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










Publication year range
1.
J Nanobiotechnology ; 22(1): 223, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702815

ABSTRACT

Cardiac muscle targeting is a notoriously difficult task. Although various nanoparticle (NP) and adeno-associated viral (AAV) strategies with heart tissue tropism have been developed, their performance remains suboptimal. Significant off-target accumulation of i.v.-delivered pharmacotherapies has thwarted development of disease-modifying cardiac treatments, such as gene transfer and gene editing, that may address both rare and highly prevalent cardiomyopathies and their complications. Here, we present an intriguing discovery: cargo-less, safe poly (lactic-co-glycolic acid) particles that drastically improve heart delivery of AAVs and NPs. Our lead formulation is referred to as ePL (enhancer polymer). We show that ePL increases selectivity of AAVs and virus-like NPs (VLNPs) to the heart and de-targets them from the liver. Serotypes known to have high (AAVrh.74) and low (AAV1) heart tissue tropisms were tested with and without ePL. We demonstrate up to an order of magnitude increase in heart-to-liver accumulation ratios in ePL-injected mice. We also show that ePL exhibits AAV/NP-independent mechanisms of action, increasing glucose uptake in the heart, increasing cardiac protein glycosylation, reducing AAV neutralizing antibodies, and delaying blood clearance of AAV/NPs. Current approaches utilizing AAVs or NPs are fraught with challenges related to the low transduction of cardiomyocytes and life-threatening immune responses; our study introduces an exciting possibility to direct these modalities to the heart at reduced i.v. doses and, thus, has an unprecedented impact on drug delivery and gene therapy. Based on our current data, the ePL system is potentially compatible with any therapeutic modality, opening a possibility of cardiac targeting with numerous pharmacological approaches.


Subject(s)
Dependovirus , Genetic Vectors , Myocardium , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Dependovirus/genetics , Animals , Nanoparticles/chemistry , Mice , Myocardium/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Humans , Mice, Inbred C57BL , Heart , Genetic Therapy/methods , Gene Transfer Techniques , Liver/metabolism , Viral Tropism , HEK293 Cells
2.
Toxicol Rep ; 9: 1713-1724, 2022.
Article in English | MEDLINE | ID: mdl-36561952

ABSTRACT

Since its discovery in the 1960 s, doxorubicin (DOX) has constantly elicited the broadest spectrum of cancerocidal activity against human cancers. However, cardiotoxicity caused by DOX directly as well as its metabolites is a great source of concern over the continuous use of DOX in chemotherapy. While the exact mechanism of DOX-induced cardiotoxicity is yet to be completely understood, recent studies indicate oxidative stress, inflammation, and several forms of cell death as key pathogenic mechanisms that underpin the etiology of doxorubicin-induced cardiotoxicity (DIC). Notably, these key mechanistic events are believed to be negatively regulated by 3,4-dihydroxybenzoic acid or protocatechuic acid (PCA)-a plant-based phytochemical with proven anti-oxidant, anti-inflammatory, and anti-apoptotic properties. Here, we review the experimental findings detailing the potential ameliorative effects of PCA under exposure to DOX. We also discuss molecular insights into the pathophysiology of DIC, highlighting the potential intervention points where the use of PCA as a veritable chemoprotective agent may ameliorate DOX-induced cardiotoxicities as well as toxicities due to other anticancer drugs like cisplatin. While we acknowledge that controlled oral administration of PCA during chemotherapy may be insufficient to eliminate all toxicities due to DOX treatment, we propose that the ability of PCA to block oxidative stress, attenuate inflammation, and abrogate several forms of cardiomyocyte cell death underlines its great promise in the amelioration of DIC.

3.
Biomolecules ; 12(10)2022 10 17.
Article in English | MEDLINE | ID: mdl-36291708

ABSTRACT

Beta-hydroxybutyrate (ßOHB), along with acetoacetate and acetone, are liver-produced ketone bodies that are increased after fasting or prolonged exercise as an alternative fuel source to glucose. ßOHB, as the main circulating ketone body, is not only a G-protein coupled receptor ligand but also a histone deacetylases inhibitor, prompting the reexamination of its role in health and disease. In this study, we compared the effects of two commercial ßOHB formulations an enantiomer R ßOHB and a racemic mixture ±ßOHB on induced pluripotent stem cell cardiac myocytes (iPS-CMs) electrophysiology. Cardiac myocytes were cultured in R ßOHB or ±ßOHB for at least ten days after lactate selection. Flouvolt or Fluo-4 was used to assay iPS-CMs electrophysiology. We found that while both formulations increased the optical potential amplitude, R ßOHB prolonged the action potential duration but ±ßOHB shortened the action potential duration. Moreover, ±ßOHB increased the peak calcium transient but R ßOHB reduced the peak calcium transient. Co-culturing with glucose or fatty acids did not ameliorate the effects, suggesting that ßOHB was more than a fuel source. The effect of ßOHB on iPS-CMs electrophysiology is most likely stereoselective, and care must be taken to evaluate the role of exogenous ßOHB in health and disease.


Subject(s)
Acetoacetates , Myocytes, Cardiac , 3-Hydroxybutyric Acid/pharmacology , Calcium , Acetone , Ligands , Ketone Bodies , Glucose/pharmacology , Histone Deacetylases , Receptors, G-Protein-Coupled , Lactates , Electrophysiology
4.
Theranostics ; 11(10): 4743-4758, 2021.
Article in English | MEDLINE | ID: mdl-33754025

ABSTRACT

Aims: Emerging evidence is demonstrating that rapid regeneration of remnant liver elicited by associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) may be attenuated in fibrotic livers. However, the molecular mechanisms responsible for this process are largely unknown. It is widely acknowledged that the TGFß1 signaling axis plays a major role in liver fibrosis. Therefore, the aims of this study were to elucidate the underlying mechanism of liver regeneration during ALPPS with or without fibrosis, specifically focusing on TGFß1 signaling. Approach: ALPPS was performed in rat models with N-diethylnitrosamine-induced liver fibrosis and no fibrosis. Functional liver remnant regeneration and expression of TGFß1 were analyzed during the ALPPS procedures. Adeno-associated virus-shTGFß1 and the small molecule inhibitor LY2157299 (galunisertib) were used separately or in combination to inhibit TGFß1 signaling in fibrotic rats. Results: Liver regeneration following ALPPS was lower in fibrotic rats than non-fibrotic rats. TGFß1 was a key mediator of postoperative regeneration in fibrotic liver. Interestingly, AAV-shTGFß1 accelerated the regeneration of fibrotic functional liver remnant and improved fibrosis, while LY2157299 only enhanced liver regeneration. Moreover, combination treatment elicited a stronger effect. Conclusions: Inhibition of TGFß1 accelerated regeneration of fibrotic liver, ameliorated liver fibrosis, and improved liver function following ALPPS. Therefore, TGFß1 is a promising therapeutic target in ALPPS to improve fibrotic liver reserve function and prognosis.


Subject(s)
Hepatectomy/methods , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Liver Regeneration/physiology , Liver/physiology , Transforming Growth Factor beta1/metabolism , Animals , Carbon Tetrachloride/toxicity , Diethylnitrosamine/toxicity , Hepatic Stellate Cells/metabolism , Ligation , Liver/drug effects , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Regeneration/drug effects , Portal Vein/surgery , Primary Cell Culture , Pyrazoles/pharmacology , Quinolines/pharmacology , Rats , Signal Transduction , Transforming Growth Factor beta1/antagonists & inhibitors
5.
Animals (Basel) ; 10(9)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887495

ABSTRACT

Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.

6.
Circ Res ; 127(2): 284-297, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32345129

ABSTRACT

RATIONALE: ZO-1 (Zonula occludens-1), a plasma membrane-associated scaffolding protein regulates signal transduction, transcription, and cellular communication. Global deletion of ZO-1 in the mouse is lethal by embryonic day 11.5. The function of ZO-1 in cardiac myocytes (CM) is largely unknown. OBJECTIVE: To determine the function of CM ZO-1 in the intact heart, given its binding to other CM proteins that have been shown instrumental in normal cardiac conduction and function. METHODS AND RESULTS: We generated ZO-1 CM-specific knockout (KO) mice using α-Myosin Heavy Chain-nuclear Cre (ZO-1cKO) and investigated physiological and electrophysiological function by echocardiography, surface ECG and conscious telemetry, intracardiac electrograms and pacing, and optical mapping studies. ZO-1cKO mice were viable, had normal Mendelian ratios, and had a normal lifespan. Ventricular morphometry and function were not significantly different between the ZO-1cKO versus control (CTL) mice, basally in young or aged mice, or even when hearts were subjected to hemodynamic loading. Atrial mass was increased in ZO-1cKO. Electrophysiological and optical mapping studies indicated high-grade atrioventricular (A-V) block in ZO-1cKO comparing to CTL hearts. While ZO-1-associated proteins such as vinculin, connexin 43, N-cadherin, and α-catenin showed no significant change with the loss of ZO-1, Connexin-45 and Coxsackie-adenovirus (CAR) proteins were reduced in atria of ZO-1cKO. Further, with loss of ZO-1, ZO-2 protein was increased significantly in ventricular CM in a presumed compensatory manner but was still not detected in the AV nodal myocytes. Importantly, the expression of the sodium channel protein NaV1.5 was altered in AV nodal cells of the ZO-1cKO versus CTL. CONCLUSIONS: ZO-1 protein has a unique physiological role in cardiac nodal tissue. This is in alignment with its known interaction with CAR and Cx45, and a new function in regulating the expression of NaV1.5 in AV node. Uniquely, ZO-1 is dispensable for function of the working myocardium.


Subject(s)
Atrioventricular Block/metabolism , Atrioventricular Node/metabolism , Ventricular Function , Zonula Occludens-1 Protein/metabolism , Animals , Atrioventricular Block/physiopathology , Atrioventricular Node/physiology , Cadherins/genetics , Cadherins/metabolism , Connexins/genetics , Connexins/metabolism , Male , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Vinculin/genetics , Vinculin/metabolism , Zonula Occludens-1 Protein/genetics , alpha Catenin/genetics , alpha Catenin/metabolism
7.
Arch Biochem Biophys ; 662: 143-150, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30543786

ABSTRACT

ß-hydroxybutyrate is the primary ketone body produced by the body during ketosis and is used to meet its metabolic demands. The healthy adult heart derives most of its energy from fatty acid oxidation. However, in certain diseases, the heart alters its substrate preference and increases its ketone body metabolism. Little is known about the effects of ßOHB on ventricular myocyte excitation-contraction coupling. Therefore, we examined the effects of ketone body metabolism on single cell excitation-contraction coupling during normoxic and hypoxic conditions. Myocytes were isolated from adult rats, cultured for 18 h in RPMI 1640, RPMI 1640 no glucose, and M199, HEPES with/without various amount of ßOHB added. To simulate hypoxia, myocytes were incubated at 1%O2, 5% CO2 for 1 h followed by incubation at atmospheric oxygen (21%O2,5% CO2) for 30 min before recordings. Recordings were obtained using an IonOptix system at 36±1ᵒ C. Myocytes were paced at 0.5, 1, 2, 3, and 4 Hz. We found that exposure to ßOHB had no effect on excitation-contraction coupling. However, culturing cells with ßOHB results in a significant increase in both contraction and calcium in RPMI 1640 media. Dose response experiments demonstrated 0.5 mM ßOHB is enough to increase myocyte contraction in the absence of glucose. However, ßOHB has no measurable effects on myocytes cultured in a nutrient rich media, M199, HEPES. Therefore, ßOHB improves single cell excitation-contraction coupling, is protective against hypoxia, and may be a beneficial adaptation for the heart during periods of nutrient scarcity and or metabolic dysregulation.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Heart Ventricles/metabolism , Ketone Bodies/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcium/metabolism , Glucose/metabolism , In Vitro Techniques , Rats , Rats, Sprague-Dawley
8.
J Mol Cell Cardiol ; 108: 86-94, 2017 07.
Article in English | MEDLINE | ID: mdl-28526246

ABSTRACT

RATIONALE: Lysosomal associated membrane protein type-2 (LAMP-2) is a highly conserved, ubiquitous protein that is critical for autophagic flux. Loss of function mutations in the LAMP-2 gene cause Danon disease, a rare X-linked disorder characterized by developmental delay, skeletal muscle weakness, and severe cardiomyopathy. We previously found that human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from Danon patients exhibited significant mitochondrial oxidative stress and apoptosis. Understanding how loss of LAMP-2 expression leads to cardiomyocyte dysfunction and heart failure has important implications for the treatment of Danon disease as well as a variety of other cardiac disorders associated with impaired autophagy. OBJECTIVE: Elucidate the pathophysiology of cardiac dysfunction in Danon disease. METHODS AND RESULTS: We created hiPSCs from two patients with Danon disease and differentiated those cells into hiPSC-CMs using well-established protocols. Danon hiPSC-CMs demonstrated an accumulation of damaged mitochondria, disrupted mitophagic flux, depressed mitochondrial respiratory capacity, and abnormal gene expression of key mitochondrial pathways. Restoring the expression of LAMP-2B, the most abundant LAMP-2 isoform in the heart, rescued mitophagic flux as well as mitochondrial health and bioenergetics. To confirm our findings in vivo, we evaluated Lamp-2 knockout (KO) mice. Impaired autophagic flux was noted in the Lamp-2 KO mice compared to WT reporter mice, as well as an increased number of abnormal mitochondria, evidence of incomplete mitophagy, and impaired mitochondrial respiration. Physiologically, Lamp-2 KO mice demonstrated early features of contractile dysfunction without overt heart failure, indicating that the metabolic abnormalities associated with Danon disease precede the development of end-stage disease and are not merely part of the secondary changes associated with heart failure. CONCLUSIONS: Incomplete mitophagic flux and mitochondrial dysfunction are noted in both in vitro and in vivo models of Danon disease, and proceed overt cardiac contractile dysfunction. This suggests that impaired mitochondrial clearance may be central to the pathogenesis of disease and a potential target for therapeutic intervention.


Subject(s)
Glycogen Storage Disease Type IIb/genetics , Glycogen Storage Disease Type IIb/metabolism , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Mitophagy/genetics , Animals , Gene Knockout Techniques , Glycogen Storage Disease Type IIb/diagnosis , Hemodynamics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Magnetic Resonance Imaging , Mice, Knockout , Mitochondria, Heart/ultrastructure , Models, Biological , Myocytes, Cardiac/metabolism
9.
Arch Biochem Biophys ; 615: 53-60, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28088328

ABSTRACT

Mutations in the human cardiac motor protein beta-myosin heavy chain (ßMHC) have been long recognized as a cause of familial hypertrophic cardiomyopathy. Recently, mutations (P830L and A1004S) in the less abundant but faster isoform alpha-myosin heavy chain (αMHC) have been linked to dilated cardiomyopathy (DCM). In this study, we sought to determine the cellular contractile phenotype associated with these point mutations. Ventricular myocytes were isolated from 2 month male Sprague Dawley rats. Cells were cultured in M199 media and infected with recombinant adenovirus containing the P830L or the A1004S mutant human αMHC at a MOI of 500 for 18 h. Uninfected cells (UI), human ßMHC (MOI 500, 18 h), and human αMHC (MOI 500, 18 h) were used as controls. Cells were loaded with fura-2 (1 µM, 15 min) after 48 h. Sarcomere shortening and calcium transients were recorded in CO2 buffered M199 media (36°±1 C) with and without 10 nM isoproterenol (Iso). The A1004S mutation resulted in decreased peak sarcomere shortening while P830L demonstrated near normal shortening kinetics at baseline. In the presence of Iso, the A1004S sarcomere shortening was identical to the ßMHC shortening while the P830L was identical to the αMHC control. All experimental groups had identical calcium transients. Despite a shared association with DCM, the P830L and A1004S αMHC mutations alter myocyte contractility in completely different ways while at the same preserving peak intracellular calcium.


Subject(s)
Calcium/metabolism , Muscle Cells/cytology , Myosin Heavy Chains/genetics , Animals , Cardiomyopathy, Dilated , Homeostasis , Humans , Hypertrophy , Isoproterenol/chemistry , Kinetics , Male , Mutagenesis , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/metabolism , Phenotype , Point Mutation , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Sarcomeres/metabolism , Ventricular Myosins/metabolism
10.
Sci Signal ; 7(326): ra48, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24847117

ABSTRACT

Thyroid hormone (TH) is essential for vertebrate development and the homeostasis of most adult tissues, including bone. TH stimulates target gene expression through the nuclear thyroid receptors TRα and TRß; however, TH also has rapid, transcription-independent (nongenomic) effects. We found a previously uncharacterized plasma membrane-bound receptor that was necessary and sufficient for nongenomic TH signaling in several cell types. We determined that this receptor is generated by translation initiation from an internal methionine of TRα, which produces a transcriptionally incompetent protein that is palmitoylated and associates with caveolin-containing plasma membrane domains. TH signaling through this receptor stimulated a pro-proliferative and pro-survival program by increasing the intracellular concentrations of calcium, nitric oxide (NO), and cyclic guanosine monophosphate (cGMP), which led to the sequential activation of protein kinase G II (PKGII), the tyrosine kinase Src, and extracellular signal-regulated kinase (ERK) and Akt signaling. Hypothyroid mice exhibited a cGMP-deficient state with impaired bone formation and increased apoptosis of osteocytes, which was rescued by a direct stimulator of guanylate cyclase. Our results link nongenomic TH signaling to a previously uncharacterized membrane-bound receptor, and identify NO synthase, guanylate cyclase, and PKGII as TH effectors that activate kinase cascades to regulate cell survival and proliferation.


Subject(s)
MAP Kinase Signaling System , Membrane Microdomains/metabolism , Osteocytes/metabolism , Receptors, Thyroid Hormone/metabolism , Thyroid Hormones/metabolism , Adult , Animals , Cells, Cultured , Humans , Hypothyroidism/genetics , Hypothyroidism/metabolism , Membrane Microdomains/genetics , Mice , Mice, Transgenic , Osteogenesis/genetics , Rats , Receptors, Thyroid Hormone/genetics , Thyroid Hormones/genetics
11.
Stem Cell Res ; 11(3): 1335-47, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24095945

ABSTRACT

Applications of human induced pluripotent stem cell derived-cardiac myocytes (hiPSC-CMs) would be strengthened by the ability to generate specific cardiac myocyte (CM) lineages. However, purification of lineage-specific hiPSC-CMs is limited by the lack of cell marking techniques. Here, we have developed an iPSC-CM marking system using recombinant adenoviral reporter constructs with atrial- or ventricular-specific myosin light chain-2 (MLC-2) promoters. MLC-2a and MLC-2v selected hiPSC-CMs were purified by fluorescence-activated cell sorting and their biochemical and electrophysiological phenotypes analyzed. We demonstrate that the phenotype of both populations remained stable in culture and they expressed the expected sarcomeric proteins, gap junction proteins and chamber-specific transcription factors. Compared to MLC-2a cells, MLC-2v selected CMs had larger action potential amplitudes and durations. In addition, by immunofluorescence, we showed that MLC-2 isoform expression can be used to enrich hiPSC-CM consistent with early atrial and ventricular myocyte lineages. However, only the ventricular myosin light chain-2 promoter was able to purify a highly homogeneous population of iPSC-CMs. Using this approach, it is now possible to develop ventricular-specific disease models using iPSC-CMs while atrial-specific iPSC-CM cultures may require additional chamber-specific markers.


Subject(s)
Cardiac Myosins/metabolism , Cell Separation/methods , Heart Ventricles/cytology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Myosin Light Chains/metabolism , Adenoviridae/genetics , Cardiac Myosins/genetics , Cell Differentiation , Cell Lineage , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Myosin Light Chains/genetics , Phenotype , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism
12.
Circ Res ; 111(9): 1125-36, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22912385

ABSTRACT

RATIONALE: Cardiomyocytes (CMs) differentiated from human pluripotent stem cells (PSCs) are increasingly being used for cardiovascular research, including disease modeling, and hold promise for clinical applications. Current cardiac differentiation protocols exhibit variable success across different PSC lines and are primarily based on the application of growth factors. However, extracellular matrix is also fundamentally involved in cardiac development from the earliest morphogenetic events, such as gastrulation. OBJECTIVE: We sought to develop a more effective protocol for cardiac differentiation of human PSCs by using extracellular matrix in combination with growth factors known to promote cardiogenesis. METHODS AND RESULTS: PSCs were cultured as monolayers on Matrigel, an extracellular matrix preparation, and subsequently overlayed with Matrigel. The matrix sandwich promoted an epithelial-to-mesenchymal transition as in gastrulation with the generation of N-cadherin-positive mesenchymal cells. Combining the matrix sandwich with sequential application of growth factors (Activin A, bone morphogenetic protein 4, and basic fibroblast growth factor) generated CMs with high purity (up to 98%) and yield (up to 11 CMs/input PSC) from multiple PSC lines. The resulting CMs progressively matured over 30 days in culture based on myofilament expression pattern and mitotic activity. Action potentials typical of embryonic nodal, atrial, and ventricular CMs were observed, and monolayers of electrically coupled CMs modeled cardiac tissue and basic arrhythmia mechanisms. CONCLUSIONS: Dynamic extracellular matrix application promoted epithelial-mesenchymal transition of human PSCs and complemented growth factor signaling to enable robust cardiac differentiation.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Collagen , Extracellular Matrix/physiology , Laminin , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Proteoglycans , Activins/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Drug Combinations , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Fibroblast Growth Factor 2/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
13.
J Clin Invest ; 121(7): 2641-50, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21646717

ABSTRACT

Histone H3 lysine 4 (H3K4me) methyltransferases and their cofactors are essential for embryonic development and the establishment of gene expression patterns in a cell-specific and heritable manner. However, the importance of such epigenetic marks in maintaining gene expression in adults and in initiating human disease is unclear. Here, we addressed this question using a mouse model in which we could inducibly ablate PAX interacting (with transcription-activation domain) protein 1 (PTIP), a key component of the H3K4me complex, in cardiac cells. Reducing H3K4me3 marks in differentiated cardiomyocytes was sufficient to alter gene expression profiles. One gene regulated by H3K4me3 was Kv channel-interacting protein 2 (Kcnip2), which regulates a cardiac repolarization current that is downregulated in heart failure and functions in arrhythmogenesis. This regulation led to a decreased sodium current and action potential upstroke velocity and significantly prolonged action potential duration (APD). The prolonged APD augmented intracellular calcium and in vivo systolic heart function. Treatment with isoproterenol and caffeine in this mouse model resulted in the generation of premature ventricular beats, a harbinger of lethal ventricular arrhythmias. These results suggest that the maintenance of H3K4me3 marks is necessary for the stability of a transcriptional program in differentiated cells and point to an essential function for H3K4me3 epigenetic marks in cellular homeostasis.


Subject(s)
Gene Expression , Histones/metabolism , Lysine/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Animals , Calcium/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins , Epigenesis, Genetic , Histones/genetics , Humans , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Methylation , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ventricular Premature Complexes
14.
Heart Rhythm ; 7(10): 1501-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20621203

ABSTRACT

BACKGROUND: The role of coronary perfusion in the maintenance of atrial fibrillation (AF) electrical sources that anchor to the posterior (wall of the) left atrium (PLA) has been incompletely investigated. We hypothesized that the PLA-pulmonary vein region is perfused by branches originating from both the right and left coronary arteries. OBJECTIVE: The purpose of this study was to evaluate whether branches originating from the right and left coronary arteries could serve as conduits to chemically ablate restricted PLA regions. METHODS: In Langendorff-perfused sheep hearts, the right anterior and left anterior atrial arteries (RAAA and LAAA) and the branches of the left circumflex artery (LCX) were identified as main coronary artery branches perfusing the atria. During sustained AF, 20-mL boluses of cold Tyrode's solution (4°C) was injected into each artery to determine changes in dominant frequency. The injection that yielded the largest dominant frequency decrease indicated the coronary branch to be subsequently perfused with ethanol. Ethanol was selectively injected into the LAAA (n = 4), LCX (n = 4), or RAAA (n = 1). RESULTS: Six of nine AF cases rapidly terminated upon ethanol perfusion. In those hearts and in eight additional preparations (n = 17), Congo red and Evans blue was subsequently perfused into the remaining atrial branches. The perfusion territories were classified as triple-vessel PLA perfusion (n = 4), LAAA-dominant PLA perfusion (n = 5), balanced double-vessel PLA perfusion (n = 5), and LCX or RAAA dominant (n = 3). CONCLUSION: PLA coronary perfusion relies on a variable contribution of right and left coronary branches. Regional irrigation of ethanol in well-delineated PLA perfusion territories enabled ablation of high-frequency sites during AF.


Subject(s)
Atrial Fibrillation/physiopathology , Coronary Circulation , Ablation Techniques , Animals , Atrial Fibrillation/surgery , Cardiac Pacing, Artificial , Catheter Ablation , Coronary Vessels/physiopathology , Ethanol/pharmacology , Heart Atria/physiopathology , Isotonic Solutions/pharmacology , Pulmonary Veins/physiopathology , Sheep, Domestic
15.
Heart Rhythm ; 6(7): 1009-17, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19560089

ABSTRACT

BACKGROUND: Both atrial stretch and combined adrenocholinergic stimulation (ACS) have been shown to favor initiation and maintenance of atrial fibrillation (AF). Their respective contributions to the electrophysiological mechanism remains, however, incompletely understood. OBJECTIVE: This study endeavored to determine the mechanism of maintenance of stretch-related AF (SRAF) in the presence and absence of ACS and to assess how focal discharges interact with rotors to modify the level of complexity in the activation patterns to perpetuate AF. METHODS: Video imaging of AF dynamics was carried out using a SRAF model in isolated sheep hearts (n = 24). Pharmacological approaches were used to (1) mimic ACS with acetylcholine (1 microM) plus isoproterenol (0.03 microM), and (2) abolish triggered activity, in response to sarcoplasmic reticulum calcium release, with caffeine (5 mM, CA) or ryanodine (10 to 40 microM, RYA). RESULTS: In the absence of ACS, on perfusion of CA or RYA, focal discharges were abolished and SRAF was terminated in most of the cases (10 of 13 experiments). In the presence of ACS, multiple drifting rotors as well as a large number of focal discharges were identified and only 1 of 11 AF episodes was terminated. CONCLUSIONS: In the absence of ACS, SRAF is maintained by high-frequency focal discharges that generate fibrillatory conduction and wave breaks. In the presence of ACS, SRAF dynamics is characterized by multiple high frequency rotors that are rendered unstable by spatially distributed focal discharges.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Atrial Fibrillation/physiopathology , Cholinergic Agents/pharmacology , Heart/drug effects , Acetylcholine/pharmacology , Animals , Atrial Fibrillation/etiology , Disease Models, Animal , Isoproterenol/pharmacology , Sheep , Video Recording
16.
Circ Arrhythm Electrophysiol ; 1(3): 175-83, 2008 Aug.
Article in English | MEDLINE | ID: mdl-19609369

ABSTRACT

BACKGROUND: The posterior left atrium (PLA) and pulmonary veins (PVs) have been shown to be critical for atrial fibrillation (AF) initiation. However, the detailed mechanisms of reentry and AF initiation by PV impulses are poorly understood. We hypothesized that PV impulses trigger reentry and AF by undergoing wavebreaks as a result of sink-to-source mismatch at specific PV-PLA transitions along the septopulmonary bundle, where there are changes in thickness and fiber direction. METHODS AND RESULTS: In 7 Langendorff-perfused sheep hearts AF was initiated by a burst of 6 pulses (CL 80 to 150ms) delivered to the left inferior or right superior PV ostium 100 to 150 ms after the sinus impulse in the presence of 0.5 micromol/L acetylcholine. The exposed septal-PLA endocardial area was mapped with high spatio-temporal resolution (DI-4-ANEPPS, 1000-fr/s) during AF initiation. Isochronal maps for each paced beat preceding AF onset were constructed to localize areas of conduction delay and block. Phase movies allowed the determination of the wavebreak sites at the onset of AF. Thereafter, the PLA myocardial wall thickness was quantified by echocardiography, and the fiber direction in the optical field of view was determined after peeling off the endocardium. Finally, isochrone, phase and conduction velocity maps were superimposed on the corresponding anatomic pictures for each of the 28 episodes of AF initiation. The longest delays of the paced PV impulses, as well as the first wavebreak, occurred at those boundaries along the septopulmonary bundle that showed sharp changes in fiber direction and the largest and most abrupt increase in myocardial thickness. CONCLUSION: Waves propagating from the PVs into the PLA originating from a simulated PV tachycardia triggered reentry and vagally mediated AF by breaking at boundaries along the septopulmonary bundle where abrupt changes in thickness and fiber direction resulted in sink-to-source mismatch and low safety for propagation.


Subject(s)
Atrial Fibrillation/physiopathology , Heart Atria/innervation , Heart Conduction System/pathology , Nerve Fibers/pathology , Pulmonary Veins/innervation , Vagus Nerve/physiopathology , Animals , Atrial Fibrillation/pathology , Disease Models, Animal , Electrophysiologic Techniques, Cardiac/methods , Heart Conduction System/physiopathology , Male , Pulmonary Veins/pathology , Sheep
17.
Circ Res ; 101(8): 839-47, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17704207

ABSTRACT

Heart failure (HF) commonly results in atrial fibrillation (AF) and fibrosis, but how the distribution of fibrosis impacts AF dynamics has not been studied. HF was induced in sheep by ventricular tachypacing (220 bpm, 6 to 7 weeks). Optical mapping (Di-4-ANEPPS, 300 frames/sec) of the posterior left atrial (PLA) endocardium was performed during sustained AF (burst pacing) in Langendorff-perfused HF (n=7, 4 micromol/L acetylcholine; n=3, no acetylcholine) and control (n=6) hearts. PLA breakthroughs were the most frequent activation pattern in both groups (72.0+/-4.6 and 90.2+/-2.7%, HF and control, respectively). However, unlike control, HF breakthroughs preferentially occurred at the PLAs periphery near the pulmonary vein ostia, and their beat-to-beat variability was greater than control (1.93+/-0.14 versus 1.47+/-0.07 changes/[beats/sec], respectively, P<0.05). On histological analysis (picrosirius red), the area of diffuse fibrosis was larger in HF (23.4+/-0.4%) than control (14.1+/-0.6%; P<0.001, n=4). Also the number and size of fibrous patches were significantly larger and their location was more peripheral in HF than control. Computer simulations using 2-dimensional human atrial models with structural and ionic remodeling as in HF demonstrated that changes in AF activation frequency and dynamics were controlled by the interaction of electrical waves with clusters of fibrotic patches of various sizes and individual pulmonary vein ostia. During AF in failing hearts, heterogeneous spatial distribution of fibrosis at the PLA governs AF dynamics and fractionation.


Subject(s)
Atrial Fibrillation/physiopathology , Atrial Function, Left/physiology , Heart Failure/physiopathology , Animals , Atrial Fibrillation/complications , Atrial Fibrillation/pathology , Fibrosis , Heart Failure/complications , Heart Failure/pathology , Sheep
18.
Heart Rhythm ; 4(7): 916-24, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17599678

ABSTRACT

BACKGROUND: Despite the availability of several mapping technologies for investigating the electrophysiologic mechanisms of atrial fibrillation (AF), an experimental tool enabling high-resolution mapping of electrical impulses on the endocardial surface of the intact left atrium is lacking. OBJECTIVE: The purpose of this report is to present a new optical mapping approach implementing a steerable cardio-endoscope in isolated hearts. METHODS: The system consists of a direct or side-view endoscope coupled to a 532-nm excitation laser for illumination and a CCD camera for imaging of potentiometric dye fluorescence (di-4-ANEPPS, 80 x 80 pixels, 200-800 frames/s). The cardio-endoscope was aimed successively at diverse posterior left atrial locations to obtain high-resolution movies of electrical wave propagation and detailed endocardial anatomic features in the presence and absence of atrial stretch. RESULTS: We present several examples of high-resolution endoscopic posterior left atrial recordings of wave propagation patterns during both sinus rhythm and AF with signal-to-noise ratio similar to conventional optical mapping systems. We demonstrate the endoscope's ability to visualize highly organized AF sources (rotors) at specific locations on the posterior left atrium and posterior left atrium-pulmonary vein junctions. We present video images of waves emanating from such sources as they propagate into pectinate muscles in the left atrial appendage. In particular, we demonstrate this approach is ideally suited for studying the effects of atrial stretch on AF dynamics. CONCLUSION: In isolated hearts, cardio-endoscopic optical mapping of electrical activity should enable comprehensive evaluation of AF activity in the posterior left atrium, the role of local anatomy on AF dynamics, and the efficacy of pharmacologic and ablative interventions.


Subject(s)
Atrial Function, Left/physiology , Endoscopy/methods , Heart Atria/anatomy & histology , Imaging, Three-Dimensional/methods , Animals , Fluorescence , Lasers , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...