Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Physiol Rep ; 4(3)2016 Feb.
Article in English | MEDLINE | ID: mdl-26869681

ABSTRACT

Isolated cardiac tissue allows investigators to study mechanisms underlying normal and pathological conditions, which would otherwise be difficult or impossible to perform in vivo. Cultured neonatal rat ventricular cardiac myocytes (NRVM) are widely used to study signaling and growth mechanisms in the heart, primarily due to the versatility, economy, and convenience of this in vitro model. However, the lack of a well-defined longitudinal cellular axis greatly hampers the ability to measure contractile function in these cells, and therefore to associate signaling with mechanical function. In these methods, we demonstrate that this limitation can be overcome by using papillary muscles isolated from neonatal rat hearts. In the methods we describe procedures for isolation of right ventricular papillary muscles from 3-day-old neonatal rats and effects of mechanical and humoral stimuli on contraction and relaxation properties of these tissues.


Subject(s)
Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Organ Culture Techniques/methods , Papillary Muscles/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Electrophysiology/methods , Rats
2.
Int J Cardiol ; 203: 145-55, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26512830

ABSTRACT

Mechanical stretch is a major determinant that leads to heart failure, which is associated with a steady increase in myocardial angiotensinogen (Aogen) expression and formation of the biological peptide angiotensin II (Ang II). c-jun NH2-terminal kinase (JNK) and p38α have been found to have opposing roles on stretch-induced Aogen gene expression in neonatal rat ventricular myocytes (NRVM). JNK negatively regulated Aogen expression in NRVM following acute stretch, whereas with prolonged stretch, JNK phosphorylation was downregulated and p38α was found responsible for upregulation of Aogen expression. However, the mechanisms responsible for regulation of these kinases, especially the cross-talk between p38 and JNK1/2, remain to be determined. In this study, a combination of pharmacologic and molecular approaches (adenovirus-mediated gene transfer) were used to examine the mechanisms by which p38 regulates JNK phosphorylation in NRVM under stretch and non-stretch conditions. Pharmacologic inhibition of p38 significantly increased JNK phosphorylation in NRVM at 15 min, whereas overexpression of wild-type p38α significantly decreased JNK phosphorylation. While p38α overexpression prevented stretch-induced JNK phosphorylation, pharmacologic p38 inhibition abolished the JNK dephosphorylation during 15-60 min of stretch. Expression of constitutively-active MKK3 (MKK3CA), the upstream activator of p38, abolished JNK phosphorylation in both basal and stretched NRVM. Pharmacologic inhibition of MAP kinase phosphatase-1 (MKP-1) or protein phosphatase-1 (PP1) increased JNK phosphorylation in NRVM, suggesting the involvement of these phosphatases on reversing stretch-induced JNK activation. Inhibition of MKP-1, but not PP1, reduced JNK phosphorylation in NRVM overexpressing MKK3CA under basal conditions (no-stretch). Inhibition of MKP-1 also enhanced stretch-induced JNK phosphorylation in NRVM at 15 to 60 min. In summary, these results indicate that MKP-1 inhibits JNK phosphorylation in stretched NRVM through p38 dependent and independent mechanisms, whereas PP1 regulates JNK through a p38-independent mechanism.


Subject(s)
Dual Specificity Phosphatase 1/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Mitogen-Activated Protein Kinase 14/physiology , Myocytes, Cardiac/physiology , Animals , Animals, Newborn , Biomechanical Phenomena , Rats , Rats, Sprague-Dawley
3.
J Clin Exp Cardiolog ; 5(6): 314, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-25485172

ABSTRACT

The role of mechanical force as an important regulator of structure and function of mammalian cells, tissues, and organs has recently been recognized. However, mechanical overload is a pathogenesis or comorbidity existing in a variety of heart diseases, such as hypertension, aortic regurgitation and myocardial infarction. Physical stimuli sensed by cells are transmitted through intracellular signal transduction pathways resulting in altered physiological responses or pathological conditions. Emerging evidence from experimental studies indicate that ß1-integrin and the angiotensin II type I (AT1) receptor play critical roles as mechanosensors in the regulation of heart contraction, growth and leading to heart failure. Integrin link the extracellular matrix and the intracellular cytoskeleton to initiate the mechanical signalling, whereas, the AT1 receptor could be activated by mechanical stress through an angiotensin-II-independent mechanism. Recent studies show that both Integrin and AT1 receptor and their downstream signalling factors including MAPKs, AKT, FAK, ILK and GTPase regulate heart function in cardiac myocytes. In this review we describe the role of mechanical sensors residing within the plasma membrane, mechanical sensor induced downstream signalling factors and its potential roles in cardiac contraction and growth.

4.
Methods Mol Biol ; 1066: 45-56, 2013.
Article in English | MEDLINE | ID: mdl-23955732

ABSTRACT

Neonatal rat ventricular myocytes (NRVM) and fibroblasts (FB) serve as in vitro models for studying fundamental mechanisms underlying cardiac pathologies, as well as identifying potential therapeutic targets. Typically, these cell types are separated using Percoll density gradient procedures. Cells located between the Percoll bands (interband cells [IBCs]), which contain less mature NRVM and a variety of non-myocytes, including coronary vascular smooth muscle cells and endothelial cells (ECs), are routinely discarded. However, we have demonstrated that IBCs readily attach to extracellular matrix-coated coverslips, plastic culture dishes, and deformable membranes to form a 2-dimensional cardiac tissue layer which quickly develops spontaneous contraction within 24 h, providing a robust coculture model for the study of cell-to-cell signaling and contractile studies. Below, we describe methods that provide good cell yield and viability of IBCs during isolation of NRVM and FB obtained from 0- to 3-day-old neonatal rat pups. Basic characterization of IBCs and methods for use in intracellular calcium and contractile experiments are also presented. This method maximizes the use of cells obtained from neonatal rat hearts.


Subject(s)
Calcium/analysis , Fibroblasts/cytology , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cells, Cultured , Centrifugation, Density Gradient , Fibroblasts/metabolism , Fibroblasts/physiology , Heart/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction
5.
Methods Mol Biol ; 1066: 57-66, 2013.
Article in English | MEDLINE | ID: mdl-23955733

ABSTRACT

Mechanical stretch is a major factor for myocardial hypertrophy and heart failure. Stretch activates mechanical sensors from cardiac myocytes, leading to a series of signal transduction cascades, which can result in cell malfunction and remodeling. It is well known that mechanical stretch also induces the release of paracrine factors from cardiac fibroblasts, as well as myocytes. Due to complicated circumstance of heart tissue, it is difficult to fully investigate the characteristics of these factors in situ. Here we describe static stretch and conditioned medium experiments as methods to examine the function of paracrine factors between primary cultured cardiac myocytes and fibroblasts.


Subject(s)
Fibroblasts/physiology , Myocytes, Cardiac/physiology , Paracrine Communication/physiology , Stress, Mechanical , Animals , Cardiomegaly , Cells, Cultured , Centrifugation, Density Gradient , Extracellular Matrix/metabolism , Heart/physiology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
6.
Int J Cardiol ; 168(4): 3884-95, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23907041

ABSTRACT

BACKGROUND: Anthrax lethal toxin (LT), secreted by Bacillus anthracis, causes severe cardiac dysfunction by unknown mechanisms. LT specifically cleaves the docking domains of MAPKK (MEKs); thus, we hypothesized that LT directly impairs cardiac function through dysregulation of MAPK signaling mechanisms. METHODS AND RESULTS: In a time-course study of LT toxicity, echocardiography revealed acute diastolic heart failure accompanied by pulmonary regurgitation and left atrial dilation in adult Sprague-Dawley rats at time points corresponding to dysregulated JNK, phospholamban (PLB) and protein phosphatase 2A (PP2A) myocardial signaling. Using isolated rat ventricular myocytes, we identified the MEK7-JNK1-PP2A-PLB signaling axis to be important for regulation of intracellular calcium (Ca(2+)(i)) handling, PP2A activation and targeting of PP2A-B56α to Ca(2+)(i) handling proteins, such as PLB. Through a combination of gain-of-function and loss-of-function studies, we demonstrated that over-expression of MEK7 protects against LT-induced PP2A activation and Ca(2+)(i) dysregulation through activation of JNK1. Moreover, targeted phosphorylation of PLB-Thr(17) by Akt improved sarcoplasmic reticulum Ca(2+)(i) release and reuptake during LT toxicity. Co-immunoprecipitation experiments further revealed the pivotal role of MEK7-JNK-Akt complex formation for phosphorylation of PLB-Thr(17) during acute LT toxicity. CONCLUSIONS: Our findings support a cardiogenic mechanism of LT-induced diastolic dysfunction, by which LT disrupts JNK1 signaling and results in Ca(2+)(i) dysregulation through diminished phosphorylation of PLB by Akt and increased dephosphorylation of PLB by PP2A. Integration of the MEK7-JNK1 signaling module with Akt represents an important stress-activated signalosome that may confer protection to sustain cardiac contractility and maintain normal levels of Ca(2+)(i) through PLB-T(17) phosphorylation.


Subject(s)
Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Calcium-Binding Proteins/metabolism , Heart Failure, Diastolic/chemically induced , Heart Failure, Diastolic/metabolism , Signal Transduction/physiology , Acute Disease , Animals , Calcium-Binding Proteins/antagonists & inhibitors , Cells, Cultured , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
7.
Int J Cardiol ; 168(1): 436-45, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23058350

ABSTRACT

BACKGROUND: The cardiac renin-angiotensin system (RAS) has been implicated in mediating myocyte hypertrophy and remodeling, although the biochemical mechanisms responsible for regulating the local RAS are poorly understood. Caveolin-1 (Cav-1)/Cav-3 double-knockout mice display cardiac hypertrophy, and in vitro disruption of lipid rafts/caveolae using methyl-ß-cyclodextrin (MßCD) abolishes cardiac protection. METHODS: In this study, neonatal rat ventricular myocytes (NRVM) were used to determine whether lipid rafts/caveolae may be involved in the regulation of angiotensinogen (Ao) gene expression, a substrate of the RAS system. RESULTS: Treatment with MßCD caused a time-dependent upregulation of Ao gene expression, which was associated with differential regulation of mitogen-activated protein (MAP) kinases ERK1/2, p38 and JNK phosphorylation. JNK was highly phosphorylated shortly after MßCD treatment (2-30 min), whereas marked activation of ERK1/2 and p38 occurred much later (2-4h). ß1D-Integrin was required for MßCD-induced activation of the MAP kinases. Pharmacologic inhibition of ERK1/2 and JNK enhanced MßCD-induced Ao gene expression, whereas p38 blockade inhibited this response. Adenovirus-mediated expression of wild-type p38α enhanced MßCD-induced Ao gene expression; conversely expression of dominant negative p38α blocked the stimulatory effects of MßCD. Expression of Cav-3 siRNA stimulated Ao gene expression, whereas overexpression of Cav-3 was inhibitory. Cav-1 and Cav-3 expression levels were found to be positively regulated by p38, but unaffected by ERK1/2 and JNK. CONCLUSION: Collectively, these studies indicate that lipid rafts/caveolae couple to Ao gene expression through a mechanism that involves ß1-integrin and the differential actions of MAP kinase family members.


Subject(s)
Angiotensinogen/biosynthesis , Caveolin 3/biosynthesis , Gene Expression Regulation , Integrin beta1/biosynthesis , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Caveolin 1/biosynthesis , Cells, Cultured , Gene Knockdown Techniques/methods , Membrane Microdomains/metabolism , RNA, Small Interfering/biosynthesis , Rats , Rats, Sprague-Dawley
8.
Cardiovasc Res ; 90(1): 88-96, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21131638

ABSTRACT

AIMS: Angiotensin II (Ang II) stimulates cardiac remodelling and fibrosis in the mechanically overloaded myocardium. Although Rho GTPases regulate several cellular processes, including myocardial remodelling, involvement in mediating mechanical stretch-induced regulation of angiotensinogen (Ao), the precursor to Ang II, remains to be determined. We, therefore, examined the role and associated signalling mechanisms of Rho GTPases (Rac1 and RhoA) in regulation of Ao gene expression in a stretch model of neonatal rat cardiac fibroblasts (CFs). METHODS AND RESULTS: CFs were plated on deformable stretch membranes. Equiaxial mechanical stretch caused significant activation of both Rac1 and RhoA within 2-5 min. Rac1 activity returned to control levels after 4 h, whereas RhoA remained at a high level of activity until the end of the stretch period (24 h). Mechanical stretch initially caused a moderate decrease in Ao gene expression, but was significantly increased at 8-24 h. RhoA had a major role in mediating both the stretch-induced inhibition of Ao at 4 h and the subsequent upregulation of Ao expression at 24 h. ß1 integrin receptor blockade by Tac ß1 expression impaired acute (2 and 15 min) stretch-induced Rac1 activation, but increased RhoA activity. Molecular experiments revealed that Ao gene expression was inhibited by Rac1 through both JNK-dependent and independent mechanisms, and stimulated by RhoA through a p38-dependent mechanism. CONCLUSION: These results indicate that stretch-induced activation of Rac1 and RhoA differentially regulates Ao gene expression by modulating p38 and JNK activation.


Subject(s)
Angiotensinogen/metabolism , Cell Shape , Fibroblasts/enzymology , Mechanotransduction, Cellular , Myocardium/enzymology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Angiotensinogen/genetics , Animals , Animals, Newborn , Cells, Cultured , Enzyme Activation , Gene Expression Regulation , Integrin beta1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Phosphorylation , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Stress, Mechanical , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
9.
Front Biosci (Landmark Ed) ; 14(6): 2307-34, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273203

ABSTRACT

Integrins are heterodimeric cell-surface molecules, which act as the principle mediators of molecular dialog between a cell and its extracellular matrix environment. In addition to their structural functions, integrins mediate signaling from the extracellular space into the cell through integrin-associated signaling and adaptor molecules such as FAK (focal adhesion kinase), ILK (integrin-linked kinase), PINCH (particularly interesting new cysteine-histidine rich protein) and Nck2 (non-catalytic (region of) tyrosine kinase adaptor protein-2). Via these molecules, integrin signaling tightly and cooperatively interacts with receptor tyrosine kinases (RTKs) signaling to regulate survival, proliferation and cell shape as well as polarity, adhesion, migration and differentiation. In the heart and blood vessels, the function and regulation of these molecules can be partially disturbed and thus contribute to cardiovascular diseases such as cardiac hypertrophy and atherosclerosis. In this review, we discuss the primary mechanisms of action and signaling of integrins in the cardiac and vascular system in normal and pathological states, as well as therapeutic strategies for targeting these systems (1).


Subject(s)
Cardiovascular Diseases/physiopathology , Integrins/physiology , Signal Transduction , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/therapy , Cell Membrane/metabolism , Humans , Integrins/metabolism , Receptor Cross-Talk
10.
Front Biosci (Landmark Ed) ; 14(6): 2335-57, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273204

ABSTRACT

Anthrax is a disease caused by infection with spores from the bacteria Bacillus anthracis. After entering the body, the spores germinate into bacteria and secrete a toxin that causes local edema and, in systemic infections, cardiovascular collapse and death. The toxin is a tripartite polypeptide, consisting of protective antigen (PA), lethal factor (LF) and edema factor (EF), which have key roles in the bacterial pathogenesis and disease progression. PA facilitates transfer of LF and EF to the cytosol. Lethal toxin is a zinc metalloproteinase, which has the capacity to inactivate mitogen-activated protein (MAP) kinase kinase (MEK) and stimulates the release of sepsis-related cytokines tumor necrosis factor-alpha and interleukin-1beta. Edema factor is a calmodulin (CaM)-dependent adenylate cyclase, which increases levels of cyclic AMP, causing impaired neutrophil function and disruption of water balance that ultimately results in massive tissue edema. Together, the toxins effectively inhibit host innate and adaptive immune responses, allowing the bacteria to grow unrestrained and overwhelming any resistance. Clinically, inhalational anthrax presents in a biphasic pattern with initial nonspecific "flu-like" symptoms nausea and vomiting 1 to 4 days after exposure, followed by severe illness with dyspnea, high fever and circulatory shock. The latter symptoms represent a terminal stage and treatment is often ineffective when started at that time. Key indicators of early anthrax cardiovascular-related pathogenesis include mediastinal widening in association with pleural effusion and edema. In this review, we describe the current understanding of anthrax toxins on cellular function in the context of cardiovascular function and discuss potential therapeutic strategies.


Subject(s)
Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Cardiovascular System/drug effects , Anthrax/physiopathology , Anthrax/therapy , Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Humans
11.
J Mol Cell Cardiol ; 45(6): 770-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18926830

ABSTRACT

The cardiac renin-angiotensin system (RAS) has been implicated in mediating myocyte hypertrophy, remodeling, and fibroblast proliferation in the hemodynamically overloaded heart. However, the intracellular signaling mechanisms responsible for regulation of angiotensinogen (Ao), a substrate of the RAS system, are largely unknown. Here we report the identification of JNK1/2 as a negative, and p38alpha as a major positive regulator of Ao gene expression. Isolated neonatal rat ventricular myocytes (NRVM) and fibroblasts (NRFB) plated on deformable membranes coated with collagen IV, were exposed to 20% equiaxial static-stretch (0-24 h). Mechanical stretch initially depressed Ao gene expression (4 h), whereas after 8 h, Ao gene expression increased in a time-dependent manner. Blockade of JNK1/2 with SP600125 increased basal Ao gene expression in NRVM (10.52+/-1.98 fold, P<0.001) and NRFB (13.32+/-2.07 fold, P<0.001). Adenovirus-mediated expression of wild-type JNK1 significantly inhibited, whereas expression of dominant-negative JNK1 and JNK2 increased basal and stretch-mediated (24 h) Ao gene expression, showing both JNK1 and JNK2 to be negative regulators of Ao gene expression in NRVM and NRFB. Blockade of p38alpha/beta by SB202190 or p38alpha by SB203580 significantly inhibited stretch-induced (24 h) Ao gene expression, whereas expression of wild-type p38alpha increased stretch-induced Ao gene expression in both NRVM (8.41+/-1.50 fold, P<0.001) and NRFB (3.39+/-0.74 fold, P<0.001). Conversely, expression of dominant-negative p38alpha significantly inhibited stretch response. Moreover, expression of constitutively active MKK6b (E) significantly stimulated Ao gene expression in the absence of stretch, indicating that p38 activation alone is sufficient to induce Ao gene expression. Taken together p38alpha was demonstrated to be a positive regulator, whereas JNK1/2 was found to be a negative regulator of Ao gene expression. Prolonged stretch diminished JNK1/2 activation, which was accompanied by a reciprocal increase in p38 activation and Ao gene expression. This suggests that a balance in JNK1/2 and p38alpha activation determines the level of Ao gene expression in myocardial cells.


Subject(s)
Angiotensinogen/biosynthesis , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Mitogen-Activated Protein Kinase 14/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Myocytes, Cardiac/metabolism , Animals , Cells, Cultured , Fibroblasts/cytology , Gene Expression Regulation/drug effects , Heart Ventricles/cytology , Heart Ventricles/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Myocardium/metabolism , Myocytes, Cardiac/cytology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/physiology , Stress, Physiological/drug effects , Stress, Physiological/physiology , Time Factors
12.
Cardiovasc Hematol Agents Med Chem ; 5(2): 109-32, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17430135

ABSTRACT

Integrins are the principle mediators of molecular dialog between a cell and its extracellular matrix environment. The unique combinations of integrin subunits determine which extracellular matrix molecules are recognized by a cell. Recent studies have demonstrated that remodeling in heart and vasculature is linked to alterations in extracellular matrix and integrin expression. The roles of integrins in controlling cellular behavior have made these molecules highly attractive drug targets. New insights into mechanisms whereby the extracellular matrix takes part in the control of smooth muscle cell proliferation and cardiac growth suggest a number of putative targets for future therapies that can be applied to increase plaque stability, prevent the clinical consequences of atherosclerosis and improve outcomes after interventional procedures such as cardiac transplantation. Therapeutic candidates include antibodies, cyclic peptides, peptidomimetics and small molecules. The integrin inhibitors Integrilin and ReoPro have been approved as blood thinners in cardiovascular disease, and newer agents are undergoing testing. Although integrin function is important in the cardiovascular system, there are wide gaps in knowledge. In this review, we discuss the primary mechanisms of action and signaling of integrins in the cardiac and vascular system in normal and pathological states, as well as therapeutic strategies for targeting these molecules in the cardiovascular system.


Subject(s)
Cardiovascular Agents/pharmacology , Cardiovascular Diseases/drug therapy , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Integrins , Cardiovascular Agents/chemistry , Cardiovascular Physiological Phenomena , Endothelium, Vascular/drug effects , Endothelium, Vascular/growth & development , Extracellular Matrix/drug effects , Humans , Integrins/antagonists & inhibitors , Integrins/drug effects , Integrins/metabolism , Randomized Controlled Trials as Topic , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...