Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
3.
Pacing Clin Electrophysiol ; 41(3): 261-266, 2018 03.
Article in English | MEDLINE | ID: mdl-29344960

ABSTRACT

BACKGROUND: Telomeres are known to provide genomic stability and telomere length has been associated with cardiovascular diseases. Moreover, a higher telomerase activity has been shown to be associated with ventricular arrhythmias (VA) in ischemic cardiomyopathy. Increasing evidence suggests that genetic variation in key telomere genes has an impact on telomerase activity. Each copy of the minor allele of SNP rs12696304, at a locus including TERC (telomerase), has been associated with ∼75 base pairs reduction in mean telomere length likely mediated by an effect on TERC expression. We investigated the impact of genetic variation of this SNP on telomerase and its association with VA in ischemic cardiomyopathy patients. METHODS AND RESULTS: Ninety ischemic cardiomyopathy patients with primary prevention implantable cardioverter defibrillators (ICDs) were recruited. Thirty-five received appropriate ICD therapy for potentially fatal VA (cases), while the remaining 55 patients did not (controls). No significant differences in baseline demographics were seen between the groups. TS was measured by qPCR, telomerase activity by TRAP assay, and SNP genotyping with Taqman probes. Telomerase was highest in C homozygous allele and had a significant association with VA in this group only (C/C,C/G,G/G; P-value 0.04, 0.33, 0.43). CONCLUSION: The present study is the first to examine the association between telomerase, a SNP at a locus including TERC, and VA in ischemic cardiomyopathy patients. Homozygosity for C-allele significantly effects telomerase expression and its association with VA in this cohort. Large-scale prospective studies are required to determine if this genetic variation predisposes patients to greater arrhythmic tendency post-MI.


Subject(s)
Arrhythmias, Cardiac/genetics , Cardiomyopathies/genetics , Myocardial Ischemia/genetics , Polymorphism, Single Nucleotide , Telomerase/genetics , Telomere/genetics , Aged , Alleles , Arrhythmias, Cardiac/enzymology , Cardiomyopathies/enzymology , Case-Control Studies , Cross-Sectional Studies , Defibrillators, Implantable , Female , Genetic Variation , Genotype , Humans , Male , Myocardial Ischemia/enzymology , Polymerase Chain Reaction , Retrospective Studies
4.
PLoS One ; 11(7): e0158232, 2016.
Article in English | MEDLINE | ID: mdl-27380410

ABSTRACT

Intracoronary injection of bone marrow mononuclear cells (BMMNC) is an emerging treatment for heart failure. Initial donor cell retention in the heart is the key to the success of this approach, but this process remains insufficiently characterized. Although it is assumed that cell size of injected cells may influence their initial retention, no scientific evidence has been reported. We developed a unique model utilizing an ex-vivo rat heart perfusion system, enabling quantitative assessment of retention of donor cells after intracoronary injection. The initial (5 minutes after intracoronary injection) retention rate of BMMNC was as low as approximately 20% irrespective of donor cell doses injected (1×106, 8×106, 4×107). Quantitative cell-size assessment revealed a positive relationship between the size of BMMNC and retention ratio; larger subpopulations of BMMNC were more preferentially retained compared to smaller ones. Furthermore, a larger cell type-bone marrow-derived mesenchymal stromal cells (median size = 11.5µm versus 7.0µm for BMMNC)-had a markedly increased retention rate (77.5±1.8%). A positive relationship between the cell size and retention ratio was also seen in mesenchymal stromal cells. Flow-cytometric studies showed expression of cell-surface proteins, including integrins and selectin-ligands, was unchanged between pre-injection BMMNC and those exited from the heart, suggesting that biochemical interaction between donor cells and host coronary endothelium is not critical for BMMNC retention. Histological analyses showed that retained BMMNC and mesenchymal stromal cells were entrapped in the coronary vasculature and did not extravasate by 60 minutes after transplantation. Whilst BMMNC did not change coronary flow after intracoronary injection, mesenchymal stromal cells reduced it, suggesting coronary embolism, which was supported by the histological finding of intravascular cell-clump formation. These data indicate that cell-size dependent, passive (mechanical), intravascular entrapment is responsible for the initial donor cell retention after intracoronary injection of BMMNC in the heart having normal vasculatures (at least).


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation/methods , Cell Size , Heart Failure/therapy , Leukocytes, Mononuclear/cytology , Animals , Bone Marrow Cells/metabolism , Cell Survival , Coronary Vessels/metabolism , Disease Models, Animal , Flow Cytometry , Graft Survival , In Vitro Techniques , Injections , Leukocytes, Mononuclear/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Rats, Sprague-Dawley
5.
J Am Heart Assoc ; 5(2)2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26896478

ABSTRACT

BACKGROUND: Transplantation of allogeneic mesenchymal stromal cells (MSCs) is a promising treatment for heart failure. We have shown that epicardial placement of cell sheets markedly increases donor cell survival and augments therapeutic effects compared with the current methods. Although immune rejection of intramyocardially injected allogeneic MSCs have been suggested, allogeneic MSCs transplanted on the heart surface (virtual space) may undergo different courses. This study aimed to elucidate immunologic response against epicardially placed allogeneic MSCs, rejection or acceptance of these cells, and their therapeutic effects for heart failure. METHODS AND RESULTS: At 4 weeks after coronary artery ligation, Lewis rats underwent epicardial placement of MSC sheets from syngeneic Lewis or allogeneic Fischer 344 rats or sham treatment. At days 3 and 10 after treatment, similar ratios (≈50% and 30%, respectively) of grafted MSCs survived on the heart surface in both MSC sheet groups. By day 28, survival of syngeneic MSCs was substantially reduced (8.9%); survival of allogeneic MSCs was more extensively reduced (0.2%), suggesting allorejection. Correspondingly, allogeneic MSCs were found to have evoked an immunologic response, albeit low level, as characterized by accumulation of CD4(+) T cells and upregulation of interleukin 6. Despite this alloimmune response, the allogeneic MSC sheet achieved myocardial upregulation of reparative factors, enhanced repair of the failing myocardium, and improved cardiac function to the equivalent degree observed for the syngeneic MSC sheet. CONCLUSIONS: Allogeneic MSCs placed on the heart surface evoked an immunologic response; however, this allowed sufficient early phase donor cell survival to induce equivalent therapeutic benefits to syngeneic MSCs. Further development of this approach toward clinical application is warranted.


Subject(s)
Heart Failure/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Myocardial Infarction/surgery , Myocardium/immunology , Regeneration , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Survival , Cells, Cultured , Disease Models, Animal , Graft Rejection/immunology , Graft Survival , Heart Failure/immunology , Heart Failure/pathology , Heart Failure/physiopathology , Interleukin-6/immunology , Male , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Rats, Inbred F344 , Rats, Inbred Lew , Recovery of Function , Stroke Volume , Time Factors , Transplantation, Homologous , Ventricular Function, Left
6.
Dev Dyn ; 245(2): 157-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26515123

ABSTRACT

BACKGROUND: Embryonic stem (ES) cells are pluripotent cells with the ability to differentiate to any cell type of the resident organism. In recent years, significant advances have been made in using these cells to obtain large numbers of cardiomyocyte (CM)-like cells for scientific research and clinical application. A vast number of protocols have emerged describing differentiation methods without the use of animal serum or extracts restrictive for use in a human clinical setting. These techniques follow a complicated procedure, which although successful, show a relatively varied yield among cell batches. RESULTS: We have designed a three-step differentiation protocol using defined reagents and a monolayer culture without feeder cells, avoiding embryoid body formation and multiple trypsin treatment, in which beating foci appeared as early as day 6 in in vitro differentiating conditions. Our results show a high yield of CM reaching approximately 60% of the differentiated cells after 13 days in vitro. CONCLUSIONS: We provide a fast, simple, reliable and reproducible protocol for inducing murine ES cells toward a CM-like phenotype comparable to available high-yield protocols, without the use of intermediate trypsinization/passage steps.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Mice
7.
PLoS One ; 10(10): e0140831, 2015.
Article in English | MEDLINE | ID: mdl-26469858

ABSTRACT

In the early vertebrate embryo, cardiac progenitor/precursor cells (CPs) give rise to cardiac structures. Better understanding their biological character is critical to understand the heart development and to apply CPs for the clinical arena. However, our knowledge remains incomplete. With the use of single-cell expression profiling, we have now revealed rapid and dynamic changes in gene expression profiles of the embryonic CPs during the early phase after their segregation from the cardiac mesoderm. Progressively, the nascent mesodermal gene Mesp1 terminated, and Nkx2-5+/Tbx5+ population rapidly replaced the Tbx5low+ population as the expression of the cardiac genes Tbx5 and Nkx2-5 increased. At the Early Headfold stage, Tbx5-expressing CPs gradually showed a unique molecular signature with signs of cardiomyocyte differentiation. Lineage-tracing revealed a developmentally distinct characteristic of this population. They underwent progressive differentiation only towards the cardiomyocyte lineage corresponding to the first heart field rather than being maintained as a progenitor pool. More importantly, Tbx5 likely plays an important role in a transcriptional network to regulate the distinct character of the FHF via a positive feedback loop to activate the robust expression of Tbx5 in CPs. These data expands our knowledge on the behavior of CPs during the early phase of cardiac development, subsequently providing a platform for further study.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Profiling/methods , Myocytes, Cardiac/metabolism , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryo, Mammalian , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Pregnancy , Transcriptome
8.
Mol Ther ; 22(10): 1864-71, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24930600

ABSTRACT

Transplantation of bone marrow mesenchymal stromal cells (MSCs) is an emerging treatment for heart failure. We have reported that epicardial placement of MSC-sheets generated using temperature-responsive dishes markedly increases donor MSC survival and augments therapeutic effects in an acute myocardial infarction (MI) model, compared to intramyocardial (IM) injection. This study aims to expand this knowledge for the treatment of ischemic cardiomyopathy, which is likely to be more difficult to treat due to mature fibrosis and chronically stressed myocardium. Four weeks after MI, rats underwent either epicardial MSC-sheet placement, IM MSC injection, or sham treatment. At day 28 after treatment, the cell-sheet group showed augmented cardiac function improvement, which was associated with over 11-fold increased donor cell survival at both days 3 and 28 compared to IM injection. Moreover, the cell-sheet group showed improved myocardial repair, in conjunction with amplified upregulation of a group of reparative factors. Furthermore, by comparing with our own previous data, this study highlighted similar dynamics and behavior of epicardially placed MSCs in acute and chronic stages after MI, while the acute-phase myocardium may be more responsive to the stimuli from donor MSCs. These proof-of-concept data encourage further development of the MSC-sheet therapy for ischemic cardiomyopathy toward clinical application.


Subject(s)
Guided Tissue Regeneration , Mesenchymal Stem Cells/cytology , Myocardial Ischemia/therapy , Pericardium , Regeneration , Animals , Cell Differentiation , Cell Survival , Disease Models, Animal , Endothelial Cells/cytology , Female , Male , Mesenchymal Stem Cell Transplantation , Myocardial Ischemia/physiopathology , Rats , Tissue Scaffolds
9.
EMBO Rep ; 15(4): 438-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24610369

ABSTRACT

Toll-like receptor 9 (TLR9) has a key role in the recognition of pathogen DNA in the context of infection and cellular DNA that is released from damaged cells. Pro-inflammatory TLR9 signalling pathways in immune cells have been well investigated, but we have recently discovered an alternative pathway in which TLR9 temporarily reduces energy substrates to induce cellular protection from stress in cardiomyocytes and neurons. However, the mechanism by which TLR9 stimulation reduces energy substrates remained unknown. Here, we identify the calcium-transporting ATPase, SERCA2 (also known as Atp2a2), as a key molecule for the alternative TLR9 signalling pathway. TLR9 stimulation reduces SERCA2 activity, modulating Ca(2+) handling between the SR/ER and mitochondria, which leads to a decrease in mitochondrial ATP levels and the activation of cellular protective machinery. These findings reveal how distinct innate responses can be elicited in immune and non-immune cells--including cardiomyocytes--using the same ligand-receptor system.


Subject(s)
Adenosine Triphosphate/biosynthesis , Fibroblasts/physiology , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Toll-Like Receptor 9/physiology , Animals , Calcium/metabolism , Calcium Signaling , Cells, Cultured , Endoplasmic Reticulum/metabolism , Mice , Mitochondria/metabolism , Protein Binding , Stress, Physiological
10.
PLoS One ; 8(10): e76908, 2013.
Article in English | MEDLINE | ID: mdl-24204700

ABSTRACT

Transplantation of unfractionated bone marrow mononuclear cells (BMCs) repairs and/or regenerates the damaged myocardium allegedly due to secretion from surviving BMCs (paracrine effect). However, donor cell survival after transplantation is known to be markedly poor. This discrepancy led us to hypothesize that dead donor BMCs might also contribute to the therapeutic benefits from BMC transplantation. High mobility group box 1 (HMGB1) is a nuclear protein that stabilizes nucleosomes, and also acts as a multi-functional cytokine when released from damaged cells. We thus studied the role of extracellular HMGB1 in the effect of BMC transplantation for heart failure. Four weeks after coronary artery ligation in female rats, syngeneic male BMCs (or PBS only as control) were intramyocardially injected with/without anti-HMGB1 antibody or control IgG. One hour after injection, ELISA showed that circulating extracellular HMGB1 levels were elevated after BMC transplantation compared to the PBS injection. Quantitative donor cell survival assessed by PCR for male-specific sry gene at days 3 and 28 was similarly poor. Echocardiography and catheterization showed enhanced cardiac function after BMC transplantation compared to PBS injection at day 28, while this effect was abolished by antibody-neutralization of HMGB1. BMC transplantation reduced post-infarction fibrosis, improved neovascularization, and increased proliferation, while all these effects in repairing the failing myocardium were eliminated by HMGB1-inhibition. Furthermore, BMC transplantation drove the macrophage polarization towards alternatively-activated, anti-inflammatory M2 macrophages in the heart at day 3, while this was abolished by HMGB1-inhibition. Quantitative RT-PCR showed that BMC transplantation upregulated expression of an anti-inflammatory cytokine IL-10 in the heart at day 3 compared to PBS injection. In contrast, neutralizing HMGB1 by antibody-treatment suppressed this anti-inflammatory expression. These data suggest that extracellular HMGB1 contributes to the effect of BMC transplantation to recover the damaged myocardium by favorably modulating innate immunity in heart failure.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , HMGB1 Protein/metabolism , Heart Failure/surgery , Leukocytes, Mononuclear/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Cell Proliferation/drug effects , Echocardiography , Extracellular Space/metabolism , Female , Gene Expression/drug effects , Genes, sry/genetics , Graft Survival , HMGB1 Protein/immunology , HMGB1 Protein/physiology , Heart Failure/metabolism , Heart Failure/physiopathology , Interleukin-10/genetics , Macrophages/metabolism , Male , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
Proc Natl Acad Sci U S A ; 110(13): 5109-14, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23479602

ABSTRACT

Toll-like receptors (TLRs) are the central players in innate immunity. In particular, TLR9 initiates inflammatory response by recognizing DNA, imported by infection or released from tissue damage. Inflammation is, however, harmful to terminally differentiated organs, such as the heart and brain, with poor regenerative capacity, yet the role of TLR9 in such nonimmune cells, including cardiomyocytes and neurons, is undefined. Here we uncover an unexpected role of TLR9 in energy metabolism and cellular protection in cardiomyocytes and neurons. TLR9 stimulation reduced energy substrates and increased the AMP/ATP ratio, subsequently activating AMP-activated kinase (AMPK), leading to increased stress tolerance against hypoxia in cardiomyocytes without inducing the canonical inflammatory response. Analysis of the expression profiles between cardiomyocytes and macrophages identified that unc93 homolog B1 (C. elegans) was a pivotal switch for the distinct TLR9 responses by regulating subcellular localization of TLR9. Furthermore, this alternative TLR9 signaling was also found to operate in differentiated neuronal cells. These data propose an intriguing model that the same ligand-receptor can concomitantly increase the stress tolerance in cardiomyocytes and neurons, whereas immune cells induce inflammation upon tissue injury.


Subject(s)
Energy Metabolism/physiology , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Toll-Like Receptor 9/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cells, Cultured , Inflammation/genetics , Inflammation/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Muscle Proteins/genetics , Myocytes, Cardiac/cytology , Nerve Tissue Proteins/genetics , Neurons/cytology , Protein Transport/physiology , Rats , Rats, Wistar , Signal Transduction/physiology , Toll-Like Receptor 9/genetics
12.
Mol Ther ; 21(4): 860-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358187

ABSTRACT

Transplantation of bone marrow-derived mesenchymal stromal cells (MSCs) is an emerging treatment for heart failure based on their secretion-mediated "paracrine effects". Feasibility of the scaffoldless cell sheet technique to enhance the outcome of cell transplantation has been reported using other cell types, though the mechanism underpinning the enhancement remains uncertain. We here investigated the role of this innovative technique to amplify the effects of MSC transplantation with a focus on the underlying factors. After coronary artery ligation in rats, syngeneic MSCs were grafted by either epicardial placement of MSC sheets generated using temperature-responsive dishes or intramyocardial (IM) injection. Markedly increased initial retention boosted the presence of donor MSCs persistently after MSC sheet placement although the donor survival was not improved. Most of the MSCs grafted by the cell sheet technique remained resided on the epicardial surface, but the epicardium quickly regressed and new vessels sprouted into the sheets, assuring the permeation of paracrine mediators from MSCs into the host myocardium. In fact, there was augmented upregulation of various paracrine effect-related genes and signaling pathways in the early phase after MSC sheet therapy. Correspondingly, more extensive paracrine effects and resultant cardiac function recovery were achieved by MSC sheet therapy. Further development of this approach towards clinical application is encouraged.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Heart Failure/therapy , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Female , Male , Mesenchymal Stem Cells/physiology , Rats
13.
Int J Cardiol ; 168(1): 261-9, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23046598

ABSTRACT

BACKGROUND: Clinical application of skeletal myoblast transplantation has been curtailed due to arrhythmogenicity and inconsistent therapeutic benefits observed in previous studies. However, these issues may be solved by the use of a new cell-delivery mode. It is now possible to generate "cell-sheets" using temperature-responsive dishes without artificial scaffolds. This study aimed to validate the safety and efficacy of epicardial placement of myoblast-sheets (myoblast-sheet therapy) in treating heart failure. METHODS AND RESULTS: After coronary artery ligation in rats, the same numbers of syngeneic myoblasts were transplanted by intramyocardial injection or cell-sheet placement. Continuous radio-telemetry monitoring detected increased ventricular arrhythmias, including ventricular tachycardia, after intramyocardial injection compared to the sham-control, while these were abolished in myoblast-sheet therapy. This effect was conjunct with avoidance of islet-like cell-cluster formation that disrupts electrical conduction, and with prevention of increased arrhythmogenic substrates due to exaggerated inflammation. Persistent ectopic donor cells were found in the lung only after intramyocardial injection, strengthening the improved safety of myoblast-sheet therapy. In addition, myoblast-sheet therapy enhanced cardiac function, corresponding to a 9.2-fold increase in donor cell survival, compared to intramyocardial injection. Both methods achieved reduced infarct size, decreased fibrosis, attenuated cardiomyocyte hypertrophy, and increased neovascular formation, in association with myocardial upregulation of a group of relevant molecules. The pattern of these beneficial changes was similar between two methods, but the degree was more substantial after myoblast-sheet therapy. CONCLUSION: The cell-sheet technique enhanced safety and therapeutic efficacy of myoblast-based therapy, compared to the current method, thereby paving the way for clinical application.


Subject(s)
Arrhythmias, Cardiac/prevention & control , Cell Culture Techniques/methods , Heart Failure/surgery , Myoblasts, Skeletal/transplantation , Myocytes, Cardiac/transplantation , Animals , Arrhythmias, Cardiac/physiopathology , Female , Heart Failure/physiopathology , Male , Myoblasts, Skeletal/physiology , Myocytes, Cardiac/physiology , Rats , Rats, Inbred Lew , Treatment Outcome
14.
Dev Dyn ; 241(10): 1584-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22911638

ABSTRACT

BACKGROUND: Deep sequencing of single cell-derived cDNAs offers novel insights into oncogenesis and embryogenesis. However, traditional library preparation for RNA-seq analysis requires multiple steps with consequent sample loss and stochastic variation at each step significantly affecting output. Thus, a simpler and better protocol is desirable. The recently developed hyperactive Tn5-mediated library preparation, which brings high quality libraries, is likely one of the solutions. RESULTS AND CONCLUSIONS: Here, we tested the applicability of hyperactive Tn5-mediated library preparation to deep sequencing of single cell cDNA, optimized the protocol, and compared it with the conventional method based on sonication. This new technique does not require any expensive or special equipment, which secures wider availability. A library was constructed from only 100 ng of cDNA, which enables the saving of precious specimens. Only a few steps of robust enzymatic reaction resulted in saved time, enabling more specimens to be prepared at once, and with a more reproducible size distribution among the different specimens. The obtained RNA-seq results were comparable to the conventional method. Thus, this Tn5-mediated preparation is applicable for anyone who aims to carry out deep sequencing for single cell cDNAs.


Subject(s)
DNA, Complementary/genetics , Gene Library , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods , Transposases , DNA Primers/genetics , Sonication/methods
15.
J Heart Lung Transplant ; 30(2): 227-33, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20971657

ABSTRACT

BACKGROUND: Intracoronary injection of bone marrow mononuclear cells (BMMNC) is a common clinical protocol of cell transplantation for heart disease, but poor engraftment of donor cells in the heart, which will limit its therapeutic efficacy, is a major issue. Initial "retention" (endothelial adherence and/or extravasation) of BMMNC immediately after intracoronary injection is a key step toward successful engraftment; however, this event has not been fully characterized. The aim of this study is to quantitatively clarify the frequency of "retention" of BMMNC after intracoronary injection, determine the impact of prior induction of ischemia-reperfusion injury on "retention" efficiency, and elucidate the underlying mechanisms focusing on adhesion molecule-mediated cell-cell interactions. METHODS: One million BMMNC collected from green fluorescent protein (GFP)-transgenic mice were injected into the coronary arteries of syngeneic wild-type mouse hearts under Langendorff perfusion. Retention efficiency was quantitatively estimated from the GFP-positive cell number flushed out into the coronary effluent. RESULTS: Whereas only 13.3 ± 1.2% of injected BMMNC were retained into normal hearts, prior induction of 30-minute ischemia and 30-minute reperfusion increased the retention efficiency to 36.5 ± 1.6% (p < 0.05, n = 8). Immunoconfocal observation further confirmed this enhanced retention after ischemia-reperfusion. Noticeably, the enhanced retention efficiency after ischemia-reperfusion treatment was diminished by administration of anti-P-selectin antibody (8.3 ± 0.8%, p < 0.05), but was not affected by inhibiting intercellular adhesion molecule-1 (39.6 ± 3.3%) or vascular cell adhesion molecule-1 (43.9 ± 2.9%). CONCLUSIONS: Retention efficiency of intracoronary-injected BMMNC was poor in a model of isolated, crystalloid-perfused murine hearts. An antecedent period of global ischemia-reperfusion increased the retention via P-selectin-dependent BMMNC-endothelial interaction.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation/methods , Cell Communication/physiology , Coronary Vessels/pathology , Endothelium, Vascular/pathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/therapy , Animals , Antigens, Ly/metabolism , Bone Marrow Cells/physiology , CD18 Antigens/metabolism , Coronary Vessels/physiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Myocardial Reperfusion Injury/physiopathology , P-Selectin/metabolism , Regional Blood Flow/physiology , Vascular Cell Adhesion Molecule-1/metabolism
16.
J Mol Cell Cardiol ; 47(2): 288-95, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19467239

ABSTRACT

Cell transplantation is an emerging therapy for treating post-infarction heart failure. Although the paracrine effect has been proposed to be an important mechanism for the therapeutic benefits, details remain largely unknown. This study compared various aspects of the paracrine effect after transplantation of either bone marrow mononuclear cells (BMC) or skeletal myoblasts (SMB) into the post-infarction chronically failing heart. Three weeks after left coronary artery ligation, adult rats received intramyocardial injection of either BMC, SMB or PBS only. Echocardiography demonstrated that injection of either cell type improved cardiac function compared to PBS injection. Interestingly, BMC injection markedly improved neovascularization in the border areas surrounding infarcts, while SMB injection decreased fibrosis in both the border and remote areas. Injection of either cell type similarly reduced hypertrophy of cardiomyocytes as assessed by cell-size planimetry using isolated cardiomyocytes. Quantitative RT-PCR revealed that, among 15 candidate mediators of paracrine effects studied, Fgf2 and Hgf were upregulated only after BMC injection, while Mmp2 and Timp4 were modulated after SMB injection. Additional investigations of signalling pathways relevant to heart failure by western blotting showed that p38 and STAT3 were temporarily activated after BMC injection, in contrast, ERK1/2 and JNK were activated after SMB injection. There was no difference in activation of Akt, PKD or Smad3 among groups. These data suggest that paracrine effects observed after cell transplantation in post-infarction heart failure were noticeably different between cell types in terms of mediators, signal transductions and consequent effects.


Subject(s)
Bone Marrow Transplantation , Heart Failure/etiology , Heart Failure/therapy , Myocardial Infarction/complications , Myocardial Infarction/therapy , Organ Specificity , Paracrine Communication , Animals , Heart Failure/pathology , Heart Failure/physiopathology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Paracrine Communication/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Systole/physiology , Ventricular Function, Left
17.
Circulation ; 118(14 Suppl): S106-14, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18824741

ABSTRACT

BACKGROUND: Inflammation plays an important role in the progress of adverse ventricular remodeling after myocardial infarction. High-mobility group box 1 (HMGB1) is a nuclear protein, which has recently been uncovered to also act as a modifier of inflammation when released. We hypothesized that HMGB1 injection could preferentially modulate local myocardial inflammation, attenuate ventricular remodeling, and subsequently improve cardiac performance of postinfarction chronic heart failure. METHODS AND RESULTS: Three weeks after left coronary artery ligation, HMGB1 (2.5 mug) or PBS was intramyocardially injected into rat hearts. At 28 days after injection, left ventricular ejection fraction was significantly improved after HMGB1 injection compared to PBS (39.3+/-1.4 versus 33.3+/-1.8%; P<0.01). Accumulation of CD45(+) inflammatory cells, two thirds of which were OX62(+) dendritic cells, in the peri-infarct area was significantly attenuated by HMGB1 injection. Dramatic changes in the expression of major proinflammatory cytokines were not detected by microarray or RT-PCR. Adverse ventricular remodeling including cardiomyocyte hypertrophy (cardiomyocyte cross-sectional area; 439+/-7 versus 458+/-6 mum(2); P<0.05) and extracellular collagen deposition (collagen volume fraction; 11.9+/-0.4 versus 15.2+/-0.6%; P<0.01) was attenuated by HMGB1 injection. Analyses of signal transduction pathways revealed that HMGB1 injection activated ERK1/2, but not p38, Akt, and Smad3. Cardiac regeneration and neovascularization were not observed. CONCLUSIONS: HMGB1 injection modulated the local inflammation in the postinfarction chronically failing myocardium, particularly via reducing the accumulation of dendritic cells. This modulated inflammation resulted in attenuated fibrosis and cardiomyocyte hypertrophy, which thereby improved global cardiac function. These data suggest that HMGB1 may be valuable for the chronic heart failure treatment.


Subject(s)
HMGB1 Protein/administration & dosage , Heart Failure/etiology , Myocardial Infarction/complications , Myocarditis/etiology , Myocarditis/pathology , Animals , Chronic Disease , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HMGB1 Protein/pharmacology , Heart/drug effects , Heart/physiopathology , Heart Failure/physiopathology , Hypertrophy , Injections , Myocardium/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Recovery of Function , Stroke Volume/drug effects , Ventricular Remodeling
18.
Circulation ; 118(14 Suppl): S138-44, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18824745

ABSTRACT

BACKGROUND: Arrhythmia occurrence is a variable but serious concern of cell therapy for treating heart failure. Using a rat postinfarction chronic heart failure model, we compared skeletal myoblast (SMB) with bone marrow cell (BMC) injection to highlight donor cell-specific, late-phase arrhythmogenesis and the underlying factors. METHODS AND RESULTS: SMBs or BMCs derived from male GFP-transgenic rats, or PBS were injected intramyocardially into female rat hearts 3 weeks after coronary artery occlusion. At 28 days after injection, echocardiography showed that the left ventricular ejection fraction was significantly improved in both the SMB and BMC groups, compared to PBS control despite poor graft survival as assessed by PCR for the male-specific gene. Radio-telemetry analysis revealed that the SMB group displayed a higher occurrence of ventricular premature contractions with an elongation of the QRS complex and the hearts were more susceptible to isopreterenol-induced ventricular tachycardia compared to the BMC and PBS groups. Western blot and immunoconfocal analysis showed that the gap junction protein, connexin43, was widely and persistently decreased in the SMB group compared to the other groups. IL-1beta was shown to be upregulated in hearts after SMB injection, and in vitro experiments demonstrated that exposure to IL-1beta caused a decrease in connexin43 and intercellular communication in cultured cardiomyocytes. CONCLUSIONS: Although cell therapy was capable of improving function of the postinfarction chronically failing heart, there was late-phase arrhythmogenicity specific to donor cell type. Global downregulation of connexin43 in the host myocardium was indicated to be an important factor underlying late-phase arrhythmogenicity after SMB transplantation.


Subject(s)
Arrhythmias, Cardiac/etiology , Connexin 43/metabolism , Coronary Occlusion/surgery , Myoblasts, Skeletal/transplantation , Myocardium/metabolism , Postoperative Complications , Animals , Animals, Genetically Modified , Bone Marrow Transplantation/methods , Cell Communication , Cells, Cultured , Coronary Occlusion/diagnostic imaging , Coronary Occlusion/physiopathology , Down-Regulation , Echocardiography , Electrocardiography , Female , Graft Survival , Injections , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats , Rats, Sprague-Dawley , Stroke Volume , Up-Regulation , Ventricular Premature Complexes/etiology
19.
PLoS One ; 3(8): e3071, 2008 Aug 27.
Article in English | MEDLINE | ID: mdl-18728781

ABSTRACT

BACKGROUND: Intramyocardial injection of skeletal myoblasts (SMB) has been shown to be a promising strategy for treating post-infarction chronic heart failure. However, insufficient therapeutic benefit and occurrence of ventricular arrhythmias are concerns. We hypothesised that the use of a retrograde intracoronary route for SMB-delivery might favourably alter the behaviour of the grafted SMB, consequently modulating the therapeutic effects and arrhythmogenicity. METHODS AND RESULTS: Three weeks after coronary artery ligation in female wild-type rats, 5x10(6) GFP-expressing SMB or PBS only (control) were injected via either the intramyocardial or retrograde intracoronary routes. Injection of SMB via either route similarly improved cardiac performance and physical activity, associated with reduced cardiomyocyte-hypertrophy and fibrosis. Grafted SMB via either route were only present in low numbers in the myocardium, analysed by real-time PCR for the Y-chromosome specific gene, Sry. Cardiomyogenic differentiation of grafted SMB was extremely rare. Continuous ECG monitoring by telemetry revealed that only intramyocardial injection of SMB produced spontaneous ventricular tachycardia up to 14 days, associated with local myocardial heterogeneity generated by clusters of injected SMB and accumulated inflammatory cells. A small number of ventricular premature contractions with latent ventricular tachycardia were detected in the late-phase of SMB injection regardless of the injection-route. CONCLUSION: Retrograde intracoronary injection of SMB provided significant therapeutic benefits with attenuated early-phase arrhythmogenicity in treating ischaemic cardiomyopathy, indicating the promising utility of this route for SMB-delivery. Late-phase arrhythmogenicity remains a concern, regardless of the delivery route.


Subject(s)
Heart Failure/etiology , Heart Failure/surgery , Myoblasts/transplantation , Myocardial Infarction/complications , Animals , Cell Transplantation/methods , Disease Models, Animal , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Heart Function Tests , Rats , Rats, Sprague-Dawley
20.
Biochem Biophys Res Commun ; 371(4): 615-20, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18413147

ABSTRACT

Side population cells have been found in various types of adult tissue including heart and are presumed to be tissue-specific stem/progenitor cells. In the present study, we confirmed the presence of cardiac side population (cSP) cells, which showed both the Hoechst 33342 efflux ability and ABCG2 expression, in adult murine heart. Flow cytometric analysis showed that more than half of cSP cells expressed the endothelial marker VE-cadherin or the smooth muscle markers, alpha-smooth muscle actin and desmin. In addition, immunohistochemical analysis demonstrated that ABCG2(+) cells were mainly localized within vascular walls. Quantitative RT-PCR analysis demonstrated that VE-cadherin(-) cSP cells progressively expressed Nkx2.5 and cardiac troponin T with time in culture. VE-cadherin(-) cSP cells also expressed mesodermal-mesenchymal-associated markers and differentiated into osteocytes and adipocytes. These results highlight the heterogeneic nature of cSP cells, consisting of vascular endothelial cells, smooth muscle cells, and mesenchymal stem/progenitor cells including potential cardiomyogenic cells.


Subject(s)
Cell Differentiation , Heart , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Actins/analysis , Actins/genetics , Actins/metabolism , Animals , Benzimidazoles/metabolism , Cadherins/analysis , Cadherins/genetics , Cadherins/metabolism , Cell Separation , Cells, Cultured , Desmin/analysis , Desmin/genetics , Desmin/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myoblasts, Cardiac/chemistry , Transcription Factors/analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Troponin T/analysis , Troponin T/genetics , Troponin T/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...