Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Theranostics ; 8(10): 2752-2764, 2018.
Article in English | MEDLINE | ID: mdl-29774073

ABSTRACT

Background: The post-myocardial infarction (MI) scar interrupts electrical impulse propagation and delays regional contraction, which contributes to ventricular dysfunction. We investigated the potential of an injectable conductive biomaterial to restore scar tissue conductivity and re-establish synchronous ventricular contraction. Methods: A conductive biomaterial was generated by conjugating conductive polypyrrole (PPY) onto chitosan (CHI) backbones. Trypan blue staining of neonatal rat cardiomyocytes (CMs) cultured on biomaterials was used to evaluate the biocompatibility of the conductive biomaterials. Ca2+ imaging was used to visualize beating CMs. A cryoablation injury rat model was used to investigate the ability of PPY:CHI to improve cardiac electrical propagation in the injured heart in vivo. Electromyography was used to evaluate conductivity of scar tissue ex vivo. Results: Cell survival and morphology were similar between cells cultured on biomaterials-coated and uncoated-control dishes. PPY:CHI established synchronous contraction of two distinct clusters of spontaneously-beating CMs. Intramyocardial PPY:CHI injection into the cryoablation-induced injured region improved electrical impulse propagation across the scarred tissue and decreased the QRS interval, whereas saline- or CHI-injected hearts continued to have delayed propagation patterns and significantly reduced conduction velocity compared to healthy controls. Ex vivo evaluation found that scar tissue from PPY:CHI-treated rat hearts had higher signal amplitude compared to those from saline- or CHI-treated rat heart tissue. Conclusions: The PPY:CHI biomaterial is electrically conductive, biocompatible and injectable. It improved synchronous contraction between physically separated beating CM clusters in vitro. Intra-myocardial injection of PPY:CHI following cardiac injury improved electrical impulse propagation of scar tissue in vivo.


Subject(s)
Action Potentials , Biocompatible Materials/chemistry , Electric Conductivity , Hydrogels/chemistry , Myocardial Contraction , Myocardial Infarction/therapy , Myocytes, Cardiac/physiology , Animals , Biocompatible Materials/therapeutic use , Cells, Cultured , Chitosan/analogs & derivatives , Female , Hydrogels/therapeutic use , Pyrroles/chemistry , Rats , Rats, Sprague-Dawley
2.
J Mol Cell Cardiol ; 94: 32-42, 2016 05.
Article in English | MEDLINE | ID: mdl-26996757

ABSTRACT

BACKGROUND: Mast cells (MCs) dynamically participate in wound healing after myocardial infarction (MI) by releasing cytokines. Indeed, MC-deficient mice undergo rapid left ventricular dilation post-MI. Mesenchymal stem cells (MSCs) are recruited to the injured region following an MI and have potential for cardiac repair. In the current study, we evaluate the effect of MCs on MSC proliferation and myogenic differentiation. METHODS AND RESULTS: MCs were cultured from mouse bone marrow and MC granulate (MCG) was extracted from MCs via freeze-thaw cycles followed by filtration. α-SMA (smooth muscle actin) expression was examined as an indicator of myogenic differentiation. MSC/MC co-culture resulted in decreased MSC differentiation indicated by α-SMA suppression in MSCs. MCG also suppressed α-SMA expression and increased MSC migration and proliferation in a dose-dependent manner. Removal of MCG rescued α-SMA expression and MSC differentiation. Platelet derived growth factor (PDGF) receptor blockade using AG1296 also rescued MSC differentiation even after MCG treatment. Real-time PCR and Western blot showed that MCG exerted its effects on MSCs via downregulation of miR-145 and miR-143, downregulation of myocardin, upregulation of Klf4, and increased Erk and Elk1 phosphorylation. CONCLUSIONS: MCs promote MSC proliferation and migration by suppressing their myogenic differentiation. MCs accomplish this via activation of the PDGF pathway, downregulation of miR-145/143, and modulation of the myocardin-Klf4 axis. These data suggest a potential role for MSC/MC interaction in the infarcted heart where MCs may inhibit MSCs from differentiation and promote their proliferation whereby increased cardiac MSC accumulation promotes eventual cardiac regeneration after MCs cease activity.


Subject(s)
Cell Differentiation , Mast Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Actins/genetics , Actins/metabolism , Animals , Biomarkers , Cell Movement , Cell Proliferation , Coculture Techniques , Cytoplasmic Granules/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Immunophenotyping , Kruppel-Like Factor 4 , Male , Mesenchymal Stem Cell Transplantation , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Regeneration , Signal Transduction , ets-Domain Protein Elk-1/metabolism
3.
Mol Imaging Biol ; 18(4): 557-68, 2016 08.
Article in English | MEDLINE | ID: mdl-26728161

ABSTRACT

PURPOSE: We generated a folate-conjugated porphyrin nanoparticle (porphysome) suitable for multimodal non-invasive active macrophage tracking post-myocardial infarction (MI). PROCEDURES: Macrophage uptake of folate-conjugated porphysomes was selective. Folate-porphysome cardiac macrophage tracking was detected in vivo using radioligand and fluorescent imaging. To track post-MI macrophage mobilization, cardiac fluorescence signal in folate-porphysome-injected mice was measured for 9 day post-MI. Active macrophage phenotype was assessed using immunohistochemistry. RESULTS: Heart active macrophage presence peaked on day 1, returned to baseline by day 3, and peaked again on day 7 post-MI. Macrophages were distributed throughout the left ventricle at day 1, but aggregated within scar tissue at day 7. Macrophage phenotype was pro-inflammatory (TNFα(+)) at day 1, whereas scar-resident macrophages expressed anti-inflammatory markers (IL-10, TGFß) at day 7. However, day 7 macrophages outside the scar expressed neither pro- nor anti-inflammatory markers. CONCLUSIONS: We established that folate-porphysomes are suitable for non-invasive imaging of macrophages and used it to investigate active macrophage behavior in the infarcted heart.


Subject(s)
Cell Tracking/methods , Heart/diagnostic imaging , Macrophages/metabolism , Myocardial Infarction/diagnosis , Myocardial Infarction/pathology , Nanoparticles/chemistry , Porphyrins/chemistry , Animals , Cellular Microenvironment , Folic Acid/chemistry , Macrophage Activation , Mice , Mice, Inbred C57BL , Phenotype , RAW 264.7 Cells
4.
J Mol Cell Cardiol ; 84: 116-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939780

ABSTRACT

Cell therapy to prevent cardiac dysfunction after myocardial infarction (MI) is less effective in aged patients because aged cells have decreased regenerative capacity. Allogeneic transplanted stem cells (SCs) from young donors are usually rejected. Maintaining transplanted SC immunoprivilege may dramatically improve regenerative outcomes. The uterus has distinct immune characteristics, and we showed that reparative uterine SCs home to the myocardium post-MI. Here, we identify immunoprivileged uterine SCs and assess their effects on cardiac regeneration after allogeneic transplantation. We found more than 20% of cells in the mouse uterus have undetectable MHC I expression by flow cytometry. Uterine MHC I((neg)) and MHC I((pos)) cells were separated by magnetic cell sorting. The MHC I((neg)) population expressed the SC markers CD34, Sca-1 and CD90, but did not express MHC II or c-kit. In vitro, MHC I((neg)) and ((pos)) SCs show colony formation and endothelial differentiation capacity. In mixed leukocyte co-culture, MHC I((neg)) cells showed reduced cell death and leukocyte proliferation compared to MHC I((pos)) cells. MHC I((neg)) and ((pos)) cells had significantly greater angiogenic capacity than mesenchymal stem cells. The benefits of intramyocardial injection of allogeneic MHC I((neg)) cells after MI were comparable to syngeneic bone marrow cell transplantation, with engraftment in cardiac tissue and limited recruitment of CD4 and CD8 cells up to 21 days post-MI. MHC I((neg)) cells preserved cardiac function, decreased infarct size and improved regeneration post-MI. This new source of immunoprivileged cells can induce neovascularization and could be used as allogeneic cell therapy for regenerative medicine.


Subject(s)
Heart/physiopathology , Regeneration , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/immunology , Uterus/cytology , Animals , Antigens, Ly/metabolism , Cell Survival/genetics , Cicatrix/complications , Cicatrix/pathology , Coculture Techniques , Extracellular Matrix/metabolism , Female , Gene Expression Regulation , Heart Function Tests , Histocompatibility Antigens Class I/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/pathology , Neovascularization, Physiologic/genetics , Transplantation, Homologous , Wound Healing/genetics
5.
Semin Thorac Cardiovasc Surg ; 26(1): 44-52, 2014.
Article in English | MEDLINE | ID: mdl-24952757

ABSTRACT

After an extensive myocardial infarction, restoration of heart function depends on the ability of the heart to promote regeneration and prevent adverse ventricular remodeling. Preclinical research demonstrated that the transplantation of healthy stem cells restored heart function, but the stem cells obtained from older animals or patients were not as efficacious as those from younger individuals. In this paper, we review the successes and limitations discovered in preclinical studies and clinical trials examining cell therapy for damaged hearts. After the modest successes of the early clinical trials, research is now exploring the benefits of enhanced stem cell therapy. Cell based gene therapy markedly improves the angiogenesis achieved. Rejuvenating aged stems cells prior to transplantation restores the functional benefits attained. Transplanting healthy allogeneic stem cells from young donors into aged individuals can restore function if rejection can be prevented. Finally, modulating the cellular environment in aged individuals permits the full functional benefits of stem cell therapy to be realized. Significant challenges remain, but these approaches show promise that cell therapy may become routine therapy to improve functional recovery of older patients after an extensive myocardial infarction.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Heart Diseases/therapy , Stem Cell Transplantation/methods , Animals , Heart Diseases/physiopathology , Humans , Treatment Outcome , Ventricular Remodeling
6.
Arterioscler Thromb Vasc Biol ; 34(2): 321-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285579

ABSTRACT

OBJECTIVE: Transgenic overexpression of the human cysteinyl leukotriene receptor 2 (CysLT2R) in murine endothelium exacerbates vascular permeability and ischemia/reperfusion injury. Here, we explore the underlying mechanisms of CysLT2R activation-mediated inflammation and delineate the relative contributions of endogenous murine CysLT2R and the transgene-derived receptor. APPROACH AND RESULTS: We created a novel mouse with only endothelial-expressed CysLT2R (endothelium-targeted overexpression mice [EC]/CysLT2R-knockout mice [KO]) by crossing EC with KO to dissect the role of endothelial CysLT2R in tissue injury. Surprisingly, we discovered that damage in EC/KO mice was not elevated (24% versus 47% EC) after ischemia/reperfusion. We examined vascular permeability and leukocyte recruitment/rolling responses in the cremaster vasculature after cysteinyl leukotriene (cysLT) stimulation. Mice possessing transgenic endothelial CysLT2R overexpression, whether EC or EC/KO, when stimulated with cysLTs, exhibited vascular hyperpermeability, declining leukocyte flux, and a transient increase in slow-rolling leukocyte fraction. Mice lacking endogenous CysLT2R (both KO [20 ± 3 cells/min] EC/KO [24 ± 3]) showed lower-rolling leukocyte flux versus wild-type (38 ± 6) and EC (35 ± 6) mice under unstimulated conditions. EC/KO mice differed from EC counterparts in that vascular hyperpermeability was not present in the absence of exogenous cysLTs. CONCLUSIONS: These results indicate that endothelial and nonendothelial CysLT2R niches have separate roles in mediating inflammatory responses. Endothelial receptor activation results in increased vascular permeability and leukocyte slow-rolling, facilitating leukocyte transmigration. Nonendothelial receptors, likely located on resident/circulating leukocytes, facilitate endothelial receptor activation and leukocyte transit. Activation of both receptor populations is required for injury exacerbation.


Subject(s)
Endothelial Cells/metabolism , Leukocytes/metabolism , Muscle, Skeletal/blood supply , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Receptors, Leukotriene/deficiency , Receptors, Leukotriene/metabolism , Animals , Capillary Permeability , Cysteine/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Humans , Leukocyte Rolling , Leukocytes/drug effects , Leukocytes/immunology , Leukotrienes/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardium/immunology , Myocardium/pathology , Receptors, Leukotriene/agonists , Receptors, Leukotriene/genetics , Time Factors
7.
FASEB J ; 26(3): 1100-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22131271

ABSTRACT

Leukotrienes have been implicated in the pathogenesis of degenerative diabetic retinopathy, with research focusing primarily on leukotriene B(4), with little attention devoted to the cysteinyl leukotrienes (cysLTs), which act through cysLT receptors (CysLT(1)R and CysLT(2)R). We demonstrate here the presence of CysLT(2)R in pericytes and endothelial cells of superficial retinal vasculature using an indirect assay by assessment of ß-galactosidase expression in CysLT(2)R-knockout (KO) mice. Retinal damage was induced in KO and wild-type (WT) mice using an established oxygen-induced retinopathy (OIR) model. CysLT(2)R expression following OIR was intensely up-regulated compared to sham-treated controls. Staining with Griffonia simplicifolia lectin revealed enhanced tissue damage (as assessed by vasoobliteration/vasoproliferation) in KO mice compared to WT controls, yet the opposite was true with respect to retinal edema. However, vascular endothelial growth factor receptor 1 (VEGFR1) transcripts were increased by OIR similarly with respect to genotype. Intravitreal application of exogenous cysLTs elicited greater vasculature leakage (assessed ex vivo) in eyes from WT mice compared to KO mice. While mRNA encoding enzymes for various components of the leukotriene cascade were detected in sham- and OIR-treated retinas, only prostaglandins and hydroxyeicosatetraenoic acids, but not leukotrienes, were detected in A23187-treated retina preparations. Together, these results implicate the CysLT(2)R in the progression of ischemic retinopathy.


Subject(s)
Disease Models, Animal , Papilledema/genetics , Receptors, Leukotriene/genetics , Retinal Diseases/genetics , Retinal Neovascularization/genetics , Albumins/metabolism , Animals , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Capillary Permeability/drug effects , Cysteine/pharmacology , Endothelium, Vascular/metabolism , Gene Expression , Hydroxyeicosatetraenoic Acids/metabolism , Immunohistochemistry , Leukotrienes/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxygen , Papilledema/metabolism , Pericytes/metabolism , Prostaglandins/metabolism , Receptors, Leukotriene/deficiency , Retina/drug effects , Retina/metabolism , Retinal Diseases/chemically induced , Retinal Diseases/metabolism , Retinal Neovascularization/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
8.
J Pharmacol Exp Ther ; 339(3): 768-78, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21903747

ABSTRACT

Cysteinyl leukotrienes (CysLTs) are potent inflammatory mediators that predominantly exert their effects by binding to cysteinyl leukotriene receptors of the G protein-coupled receptor family. CysLT receptor 2 (CysLT(2)R), expressed in endothelial cells of some vascular beds, has been implicated in a variety of cardiovascular functions. Endothelium-specific overexpression of human CysLT(2)R in transgenic mice (hEC-CysLT(2)R) greatly increases myocardial infarction damage. Investigation of this receptor, however, has been hindered by the lack of selective pharmacological antagonists. Here, we describe the characterization of 3-(((3-carboxycyclohexyl)amino)carbonyl)-4-(3-(4-(4-phenoxybutoxy)phenyl)-propoxy)benzoic acid (BayCysLT(2)) and explore the selective effects of this compound in attenuating myocardial ischemia/reperfusion damage and vascular leakage. Using a recently developed ß-galactosidase-ß-arrestin complementation assay for CysLT(2)R activity (Mol Pharmacol 79:270-278, 2011), we determined BayCysLT(2) to be ∼20-fold more potent than the nonselective dual CysLT receptor 1 (CysLT(1)R)/CysLT(2)R antagonist 4-(((1R,2E,4E,6Z,9Z)-1-((1S)-4-carboxy-1-hydroxybutyl)-2,4,6,9-pentadecatetraen-1-yl)thio)benzoic acid (Bay-u9773) (IC(50) 274 nM versus 4.6 µM, respectively). Intracellular calcium mobilization in response to cysteinyl leukotriene administration showed that BayCysLT(2) was >500-fold more selective for CysLT(2)R compared with CysLT(1)R. Intraperitoneal injection of BayCysLT(2) in mice significantly attenuated leukotriene D(4)-induced Evans blue dye leakage in the murine ear vasculature. BayCysLT(2) administration either before or after ischemia/reperfusion attenuated the aforementioned increased myocardial infarction damage in hEC-CysLT(2)R mice. Finally, decreased neutrophil infiltration and leukocyte adhesion molecule mRNA expression were observed in mice treated with antagonist compared with untreated controls. In conclusion, we present the characterization of a potent and selective antagonist for CysLT(2)R that is useful for discerning biological activities of this receptor.


Subject(s)
Capillary Permeability/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Leukotriene Antagonists/pharmacology , Leukotriene D4/antagonists & inhibitors , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Phthalic Acids/pharmacology , Receptors, Leukotriene/metabolism , SRS-A/analogs & derivatives , Animals , Arrestins/analysis , Disease Models, Animal , Drug Evaluation, Preclinical , Ear/blood supply , Humans , Mice , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Peroxidase/metabolism , SRS-A/pharmacology , beta-Arrestins , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...