Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Circ Res ; 117(12): 995-1000, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26429802

ABSTRACT

RATIONALE: The lack of measurable single-cell contractility of human-induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CMs) currently limits the utility of hiPSC-CMs for evaluating contractile performance for both basic research and drug discovery. OBJECTIVE: To develop a culture method that rapidly generates contracting single hiPSC-CMs and allows quantification of cell shortening with standard equipment used for studying adult CMs. METHODS AND RESULTS: Single hiPSC-CMs were cultured for 5 to 7 days on a 0.4- to 0.8-mm thick mattress of undiluted Matrigel (mattress hiPSC-CMs) and compared with hiPSC-CMs maintained on a control substrate (<0.1-mm thick 1:60 diluted Matrigel, control hiPSC-CMs). Compared with control hiPSC-CMs, mattress hiPSC-CMs had more rod-shape morphology and significantly increased sarcomere length. Contractile parameters of mattress hiPSC-CMs measured with video-based edge detection were comparable with those of freshly isolated adult rabbit ventricular CMs. Morphological and contractile properties of mattress hiPSC-CMs were consistent across cryopreserved hiPSC-CMs generated independently at another institution. Unlike control hiPSC-CMs, mattress hiPSC-CMs display robust contractile responses to positive inotropic agents, such as myofilament calcium sensitizers. Mattress hiPSC-CMs exhibit molecular changes that include increased expression of the maturation marker cardiac troponin I and significantly increased action potential upstroke velocity because of a 2-fold increase in sodium current (INa). CONCLUSIONS: The Matrigel mattress method enables the rapid generation of robustly contracting hiPSC-CMs and enhances maturation. This new method allows quantification of contractile performance at the single-cell level, which should be valuable to disease modeling, drug discovery, and preclinical cardiotoxicity testing.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Cell Differentiation/drug effects , Cells, Cultured , Collagen/administration & dosage , Drug Combinations , Humans , Induced Pluripotent Stem Cells/drug effects , Laminin/administration & dosage , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , Proteoglycans/administration & dosage
2.
Pulm Circ ; 5(3): 481-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26401249

ABSTRACT

Little is known about the right ventricular (RV) proteome in human heart failure (HF), including possible differences compared to the left ventricular (LV) proteome. We used 2-dimensional differential in-gel electrophoresis (pH: 4-7, 10-150 kDa), followed by liquid chromatography tandem mass spectrometry, to compare the RV and LV proteomes in 12 explanted human hearts. We used Western blotting and multiple-reaction monitoring for protein verification and RNA sequencing for messenger RNA and protein expression correlation. In all 12 hearts, the right ventricles (RVs) demonstrated differential expression of 11 proteins relative to the left ventricles (LVs), including lesser expression of CRYM, TPM1, CLU, TXNL1, and COQ9 and greater expression of TNNI3, SAAI, ERP29, ACTN2, HSPB2, and NDUFS3. Principal-components analysis did not suggest RV-versus-LV proteome partitioning. In the nonischemic RVs (n = 6), 7 proteins were differentially expressed relative to the ischemic RVs (n = 6), including increased expression of CRYM, B7Z964, desmin, ANXA5, and MIME and decreased expression of SERPINA1 and ANT3. Principal-components analysis demonstrated partitioning of the nonischemic and ischemic RV proteomes, and gene ontology analysis identified differences in hemostasis and atherosclerosis-associated networks. There were no proteomic differences between RVs with echocardiographic dysfunction (n = 8) and those with normal function (n = 4). Messenger RNA and protein expression did not correlate consistently, suggesting a major role for RV posttranscriptional protein expression regulation. Differences in contractile, cytoskeletal, metabolic, signaling, and survival pathways exist between the RV and the LV in HF and may be related to the underlying HF etiology and differential posttranscriptional regulation.

3.
Development ; 141(2): 335-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24353062

ABSTRACT

Organ development is a highly regulated process involving the coordinated proliferation and differentiation of diverse cellular populations. The pathways regulating cell proliferation and their effects on organ growth are complex and for many organs incompletely understood. In all vertebrate species, the cardiac natriuretic peptides (ANP and BNP) are produced by cardiomyocytes in the developing heart. However, their role during cardiogenesis is not defined. Using the embryonic zebrafish and neonatal mammalian cardiomyocytes we explored the natriuretic peptide signaling network during myocardial development. We observed that the cardiac natriuretic peptides ANP and BNP and the guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2 are functionally redundant during early cardiovascular development. In addition, we demonstrate that low levels of the natriuretic peptides preferentially activate Npr3, a receptor with Gi activator sequences, and increase cardiomyocyte proliferation through inhibition of adenylate cyclase. Conversely, high concentrations of natriuretic peptides reduce cardiomyocyte proliferation through activation of the particulate guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2, and activation of protein kinase G. These data link the cardiac natriuretic peptides in a complex hierarchy modulating cardiomyocyte numbers during development through opposing effects on cardiomyocyte proliferation mediated through distinct cyclic nucleotide signaling pathways.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cell Proliferation , Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Knockdown Techniques , Heart/embryology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
4.
Biochem Biophys Res Commun ; 425(4): 830-5, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22892129

ABSTRACT

Ankyrin repeat domain 1 protein (Ankrd1), also known as cardiac ankyrin repeat protein (CARP), increases dramatically after tissue injury, and its overexpression improves aspects of wound healing. Reports that Ankrd1/CARP protein stability may affect cardiovascular organization, together with our findings that the protein is crucial to stability of the cardiomyocyte sarcomere and increased in wound healing, led us to compare the contribution of Ankrd1/CARP stability to its abundance. We found that the 26S proteasome is the dominant regulator of Ankrd1/CARP degradation, and that Ankrd1/CARP half-life is significantly longer in cardiomyocytes (h) than endothelial cells (min). In addition, higher endothelial cell density decreased the abundance of the protein without affecting steady state mRNA levels. Taken together, our data and that of others indicate that Ankrd1/CARP is highly regulated at multiple levels of its expression. The striking difference in protein half-life between a muscle and a non-muscle cell type suggests that post-translational proteolysis is correlated with the predominantly structural versus regulatory role of the protein in the two cell types.


Subject(s)
Endothelium, Vascular/metabolism , Heart Ventricles/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Animals , Cell Count , Cells, Cultured , Half-Life , Heart Ventricles/cytology , Humans , Microvessels/metabolism , Proteolysis , Rats
5.
Am J Physiol Heart Circ Physiol ; 303(2): H156-67, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22636675

ABSTRACT

The isolated retrograde-perfused Langendorff heart and the isolated ejecting heart have, over many decades, resulted in fundamental discoveries that form the underpinnings of our current understanding of the biology and physiology of the heart. These two experimental methodologies have proven invaluable in studying pharmacological effects on myocardial function, metabolism, and vascular reactivity and in the investigation of clinically relevant disease states such as ischemia-reperfusion injury, diabetes, obesity, and heart failure. With the advent of the genomics era, the isolated mouse heart preparation has gained prominence as an ex vivo research tool for investigators studying the impact of gene modification in the intact heart. This review summarizes the historical development of the isolated heart and provides a practical guide for the establishment of the Langendorff and ejecting heart preparations with a particular emphasis on the murine heart. In addition, current applications and novel methods of recording cardiovascular parameters in the isolated heart preparation will be discussed. With continued advances in methodological recordings, the isolated mouse heart preparation will remain physiologically relevant for the foreseeable future, serving as an integral bridge between in vitro assays and in vivo approaches.


Subject(s)
Heart/physiology , Stroke Volume , Animals , Anura , Cardiac Electrophysiology/history , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Mice , Mice, Mutant Strains , Myocardial Contraction/physiology , Perfusion/methods , Tissue and Organ Harvesting/history
6.
PLoS One ; 7(4): e35743, 2012.
Article in English | MEDLINE | ID: mdl-22532871

ABSTRACT

Doxorubicin (Adriamycin) is an effective anti-cancer drug, but its clinical usage is limited by a dose-dependent cardiotoxicity characterized by widespread sarcomere disarray and loss of myofilaments. Cardiac ankyrin repeat protein (CARP, ANKRD1) is a transcriptional regulatory protein that is extremely susceptible to doxorubicin; however, the mechanism(s) of doxorubicin-induced CARP depletion and its specific role in cardiomyocytes have not been completely defined. We report that doxorubicin treatment in cardiomyocytes resulted in inhibition of CARP transcription, depletion of CARP protein levels, inhibition of myofilament gene transcription, and marked sarcomere disarray. Knockdown of CARP with small interfering RNA (siRNA) similarly inhibited myofilament gene transcription and disrupted cardiomyocyte sarcomere structure. Adenoviral overexpression of CARP, however, was unable to rescue the doxorubicin-induced sarcomere disarray phenotype. Doxorubicin also induced depletion of the cardiac transcription factor GATA4 in cardiomyocytes. CARP expression is regulated in part by GATA4, prompting us to examine the relationship between GATA4 and CARP in cardiomyocytes. We show in co-transfection experiments that GATA4 operates upstream of CARP by activating the proximal CARP promoter. GATA4-siRNA knockdown in cardiomyocytes inhibited CARP expression and myofilament gene transcription, and induced extensive sarcomere disarray. Adenoviral overexpression of GATA4 (AdV-GATA4) in cardiomyocytes prior to doxorubicin exposure maintained GATA4 levels, modestly restored CARP levels, and attenuated sarcomere disarray. Interestingly, siRNA-mediated depletion of CARP completely abolished the Adv-GATA4 rescue of the doxorubicin-induced sarcomere phenotype. These data demonstrate co-dependent roles for GATA4 and CARP in regulating sarcomere gene expression and maintaining sarcomeric organization in cardiomyocytes in culture. The data further suggests that concurrent depletion of GATA4 and CARP in cardiomyocytes by doxorubicin contributes in large part to myofibrillar disarray and the overall pathophysiology of anthracycline cardiomyopathy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cardiomyopathies/metabolism , Doxorubicin/pharmacology , GATA4 Transcription Factor/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/drug effects , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Sarcomeres/drug effects , Signal Transduction/physiology , Animals , Cardiomyopathies/chemically induced , GATA4 Transcription Factor/genetics , Male , Muscle Proteins/genetics , Myocytes, Cardiac/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Sarcomeres/metabolism
7.
Am J Physiol Heart Circ Physiol ; 300(4): H1311-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21239627

ABSTRACT

Endothelial progenitor cells (EPCs) are mobilized into the vascular space and home to damaged tissues, where they promote repair in part through a process of angiogenesis. Neuregulins (NRGs) are ligands in the epidermal growth factor family that signal through type I receptor tyrosine kinases in the erbB family (erbB2, erbB3, and erbB4) and regulate endothelial cell biology, promoting angiogenesis. Stimuli such as ischemia and exercise that promote EPC mobilization also induce cleavage and release of transmembrane NRG from cardiac microvascular endothelial cells (CMECs). We hypothesized that NRG/erbB signaling may regulate EPC biology. Using an embryonic (e)EPC cell line that homes to and repairs injured myocardium, we were able to detect erbB2 and erbB3 transcripts. Identical receptor expression was found in EPCs isolated from rat bone marrow and human whole blood. NRG treatment of eEPCs induces phosphorylation of kinases including Akt, GSK-3ß, and Erk1/2 and the nuclear accumulation and transcriptional activation of ß-catenin. NRG does not induce eEPC proliferation or migration but does protect eEPCs against serum deprivation-induced apoptosis. These results suggest a role for tissue-derived NRG in the regulation of EPC survival.


Subject(s)
Embryonic Stem Cells/physiology , Neuregulin-1/physiology , Animals , Bone Marrow Cells/metabolism , Cell Survival , Cells, Cultured , Embryonic Stem Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Leukocytes, Mononuclear/metabolism , Mice , Neuregulin-1/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-3/biosynthesis , beta Catenin/metabolism
8.
Prog Cardiovasc Dis ; 53(2): 105-13, 2010.
Article in English | MEDLINE | ID: mdl-20728697

ABSTRACT

Anthracycline antibiotics have saved the lives of many cancer victims in the 50 plus years since their discovery. A major limitation of their use is the dose-limiting cardiotoxicity. Efforts focusing on understanding the biochemical basis for anthracycline cardiac effects have provided several strategies currently in clinical use: limit dose exposure, encapsulate anthracyclines in liposomes to reduce myocardial uptake, administer concurrently with the iron chelator dexrazoxane to reduce free iron-catalyzed reactive oxygen species formation; and modify anthracycline structure in an effort to reduce myocardial toxicity. Despite these efforts, anthracycline-induced heart failure continues to occur with consequences for both morbidity and mortality. Our inability to predict and prevent anthracycline cardiotoxicity is, in part, due to the fact that the molecular and cellular mechanisms remain controversial and incompletely understood. Studies examining the effects of anthracyclines in cardiac myocytes in vitro and small animals in vivo have demonstrated several forms of cardiac injury, and it remains unclear how these translate to the clinical setting. Given the clinical evidence that myocyte death occurs after anthracycline exposure in the form of elevations in serum troponin, myocyte cell death seems to be a probable mechanism for anthracycline-induced cardiac injury. Other mechanisms of myocyte injury include the development of cellular "sarcopenia" characterized by disruption of normal sarcomere structure. Anthracyclines suppress expression of several cardiac transcription factors, and this may play a role in the development of myocyte death as well as sarcopenia. Degradation of the giant myofilament protein titin may represent an important proximal step that leads to accelerated myofilament degradation. An interesting interaction has been noted clinically between anthracyclines and newer cancer therapies that target the erbB2 receptor tyrosine kinase. There is now evidence that erbB2 signaling in response to the ligand neuregulin regulates anthracycline uptake into cells via the multidrug-resistance protein. Therefore, up-regulation of cardiac neuregulin signaling may be one strategy to limit myocardial anthracycline injury. Moreover, assessing an individual's risk for anthracycline injury may be improved by having some measure of endogenous activity of this and other myocardial protective signals.


Subject(s)
Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Heart Diseases/chemically induced , Myocytes, Cardiac/drug effects , Neoplasms/drug therapy , Animals , Cell Death , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/prevention & control , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Receptor, ErbB-2/metabolism , Sarcomeres/drug effects , Sarcomeres/metabolism , Signal Transduction/drug effects
9.
Am J Physiol Heart Circ Physiol ; 297(6): H2144-53, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19801492

ABSTRACT

The female sex has been associated with increased resistance to acute myocardial ischemia-reperfusion (I/R) injury. While enhanced antioxidant capacity has been implicated in female cardioprotection, there is little evidence to support this assumption. Previous studies have shown an important role of cellular glutathione peroxidase (GPx1) in protection of the myocardium from I/R injury. Whether GPx1 is mechanistic in the protection of female myocardium, post-I/R, has not been examined. We utilized a murine model with homozygous deletion of GPx1 and examined its impact on postischemic myocardial recovery in male and female mice. Following I/R, male GPx1(-/-) hearts were more susceptible to contractile and diastolic dysfunction, and this was associated with increased protein carbonyls, a marker of oxidative stress. In contrast, GPx1 deficiency in female hearts did not exacerbate dysfunction or oxidative stress post-I/R. Both wild-type and GPx1(-/-) female hearts exhibited reduced creatine kinase leakage and a more favorable ascorbate redox status compared with males. Following I/R, female GPx1(-/-) hearts showed a comparable decrease in glutathione redox status as their male counterparts; however, they exhibited a greater decrease in nitrate-to-nitrite ratio, suggesting a higher consumption of nitrate in female GPx1(-/-) hearts. Our findings demonstrate that GPx1 is critical for cardioprotection during I/R in male, but not female, mice. The maintenance of cardioprotection in female mice lacking GPx1 post-I/R may be due to an improved ascorbate redox homeostasis and enhanced nitrate-to-nitrite conversion, which would predictably be accompanied by enhanced production of cardioprotective nitric oxide.


Subject(s)
Antioxidants/metabolism , Glutathione Peroxidase/deficiency , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Oxidative Stress , Animals , Ascorbic Acid/metabolism , Creatine Kinase/metabolism , Disease Models, Animal , Female , Glutathione/metabolism , Glutathione Peroxidase/genetics , Male , Mice , Mice, Knockout , Myocardial Contraction , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Nitrates/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Oxidation-Reduction , Recovery of Function , Sex Factors , Time Factors , Ventricular Function, Left , Glutathione Peroxidase GPX1
10.
Circulation ; 119(15): 2058-68, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19349318

ABSTRACT

BACKGROUND: Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor superfamily, is a multifunctional cytokine known to regulate cellular functions in contexts of injury and disease through its receptor, fibroblast growth factor-inducible molecule 14 (Fn14). Although many of the processes and downstream signals regulated by the TWEAK/Fn14 pathway have been implicated in the development of cardiac dysfunction, the role of TWEAK in the cardiovascular system is completely unknown. METHODS AND RESULTS: Herein, we demonstrate that mouse and human cardiomyocytes express the TWEAK receptor Fn14. Furthermore, we determine that elevated circulating levels of TWEAK, induced via transgenic or adenoviral-mediated gene expression in mice, result in dilated cardiomyopathy with subsequent severe cardiac dysfunction. This phenotype was mediated exclusively by the Fn14 receptor, independent of tumor necrosis factor-alpha, and was associated with cardiomyocyte elongation and cardiac fibrosis but not cardiomyocyte apoptosis. Moreover, we find that circulating TWEAK levels were differentially upregulated in patients with idiopathic dilated cardiomyopathy compared with other forms of heart disease and normal control subjects. CONCLUSIONS: Our data suggest that TWEAK/Fn14 may be important in regulating myocardial structural remodeling and function and may play a role in the pathogenesis of dilated cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factors/physiology , Animals , Apoptosis , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cell Size , Coronary Disease/metabolism , Coronary Disease/pathology , Cytokine TWEAK , Female , Fibrosis , Heart Failure/etiology , Heart Failure/pathology , Humans , Hypertension/complications , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Middle Aged , Phenotype , Recombinant Fusion Proteins/physiology , TWEAK Receptor , Transduction, Genetic , Tumor Necrosis Factors/blood , Tumor Necrosis Factors/genetics
11.
Am J Physiol Heart Circ Physiol ; 294(3): H1335-47, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18192223

ABSTRACT

In contrast to the other heterotrimeric GTP-binding proteins (G proteins) Gs and Gi, the functional role of G o is still poorly defined. To investigate the role of G alpha o in the heart, we generated transgenic mice with cardiac-specific expression of a constitutively active form of G alpha o1* (G alpha o*), the predominant G alpha o isoform in the heart. G alpha o expression was increased 3- to 15-fold in mice from 5 independent lines, all of which had a normal life span and no gross cardiac morphological abnormalities. We demonstrate enhanced contractile function in G alpha o* transgenic mice in vivo, along with increased L-type Ca2+ channel current density, calcium transients, and cell shortening in ventricular G alpha o*-expressing myocytes compared with wild-type controls. These changes were evident at baseline and maintained after isoproterenol stimulation. Expression levels of all major Ca2+ handling proteins were largely unchanged, except for a modest reduction in Na+/Ca2+ exchanger in transgenic ventricles. In contrast, phosphorylation of the ryanodine receptor and phospholamban at known PKA sites was increased 1.6- and 1.9-fold, respectively, in G alpha o* ventricles. Density and affinity of beta-adrenoceptors, cAMP levels, and PKA activity were comparable in G alpha o* and wild-type myocytes, but protein phosphatase 1 activity was reduced upon G alpha o* expression, particularly in the vicinity of the ryanodine receptor. We conclude that G alpha o* exerts a positive effect on Ca2+ cycling and contractile function. Alterations in protein phosphatase 1 activity rather than PKA-mediated phosphorylation might be involved in hyperphosphorylation of key Ca2+ handling proteins in hearts with constitutive G alpha o activation.


Subject(s)
Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/biosynthesis , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Myocardium/metabolism , Actin Cytoskeleton/physiology , Adenoviridae Infections/pathology , Adrenergic beta-Agonists/pharmacology , Animals , Animals, Genetically Modified , Blotting, Northern , Blotting, Western , Calcium Channels, L-Type/physiology , Calcium Signaling/physiology , Cell Separation , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytochromes c/biosynthesis , Isoproterenol/pharmacology , Mice , Myocytes, Cardiac/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/physiology
12.
Biophys J ; 94(9): 3577-89, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18212018

ABSTRACT

Troponin C (TnC) belongs to the superfamily of EF-hand (helix-loop-helix) Ca(2+)-binding proteins and is an essential component of the regulatory thin filament complex. In a patient diagnosed with idiopathic dilated cardiomyopathy, we identified two novel missense mutations localized in the regulatory Ca(2+)-binding Site II of TnC, TnC((E59D,D75Y)). Expression of recombinant TnC((E59D,D75Y)) in isolated rat cardiomyocytes induced a marked decrease in contractility despite normal intracellular calcium homeostasis in intact cardiomyocytes and resulted in impaired myofilament calcium responsiveness in Triton-permeabilized cardiomyocytes. Expression of the individual mutants in cardiomyocytes showed that TnC(D75Y) was able to recapitulate the TnC((E59D,D75Y)) phenotype, whereas TnC(E59D) was functionally benign. Force-pCa relationships in TnC((E59D,D75Y)) reconstituted rabbit psoas fibers and fluorescence spectroscopy of TnC((E59D,D75Y)) labeled with 2-[(4'-iodoacetamide)-aniline]naphthalene-6-sulfonic acid showed a decrease in myofilament Ca(2+) sensitivity and Ca(2+) binding affinity, respectively. Furthermore, computational analysis of TnC showed the Ca(2+)-binding pocket as an active region of concerted motions, which are decreased markedly by mutation D75Y. We conclude that D75Y interferes with proper concerted motions within the regulatory Ca(2+)-binding pocket of TnC that hinders the relay of the thin filament calcium signal, thereby providing a primary stimulus for impaired cardiomyocyte contractility. This in turn may trigger pathways leading to aberrant ventricular remodeling and ultimately a dilated cardiomyopathy phenotype.


Subject(s)
Calcium/metabolism , Movement/physiology , Mutation, Missense , Myocardial Contraction/genetics , Myocytes, Cardiac/metabolism , Troponin C/genetics , Troponin C/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Cell Membrane Permeability , Gene Expression Regulation , Humans , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Protein Binding , Psoas Muscles/cytology , Psoas Muscles/metabolism , Rabbits , Sarcomeres/genetics , Sarcomeres/metabolism , Substrate Specificity , Troponin C/chemistry
13.
Cell Motil Cytoskeleton ; 64(9): 718-25, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17615571

ABSTRACT

A number of techniques have been developed to monitor contractile function in isolated cardiac myocytes. While invaluable observations have been gained from these methodologies in understanding the contractile processes of the heart, they are invariably limited by their in vitro conditions. The present challenge is to develop innovative assays to mimic the in vivo milieu so as to allow a more physiological assessment of cardiac myocyte contractile forces. Here we demonstrate the use of a silicone elastomer, poly(dimethylsiloxane) (PDMS), to simultaneously orient adult cardiac myocytes in primary culture and measure the cellular forces in a three-dimensional substrate. The realignment of adult cardiac myocytes in long-term culture (7 days) was achieved due to directional reassembly of the myofibrils along the parallel polymeric sidewalls. The cellular mechanical forces were recorded in situ by observing the deformation of the micropillars embedded in the substrate. By coupling the cellular mechanical force measurements with on-chip cell orientation, this novel assay is expected to provide a means of a more physiological assessment of single cardiac myocyte contractile function and may facilitate the future development of in vitro assembled functional cardiac tissue.


Subject(s)
Dimethylpolysiloxanes , Muscle Contraction , Myocytes, Cardiac/cytology , Nylons , Animals , Cell Culture Techniques/instrumentation , Cells, Cultured , Male , Rats , Rats, Wistar
14.
Cardiovasc Toxicol ; 7(2): 114-21, 2007.
Article in English | MEDLINE | ID: mdl-17652815

ABSTRACT

The molecular and cellular mechanisms that cause cumulative dose-dependent anthracycline-cardiotoxicity remain controversial and incompletely understood. Studies examining the effects of anthracyclines in cardiac myocytes inA vitro have demonstrated several forms of cellular injury. Cell death in response to anthracyclines can be observed by one of several mechanisms including apoptosis and necrosis. Cell death by apoptosis can be inhibited by dexrazoxane, the iron chelator that is known to prevent clinical development of heart failure at high cumulative anthracycline exposure. Together with clinical evidence for myocyte death after anthracycline exposure, in the form of elevations in serum troponin, make myocyte cell death a probable mechanism for anthracycline-induced cardiac injury. Other mechanisms of myocyte injury include the development of cellular \'sarcopenia\' characterized by disruption of normal sarcomere structure. Anthracyclines suppress expression of several cardiac transcription factors, and this may play a role in the development of myocyte death as well as sarcopenia. Degradation of the giant myofilament protein titin may represent an important proximal step that leads to accelerated myofilament degradation. Titin is an entropic spring element in the sarcomere that regulates length-dependent calcium sensitivity. Thus titin degradation may lead to impaired diastolic as well as systolic dysfunction, as well as potentiate the effect of suppression of transcription of sarcomere proteins. An interesting interaction has been noted clinically between anthracyclines and newer cancer therapies that target the erbB2 receptor tyrosine kinase. Studies of erbB2 function in viro suggest that signaling through erbB2 by the growth factor neuregulin may regulate cardiac myocyte sarcomere turnover, as well as myocyte-myocyte/myocyte-matrix force coupling. A combination of further in vitro studies, with more careful monitoring of cardiac function after exposure to these cancer therapies, may help to understand to what extent these mechanisms are at work during clinical exposure of the heart to these important pharmaceuticals.


Subject(s)
Anthracyclines/toxicity , Antibiotics, Antineoplastic/toxicity , Heart Diseases/chemically induced , Heart Diseases/pathology , Animals , Antibodies, Monoclonal/toxicity , Antibodies, Monoclonal, Humanized , Cell Death/drug effects , Heart Diseases/metabolism , Humans , Myocytes, Cardiac/drug effects , Sarcomeres/drug effects , Trastuzumab
15.
Biosens Bioelectron ; 22(8): 1688-93, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-16962309

ABSTRACT

Cardiac hypertrophy is an established and independent risk factor for the development of heart failure and sudden cardiac death. At the level of individual cardiac myocytes (heart muscle cells), the cell morphology alters (increase in cell size and myofibrillar re-organization) and protein synthesis is activated. In this paper, a novel cardiomyocyte-based impedance sensing system with the assistance of dielectrophoresis cell concentration is reported to monitor the dynamic process of endothelin-1-induced cardiomyocyte hypertrophy. A dielectrophoresis (DEP) microfluidic device is fabricated capable of concentrating cells from a dilute sample to form a confluent cell monolayer on the surface of microelectrodes. This device can increase the sensitivity of the impedance system and also has the potential to reduce the time for detection by a significant factor. To examine the feasibility of this impedance sensing system, cardiomyocytes are treated with endothelin-1 (ET-1), a known hypertrophic agent. ET-1 induces a continuous rise in cardiomyocyte impedance, which we interpret as strengthening of cellular attachments to the surface substrate. An equivalent circuit model is introduced to fit the impedance spectrum to fully understand the impedance sensing system.


Subject(s)
Biosensing Techniques/instrumentation , Cardiomegaly/diagnosis , Endothelin-1/pharmacology , Myocytes, Cardiac/pathology , Animals , Electric Impedance , Male , Microfluidic Analytical Techniques , Rats , Rats, Wistar
16.
Exp Physiol ; 91(4): 773-80, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16675497

ABSTRACT

Chronic tachycardia in patients and rapid pacing in animal models induce myocardial dysfunction and initiate a cascade of compensatory adaptations that are ultimately unsustainable, leading to ventricular enlargement and failure. The molecular pathogenesis during the early stages of tachycardia-induced cardiomyopathy, however, remains unclear. We utilized our previously reported cell culture pacing system to directly assess phosphatidylinositol-3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK) signalling of adult rat ventricular myocytes (ARVM) in response to rapid electrical stimulation. Freshly isolated ARVMs were maintained quiescent (0 Hz), or continuously stimulated at 5 (normofrequency) and 8 Hz (rapid frequency). Pacing resulted in an increase in mitochondrial respiration, assessed by mitochondrial uptake of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) at 48 h. Rapid pacing at 8 Hz significantly increased cell injury and death as assessed by Trypan Blue uptake, creatine phosphokinase release, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) assay. Pacing at 5 Hz induced early, but weak, activation of Akt and protein kinase 38 (p38). Rapid pacing further augmented the early activation of Akt and p38, and induced extracellular signal-related kinase (Erk) and c-jun amino terminal kinase (JNK) activation. Incubation of ARVM with PI3K inhibitor LY294002 resulted in a twofold increase of TUNEL-positive cells under all pacing conditions examined. In conclusion, rapid pacing has immediate and detrimental consequences for cardiomyocyte survival, with pro-apoptotic pathways (e.g. JNK, p38) able to overwhelm antiapoptotic signalling (PI3K/Akt, Erk). The rapid pacing methodology described in this report will be particularly useful in determination of cell signalling pathways associated with tachycardia-induced cardiomyopathy.


Subject(s)
Electric Stimulation/methods , Heart Ventricles/cytology , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Ventricular Function , Animals , Apoptosis/physiology , Cell Survival , Cells, Cultured , Enzyme Activation , Male , Rats , Rats, Sprague-Dawley
17.
Mol Interv ; 5(3): 163-71, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15994456

ABSTRACT

Anthracyclines remain a mainstay of chemotherapy in spite of their well-recognized cardiotoxicity. Recent experience with trastuzumab (Herceptin) and anthracycline therapy has prompted a detailed analysis of the function of erbB2 in the heart. These studies demonstrate a cardioprotective effect of neuregulin, the endogenous ligand for the erbB4/erbB2 heterodimeric receptor complex. Although the mechanisms of cytoprotection remain incompletely understood, these studies have triggered the question of whether physiological manipulation of cardioprotective pathways that involve erbB can be used to improve outcome in patients treated with anthracyclines. The local activation of cardioprotection by cardiovascular exercise may be such a manipulation and warrants further investigation.


Subject(s)
Anthracyclines/toxicity , Antibiotics, Antineoplastic/toxicity , Heart/drug effects , Preventive Medicine , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Humans , Trastuzumab
19.
J Biol Chem ; 279(9): 8290-9, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14676206

ABSTRACT

Titin, the largest myofilament protein, serves as a template for sarcomere assembly and acts as a molecular spring to contribute to diastolic function. Titin is known to be extremely susceptible to calcium-dependent protease degradation in vitro. We hypothesized that titin degradation is an early event in doxorubicin-induced cardiac injury and that titin degradation occurs by activation of the calcium-dependent proteases, the calpains. Treatment of cultured adult rat cardiomyocytes with 1 or 3 micromol/liter doxorubicin for 24 h resulted in degradation of titin in myocyte lysates, which was confirmed by a reduction in immunostaining of an antibody to the spring-like (PEVK) domain of titin at the I-band of the sarcomere. The elastic domain of titin appears to be most susceptible to proteolysis because co-immunostaining with an antibody to titin at the M-line was preserved, suggesting targeted proteolysis of the spring-like domain of titin. Doxorubicin treatment for 1 h resulted in approximately 3-fold increase in calpain activity, which remained elevated at 48 h. Co-treatment with calpain inhibitors resulted in preservation of titin, reduction in myofibrillar disarray, and attenuation of cardiomyocyte necrosis but not apoptosis. Co-treatment with a caspase inhibitor did not prevent the degradation of titin, which precludes caspase-3 as an early mechanism of titin proteolysis. We conclude that calpain activation is an early event after doxorubicin treatment in cardiomyocytes and appears to target the degradation of titin. Proteolysis of the spring-like domain of titin may predispose cardiomyocytes to diastolic dysfunction, myofilament instability, and cell death by necrosis.


Subject(s)
Anthracyclines/pharmacology , Calpain/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Protein Kinases/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Calpain/antagonists & inhibitors , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cells, Cultured , Connectin , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Heart Ventricles , Male , Muscle Cells/metabolism , Necrosis , Rats , Rats, Sprague-Dawley
20.
Am J Physiol Cell Physiol ; 286(2): C222-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14522821

ABSTRACT

The purpose of this study is to test the hypothesis that mechanical and electrical activity in adult rat ventricular myocytes (ARVM) alters responses to proapoptotic and prosurvival ligands. The effects of electrical stimulation on myocyte survival, stress signaling, response to beta-adrenergic receptor (beta-AR)-stimulated apoptosis, and neuregulin-1beta (NRG) were examined. Electrical stimulation (6.6 V/cm; 0, 2, and 5 Hz; 2-ms duration; alternating polarity) of ARVM resulted in more than 70% capture. Although ARVM paced for 48 h showed higher mitochondrial uptake of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (P < 0.05, 0 vs. 2 and 5 Hz), electrical stimulation had little effect on cell survival assessed by trypan blue uptake, CPK release, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining. Electrical stimulation for 24 h did not induce stress response (heat shock protein 70, 90) nor stress kinase (Erk, JNK, p38) activation. NRG stimulation of Erk and Akt was similar between paced and quiescent cells. Pacing sensitized myocytes to beta-AR-stimulated JNK phosphorylation and cell death with 0.1 microM norepinephrine (NE) in paced myocytes causing equivalent cytotoxicity to 10 microM NE in quiescent cells. NRG suppressed beta-AR-induced apoptosis through a phosphatidylinositol-3-kinase-dependent pathway in both paced and quiescent cells, although it is overwhelmed by high-NE concentration in paced cells. Thus myocyte contractility modulates both NE cytotoxicity as well as the cytoprotective effect of NRG. These results demonstrate the feasibility and importance of using electrically paced cardiomyocytes in primary culture when examining the signaling pathways of cell survival.


Subject(s)
Apoptosis/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Neuregulin-1/pharmacology , Norepinephrine/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Electric Stimulation , Electrophysiologic Techniques, Cardiac , Enzyme Activation/physiology , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heart Ventricles , JNK Mitogen-Activated Protein Kinases , Mitochondria, Heart/physiology , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Isoforms/pharmacology , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL