is usually within normal limits in these patients, we hypothesized that failing/hypertrophied human myocardium might have a defect in thyroid hormone signaling due to alterations in expression of thyroid hormone receptors (TRs). METHODS AND RESULTS: To examine this hypothesis, we used RNase protection assay to measure mRNA levels of TRs in failing left ventricles that exhibited a fetal pattern of gene expression, ie, decreased expression of alpha-MHC with increased beta-MHC expression compared with left ventricles from age-matched controls. We detected expression of TR-alpha(1), -alpha(2), and -beta(1) isoforms in human left ventricles. In failing left ventricles, TR-alpha(1) was downregulated, whereas TR-alpha(2), a splice variant that does not bind thyroid hormone but inhibits responses to liganded TRs, was increased. Expression levels of TR-beta(1) did not differ significantly between the 2 groups. According to linear regression analysis, expression levels of TR-alpha(1) and -alpha(2) were positively and negatively correlated with those of alpha-MHC, respectively. CONCLUSIONS: We conclude that decreases in TR-alpha(1) and increases in TR-alpha(2) may lead to local attenuation of thyroid hormone signaling in the failing human heart and that the resulting tissue-specific hypothyroidism is a candidate for the molecular mechanism that induces fetal gene expression in the failing human ventricle.


Subject(s)
Gene Expression , Heart Diseases/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction/genetics , Adult , Atrial Natriuretic Factor/genetics , Female , Fetal Proteins/genetics , Humans , Major Histocompatibility Complex/genetics , Male , Middle Aged , RNA, Messenger/biosynthesis , Receptors, Thyroid Hormone/biosynthesis
9.
J Mol Cell Cardiol ; 32(7): 1341-52, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10860774

ABSTRACT

Current research from both clinical and basic science perspectives indicates that cytokines play an important role in the genesis of cardiovascular pathology. Specifically, levels of cytokines such as interleukin-1 (IL-1), tumor necrosis factor- alpha (TNF- alpha), and interleukin-6 (IL-6) have been found to be elevated in both acute myocardial injury as well as situations of chronic dysfunction. Further, therapies directed primarily at interfering with cytokine action have suggested that such an immunomodulatory approach may be beneficial in some of these circumstances of myocardial injury. We recently reported that IL-1 beta induces a hypertrophic state in cultured neonatal rat cardiac myocytes that differs from other well described hypertrophic phenotypes in terms of myocardial gene expression (such as skeletal alpha -actin, sACT), an effect that appeared to co-localize with that of the negative regulator yin yang-1 (YY1).(1)In the present study, we further localize the area in the sACT promoter responsible for the IL-1 effect. These investigations indicate that sequences in and around the third upstream serum response element (SRE3) bind YY1 and are required for IL-1 beta mediated repression. This element is also capable of transferring both IL-1 beta and YY1-mediated transcriptional repression to a heterologous promoter. In support of an IL-1 beta induced post-translational modification of YY1 that results in an increase in DNA-binding activity,(32)P-labeling experiments reveal an increase in phosphorylated YY1 in IL-1 beta treated cells and phosphatase-treated myocyte nuclear proteins lose their ability to bind to the YY1 site. In summary, these results provide evidence that sequences within the SRE3 of the skeletal actin promoter represent an IL-1 beta response element and suggest that IL-1 beta activates the negative transcription factor YY1 by both transcriptional and post-transcriptional mechanisms.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-1/pharmacology , Interleukin-1/physiology , Myocardium/metabolism , Transcription Factors/metabolism , Actins/genetics , Animals , Animals, Newborn , Binding Sites , Blotting, Western , Cells, Cultured , DNA/metabolism , DNA-Binding Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Erythroid-Specific DNA-Binding Factors , Mice , Muscle, Skeletal/metabolism , Phosphorylation , Plasmids , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic , Transfection , YY1 Transcription Factor , Zinc Fingers
10.
Cardiovasc Res ; 46(1): 139-50, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727662

ABSTRACT

OBJECTIVE: Stress proteins (heat shock proteins, HSPs) are molecular chaperones that have been shown to enhance the survival of cells exposed to environmental stress. We sought to investigate the effects of hypoxia on the levels of HSP27 and heme oxygenase-1 (HO-1 or HSP32) in an established model of rat neonatal cardiac myocytes in culture. METHODS: Myocytes were subjected to hypoxia (<0.5% O(2) for 16 h). Studies of cell viability and nuclear morphology showed no evidence of cell death under these conditions. RESULTS: Messenger RNA analysis demonstrated constitutive expression of HSP27 and low levels of HO-1. Hypoxia strongly induced HO-1 mRNA without affecting HSP27 mRNA. In parallel to mRNA levels, hypoxia increased HO-1 protein level without affecting HSP27. To further assess the signaling pathways implicated in HO-1 induction, we used inhibition experiments. The tyrosine kinase inhibitor tyrphostin and the mitogen-activated protein kinase inhibitor PD98059 did not prevent HO-1 induction, while the protein kinase C inhibitor chelerythrine partially blocked this response. The p38 stress-activated kinase inhibitor SB203580 was the most potent in suppressing hypoxia-induced HO-1. In vitro kinase assays, cell labeling and immunoprecipitation showed activation of signaling pathways downstream of p38 stress-activated kinase as revealed by an increase in phosphorylation of MAPKAPK-2/3 kinases and HSP27. CONCLUSIONS: These data show a differential pattern of hypoxia-induced HSP expression and implicate the stress kinase in HO-1 induction. Thus, selective regulation of HSP levels may play a role in the cardioprotective mechanisms that participate in the adaptive response to hypoxia-induced stress.


Subject(s)
Heat-Shock Proteins/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Myocardium/metabolism , Oxidative Phosphorylation , Receptors, Estrogen/metabolism , Signal Transduction , Alkaloids , Analysis of Variance , Animals , Benzophenanthridines , Cell Hypoxia , Cell Survival , Cells, Cultured , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Heat-Shock Proteins/genetics , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Imidazoles/pharmacology , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phenanthridines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/pharmacology , RNA, Messenger/analysis , Rats , Receptors, Estrogen/genetics , Tyrphostins/pharmacology
11.
Dermatol Online J ; 6(1): 2, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11328612

ABSTRACT

The Ultraviolet Index was developed in the United States in 1994 following successful use of ultraviolet (UV) alerts in other countries. This daily National Weather Service prediction is a calculation which integrates five data elements to yield the amount of UV radiation impacting the surface (1m2) at solar noon in 58 of the largest US population centers. This simple numeric prediction is then categorized by the Environmental Protection Agency into five "exposure levels" with protective actions recommended for each level. This information is disseminated through the media. Daily reminders seem to affect awareness and behavior in Canada, but US surveys indicate the need for better understanding through educational graphics. Comparing the UV Index to a precipitation prediction has merit in that it links a familiar daily prediction with implied appropriate protective measures. Graphics link the ideas that "when it rains it pours and when it shines it radiates." Beginning in schools, camps, and dermatology meetings, using the rain/shine analogy, a wider exposure to the Ultraviolet Index is proposed.


Subject(s)
Health Promotion , Ultraviolet Rays , Humans , Skin Neoplasms/prevention & control , Sunburn/prevention & control , United States , Weather
12.
Heart ; 81(3): 303-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10026358

ABSTRACT

OBJECTIVE: To determine whether heart failure in rats is associated with altered expression of the skeletal muscle sarcoplasmic reticulum Ca2+-ATPase (SERCA). METHODS: SERCA protein and mRNA were examined in the soleus muscles of eight female rats with heart failure induced by coronary artery ligation, six weeks after the procedure (mean (SEM) left ventricular end diastolic pressure 20.4 (2.2) mm Hg) and in six sham operated controls by western and northern analyses, respectively. RESULTS: SERCA-2a isoform protein was reduced by 16% (112 000 (4000) v 134 000 (2000) arbitrary units, p < 0.001), and SERCA-2a messenger RNA was reduced by 59% (0.24 (0. 06) v 0.58 (0.02) arbitrary units, p < 0.001). Although rats with heart failure had smaller muscles (0.54 mg/g v 0.66 mg/g body weight), no difference in locomotor activity was observed. CONCLUSIONS: These results may explain the previously documented abnormalities in calcium handling in skeletal muscle from animals with the same model of congestive heart failure, and could be responsible for the accelerated muscle fatigue characteristic of patients with heart failure.


Subject(s)
Calcium-Transporting ATPases/metabolism , Heart Failure/enzymology , Muscle, Skeletal/enzymology , Myocardial Infarction/complications , Sarcoplasmic Reticulum/enzymology , Animals , Calcium-Transporting ATPases/genetics , DNA/analysis , Female , Gene Expression , Heart Failure/etiology , Muscle, Skeletal/ultrastructure , Myocardial Infarction/enzymology , Rats , Rats, Sprague-Dawley
13.
J Mol Cell Cardiol ; 31(12): 2087-99, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10640438

ABSTRACT

Pro-inflammatory cytokines may be important in the pathophysiological responses of the heart. We investigated the activation of the three mitogen-activated protein kinase (MAPK) subfamilies ¿c-Jun N-terminal kinases (JNKs), p38-MAPKs and extracellularly-responsive kinases (ERKs) by interleukin-1 beta (IL-1 beta) or tumour necrosis factor alpha (TNF alpha) in primary cultures of myocytes isolated from neonatal rat ventricles. Both cytokines stimulated a rapid (maximal within 10 min) increase in JNK activity. Although activation of JNKs by IL-1 beta was transient returning to control values within 1 h, the response to TNF alpha was sustained. IL-1 beta and TNF alpha also stimulated p38-MAPK phosphorylation, but the response to IL-1 beta was consistently greater than TNF alpha. Both cytokines activated ERKs, but to a lesser degree than that induced by phorbol esters. The transcription factors, c-Jun and ATF2, are phosphorylated by the MAPKs and are implicated in the upregulation of c-Jun. IL-1 beta and TNF alpha stimulated the phosphorylation of c-Jun and ATF2. However, IL-1 beta induced a greater increase in c-Jun protein. Inhibitors of protein kinase C (PKC) (Ro318220, GF109203X) and the ERK cascade (PD98059) attenuated the increase in c-Jun induced by IL-1 beta, but LY294002 (an inhibitor of phosphatidylinositol 3' kinase) and SB203580 (an inhibitor of p38-MAPK, which also inhibits certain JNK isoforms) had no effect. These data illustrate that some of the pathological effects of IL-1 beta and TNF alpha may be mediated through the MAPK cascades, and that the ERK cascade, rather than JNKs or p38-MAPKs, are implicated in the upregulation of c-Jun by IL-1 beta.


Subject(s)
Interleukin-1/pharmacology , MAP Kinase Signaling System/drug effects , Myocardium/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Activating Transcription Factor 2 , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transcription Factors/metabolism , Up-Regulation/drug effects
14.
J Biol Chem ; 273(40): 25796-803, 1998 Oct 02.
Article in English | MEDLINE | ID: mdl-9748252

ABSTRACT

Although responsible for only approximately one-third of the overall myocardial mass, the interstitial fibroblasts of the heart serve a fundamental role in establishing the functional integrity of myocardium and are the major source of myocardial extracellular matrix production. Their importance in clinical medicine is underscored by the observation that fibroblast numbers increase in response to several pathologic circumstances that are associated with an increase in extracellular matrix production, such as long standing hypertension and myocardial injury/infarction. Up to the present time, however, there has been little information available on either the kinetics of the cardiac fibroblast cell cycle, or the fundamental mechanisms that regulate its entry into and exit from the cell cycle. Previous work from our laboratory examining the effects of interleukin (IL)-1beta on myocardial growth and gene expression in culture indicated that cardiac fibroblasts have a diminished capacity to synthesize DNA in response to mitogen in the presence of this cytokine. The mechanism of IL-1beta action was not clear, however, and could have resulted from action at several different points in the cell cycle. The investigations described in this report indicate that IL-1beta exerts its effect on the fibroblast cell cycle at multiple levels through altering the expression of cardiac fibroblast cyclins, cyclin-dependent kinases, and their inhibitors, which ultimately affect the phosphorylation of the retinoblastoma gene product.


Subject(s)
Cell Cycle/drug effects , Interleukin-1/pharmacology , Myocardium/metabolism , Animals , Cell Division/drug effects , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Fibroblasts , Flow Cytometry , G1 Phase/physiology , Mitogens/pharmacology , Phosphorylation/drug effects , Rats , Retinoblastoma Protein/metabolism , S Phase/physiology
15.
J Mol Cell Cardiol ; 30(8): 1615-30, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9737947

ABSTRACT

The myocardial molecular and cellular responses to hemodynamic and other hypertrophic stimuli have been characterized extensively, but less is known of the alterations in gene expression during the evolution of heart failure following myocardial infarction, and specifically those affecting the cardiac myocytes. Therefore, the present study was undertaken to test the hypothesis that post-infarction heart failure and remodeling in the rat is associated with a distinct myocyte molecular phenotype. To address this question, hemodynamic measurements were performed in vivo; and myocytes isolated from the non-infarcted myocardium 1 day, 1 week, and 6 weeks post-coronary artery ligation in post-infarct rats and sham controls. Myocyte size, mRNA levels for immediate early genes, contractile proteins, and sarcoplasmic reticulum Ca2+-ATPase (SERCA) and phospholamban were assayed by Northern analyses, and SERCA and phospholamban proteins were examined by Western blotting. Hemodynamic evidence of heart failure was present at all post-infarct time points. Myocyte size was increased significantly at 6 weeks. c-myc expression was increased at 1 day and 1 week in the infarcted rats, but returned to baseline by 6 weeks. Atrial natriuretic peptide and VEGF mRNAs were elevated at 1 and 6 weeks. Both beta-myosin heavy chain and skeletal alpha-actin expression were increased at all post-MI time points. In contrast, neither changes in the expression of the calcium-handling proteins (SERCA and phospholamban) were not observed, nor was there a change in TGFbeta1 or TGFbeta3. These results demonstrate that in rats with post-MI heart failure, there was an immediate induction of the fetal/embryonic transcriptional gene program which preceded myocyte hypertrophy and appeared to persist longer than in pressure-overload models. In further contrast to pressure-overload, expression of sarcoplasmic reticulum Ca2+-ATPase and phospholamban, was not altered despite a comparable degree of cellular hypertrophy and more severe hemodynamic decompensation. These findings suggest that there may be important differences in the regulatory mechanisms underlying these two forms of myocardial hypertrophy and heart failure.


Subject(s)
Gene Expression Regulation , Heart Failure/pathology , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardium/pathology , Actins/genetics , Actins/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Cell Size , Contractile Proteins/genetics , Contractile Proteins/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Genes, fos , Genes, myc , Heart Failure/genetics , Heart Failure/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Male , Myocardial Infarction/genetics , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , RNA/analysis , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Am J Physiol ; 275(1): H250-8, 1998 07.
Article in English | MEDLINE | ID: mdl-9688921

ABSTRACT

Growing evidence suggests that cardiac nonmyocyte cells may play an important regulatory role in the response to myocardial overload and injury via altered expression of paracrine products, such as cytokines and growth factors, but information concerning the cell-specific changes in the expression of these substances in heart-failure models is limited. Therefore, cardiac nonmyocytes were isolated from rats 1 day and 1 and 6 wk after left coronary artery ligation with resulting hemodynamic evidence of heart failure and in sham-operated control animals. mRNAs for tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1beta, IL-6, transforming growth factors (TGF)-beta1 and TGF-beta3, and type I and type III collagen were measured by Northern analyses. The temporal and quantitative relationships between the expression of these cytokines and collagen and myocyte hypertrophy were determined. mRNA expression of IL-1beta was increased by 1.3-fold at 1 day and 1 wk, and expression of TNF-alpha, IL-1beta, IL-6, TGF-beta1, and TGF-beta3 were increased by 1.4- to 2.1-fold at the 1-wk time point before returning toward baseline at 6 wk. There were significant correlations between the expression of these cytokines and the expression of types I and III collagen, which also peaked at 1 wk. Myocyte hypertrophy was seen first at 6 wk. These observations are consistent with a hypothesis that nonmyocyte cells play a regulatory role in the extracellular matrix changes during postinfarction remodeling and highlight the importance of examining cell-specific changes in gene expression and elucidating the role of cell-to-cell interactions within the myocardium.


Subject(s)
Cytokines/biosynthesis , Heart Failure/immunology , Hemodynamics , Myocardial Infarction/immunology , Myocardium/immunology , Transcription, Genetic , Animals , Blood Pressure , Collagen/biosynthesis , Heart Failure/etiology , Heart Failure/physiopathology , Heart Rate , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Male , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Myocardium/metabolism , Organ Specificity , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Regression Analysis , Time Factors , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
17.
Circ Res ; 82(5): 576-86, 1998 Mar 23.
Article in English | MEDLINE | ID: mdl-9529162

ABSTRACT

Adhesion molecules mediate inflammatory myocardial injury after ischemia/reperfusion. Cytokine release and hypoxia are features of acute ischemia that may influence expression of these molecules. Accordingly, we studied intercellular adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM) responses to cytokines and acute hypoxia in cultured myocardial cells. Northern blot analysis and immunoassay showed that the proinflammatory cytokines interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha stimulated concentration-dependent increases in ICAM and VCAM mRNA and protein. In both cardiac myocytes and fibroblasts, pretreatment with a specific inhibitor of nuclear transcription factor-kappaB (NF-kappaB) prevented cytokine induction of both molecules. We also found that inhibition of tyrosine kinase and p38/RK (stress-activated protein kinase) pathways prevented IL-1beta-induced ICAM and VCAM protein synthesis, whereas extracellular signal-regulated protein kinase (ERK1/ERK2) inhibition did not. Neither hypoxia (0% O2 for 6 hours) alone nor hypoxia/reoxygenation had any significant effect on ICAM and VCAM mRNA. However, hypoxia did enhance IL-1beta-induced ICAM mRNA expression in myocytes. As a possible mechanism of this synergistic action on CAM expression, hypoxia induced a time-dependent increase in the DNA binding activity of both NF-kappaB and activator protein-1 (AP-1), two transcription factors important for cell adhesion molecule expression. In contrast to the enhanced ICAM mRNA induced by IL-1beta during hypoxia, however, protein levels for this adhesion molecule were unchanged beyond IL-1beta-stimulated levels, suggesting posttranscriptional and/or posttranslational control mechanisms. We conclude that cytokines regulate ICAM and VCAM mRNA and protein in both cardiac myocytes and fibroblasts. Furthermore, adhesion molecule induction requires translocation of at least two transcription factors, NF-kappaB and AP-1.


Subject(s)
Intercellular Adhesion Molecule-1/genetics , Interleukin-1/pharmacology , Myocardium/cytology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Animals , Cell Hypoxia/physiology , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation/drug effects , Intercellular Adhesion Molecule-1/metabolism , Monocytes/cytology , Myocardium/chemistry , Myocardium/enzymology , NF-kappa B/metabolism , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Time Factors , Transcription Factor AP-1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
18.
Cardiovasc Res ; 40(2): 352-63, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9893729

ABSTRACT

OBJECTIVE: We sought to determine whether angiotensin II (Ang II) promotes hypertrophy of cardiac directly or via paracrine mechanisms mediated by cardiac fibroblasts. METHODS: We studied neonatal rat cardiac myocytes and fibroblasts in culture as a model system. Paracrine effects of Ang II were identified using conditioned medium and co-culture experiments. RESULTS: Ang II type 1 (AT1) receptors responsible for myocyte growth localized to fibroblasts in radioligand binding, emulsion autoradiography, Western analysis, and immunofluorescence staining experiments. The bulk of AT1 receptor binding in myocyte cultures (1343 +/- 472 sites/cell) was to Ang II receptors on contaminating fibroblasts (9747 +/- 2126 sites/cell). Ang II induced significant paracrine trophic effects on myocytes in conditioned medium (40% increase in protein synthesis over control) and co-culture (4-fold increase over control) experiments. TGF-beta 1 and endothelin-1 were paracrine mediators of hypertrophy in neutralization experiments. CONCLUSIONS: Ang II stimulates cardiac myocyte hypertrophy via paracrine release of TGF-beta 1 and endothelin-1 from cardiac fibroblasts in a neonatal rat cell culture model.


Subject(s)
Angiotensin II/pharmacology , Endothelin-1/metabolism , Myocardium/pathology , Paracrine Communication , Transforming Growth Factor beta/metabolism , Vasoconstrictor Agents/pharmacology , Animals , Animals, Newborn , Blotting, Northern , Blotting, Western , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Endothelin-1/analysis , Fibroblasts/drug effects , Fibroblasts/metabolism , Microscopy, Fluorescence , Myocardium/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/genetics , Receptors, Angiotensin/metabolism , Transforming Growth Factor beta/analysis
19.
Circulation ; 96(6): 1937-43, 1997 Sep 16.
Article in English | MEDLINE | ID: mdl-9323084

ABSTRACT

BACKGROUND: We wished to determine whether the cytokine-inducible nitric oxide synthase (iNOS) pathway is modulated by chronic hypoxia in vitro. METHODS AND RESULTS: We investigated the effects of the proinflammatory cytokine interleukin (IL)-1beta on expression of iNOS mRNA, iNOS protein, and NO production in cultured neonatal rat cardiomyocytes subjected to 1% O2 for 48 hours. Among several cytokines tested, IL-1beta was the most effective in stimulating NO production, which was maximum at 48 hours. In parallel, IL-1beta induced expression of both iNOS mRNA and protein. Hypoxia alone had no effect on NO production, iNOS gene expression, or protein induction. However, chronic hypoxia decreased IL-1beta-stimulated NO production, mRNA expression, and protein level in cardiac myocytes. Radioligand binding and electrophoretic mobility shift assays showed that during chronic hypoxia, IL-1 receptor density and activity of the transcription factor NF-kappaB induced by IL-1beta were decreased, which may account at least in part for the decrease in iNOS expression. CONCLUSIONS: These data indicate that IL-1beta induces iNOS gene expression, de novo synthesis of iNOS protein, and NO generation in neonatal rat cardiomyocytes and that chronic hypoxia appears to be a potent negative regulator of iNOS expression in these cells.


Subject(s)
Hypoxia/metabolism , Interleukin-1/pharmacology , Muscle Fibers, Skeletal/enzymology , Myocardium/cytology , Nitric Oxide Synthase/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Chronic Disease , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Genistein , Interleukin 1 Receptor Antagonist Protein , Isoflavones/pharmacology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Myocardium/chemistry , Myocardium/enzymology , NF-kappa B/metabolism , Naphthalenes/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/antagonists & inhibitors , Sialoglycoproteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , omega-N-Methylarginine/pharmacology
20.
Circ Res ; 81(3): 396-403, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9285642

ABSTRACT

Signaling mediated by the angiotensin (Ang) II and alpha1-adrenergic receptor (alpha1-AR) pathways is important for cardiovascular homeostasis. However, it is unknown whether Ang II has any direct effect on alpha1-AR expression and signaling in cardiac myocytes. In the present study, we determined alpha1-AR subtype mRNA levels by RNase protection; receptor density by competition binding with 5-methylurapidil; and alpha1-AR-mediated c-fos expression by Northern blot analysis. We found that Ang II had no effect on alpha1b- and alpha1d-AR mRNA levels but decreased the alpha1a-AR mRNA level in a time- and dose-dependent manner. The maximal effect occurred at 6 hours with 100 nmol/L Ang II (40.0+/-8.2% reduction, n=4, P<.01). The decrease in alpha1a-AR mRNA level induced by Ang II is mediated by the Ang II AT1 receptor subtype and is associated with decreased stability of alpha1a-AR mRNA. Corresponding to the changes in the alpha1a-AR mRNA level, Ang II (100 nmol/L, 24 hours) reduced the density of high-affinity sites for 5-methylurapidil (alpha1A-AR) by 29% (56.5+/-6.4 versus 79.0+/-11.6 fmol/mg protein, n=4, P<.05). Alpha1-AR-stimulated c-fos induction, which could be blocked by 5-methylurapidil but not by chloroethylclonidine, was attenuated by Ang II preincubation (100 nmol/L, 24 hours). We conclude that there is previously undescribed cross talk between AT1 receptors and alpha1-ARs. Ang II selectively downregulates alpha1a-AR subtype mRNA and its corresponding receptor as well as alpha1a-AR-mediated expression of the immediate-early gene c-fos in cardiac myocytes.


Subject(s)
Angiotensin II/pharmacology , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Angiotensin/metabolism , Animals , Animals, Newborn , Cells, Cultured , Down-Regulation/drug effects , Genes, fos/drug effects , Myocardium/cytology , Myocardium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...