Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Can J Physiol Pharmacol ; 96(3): 304-307, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28926719

ABSTRACT

Doxorubicin (Dox) is an effective anticancer drug. Unfortunately, it causes cardiac and muscle toxicity due to increased oxidative stress and inflammation; however, it remains unknown whether Dox induces "pyroptosis" - an inflammation-mediated cell death. We investigated whether Dox induces pyroptosis in mouse soleus muscle (Sol 8) cells in vitro and to show the protective effect of embryonic stem cell exosomes (ES-exos) on pyroptosis. Dox and inflammation-induced in vitro model was generated. Pyroptosis was confirmed using immunohistochemistry (with putative markers caspase-1, IL-1ß, and pro-inflammatory cytokine IL-18) and Western blotting of caspase-1 and IL-1ß. The results show significant increase in the expression of caspase-1, IL-1ß, and IL-18 following treatment with Dox, which was inhibited by ES-exos but not mouse embryonic fibroblast exosomes. Moreover, GW4869 compound inhibited functional activity of ES-exos, suggesting these vesicles are key players in the inhibition of pyroptosis. These results suggest that Dox induces inflammatory pyroptosis in Sol 8 cells, which is attenuated by ES-exos in vitro.


Subject(s)
Doxorubicin/adverse effects , Embryonic Stem Cells/cytology , Exosomes/metabolism , Muscle Cells/cytology , Muscle Cells/drug effects , Pyroptosis/drug effects , Animals , Cell Line , Inflammation/chemically induced , Inflammation/pathology , Mice
2.
Can J Physiol Pharmacol ; 95(3): 288-294, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28238274

ABSTRACT

The current study investigates whether inhibiting the Notch-1 signaling pathway in primary human monocytes enhances M2 macrophage differentiation. We generated a primary human monocyte cell culture model to understand the effect of the Notch-1 signaling pathway. Monocytes were treated with Notch-1 inhibitors DAPT or siRNA. Our data show that there was a significant increase in the M1 macrophage population demonstrated by iNOS marker in the primary human monocytes treated with apoptotic-conditioned medium (ACM). Next, the levels of pro-inflammatory cytokines IL-6 and MCP-1, as well as TNF-α, increased in ACM media (p < 0.05). Furthermore, M1 macrophages and pro-inflammatory cytokines were reduced following DAPT or siRNA treatment. Comparatively, there was a significant increase in M2 macrophages, as demonstrated by an increase in CD206 and arginase-1 positive cells treated with DAPT or siRNA (p < 0.05). Furthermore, a significant increase in the associated anti-inflammatory cytokines IL-10 and IL-1RA was also observed with respect to control groups (p < 0.05). We conclude that blocking the Notch-1 pathway with DAPT or siRNA attenuates pro-inflammatory cytokines, enhances M2 macrophage differentiation, and increases anti-inflammatory cytokines in primary human monocytes. As a result, Notch-1 pathway inhibition has potential therapeutic applications of inflammatory disease.


Subject(s)
Cell Differentiation , Macrophages/metabolism , Monocytes/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Apoptosis , Arginase/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/metabolism , Diamines/pharmacology , Humans , Inflammation Mediators/metabolism , Lectins, C-Type/metabolism , Macrophages/drug effects , Mannose Receptor , Mannose-Binding Lectins/metabolism , Monocytes/drug effects , Monocytes/pathology , Nitric Oxide Synthase Type II/metabolism , Phenotype , Primary Cell Culture , RNA Interference , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Thiazoles/pharmacology , Transfection
3.
Mol Cell Biochem ; 425(1-2): 77-84, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27804049

ABSTRACT

Fibroblast growth factors (FGFs) comprise a large family of signaling molecules that involve cell patterning, mobilization, differentiation, and proliferation. Various FGFs, including FGF-1, FGF-2, and FGF-5, have been shown to play a role in cytoprotection during adverse cardiac events; however, whether FGF-8 is a cytoprotective remains unclear. The current study was designed to evaluate the effect of FGF-8 treatment on oxidative stress-induced apoptosis in H9c2 cells. Cells were divided into three groups: control, H2O2 (400 µm H2O2), and H2O2 + FGF-8 (4 ng/ml FGF-8). Our results suggest apoptosis was significantly (p < 0.05) enhanced in the H2O2 group relative to control. Moreover, a significant (p < 0.05) decline in apoptosis was observed in the H2O2 + FGF-8 group compared to H2O2-treated cells as evidenced by TUNEL staining, a cell death detection ELISA, and cell viability. Levels of downstream apoptotic mediators, caspase-3 and caspase-9, were significantly (p < 0.05) upregulated following H2O2 treatment but were abrogated following FGF-8 application. Expression levels of Forkhead box protein O1 (FoxO-1), MnSOD, catalase, pAKT, and p-mTOR were significantly (p < 0.05) reduced in the H2O2 group (p < 0.05). Notably, these levels were significantly (p < 0.05) reversed following FGF-8 treatment. Our data, for the first time, suggest FGF-8 is an anti-apoptotic mediator in oxidative-stressed H9c2 cells. Furthermore, our data demonstrate that apoptotic inhibition by FGF-8 is consequent to FoxO-1 oxidative detoxification as well as augmentation to the PI3K/AKT cell survival pathway.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factor 8/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Cell Line , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Nerve Tissue Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats
4.
PLoS One ; 11(1): e0147897, 2016.
Article in English | MEDLINE | ID: mdl-26824441

ABSTRACT

Inflammation plays a fundamental role in the inception and development of atherosclerosis (ATH). Mechanisms of inflammation include the infiltration of monocytes into the injured area and subsequent differentiation into either pro-inflammatory M1 macrophages or anti-inflammatory M2 macrophages. We have previously published data suggesting bone morphogenetic protein-7 (BMP-7) enhances M2 macrophage differentiation and anti-inflammatory cytokine secretion in vitro. In this regard, we hypothesized BMP-7 would inhibit plaque formation in an animal model of ATH through monocytic plasticity mediation. ATH was generated in male and female Apo E(-/-) mice via partial left carotid artery (PLCA) ligation and mice were divided into 3 groups: Sham, PLCA, and PLCA+BMP-7 (200 ug/kg; i.v.). Our data suggest that BMP-7 inhibits plaque formation and increases arterial systolic velocity. Furthermore, we report inhibition of monocyte infiltration and a decrease in associated pro-inflammatory cytokines (MCP-1, TNF-α, and IL-6) in the PLCA+BMP-7 mice. In contrast, our data suggest a significant (p<0.05) increase in M2 macrophage populations with consequential enhanced anti-inflammatory cytokine (IL-1RA, IL-10, and Arginase 1) expression following BMP-7 treatment. We have also observed that mechanisms promoting monocyte into M2 macrophage differentiation by BMP-7 involve the upregulation and activation of the BMP-7 receptor (BMP-7RII). In conclusion, we report that BMP-7 has the potential to mediate cellular plasticity and mitigate the inflammatory immune response, which results in decreased plaque formation and improved blood velocity.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Bone Morphogenetic Protein 7/pharmacology , Macrophages/drug effects , Monocytes/drug effects , Plaque, Atherosclerotic/drug therapy , Protective Agents/pharmacology , Animals , Apolipoproteins E/deficiency , Arginase/genetics , Arginase/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blood Flow Velocity/drug effects , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Carotid Arteries/pathology , Carotid Arteries/surgery , Cell Differentiation , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Ligation , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Methods Mol Biol ; 1299: 145-52, 2015.
Article in English | MEDLINE | ID: mdl-25836581

ABSTRACT

Embryonic stem (ES) cells are pluripotent stem cells capable of self-renewal and have broad differentiation potential yielding cell types from all three germ layers. In the absence of differentiation inhibitory factors, when cultured in suspension, ES cells spontaneously differentiate and form three-dimensional cell aggregates termed embryoid bodies (EBs). Although various methods exist for the generation of EBs, the hanging drop method offers reproducibility and homogeneity from a predetermined number of ES cells. Herein, we describe the in vitro differentiation of mouse embryonic stem cells into cardiac myocytes using the hanging drop method and immunocytochemistry to identify cardiomyogenic differentiation. In brief, ES cells, placed in droplets on the lid of culture dishes following a 2-day incubation, yield embryoid bodies, which are resuspended and plated. 1-2 weeks following plating of the EBs, spontaneous beating areas can be observed and staining for specific cardiac markers can be achieved.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Biomarkers , Cells, Cultured , Embryoid Bodies , Immunohistochemistry , Mice , Myocytes, Cardiac/metabolism
6.
PLoS One ; 10(3): e0120739, 2015.
Article in English | MEDLINE | ID: mdl-25768089

ABSTRACT

Inflammation has been implicated as a perpetrator of diabetes and its associated complications. Monocytes, key mediators of inflammation, differentiate into pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages upon infiltration of damaged tissue. However, the inflammatory cell types, which propagate diabetes progression and consequential adverse disorders, remain unclear. The current study was undertaken to assess monocyte infiltration and the role of fibroblast growth factor-9 (FGF-9) on monocyte to macrophage differentiation and cardioprotection in the diabetic infarcted heart. Db/db diabetic mice were assigned to sham, myocardial infarction (MI), and MI+FGF-9 groups. MI was induced by permanent coronary artery ligation and animals were subjected to 2D transthoracic echocardiography two weeks post-surgery. Immunohistochemical and immunoassay results from heart samples collected suggest significantly increased infiltration of monocytes (Mean ± SEM; MI: 2.02% ± 0.23% vs. Sham 0.75% ± 0.07%; p<0.05) and associated pro-inflammatory cytokines (TNF-α, MCP-1, and IL-6), adverse cardiac remodeling (Mean ± SEM; MI: 33% ± 3.04% vs. Sham 2.2% ± 0.33%; p<0.05), and left ventricular dysfunction (Mean ± SEM; MI: 35.4% ± 1.25% vs. Sham 49.19% ± 1.07%; p<0.05) in the MI group. Importantly, treatment of diabetic infarcted myocardium with FGF-9 resulted in significantly decreased monocyte infiltration (Mean ± SEM; MI+FGF-9: 1.39% ± 0.1% vs. MI: 2.02% ± 0.23%; p<0.05), increased M2 macrophage differentiation (Mean ± SEM; MI+FGF-9: 4.82% ± 0.86% vs. MI: 0.85% ± 0.3%; p<0.05) and associated anti-inflammatory cytokines (IL-10 and IL-1RA), reduced adverse remodeling (Mean ± SEM; MI+FGF-9: 11.59% ± 1.2% vs. MI: 33% ± 3.04%; p<0.05), and improved cardiac function (Fractional shortening, Mean ± SEM; MI+FGF-9: 41.51% ± 1.68% vs. MI: 35.4% ± 1.25%; p<0.05). In conclusion, our data suggest FGF-9 possesses novel therapeutic potential in its ability to mediate monocyte to M2 differentiation and confer cardiac protection in the post-MI diabetic heart.


Subject(s)
Cell Differentiation/physiology , Diabetes Complications/physiopathology , Fibroblast Growth Factor 9/metabolism , Monocytes/physiology , Myocardial Infarction/physiopathology , Analysis of Variance , Animals , Atrial Remodeling/physiology , Cytokines , DNA Primers/genetics , Echocardiography , Enzyme-Linked Immunosorbent Assay , Heart Function Tests , Immunohistochemistry , Macrophages/cytology , Mice , Mice, Inbred NOD , Myocardial Infarction/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
Am J Physiol Heart Circ Physiol ; 307(11): H1634-42, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25260616

ABSTRACT

Macrophage polarization is emerging as an important area of research for the development of novel therapeutics to treat inflammatory diseases. Within the current study, the role of Notch1R in macrophage differentiation was investigated as well as downstream effects in THP-1 monocytes cultured in "inflammation mimicry" media. Interference of Notch signaling was achieved using either the pharmaceutical inhibitor DAPT or Notch1R small interfering RNA (siRNA), and Notch1R expression, macrophage phenotypes, and anti- and proinflammatory cytokine expression were evaluated. Data presented show that Notch1R expression on M1 macrophages as well as M1 macrophage differentiation is significantly elevated during cellular stress (P < 0.05). However, under identical culture conditions, interference to Notch signaling via Notch1R inhibition mitigated these results as well as promoted M2 macrophage differentiation. Moreover, when subjected to cellular stress, macrophage secretion of proinflammatory cytokines was significantly heightened (P < 0.05). Importantly, Notch1R inhibition not only diminished proinflammatory cytokine secretion but also enhanced anti-inflammatory protein release (P < 0.05). Our data suggest that Notch1R plays a pivotal role in M1 macrophage differentiation and heightened inflammatory responses. Therefore, we conclude that inhibition of Notch1R and subsequent downstream signaling enhances monocyte to M2 polarized macrophage outcomes and promotes anti-inflammatory mediation during cellular stress.


Subject(s)
Macrophages/physiology , Monocytes/physiology , Receptor, Notch1/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cell Polarity/physiology , Culture Media, Conditioned , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/drug effects , Monocytes/drug effects , RNA, Small Interfering/genetics , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction/drug effects
8.
PLoS One ; 8(9): e75580, 2013.
Article in English | MEDLINE | ID: mdl-24086577

ABSTRACT

Thymosin ß4 (Tß4), a small G-actin sequestering peptide, mediates cell proliferation, migration, and angiogenesis. Whether embryonic stem (ES) cells, overexpressing Tß4, readily differentiate into cardiac myocytes in vitro and in vivo and enhance cardioprotection following transplantation post myocardial infarction (MI) remains unknown. Accordingly, we established stable mouse ES cell lines, RFP-ESCs and Tß4-ESCs, expressing RFP and an RFP-Tß4 fusion protein, respectively. In vitro, the number of spontaneously beating embryoid bodies (EBs) was significantly increased in Tß4-ESCs at day 9, 12 and 15, compared with RFP-ESCs. Enhanced expression of cardiac transcriptional factors GATA-4, Mef2c and Txb6 in Tß4-EBs, as confirmed with real time-PCR analysis, was accompanied by the increased number of EB areas stained positive for sarcomeric α-actin in Tß4-EBs, compared with the RFP control, suggesting a significant increase in functional cardiac myocytes. Furthermore, we transplanted Tß4-ESCs into the infarcted mouse heart and performed morphological and functional analysis 2 weeks after MI. There was a significant increase in newly formed cardiac myocytes associated with the Notch pathway, a decrease in apoptotic nuclei mediated by an increase in Akt and a decrease in levels of PTEN. Cardiac fibrosis was significantly reduced, and left ventricular function was significantly augmented in the Tß4-ESC transplanted group, compared with controls. It is concluded that genetically modified Tß4-ESCs, potentiates their ability to turn into cardiac myocytes in vitro as well as in vivo. Moreover, we also demonstrate that there was a significant decrease in both cardiac apoptosis and fibrosis, thus improving cardiac function in the infarcted heart.


Subject(s)
Embryonic Stem Cells/physiology , Muscle Development/physiology , Myocardial Infarction/physiopathology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thymosin/metabolism , Ventricular Remodeling/physiology , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Line , Embryonic Stem Cells/metabolism , Fibrosis/metabolism , Fibrosis/physiopathology , Heart/physiopathology , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Stem Cell Transplantation , Transcription Factors/metabolism , Ventricular Function, Left/physiology
9.
Can J Physiol Pharmacol ; 90(7): 947-51, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22720873

ABSTRACT

It was hypothesized that monocyte treatment with bone morphogenetic protein 7 (BMP7) would significantly enhance monocyte polarization into M2 macrophages as well as increasing the levels of anti-inflammatory cytokines. In a cell culture system using monocytes (human acute monocytic leukemia cell line THP-1), we studied the effects of BMP7 on monocytes polarizing into M2 macrophages. The data demonstrate that THP-1 cells contain a BMP type II receptor (BMPR2), and that its activation is significantly (p < 0.05) increased following treatment with BMP7. Furthermore, there was an increase of M2 macrophages, BMPR2, and anti-inflammatory cytokines interleukin (IL)-10 and IL-1ra compared with the respective controls. Moreover, treatment with BMP7 caused a significant (p < 0.05) decrease in the levels of pro-inflammatory cytokines IL-6, tumour necrosis factor (TNF-α), and monocyte chemotactic protein-1 (MCP-1), compared with the controls. In conclusion, we suggest for the first time that BMP7 has a unique potential to polarize monocytes into M2 macrophages, required for tissue repair, which will have significant applications for the treatment of atherosclerosis.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Macrophages/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Humans , Inflammation/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Monocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Cell Transplant ; 21(9): 1919-30, 2012.
Article in English | MEDLINE | ID: mdl-22449713

ABSTRACT

Doxorubicin (DOX) is an effective antineoplastic agent used for the treatment of a variety of cancers. Unfortunately, its use is limited as this drug induces cardiotoxicity and heart failure as a side effect. There is no report that describes whether transplanted embryonic stem (ES) cells or their conditioned medium (CM) in DOX-induced cardiomyopathy (DIC) can repair and regenerate myocardium. Therefore, we transplanted ES cells or CM in DIC to examine apoptosis, fibrosis, cytoplasmic vacuolization, and myofibrillar loss and their associated Akt and ERK pathway. Moreover, we also determined activation of endogenous c-kit(+ve) cardiac stem cells (CSCs), levels of HGF and IGF-1, growth factors required for c-kit cell activation, and their differentiation into cardiac myocytes, which also contributes in cardiac regeneration and improved heart function. We generated DIC in C57Bl/6 mice (cumulative dose of DOX 12 mg/kg body weight, IP), and animals were treated with ES cells, CM, or cell culture medium in controls. Two weeks post-DIC, ES cells or CM transplanted hearts showed a significant (p < 0.05) decrease in cardiac apoptotic nuclei and their regulation with Akt and ERK pathway. Cardiac fibrosis observed in the ES cell or CM groups was significantly less compared with DOX and cell culture medium groups (p < 0.05). Next, cytoplasmic vacuolization and myofibrillar loss was reduced (p < 0.05) following treatment with ES cells or CM. Moreover, our data also demonstrated increased levels of c-kit(+ve) CSCs in ES cells or CM hearts and differentiated cardiac myocytes from these CSCs, suggesting endogenous cardiac regeneration. Importantly, the levels of HFG and IGF-1 were significantly increased in ES cells or CM transplanted hearts. In conclusion, we reported that transplanted ES cells or CM in DIC hearts significantly decreases various adverse pathological mechanisms as well as enhances cardiac regeneration that effectively contributes to improved heart function.


Subject(s)
Cardiomyopathies/chemically induced , Cardiomyopathies/surgery , Doxorubicin/adverse effects , Embryonic Stem Cells/transplantation , Stem Cell Transplantation/methods , Animals , Apoptosis/drug effects , Cardiomyopathies/pathology , Cell Differentiation/physiology , Disease Models, Animal , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Fibrosis/chemically induced , Fibrosis/pathology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
11.
Mol Pharm ; 8(5): 1573-81, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21542647

ABSTRACT

Cardiac myocyte differentiation reported thus far is from iPS cells generated from mouse and human fibroblasts. However, there is no article on the generation of iPS cells from cardiac ventricular specific cell types such as H9c2 cells. Therefore, whether transduced H9c2 cells, originally isolated from embryonic cardiac ventricular tissue, will be able to generate iPS cells and have the potential to repair and regenerate infarcted myocardium remains completely elusive. We transduced H9c2 cells with four stemness factors, Oct3/4, Sox2, Klf4, and c-Myc, and successfully reprogrammed them into iPS cells. These iPS cells were able to differentiate into beating cardiac myocytes and positively stained for cardiac specific sarcomeric α-actin and myosin heavy chain proteins. Following transplantation in the infarcted myocardium, there were newly differentiated cardiac myocytes and formation of gap junction proteins at 2 weeks post-myocardial infarction (MI), suggesting newly formed cardiac myocytes were integrated into the native myocardium. Furthermore, transplanted iPS cells significantly (p < 0.05) inhibited apoptosis and fibrosis and improved cardiac function compared with MI and MI+H9c2 cell groups. Moreover, our iPS cell derived cardiac myocyte differentiation in vitro and in vivo was comparable to embryonic stem cells in the present study. In conclusion we report for the first time that we have H9c2 cell-derived iPS cells which contain the potential to differentiate into cardiac myocytes in the cell culture system and repair and regenerate infarcted myocardium with improved cardiac function in vivo.


Subject(s)
Heart/physiology , Heart/physiopathology , Induced Pluripotent Stem Cells/transplantation , Myocardial Infarction/therapy , Regeneration , Animals , Apoptosis , Cell Differentiation , Cell Line , Connexins/metabolism , Female , Fibrosis/prevention & control , Heart Function Tests , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , Myoblasts, Cardiac/transplantation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Rats , Transduction, Genetic
12.
Am J Physiol Heart Circ Physiol ; 300(4): H1442-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21297031

ABSTRACT

We investigated whether factors released from mouse embryonic stem (ES) cells primed with and without transforming growth factor (TGF)-ß2 inhibit iodoacetic acid (IAA)- and H(2)O(2)-induced apoptosis in the cell culture system as well as after transplantation in the infarcted heart. We generated conditioned media (CMs) from ES cells primed with and without TGF-ß2 and determined their effects on IAA- and H(2)O(2)-induced apoptosis in H9c2 cells. We also transplanted both ES-CMs in the infarcted heart to determine the effects on apoptosis and cardiac function after myocardial infarction (MI) at day (D)1 and D14. Terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL) staining, apoptotic ELISA, and cell viability data demonstrated significantly (P < 0.05) reduced apoptosis with ES-CM compared with controls in both cell culture models. Moreover, TGF-ß2-primed ES-CM (T-ES-CM) demonstrated enhanced beneficial effects, with further reduced (P < 0.05) apoptosis compared with ES-CM, suggesting the a presence of additional cytoprotective released factors after TGF-ß2 treatment. Next, our in vivo apoptosis data suggested significant decrease in apoptosis with both ES-CMs compared with MI alone at D1 and D14. Notably, T-ES-CM demonstrated significant (P < 0.05) inhibition of apoptosis and fibrosis with improved cardiac function compared with ES-CM at D14, whereas no such effects were observed at D1. Next, we confirmed that apoptosis is mediated through a prosurvival Akt pathway. Moreover, we determined that after TGF-ß2 treatment there was a two- to fivefold increase in cytoprotective released factors (interleukin-10, stem cell factor, tissue inhibitor of matrix metalloproteinase-1, and VEGF) with T-ES-CM compared with ES-CM. In conclusion, we suggest that factors released from ES cells with and without TGF-ß2 treatment contain antiapoptotic factors that inhibit apoptosis in vitro and in vivo. We also suggest that T-ES-CM demonstrates additional beneficial effects that provide useful information for future therapeutic applications in regenerative medicine.


Subject(s)
Apoptosis/drug effects , Cytoprotection/drug effects , Embryonic Stem Cells/metabolism , Myocardial Infarction/drug therapy , Transforming Growth Factor beta2/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Female , Hydrogen Peroxide/pharmacology , Indoleacetic Acids/pharmacology , Interleukin-10/metabolism , Male , Matrix Metalloproteinase 1/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/therapy , Stem Cell Factor/metabolism , Stem Cell Transplantation , Tissue Inhibitor of Metalloproteinase-1/metabolism , Vascular Endothelial Growth Factors/metabolism
13.
Antioxid Redox Signal ; 13(12): 1857-65, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20331412

ABSTRACT

We examined whether factors released from embryonic stem (ES) cells inhibit cardiac and vascular cell apoptosis and stimulate endogenous progenitor cells that enhance neovascularization with improved cardiac function. We generated and transplanted ES-conditioned medium (CM) in the infarcted heart to examine effects on cardiac and vascular apoptosis, activation of endogenous c-kit and FLK-1(+ve) cells, and their role in cardiac neovascularization. TUNEL, caspase-3 activity, immunohistochemistry, H&E, and Masson's trichrome stains were used to determine the effect of transplanted ES-CM on cardiac apoptosis and neovascularization. TUNEL staining and caspase-3 activity confirm significantly (p < 0.05) reduced apoptosis in MI+ES-CM compared with MI+ cell culture medium. Immunohistochemistry demonstrated increased (p < 0.05, 53%) c-kit(+ve) and FLK-1(+ve) positive cells, as well as increased (p < 0.05, 67%) differentiated CD31-positive cells in ES-CM groups compared with respective controls. Furthermore, significantly (p < 0.05) increased coronary artery vessels were observed in ES-CM transplanted hearts compared with control. Heart function was significantly improved following ES-CM transplantation. Next, we observed significantly increased (p < 0.05) levels of c-kit activation proteins (HGF and IGF-1), anti-apoptosis factors (IGF-1 and total antioxidants), and neovascularization protein (VEGF). In conclusion, we suggest that ES-CM following transplantation in the infarcted heart inhibits apoptosis, activates cardiac endogenous c-kit and FLK-1(+ve) cells, and differentiates them into endothelial cells (ECs) that enhances neovascularization with improved cardiac function.


Subject(s)
Culture Media, Conditioned/pharmacology , Embryonic Stem Cells/metabolism , Myocardial Infarction/metabolism , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Culture Media, Conditioned/metabolism , Echocardiography , Enzyme-Linked Immunosorbent Assay , Female , In Situ Nick-End Labeling , Insulin-Like Growth Factor I/metabolism , Male , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism
14.
Am J Physiol Heart Circ Physiol ; 295(2): H907-13, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18552162

ABSTRACT

We recently reported that embryonic stem cells-conditioned medium (ES-CM) contains antiapoptotic factors that inhibit apoptosis in the cardiac myoblast H9c2 cells. However, the mechanisms of inhibited apoptosis remain elusive. In this report, we provide evidence for the novel mechanisms involved in the inhibition of apoptosis provided by ES-CM. ES-CM from mouse ES cells was generated. Apoptosis was induced after exposure with H(2)O(2) (400 mum) in H9c2 cells followed by the replacement with ES-CM or culture medium. H9c2 cells treated with H(2)O(2) were exposed to ES-CM, and ES-CM plus cell survival protein phosphatidylinositol 3-kinase/Akt inhibitor, LY-294002, or extracellular signal-regulated kinase (ERK1/2) inhibitor, PD-98050. After 24 h, H9c2 cells treated with ES-CM demonstrated a significant increase in cell survival. ES-CM significantly inhibited (P < 0.05) apoptosis determined by terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling staining, apoptotic ELISA, and caspase-3 activity. Importantly, enhanced cell survival and inhibited apoptosis with ES-CM was abolished with LY-294002. In contrast, PD-98050 shows no effect on ES-CM-increased cell survival. Furthermore, H(2)O(2)-induced apoptosis is associated with decreased levels of phosphorylated (p)Akt activity. Following treatment with ES-CM, we observed a decrease in apoptosis with an increase in pAkt, and the increased activity was attenuated with the Akt inhibitor, suggesting that the Akt pathway is involved in the decreased apoptosis and cell survival provided by ES-CM. In contrast, we observed no change in ES-CM-decreased apoptosis or pERK with PD-98050. In conclusion, we suggest that ES-CM inhibited apoptosis and is mediated by Akt but not the ERK pathway.


Subject(s)
Apoptosis , Embryonic Stem Cells/metabolism , Myoblasts/enzymology , Myocytes, Cardiac/enzymology , Paracrine Communication , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Caspase 3/metabolism , Cell Line , Cell Survival , Culture Media, Conditioned/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , In Situ Nick-End Labeling , Mice , Myoblasts/pathology , Myocytes, Cardiac/pathology , Oxidants/toxicity , Paracrine Communication/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...