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










Publication year range
1.
Circ Res ; 127(3): e94-e106, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32290757

ABSTRACT

RATIONALE: The heartbeat is organized by the cardiac conduction system (CCS), a specialized network of cardiomyocytes. Patterning of the CCS into atrial node versus ventricular conduction system (VCS) components with distinct physiology is essential for the normal heartbeat. Distinct node versus VCS physiology has been recognized for more than a century, but the molecular basis of this regional patterning is not well understood. OBJECTIVE: To study the genetic and genomic mechanisms underlying node versus VCS distinction and investigate rhythm consequences of failed VCS patterning. METHODS AND RESULTS: Using mouse genetics, we found that the balance between T-box transcriptional activator, Tbx5, and T-box transcriptional repressor, Tbx3, determined the molecular and functional output of VCS myocytes. Adult VCS-specific removal of Tbx5 or overexpression of Tbx3 re-patterned the fast VCS into slow, nodal-like cells based on molecular and functional criteria. In these cases, gene expression profiling showed diminished expression of genes required for VCS-specific fast conduction but maintenance of expression of genes required for nodal slow conduction physiology. Action potentials of Tbx5-deficient VCS myocytes adopted nodal-specific characteristics, including increased action potential duration and cellular automaticity. Removal of Tbx5 in vivo precipitated inappropriate depolarizations in the atrioventricular (His)-bundle associated with lethal ventricular arrhythmias. TBX5 bound and directly activated cis-regulatory elements at fast conduction channel genes required for fast physiological characteristics of the VCS action potential, defining the identity of the adult VCS. CONCLUSIONS: The CCS is patterned entirely as a slow, nodal ground state, with a T-box dependent, physiologically dominant, fast conduction network driven specifically in the VCS. Disruption of the fast VCS gene regulatory network allowed nodal physiology to emerge, providing a plausible molecular mechanism for some lethal ventricular arrhythmias.


Subject(s)
Arrhythmias, Cardiac/metabolism , Atrioventricular Node/metabolism , Heart Ventricles/metabolism , T-Box Domain Proteins/metabolism , Transcription, Genetic , Action Potentials , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Atrioventricular Node/physiopathology , Body Patterning , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Heart Rate , Heart Ventricles/physiopathology , Humans , Male , Mice, Knockout , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , Time Factors
2.
Cell Rep ; 30(9): 3105-3116.e4, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130910

ABSTRACT

The mammalian heart is incapable of regenerating a sufficient number of cardiomyocytes to ameliorate the loss of contractile muscle after acute myocardial injury. Several reports have demonstrated that mononucleated cardiomyocytes are more responsive than are binucleated cardiomyocytes to pro-proliferative stimuli. We have developed a strategy to isolate and characterize highly enriched populations of mononucleated and binucleated cardiomyocytes at various times of development. Our results suggest that an E2f/Rb transcriptional network is central to the divergence of these two populations and that remnants of the differences acquired during the neonatal period remain in adult cardiomyocytes. Moreover, inducing binucleation by genetically blocking the ability of cardiomyocytes to complete cytokinesis leads to a reduction in E2f target gene expression, directly linking the E2f pathway with nucleation. These data identify key molecular differences between mononucleated and binucleated mammalian cardiomyocytes that can be used to leverage cardiomyocyte proliferation for promoting injury repair in the heart.


Subject(s)
Cell Nucleus/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Base Sequence , Cell Nucleus/ultrastructure , Cell Proliferation , Cell Separation , Down-Regulation/genetics , E2F Transcription Factors/metabolism , Flow Cytometry , G1 Phase , Mice, Knockout , Myocytes, Cardiac/ultrastructure , Proto-Oncogene Proteins/metabolism , Regeneration , Retinoblastoma Protein/metabolism , S Phase
3.
Circ Res ; 126(12): 1685-1702, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32212902

ABSTRACT

RATIONALE: The heart undergoes dramatic developmental changes during the prenatal to postnatal transition, including maturation of cardiac myocyte energy metabolic and contractile machinery. Delineation of the mechanisms involved in cardiac postnatal development could provide new insight into the fetal shifts that occur in the diseased heart and unveil strategies for driving maturation of stem cell-derived cardiac myocytes. OBJECTIVE: To delineate transcriptional drivers of cardiac maturation. METHODS AND RESULTS: We hypothesized that ERR (estrogen-related receptor) α and γ, known transcriptional regulators of postnatal mitochondrial biogenesis and function, serve a role in the broader cardiac maturation program. We devised a strategy to knockdown the expression of ERRα and γ in heart after birth (pn-csERRα/γ [postnatal cardiac-specific ERRα/γ]) in mice. With high levels of knockdown, pn-csERRα/γ knockdown mice exhibited cardiomyopathy with an arrest in mitochondrial maturation. RNA sequence analysis of pn-csERRα/γ knockdown hearts at 5 weeks of age combined with chromatin immunoprecipitation with deep sequencing and functional characterization conducted in human induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CM) demonstrated that ERRγ activates transcription of genes involved in virtually all aspects of postnatal developmental maturation, including mitochondrial energy transduction, contractile function, and ion transport. In addition, ERRγ was found to suppress genes involved in fibroblast activation in hearts of pn-csERRα/γ knockdown mice. Disruption of Esrra and Esrrg in mice during fetal development resulted in perinatal lethality associated with structural and genomic evidence of an arrest in cardiac maturation, including persistent expression of early developmental and noncardiac lineage gene markers including cardiac fibroblast signatures. Lastly, targeted deletion of ESRRA and ESRRG in hiPSC-CM derepressed expression of early (transcription factor 21 or TCF21) and mature (periostin, collagen type III) fibroblast gene signatures. CONCLUSIONS: ERRα and γ are critical regulators of cardiac myocyte maturation, serving as transcriptional activators of adult cardiac metabolic and structural genes, an.d suppressors of noncardiac lineages including fibroblast determination.


Subject(s)
Heart/embryology , Myocytes, Cardiac/metabolism , Receptors, Estrogen/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Gene Expression Regulation, Developmental , Heart/growth & development , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Mitochondria, Heart/metabolism , Myocytes, Cardiac/cytology , Receptors, Estrogen/genetics , Signal Transduction , ERRalpha Estrogen-Related Receptor
4.
Pol Merkur Lekarski ; 48(288): 399-405, 2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33387426

ABSTRACT

The etiology of preterm premature rupture of membranes (PPROM), which is responsible for approximately 30% cases of preterm birth (PTB) is not yet fully understood. AIM: The aim of the study was to create a mathematical model for prognostication of PPROM based on the anamnesis, clinical data, laboratory findings and genetics predictors. MATERIALS AND METHODS: The study involved 80 women with PPROM (between 26 and 34 weeks of gestation) and 50 women having term birth (>37 weeks of gestation) of Zaporizhzhia region of Ukraine. Anamnesis, clinical, laboratory data and single nucleotide polymorphism sequencing of interleukin1 ß (IL1ß), tumor necrosis factor α(TNFα), interleukin4 (IL4), interleukin10 (IL10) and Relaxin 2 (RLN2) genes has been analyzed. Receiver operating characteristic analysis and multivariate logistic regression were used to PPROM predictors identification. RESULTS: We have identified prognostic anamnestic (history of preterm birth), clinical (cervical insuffiency, compromised uteroplacental and fetal circulation), microbiological (vaginal dysbiosis) and hematological criteria for intra-amniotic contamination and further development of PPROM and PTB: WBC>12.3×109/L, GRAN>76%, LYM<19%, neutrophil lymphocyte ratio>3.87, Kalph-Kaliph leukocyte index of intoxication (LII) >3.4, Ostrovsky LII >2.8. Also we have found that GG genotype of IL10 gene polymorphism (rs1800872) leads to a 12.5-fold and CT genotype of RLN2 gene polymorphism (rs4742076) leads to a 17.0-fold increase in risk for PPROM. CONCLUSIONS: The prognostic model that we have suggested is an adequate and convenient instrument for practical medical use, which allows for assessment of PPROM probability with a 85% sensitivity and a 72% specificity.


Subject(s)
Fetal Membranes, Premature Rupture , Premature Birth , Female , Fetal Membranes, Premature Rupture/epidemiology , Fetal Membranes, Premature Rupture/genetics , Genotype , Humans , Infant, Newborn , Polymorphism, Single Nucleotide , Pregnancy , Premature Birth/epidemiology , Ukraine
6.
Physiol Rep ; 3(9)2015 Sep.
Article in English | MEDLINE | ID: mdl-26400986

ABSTRACT

Cardiac melanocyte-like cells (CMLCs) contribute to atrial arrhythmias when missing the melanin synthesis enzyme dopachrome tautomerase (Dct). While scavenging reactive oxygen species (ROS) in Dct-null mice partially suppressed atrial arrhythmias, it remains unclear if CMLCs influence atrial ROS and structure or if the electrical response of CMLCs to ROS differs from that of atrial myocytes. This study is designed to determine if CMLCs contribute to overall atrial oxidative stress or structural remodeling, and if ROS affects the electrophysiology of CMLCs differently than atrial myocytes. Immunohistochemical analysis showed higher expression of the oxidative marker 8-hydroxy-2'-deoxyguanosine in Dct-null atria versus Dct-heterozygous (Dct-het) atria. Exposing isolated CMLCs from Dct-het and Dct-null mice to hydrogen peroxide increased superoxide anion more in Dct-null CMLCs. Trichrome staining showed increased fibrosis in Dct-null atria, and treating Dct-null mice with the ROS scavenger Tempol reduced atrial fibrosis. Action potential recordings from atrial myocytes and isolated Dct-het and Dct-null CMLCs in response to hydrogen peroxide showed that the EC50 for action potential duration (APD) prolongation of Dct-null CMLCs was 8.2 ± 1.7 µmol/L versus 16.8 ± 2.0 µmol/L for Dct-het CMLCs, 19.9 ± 2.1 µmol/L for Dct-null atrial myocytes, and 20.5 ± 1.9 µmol/L for Dct-het atrial myocytes. However, APD90 was longer in CMLCs versus atrial myocytes in response to hydrogen peroxide. Hydrogen peroxide also induced more afterdepolarizations in CMLCs compared to atrial myocytes. These studies suggest that Dct within CMLCs contributes to atrial ROS balance and remodeling. ROS prolongs APD to a greater extent and induces afterdepolarizations more frequently in CMLCs than in atrial myocytes.

7.
Sci Transl Med ; 7(279): 279ra38, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25787764

ABSTRACT

In contrast to lower vertebrates, the mammalian heart has limited capacity to regenerate after injury in part due to ineffective reactivation of cardiomyocyte proliferation. We show that the microRNA cluster miR302-367 is important for cardiomyocyte proliferation during development and is sufficient to induce cardiomyocyte proliferation in the adult and promote cardiac regeneration. In mice, loss of miR302-367 led to decreased cardiomyocyte proliferation during development. In contrast, increased miR302-367 expression led to a profound increase in cardiomyocyte proliferation, in part through repression of the Hippo signal transduction pathway. Postnatal reexpression of miR302-367 reactivated the cell cycle in cardiomyocytes, resulting in reduced scar formation after experimental myocardial infarction. However, long-term expression of miR302-367 induced cardiomyocyte dedifferentiation and dysfunction, suggesting that persistent reactivation of the cell cycle in postnatal cardiomyocytes is not desirable. This limitation can be overcome by transient systemic application of miR302-367 mimics, leading to increased cardiomyocyte proliferation and mass, decreased fibrosis, and improved function after injury. Our data demonstrate the ability of microRNA-based therapeutic approaches to promote mammalian cardiac repair and regeneration through the transient activation of cardiomyocyte proliferation.


Subject(s)
MicroRNAs/metabolism , Myocytes, Cardiac/cytology , Protein Serine-Threonine Kinases/metabolism , Alleles , Animals , Cell Cycle , Cell Proliferation , Cell Separation , Cells, Cultured , Flow Cytometry , HEK293 Cells , Heart/physiology , Hippo Signaling Pathway , Humans , Lentivirus/genetics , Lipids/chemistry , Male , Mice , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Regeneration , Signal Transduction
8.
Mol Cell Biol ; 35(7): 1281-98, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25624346

ABSTRACT

Almost all cellular functions are powered by a continuous energy supply derived from cellular metabolism. However, it is little understood how cellular energy production is coordinated with diverse energy-consuming cellular functions. Here, using the cardiac muscle system, we demonstrate that nuclear receptors estrogen-related receptor α (ERRα) and ERRγ are essential transcriptional coordinators of cardiac energy production and consumption. On the one hand, ERRα and ERRγ together are vital for intact cardiomyocyte metabolism by directly controlling expression of genes important for mitochondrial functions and dynamics. On the other hand, ERRα and ERRγ influence major cardiomyocyte energy consumption functions through direct transcriptional regulation of key contraction, calcium homeostasis, and conduction genes. Mice lacking both ERRα and cardiac ERRγ develop severe bradycardia, lethal cardiomyopathy, and heart failure featuring metabolic, contractile, and conduction dysfunctions. These results illustrate that the ERR transcriptional pathway is essential to couple cellular energy metabolism with energy consumption processes in order to maintain normal cardiac function.


Subject(s)
Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Myocardium/metabolism , Myocardium/pathology , Receptors, Estrogen/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cells, Cultured , Female , Gene Deletion , Gene Expression Regulation , Heart/physiology , Heart/physiopathology , Ion Channels/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Receptors, Estrogen/genetics , ERRalpha Estrogen-Related Receptor
9.
Circulation ; 126(9): 1058-66, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22837163

ABSTRACT

BACKGROUND: Notch signaling has previously been shown to play an essential role in regulating cell fate decisions and differentiation during cardiogenesis in many systems including Drosophila, Xenopus, and mammals. We hypothesized that Notch may also be involved in directing the progressive lineage restriction of cardiomyocytes into specialized conduction cells. METHODS AND RESULTS: In hearts where Notch signaling is activated within the myocardium from early development onward, Notch promotes a conduction-like phenotype based on ectopic expression of conduction system-specific genes and cell autonomous changes in electrophysiology. With the use of an in vitro assay to activate Notch in newborn cardiomyocytes, we observed global changes in the transcriptome, and in action potential characteristics, consistent with reprogramming to a conduction-like phenotype. CONCLUSIONS: Notch can instruct the differentiation of chamber cardiac progenitors into specialized conduction-like cells. Plasticity remains in late-stage cardiomyocytes, which has potential implications for engineering of specialized cardiovascular tissues.


Subject(s)
Atrioventricular Node/cytology , Gene Expression Regulation, Developmental , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Receptor, Notch1/physiology , Action Potentials , Adenoviridae/genetics , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage , Contactin 2/biosynthesis , Contactin 2/genetics , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Myocytes, Cardiac/ultrastructure , NAV1.5 Voltage-Gated Sodium Channel , Neuronal Plasticity , Patch-Clamp Techniques , Phenotype , Purkinje Fibers/cytology , Receptor, Notch1/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction/physiology , Sodium Channels/biosynthesis , Sodium Channels/genetics , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Transcription Factor HES-1 , Transcription Factors/biosynthesis , Transcription Factors/genetics
10.
J Mol Cell Cardiol ; 50(3): 451-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21147122

ABSTRACT

At each heartbeat, cardiac myocytes are activated by a cytoplasmic Ca(2+) transient in great part due to Ca(2+) release from the sarcoplasmic reticulum via ryanodine receptors (RyRs) clustered within calcium release units (peripheral couplings/dyads). A Ca(2+) transient also occurs in the nucleoplasm, following the cytoplasmic transient with some delay. Under conditions where the InsP3 production is stimulated, these Ca(2+) transients are regulated actively, presumably by an additional release of Ca(2+) via InsP3 receptors (InsP3Rs). This raises the question whether InsP3Rs are appropriately located for this effect and whether sources of InsP3 and Ca(2+) are available for their activation. We have defined the structural basis for InsP3R activity at the nucleus, using immunolabeling for confocal microscopy and freeze-drying/shadowing, T tubule "staining" and thin sectioning for electron microscopy. By these means we establish the presence of InsP3R at the outer nuclear envelope and show a close spatial relationship between the nuclear envelope, T tubules (a likely source of InsP3) and dyads (the known source of Ca(2+)). The frequency, distribution and distance from the nucleus of T tubules and dyads appropriately establish local perinuclear Ca(2+) microdomains in cardiac myocytes.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Cardiac/metabolism , Nuclear Envelope/metabolism , Animals , Calcium Signaling/physiology , Cell Nucleus/metabolism , Mice , Microscopy, Confocal/methods , Microscopy, Electron/methods , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
11.
Circ Cardiovasc Imaging ; 4(1): 33-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21059858

ABSTRACT

BACKGROUND: Pluripotent stem cells represent one promising source for cellular cardiomyoplasty. In this study, we used cardiac magnetic resonance to examine the ability of highly enriched cardiomyocytes (CMs) derived from murine embryonic stem cells (ESC) to form grafts and improve contractile function of infarcted rat hearts. METHODS AND RESULTS: Highly enriched ESC-CMs were obtained by inducing cardiac differentiation of ESCs stably expressing a cardiac-restricted puromycin resistance gene. At the time of transplantation, enriched ESC-CMs expressed cardiac-specific markers and markers of developing CMs, but only 6% of them were proliferating. A growth factor-containing vehicle solution or ESC-CMs (5 to 10 million) suspended in the same solution was injected into athymic rat hearts 1 week after myocardial infarction. Initial infarct size was measured by cardiac magnetic resonance 1 day after myocardial infarction. Compared with vehicle treatment, treatment with ESC-CMs improved global systolic function 1 and 2 months after injection and significantly increased contractile function in initially infarcted areas and border zones. Immunohistochemistry confirmed successful engraftment and the persistence of α-actinin-positive ESC-CMs that also expressed α-smooth muscle actin. Connexin-43-positive sites were observed between grafted ESC-CMs but only rarely between grafted and host CMs. No teratomas were observed in any of the animals. CONCLUSIONS: Highly enriched and early-stage ESC-CMs were safe, formed stable grafts, and mediated a long-term recovery of global and regional myocardial contractile function after infarction.


Subject(s)
Embryonic Stem Cells/transplantation , Myocardial Contraction , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Stem Cell Transplantation/methods , Ventricular Dysfunction, Left/prevention & control , Analysis of Variance , Animals , Cells, Cultured , Disease Models, Animal , Female , Heart Ventricles/pathology , Magnetic Resonance Imaging/methods , Mice , Myocardial Infarction/complications , Rats , Time , Ventricular Dysfunction, Left/etiology
12.
J Biol Chem ; 285(34): 26494-505, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20566649

ABSTRACT

Mitochondrial membrane potential loss has severe bioenergetic consequences and contributes to many human diseases including myocardial infarction, stroke, cancer, and neurodegeneration. However, despite its prominence and importance in cellular energy production, the basic mechanism whereby the mitochondrial membrane potential is established remains unclear. Our studies elucidate that complex II-driven electron flow is the primary means by which the mitochondrial membrane is polarized under hypoxic conditions and that lack of the complex II substrate succinate resulted in reversible membrane potential loss that could be restored rapidly by succinate supplementation. Inhibition of mitochondrial complex I and F(0)F(1)-ATP synthase induced mitochondrial depolarization that was independent of the mitochondrial permeability transition pore, Bcl-2 (B-cell lymphoma 2) family proteins, or high amplitude swelling and could not be reversed by succinate. Importantly, succinate metabolism under hypoxic conditions restores membrane potential and ATP levels. Furthermore, a reliance on complex II-mediated electron flow allows cells from mitochondrial disease patients devoid of a functional complex I to maintain a mitochondrial membrane potential that conveys both a mitochondrial structure and the ability to sequester agonist-induced calcium similar to that of normal cells. This finding is important as it sets the stage for complex II functional preservation as an attractive therapy to maintain mitochondrial function during hypoxia.


Subject(s)
Calcium/physiology , Electron Transport Complex II/physiology , Hypoxia , Membrane Potential, Mitochondrial , Proto-Oncogene Proteins c-bcl-2/physiology , Adenosine Triphosphate , Animals , Humans , Mice , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Proton-Translocating ATPases/physiology , Rats , Succinic Acid/pharmacology
13.
J Clin Invest ; 119(11): 3420-36, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19855129

ABSTRACT

Atrial fibrillation is the most common clinical cardiac arrhythmia. It is often initiated by ectopic beats arising from the pulmonary veins and atrium, but the source and mechanism of these beats remains unclear. The melanin synthesis enzyme dopachrome tautomerase (DCT) is involved in intracellular calcium and reactive species regulation in melanocytes. Given that dysregulation of intracellular calcium and reactive species has been described in patients with atrial fibrillation, we investigated the role of DCT in this process. Here, we characterize a unique DCT-expressing cell population within murine and human hearts that populated the pulmonary veins, atria, and atrioventricular canal. Expression profiling demonstrated that this population expressed adrenergic and muscarinic receptors and displayed transcriptional profiles distinct from dermal melanocytes. Adult mice lacking DCT displayed normal cardiac development but an increased susceptibility to atrial arrhythmias. Cultured primary cardiac melanocyte-like cells were excitable, and those lacking DCT displayed prolonged repolarization with early afterdepolarizations. Furthermore, mice with mutations in the tyrosine kinase receptor Kit lacked cardiac melanocyte-like cells and did not develop atrial arrhythmias in the absence of DCT. These data suggest that dysfunction of melanocyte-like cells in the atrium and pulmonary veins may contribute to atrial arrhythmias.


Subject(s)
Arrhythmias, Cardiac/enzymology , Intramolecular Oxidoreductases/metabolism , Melanocytes/enzymology , Myocardium/cytology , Pulmonary Veins/cytology , Animals , Arrhythmias, Cardiac/genetics , Electrophysiological Phenomena , Free Radical Scavengers/metabolism , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Humans , Intramolecular Oxidoreductases/genetics , Melanocytes/ultrastructure , Mice , Mice, Knockout , Myocardium/pathology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Adrenergic/metabolism , Receptors, Muscarinic/metabolism
14.
Proc Natl Acad Sci U S A ; 106(18): 7548-52, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19376970

ABSTRACT

Nonsteroidal anti-inflammatory drugs selective for inhibition of COX-2 increase heart failure and elevate blood pressure. The COX-2 gene was floxed and crossed into merCremer mice under the alpha-myosin heavy-chain promoter. Tamoxifen induced selective deletion of COX-2 in cardiomyocytes depressed cardiac output, and resulted in weight loss, diminished exercise tolerance, and enhanced susceptibility to induced arrhythmogenesis. The cardiac dysfunction subsequent to pressure overload recovered progressively in the knockouts coincident with increasing cardiomyocyte hypertrophy and interstitial and perivascular fibrosis. Inhibition of COX-2 in cardiomyocytes may contribute to heart failure in patients receiving nonsteroidal anti-inflammatory drugs specific for inhibition of COX-2.


Subject(s)
Cyclooxygenase 2/physiology , Heart Rate , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cyclooxygenase 2/genetics , Gene Deletion , Heart Rate/genetics , Hypertrophy/chemically induced , Hypertrophy/enzymology , Mice , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology
15.
J Mol Cell Cardiol ; 45(6): 715-23, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18926829

ABSTRACT

Atrial fibrosis influences the development of atrial fibrillation (AF), particularly in the setting of structural heart disease where angiotensin-inhibition is partially effective for reducing atrial fibrosis and AF. Histone-deacetylase inhibition reduces cardiac hypertrophy and fibrosis, so we sought to determine if the HDAC inhibitor trichostatin A (TSA) could reduce atrial fibrosis and arrhythmias. Mice over-expressing homeodomain-only protein (HopX(Tg)), which recruits HDAC activity to induce cardiac hypertrophy were investigated in 4 groups (aged 14-18 weeks): wild-type (WT), HopX(Tg), HopX(Tg) mice treated with TSA for 2 weeks (TSA-HopX) and wild-type mice treated with TSA for 2 weeks (TSA-WT). These groups were characterized using invasive electrophysiology, atrial fibrosis measurements, atrial connexin immunocytochemistry and myocardial angiotensin II measurements. Invasive electrophysiologic stimulation, using the same attempts in each group, induced more atrial arrhythmias in HopX(Tg) mice (48 episodes in 13 of 15 HopX(Tg) mice versus 5 episodes in 2 of 15 TSA-HopX mice, P<0.001; versus 9 episodes in 2 of 15 WT mice, P<0.001; versus no episodes in any TSA-WT mice, P<0.001). TSA reduced atrial arrhythmia duration in HopX(Tg) mice (1307+/-289 ms versus 148+/-110 ms, P<0.01) and atrial fibrosis (8.1+/-1.5% versus 3.9+/-0.4%, P<0.001). Atrial connexin40 was lower in HopX(Tg) compared to WT mice, and TSA normalized the expression and size distribution of connexin40 gap junctions. Myocardial angiotensin II levels were similar between WT and HopX(Tg) mice (76.3+/-26.0 versus 69.7+/-16.6 pg/mg protein, P=NS). Therefore, it appears HDAC-inhibition reverses atrial fibrosis, connexin40 remodeling and atrial arrhythmia vulnerability independent of angiotensin II in cardiac hypertrophy.


Subject(s)
Arrhythmias, Cardiac/enzymology , Enzyme Inhibitors/adverse effects , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Homeodomain Proteins/biosynthesis , Hydroxamic Acids/adverse effects , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Cardiomegaly/congenital , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Connexins/genetics , Connexins/metabolism , Enzyme Inhibitors/pharmacology , Fibrosis , Homeodomain Proteins/genetics , Hydroxamic Acids/pharmacology , Mice , Mice, Transgenic , Time Factors , Gap Junction alpha-5 Protein
16.
J Mol Cell Cardiol ; 44(3): 597-606, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18201716

ABSTRACT

Cardiac-specific deletion of the murine gene (Cdh2) encoding the cell adhesion molecule, N-cadherin, results in disassembly of the intercalated disc (ICD) structure and sudden arrhythmic death. Connexin 43 (Cx43)-containing gap junctions are significantly reduced in the heart after depleting N-cadherin, therefore we hypothesized that animals expressing half the normal levels of N-cadherin would exhibit an intermediate phenotype. We examined the effect of N-cadherin haploinsufficiency on Cx43 expression and susceptibility to induced arrhythmias in mice either wild-type or heterozygous for the Cx43 (Gja1)-null allele. An increase in hypophosphorylated Cx43 accompanied by a modest decrease in total Cx43 protein levels was observed in the N-cadherin heterozygous mice. Consistent with these findings N-cadherin heterozygotes exhibited increased susceptibility to ventricular arrhythmias compared to wild-type mice. Quantitative immunofluorescence microscopy revealed a reduction in size of large Cx43-containing plaques in the N-cadherin heterozygous animals compared to wild-type. Gap junctions were further decreased in number and size in the N-cad/Cx43 compound heterozygous mice with increased arrhythmic susceptibility compared to the single mutants. The scaffold protein, ZO-1, was reduced at the ICD in N-cadherin heterozygous cardiomyocytes providing a possible explanation for the reduction in Cx43 plaque size. These data provide further support for the intimate relationship between N-cadherin and Cx43 in the heart, and suggest that germline mutations in the human N-cadherin (Cdh2) gene may predispose patients to increased risk of cardiac arrhythmias.


Subject(s)
Arrhythmias, Cardiac/metabolism , Cadherins/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Blotting, Western , Cadherins/genetics , Cell Communication/genetics , Cells, Cultured , Connexin 43/genetics , Electrophysiology , Fluorescent Antibody Technique , Heterozygote , Membrane Proteins/metabolism , Mice , Muscle Cells/metabolism , Mutation , Myocardium/metabolism , Myocardium/pathology , Phosphoproteins/metabolism , Zonula Occludens-1 Protein , beta Catenin/metabolism
17.
Nat Cell Biol ; 7(10): 1021-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16179951

ABSTRACT

Members of the Bcl-2 protein family modulate outer mitochondrial membrane permeability to control apoptosis. However, these proteins also localize to the endoplasmic reticulum (ER), the functional significance of which is controversial. Here we provide evidence that anti-apoptotic Bcl-2 proteins regulate the inositol 1,4,5-trisphosphate receptor (InsP(3)R) ER Ca(2+) release channel resulting in increased cellular apoptotic resistance and enhanced mitochondrial bioenergetics. Anti-apoptotic Bcl-X(L) interacts with the carboxyl terminus of the InsP(3)R and sensitizes single InsP(3)R channels in ER membranes to low [InsP(3)], enhancing Ca(2+) and InsP(3)-dependent regulation of channel activity in vitro and in vivo, reducing ER Ca(2+) content and stimulating mitochondrial energetics. The pro-apoptotic proteins Bax and tBid antagonize this effect by blocking the biochemical interaction of Bcl-X(L) with the InsP(3)R. These data support a novel model in which Bcl-X(L) is a direct effector of the InsP(3)R, increasing its sensitivity to InsP(3) and enabling ER Ca(2+) release to be more sensitively coupled to extracellular signals. As a consequence, cells are protected against apoptosis by a more sensitive and dynamic coupling of ER to mitochondria through Ca(2+)-dependent signal transduction that enhances cellular bioenergetics and preserves survival.


Subject(s)
Apoptosis/physiology , Calcium Channels/metabolism , Endoplasmic Reticulum/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Calcium Channels/drug effects , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum/drug effects , Humans , Inositol 1,4,5-Trisphosphate Receptors , Receptors, Cytoplasmic and Nuclear/drug effects , Time Factors , bcl-X Protein/pharmacology
18.
J Gen Physiol ; 122(5): 569-81, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14581583

ABSTRACT

The inositol 1,4,5-trisphosphate (InsP3) receptor (InsP3R), a Ca2+-release channel localized to the endoplasmic reticulum, plays a critical role in generating complex cytoplasmic Ca2+ signals in many cell types. Three InsP3R isoforms are expressed in different subcellular locations, at variable relative levels with heteromultimer formation in different cell types. A proposed reason for this diversity of InsP3R expression is that the isoforms are differentially inhibited by high cytoplasmic free Ca2+ concentrations ([Ca2+]i), possibly due to their different interactions with calmodulin. Here, we have investigated the possible roles of calmodulin and bath [Ca2+] in mediating high [Ca2+]i inhibition of InsP3R gating by studying single endogenous type 1 InsP3R channels through patch clamp electrophysiology of the outer membrane of isolated Xenopus oocyte nuclei. Neither high concentrations of a calmodulin antagonist nor overexpression of a dominant-negative Ca2+-insensitive mutant calmodulin affected inhibition of gating by high [Ca2+]i. However, a novel, calmodulin-independent regulation of [Ca2+]i inhibition of gating was revealed: whereas channels recorded from nuclei kept in the regular bathing solution with [Ca2+] approximately 400 nM were inhibited by 290 muM [Ca2+]i, exposure of the isolated nuclei to a bath solution with ultra-low [Ca2+] (<5 nM, for approximately 300 s) before the patch-clamp experiments reversibly relieved Ca2+ inhibition, with channel activities observed in [Ca2+]i up to 1.5 mM. Although InsP3 activates gating by relieving high [Ca2+]i inhibition, it was nevertheless still required to activate channels that lacked high [Ca2+]i inhibition. Our observations suggest that high [Ca2+]i inhibition of InsP3R channel gating is not regulated by calmodulin, whereas it can be disrupted by environmental conditions experienced by the channel, raising the possibility that presence or absence of high [Ca2+]i inhibition may not be an immutable property of different InsP3R isoforms. Furthermore, these observations support an allosteric model in which Ca2+ inhibition of the InsP3R is mediated by two Ca2+ binding sites, only one of which is sensitive to InsP3.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Calcium/pharmacology , Dose-Response Relationship, Drug , Female , Inositol 1,4,5-Trisphosphate Receptors , Rats , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...