Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Circ Heart Fail ; 17(1): e010813, 2024 01.
Article in English | MEDLINE | ID: mdl-38179791

ABSTRACT

BACKGROUND: Metabolomics has become a valuable tool for identifying potential new biomarkers and metabolic profiles. It has the potential to improve the diagnosis and prognosis of different phenotypes of heart failure. To generate a distinctive metabolic profile, we assessed and compared the metabolic phenotypes of patients with acute decompensated heart failure (ADHF), patients with chronic heart failure (CHF), and healthy controls. METHODS: Plasma metabolites were analyzed by liquid-chromatography mass spectrometry/mass spectrometry and the MxP Quant 500 kit in 15 patients with ADHF, 50 patients with CHF (25 with dilated cardiomyopathy, 25 with ischemic cardiomyopathy), and 13 controls. RESULTS: Of all metabolites identified to be significantly altered, 3-indolepropionic acid and 1-methyl histidine showed the highest concentration differences in ADHF and CHF compared with control. Area under the curve-receiver operating characteristic analysis showed an area under the curve ≥0.8 for 3-indolepropionic acid and 1-methyl histidine, displaying good discrimination capabilities between control and patient cohorts. Additionally, symmetrical dimethylarginine (mean, 1.97±0.61 [SD]; P=0.01) was identified as a suitable biomarker candidate for ADHF and kynurenine (mean, 1.69±0.39 [SD]; P=0.009) for CHF when compared with control, both demonstrating an area under the curve ≥0.85. CONCLUSIONS: Our study provides novel insights into the metabolic differences between ADHF and CHF and healthy controls. We here identify new metabolites for potential diagnostic and prognostic purposes.


Subject(s)
Heart Failure , Histidine , Indoles , Propionates , Humans , Stroke Volume , Heart Failure/diagnosis , Chronic Disease , Biomarkers
2.
Cells ; 10(12)2021 12 09.
Article in English | MEDLINE | ID: mdl-34943991

ABSTRACT

A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.


Subject(s)
Drug Development , Heart Diseases/drug therapy , Induced Pluripotent Stem Cells/drug effects , Cell Differentiation/drug effects , Heart Diseases/pathology , Humans , Myocytes, Cardiac/drug effects
3.
Int J Mol Sci ; 22(21)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34769283

ABSTRACT

Doxorubicin (Dox) is a chemotherapeutic agent with cardiotoxicity associated with profibrotic effects. Dox increases ceramide levels with pro-inflammatory effects, cell death, and fibrosis. The purpose of our study was to identify the underlying ceramide signaling pathways. We aimed to characterize the downstream effects on cell survival, metabolism, and fibrosis. Human fibroblasts (hFSF) were treated with 0.7 µM of Dox or transgenically overexpressed ceramide synthase 2 (FLAG-CerS2). Furthermore, cells were pre-treated with MitoTempo (MT) (2 h, 20 µM) or Fumonisin B1 (FuB) (4 h, 100 µM). Protein expression was measured by Western blot or immunofluorescence (IF). Ceramide levels were determined with mass spectroscopy (MS). Visualizations were conducted using laser scanning microscopy (LSM) or electron microscopy. Mitochondrial activity was measured using seahorse analysis. Dox and CerS2 overexpression increased CerS2 protein expression. Coherently, ceramides were elevated with the highest peak for C24:0. Ceramide- induced mitochondrial ROS production was reduced with MT or FuB preincubation. Mitochondrial homeostasis was reduced and accompanied by reduced ATP production. Our data show that the increase in pro-inflammatory ceramides is an essential contributor to Dox side-effects. The accumulation of ceramides resulted in a lipotoxic shift and subsequently mitochondrial structural and functional damage, which was partially reversible following inhibition of ceramide synthesis.


Subject(s)
Ceramides/metabolism , Doxorubicin/adverse effects , Foreskin/pathology , Membrane Proteins/genetics , Sphingosine N-Acyltransferase/genetics , Tumor Suppressor Proteins/genetics , Adenosine Triphosphate/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Foreskin/cytology , Foreskin/drug effects , Humans , Male , Mass Spectrometry , Membrane Proteins/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation
4.
Biochim Biophys Acta Mol Cell Res ; 1867(11): 118796, 2020 11.
Article in English | MEDLINE | ID: mdl-32663504

ABSTRACT

Ca2+-activated potassium (KCa) channels of small and intermediate conductance influence proliferation, apoptosis, and cell metabolism. We analysed whether prolonged activation of KCa channels by zoxazolamine (ZOX) induces differentiation of mouse embryonic stem (ES) cells towards cardiomyocytes. ZOX treatment of ES cells dose-dependent increased the number and diameter of cardiac foci, the frequency of contractions as well as mRNA expression of the cardiac transcription factor Nkx-2.5, the cardiac markers cardiac troponin I (cTnI), α-myosin heavy chain (α-MHC), ventricular myosin light chain-2 (MLC2v), and the pacemaker hyperpolarization-activated, cyclic nucleotide-gated 4 channel (HCN4). ZOX induced hyperpolarization of membrane potential due to activation of IKCa, raised intracellular Ca2+ concentration ([Ca2+]i) and nitric oxide (NO) in a Ca2+-dependent manner. The Ca2+ response to ZOX was inhibited by chelation of Ca2+ with BAPTA-AM, release of Ca2+ from intracellular stores by thapsigargin and the phospholipase C (PLC) antagonist U73,122. Moreover, the ZOX-induced Ca2+ response was blunted by the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) as well as the specific P2Y1 antagonist MRS 2,179, suggesting purinergic receptor-stimulated signal transduction. Consequently, ZOX initiated ATP release from differentiating ES cells, which was inhibited by the chloride channel inhibitor NPPB and the gap junction inhibitor carbenoxolone (CBX). The stimulation of cardiomyogenesis by ZOX was blunted by the nitric oxide synthase (NOS) inhibitor l-NAME, as well as CBX and NPPB. In summary, our data suggest that ZOX enhances cardiomyogenesis of ES cells by ATP release presumably through gap junctional hemichannels, purinergic receptor activation and intracellular Ca2+ response, thus promoting NO generation.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Heart/growth & development , Muscle Development/genetics , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Calcium Signaling/genetics , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Homeobox Protein Nkx-2.5/genetics , Humans , Mice , Myocytes, Cardiac/drug effects , Myosin Heavy Chains/genetics , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Troponin I/genetics , Zoxazolamine/pharmacology
5.
Basic Res Cardiol ; 115(4): 37, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32424548

ABSTRACT

Human-induced pluripotent stem cells (h-iPSCs) are a unique in vitro model for cardiovascular research. To realize the potential applications of h-iPSCs-derived cardiomyocytes (CMs) for drug testing or regenerative medicine and disease modeling, characterization of the metabolic features is critical. Here, we show the transcriptional profile during stages of cardiomyogenesis of h-iPSCs-derived CMs. CM differentiation was not only characterized by the expression of mature structural components (MLC2v, MYH7) but also accompanied by a significant increase in mature metabolic gene expression and activity. Our data revealed a distinct substrate switch from glucose to fatty acids utilization for ATP production. Basal respiration and respiratory capacity in 9 days h-iPSCs-derived CMs were glycolysis-dependent with a shift towards a more oxidative metabolic phenotype at 14 and 28 day old CMs. Furthermore, mitochondrial analysis characterized the early and mature forms of mitochondria during cardiomyogenesis. These results suggest that changes in cellular metabolic phenotype are accompanied by increased O2 consumption and ATP synthesis to fulfill the metabolic needs of mature CMs activity. To further determine functionality, the physiological response of h-iPSCs-derived CMs to ß-adrenergic stimulation was tested. These data provide a unique in vitro human heart model for the understanding of CM physiology and metabolic function which may provide useful insight into metabolic diseases as well as novel therapeutic options.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Cells, Cultured , Humans
6.
Reprod Toxicol ; 65: 46-58, 2016 10.
Article in English | MEDLINE | ID: mdl-27346840

ABSTRACT

Man-made magnetic fields (MFs) may exert adverse effects on mammalian embryonic development. Herein, we analysed the effect of 10mT 50Hz sinusoidal (AC) or static (DC) MFs versus 1mT MFs on embryonic development of mice. Exposure for 20days during gestation to 10mT MFs increased resorptions and dead fetuses, decreased crown-rump length and fresh weight, reduced blood vessel differentiation and caused histological changes, accompanied with diminished vascular endothelial growth factor (VEGF) protein expression in several organs. In embryonic stem (ES) cell-derived embryoid bodies exposure towards 10mT MFs increased reactive oxygen species (ROS), decreased vascular marker as well as VEGF expression and enhanced apoptosis. In conclusion, our combined data from in vivo and in vitro experiments identified VEGF as an important mediator during embryonic development that can be influenced by high strength MFs, which in consequence leads to severe abnormalities in fetus organs and blood vessel formation.


Subject(s)
Embryonic Development , Magnetic Fields , Neovascularization, Physiologic , Animals , Cell Line , Crown-Rump Length , Embryo Loss , Embryo, Mammalian , Embryoid Bodies/metabolism , Eye/embryology , Eye/metabolism , Female , Liver/embryology , Liver/metabolism , Lung/embryology , Lung/metabolism , Mice , Mice, Inbred BALB C , Mouse Embryonic Stem Cells/metabolism , Pregnancy , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/metabolism , Weight Gain
7.
Cell Tissue Res ; 364(1): 159-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26553657

ABSTRACT

The impact of reactive oxygen species and phosphoinositide 3-kinase (PI3K) in differentiating embryonic stem (ES) cells is largely unknown. Here, we show that the silencing of the PI3K catalytic subunit p110α and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 (NOX1) by short hairpin RNA or pharmacological inhibition of NOX and ras-related C3 botulinum toxin substrate 1 (Rac1) abolishes superoxide production by vascular endothelial growth factor (VEGF) in mouse ES cells and in ES-cell-derived fetal liver kinase-1(+) (Flk-1(+)) vascular progenitor cells, whereas the mitochondrial complex I inhibitor rotenone does not have an effect. Silencing p110α or inhibiting Rac1 arrests vasculogenesis at initial stages in embryoid bodies, even under VEGF treatment, as indicated by platelet endothelial cell adhesion molecule-1 (PECAM-1)-positive areas and branching points. In the absence of p110α, tube-like structure formation on matrigel and cell migration of Flk-1(+) cells in scratch migration assays are totally impaired. Silencing NOX1 causes a reduction in PECAM-1-positive areas, branching points, cell migration and tube length upon VEGF treatment, despite the expression of vascular differentiation markers. Interestingly, silencing p110α but not NOX1 inhibits the activation of Rac1, Ras homologue gene family member A (RhoA) and Akt leading to the abrogation of VEGF-induced lamellipodia structure formation. Thus, our data demonstrate that the PI3K p110α-Akt/Rac1 and NOX1 signalling pathways play a pivotal role in VEGF-induced vascular differentiation and cell migration. Rac1, RhoA and Akt phosphorylation occur downstream of PI3K and upstream of NOX1 underscoring a role of PI3K p110α in the regulation of cell polarity and migration.


Subject(s)
Cell Differentiation/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Mouse Embryonic Stem Cells/enzymology , NADH, NADPH Oxidoreductases/metabolism , Neovascularization, Physiologic/drug effects , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Differentiation/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Mice , Mouse Embryonic Stem Cells/cytology , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , Neovascularization, Physiologic/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Free Radic Biol Med ; 89: 1203-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26524400

ABSTRACT

OBJECTIVE: Cell injury releases nucleic acids supporting inflammation and stem cell activation. Here, the impact of extracellular ribonucleic acid, especially transfer RNA (ex-tRNA), on vasculogenesis and leukopoiesis of mouse embryonic stem (ES) cells was investigated. APPROACH AND RESULTS: ex-tRNA, whole cell RNA and ribosomal RNA (ex-rRNA) but not DNA increased CD31-positive vascular structures in embryoid bodies. Ex-tRNA and ex-rRNA increased numbers of VEGFR2(+), CD31(+) and VE-cadherin(+) vascular cells as well as CD18(+), CD45(+) and CD68(+) cells, indicating leukocyte/macrophage differentiation. This was paralleled by mRNA and protein expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor-165 (VEGF165) and neuropilin 1 (NRP1), phosphorylation of phosphatidyl inositol 3-kinase (PI3K) and VEGF receptor 2 (VEGFR2) as well as mRNA expression of α-smooth muscle actin (α-SMA). ex-tRNA was taken up by endosomes, increased expression of the pro-angiogenic semaphorin B4 receptor plexin B1 as well as the ephrin-type B receptor 4 (EphB4) and ephrinB2 ligand and enhanced cell migration, which was inhibited by the VEGFR2 antagonist SU5614 and the PI3K inhibitor LY294002. This likewise abolished the effects of ex-tRNA on vasculogenesis and leukopoiesis of ES cells. Ex-tRNA increased NOX1, NOX2, NOX4 and DUOX2 mRNA and boosted the generation of superoxide and hydrogen peroxide which was inhibited by radical scavengers, the NADPH oxidase inhibitors apocynin, VAS2870, ML171, and plumbagin as well as shRNA silencing of NOX1 and NOX4. CONCLUSIONS: Our findings indicate that ex-tRNA treatment induces vasculogenesis and leukopoiesis of ES cells via superoxide/hydrogen peroxide generated by NADPH oxidase and activation of VEGFR2 and PI3K.


Subject(s)
Embryonic Stem Cells/pathology , Leukopoiesis/genetics , Neovascularization, Physiologic/genetics , RNA, Ribosomal/genetics , RNA, Transfer/genetics , Animals , Blotting, Western , Cell Differentiation , Cell Movement , Cells, Cultured , Embryonic Stem Cells/metabolism , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Mice , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Purinergic Signal ; 11(4): 491-506, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26395809

ABSTRACT

Purinergic signaling may be involved in embryonic development of the heart. In the present study, the effects of purinergic receptor stimulation on cardiomyogenesis of mouse embryonic stem (ES) cells were investigated. ADP or ATP increased the number of cardiac clusters and cardiac cells, as well as beating frequency. Cardiac-specific genes showed enhanced expression of α-MHC, MLC2v, α-actinin, connexin 45 (Cx45), and HCN4, on both gene and protein levels upon ADP/ATP treatment, indicating increased cardiomyogenesis and pacemaker cell differentiation. Real-time RT-PCR analysis of purinergic receptor expression demonstrated presence of P2X1, P2X4, P2X6, P2X7, P2Y1, P2Y2, P2Y4, and P2Y6 on differentiating ES cells. ATP and ADP as well as the P2X agonists ß,γ-methylenadenosine 5'-triphosphate (ß,γ-MetATP) and 8-bromoadenosine 5'-triphosphate (8-Br-ATP) but not UTP or UDP transiently increased the intracellular calcium concentration ([Ca(2+)](i)) as evaluated by the calcium indicator Fluo-4, whereas no changes in membrane potential were observed. [Ca(2+)](i) transients induced by ADP/ATP were abolished by the phospholipase C-ß (PLC-ß) inhibitor U-73122, suggesting involvement of metabotropic P2Y receptors. Furthermore, partial inhibition of [Ca(2+)](i) transients was achieved in presence of MRS2179, a selective P2Y1 receptor antagonist, whereas PPADS, a non-selective P2 receptor inhibitor, completely abolished the [Ca(2+)](i) response. Consequently, cardiomyocyte differentiation was decreased upon long term co-incubation of cells with ADP and P2 receptor antagonists. In summary, activation of purinoceptors and the subsequent [Ca(2+)](i) transients enhance the differentiation of ES cells toward cardiomyocytes. Purinergic receptor stimulation may be a promising strategy to drive the fate of pluripotent ES cells into a particular population of cardiomyocytes.


Subject(s)
Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Embryonic Stem Cells/drug effects , Muscle Development/drug effects , Myocytes, Cardiac/drug effects , Adenosine Triphosphate/antagonists & inhibitors , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Female , Gene Expression/drug effects , Membrane Potentials/drug effects , Mice , Myocardial Contraction/drug effects , Pregnancy , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2X/biosynthesis , Receptors, Purinergic P2X/drug effects , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2Y1/drug effects , Uridine Diphosphate/pharmacology , Uridine Triphosphate/pharmacology
10.
Cell Tissue Res ; 358(2): 443-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25130141

ABSTRACT

The ß-adrenoceptor antagonist Propranolol has been successfully used to treat infantile hemangioma. However, its mechanism of action is so far unknown. The hypothesis of this research was that ß-adrenoceptor antagonists may interfere with endothelial cell differentiation of stem cells. Specifically, the effects of the non-specific ß-adrenergic receptor (ß-adrenoceptor) antagonist Propranolol, the ß1-adrenoceptor-specific antagonist Atenolol and the ß2-adrenoceptor-specific antagonist ICI118,551 on vasculogenesis of mouse embryonic stem (ES) cells were investigated. All three ß-blockers dose-dependently downregulated formation of capillary structures in ES cell-derived embryoid bodies and decreased the expression of the vascular cell markers CD31 and VE-cadherin. Furthermore, ß-blockers downregulated the expression of fibroblast growth factor-2 (FGF-2), hypoxia inducible factor-1α (HIF-1α), vascular endothelial growth factor 165 (VEGF165), VEGF receptor 2 (VEGF-R2) and phospho VEGF-R2, as well as neuropilin 1 (NRP1) and plexin-B1 which are essential modulators of embryonic angiogenesis with additional roles in vessel remodelling and arteriogenesis. Under conditions of ß-adrenoceptor inhibition, the endogenous generation of nitric oxide (NO) as well as the phosphorylation of endothelial nitric oxide synthase (eNOS) was decreased in embryoid bodies, whereas an increase in NO generation was observed with the NO donor S-nitroso-N-acetyl-D,L-penicillamine (SNAP). Consequently, vasculogenesis of ES cells was restored upon treatment of differentiating ES cells with ß-adrenoceptor antagonists in the presence of NO donor. In summary, our data suggest that ß-blockers impair vasculogenesis of ES cells by interfering with NO generation which could be the explanation for their anti-angiogenic effects in infantile hemangioma.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Down-Regulation/drug effects , Embryonic Stem Cells/physiology , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Adrenergic beta-Antagonists/administration & dosage , Animals , Atenolol/administration & dosage , Atenolol/pharmacology , Biomarkers/metabolism , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Embryonic Stem Cells/drug effects , Mice , Nitric Oxide Synthase Type III/metabolism , Propanolamines/administration & dosage , Propanolamines/pharmacology , Propranolol/administration & dosage , Propranolol/pharmacology , Receptors, Adrenergic, beta/metabolism , S-Nitroso-N-Acetylpenicillamine/administration & dosage , S-Nitroso-N-Acetylpenicillamine/pharmacology , Time Factors
11.
Hum Mutat ; 35(4): 497-504, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478229

ABSTRACT

Hereditary axonopathies are frequently caused by mutations in proteins that reside in the endoplasmic reticulum (ER). Which of the many ER functions are pathologically relevant, however, remains to be determined. REEP1 is an ER protein mutated in hereditary spastic paraplegia (HSP) and hereditary motor neuropathy (HMN). We found that HSP-associated missense variants at the N-terminus of REEP1 abolish ER targeting, whereas two more central variants are either rare benign SNPs or confer pathogenicity via a different mechanism. The mis-targeted variants accumulate at lipid droplets (LDs). N-terminal tagging, deletion of the N-terminus, and expression of a minor REEP1 isoform had the same effect. We also confirmed an increase in LD size upon cooverexpression of atlastins and REEP1. Neither wild-type REEP1, LD-targeted HSP variants, nor a non-LD-targeted HMN variant reproduced this effect when expressed alone. We conclude that the N-terminus of REEP1 is necessary for proper targeting to and/or retention in the ER. The protein's potential to also associate with LDs corroborates a synergistic effect with atlastins on LD size. Interestingly, LD size is also altered upon knockdown of seipin, mutations of which also cause HSP and HMN. Regulation of LDs may thus be an ER function critical for long-term axonal maintenance.


Subject(s)
Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Animals , Cell Line, Tumor , DNA Mutational Analysis , Genetic Variation , HeLa Cells , Humans , Mice , Muscular Atrophy, Spinal/genetics , Mutation , Spastic Paraplegia, Hereditary/genetics
12.
Stem Cells Dev ; 23(4): 333-51, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24134622

ABSTRACT

The plasticity of human adipose tissue-derived stem cells (hASCs) is promising, but differentiation in vitro toward endothelial cells is poorly understood. Flow cytometry demonstrated that hASCs isolated from excised fat tissue were positive for CD29, CD44, CD70, CD90, CD105, and CD166 and negative for the endothelial marker CD31, and the hematopoietic cell markers CD34 and CD133. hASCs differentiated into adipocytes after cultivation in adipogenic medium. Exposure of hASCs for 10 days under hypoxia (3% oxygen) in combination with leptin increased the percentage of CD31(+) endothelial cells as well as CD31, VE-Cadherin, Flk-1, Tie2, von Willebrand factor, and endothelial cell nitric oxide synthase mRNA expression. This was enhanced on co-incubation of vascular endothelial growth factor (VEGF) and leptin, whereas VEGF alone was not sufficient. Moreover, hASCs cultured on a matrigel surface under hypoxia/VEGF/leptin, showed a stable branching network. Hypoxic conditions significantly decreased apoptosis as evaluated by cleaved caspase-3, and increased prolyl hydroxylase domain 3 mRNA expression. Hypoxia increased expression of VEGF as well as leptin transcripts, which were significantly inhibited on co-incubation with either VEGF or leptin or a combination of both. Furthermore, leptin treatment of hypoxic cells increased the expression of the long/signaling form of the leptin receptor (ObRL), which was augmented on co-incubation with VEGF. The observed endothelial differentiation was dependent on the Akt pathway, as co-administration with Akt inhibitor abolished the observed effects. In conclusion, our data demonstrate that hASCs can be efficiently differentiated to endothelial cells by mimicking the hypoxic and pro-angiogenic microenvironment of adipose tissue.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation , Endothelium, Vascular/cytology , Leptin/physiology , Vascular Endothelial Growth Factor A/physiology , Abdominal Fat/cytology , Aged , Apoptosis , Biomarkers/metabolism , Cell Hypoxia , Cells, Cultured , Humans , Middle Aged , Neovascularization, Physiologic , Transcriptome
13.
Stem Cells Dev ; 22(14): 1977-90, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23441986

ABSTRACT

Embryonic stem (ES) cells are a powerful model for the development of cells responsible for the cellular immune response. Therefore, we analyzed the defense and phagocytic capacity of embryoid bodies (EBs) derived from ES cells using in the vitro inflammatory conditions caused by Escherichia coli. Further, we used this phagocytic activity to purify activated immune cells. Our data show that spontaneously differentiated 18-day-old EBs of the cell line CGR8 contained immune cells, which were positive for CD45, CD68, CD11b, F4/80, and CD19. Exposure of these EBs to E. coli with defined infection doses of bacterial colony-forming units (CFUs) led to a significant time-dependent reduction of CFUs, indicating the immune responses exerted by EBs. This was paralleled by an upregulation of inflammatory cytokines, that is, IL-1ß and TNF-α. Western blot analysis of infected EBs indicated an upregulation of CD14 and cytochrome b-245 heavy chain (NOX2). Silencing of NOX2 significantly reduced the antibacterial capacity of EBs, which was partially explained by reduction of F4/80-positive cells. To identify, isolate, and further cultivate phagocytic active cells from differentiated EBs, a cocultivation assay of differentiated ES cells with green fluorescent protein (GFP)-labeled E. coli was established. Colocalization of GFP-labeled E. coli with cells positive for CD45, CD68, and F4/80 revealed time-dependent phagocytotic uptake, which was underlined by colocalization with the LysoTracker-Red(®) dye as well as preincubation with cytochalasin D. In conclusion, a primitive immune response with efficient phagocytosis was responsible for the antibacterial capacity of differentiated EBs.


Subject(s)
Embryoid Bodies/immunology , Escherichia coli/growth & development , Phagocytosis/immunology , Amines , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Cell Differentiation , Cell Line , Coculture Techniques , Colony Count, Microbial , Cytochalasin D , Embryoid Bodies/cytology , Escherichia coli/immunology , Flow Cytometry , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology
14.
Int J Cardiol ; 167(3): 798-808, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-22465345

ABSTRACT

AIMS: To investigate the effects of static magnetic fields (MFs) on cardiomyogenesis of mouse embryonic stem (ES) cell-derived embryoid bodies and Flk-1(+) cardiac progenitor cells and to assess the impact of cytosolic calcium [Ca(2+)]c and reactive oxygen species (ROS). METHODS AND RESULTS: Embryoid bodies and ES cell-derived Flk-1(+) cardiovascular progenitor cells were exposed to static MFs. The expression of cardiac genes was evaluated by RT-PCR; sarcomeric structures were assessed by immunohistochemistry; intracellular ROS and [Ca(2+)]c of ES cells were examined by H2DCF-DA- and fluo-4-based microfluorometry. Treatment of embryoid bodies with MFs dose-dependent increased the number of contracting foci and cardiac areas as well as mRNA expression of the cardiac genes MLC2a, MLC2v, α-MHC and ß-MHC. In Flk-1(+) cells MFs (1 mT) elevated both [Ca(2+)]c and ROS, increased expression of the cardiogenic transcription factors Nkx-2.5 and GATA-4 as well as cardiac genes. This effect was due to Ca(2+) influx, since extracellular Ca(2+) chelation abrogated ROS production and MF-induced cardiomyogenesis. Furthermore absence of extracellular calcium impaired sarcomere structures. Neither the phospholipase C inhibitor U73122 nor thapsigargin inhibited MF-induced increase in [Ca(2+)]c excluding involvement of intracellular calcium stores. ROS were generated through NAD(P)H oxidase, since NOX-4 but not NOX-1 and NOX-2 mRNA was upregulated upon MF exposure. Ablation of NOX-4 by sh-RNA and treatment with the NAD(P)H oxidase inhibitor diphenylen iodonium (DPI) totally abolished MF-induced cardiomyogenesis. CONCLUSION: The ability of static MFs to enhance cardiomyocyte differentiation of ES cells allows high throughput generation of cardiomyocytes without pharmacological or genetic modification.


Subject(s)
Calcium/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Magnetic Fields , Myocytes, Cardiac/cytology , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Base Sequence , Cell Line , Embryonic Stem Cells/metabolism , Male , Mice , Molecular Sequence Data , Myocytes, Cardiac/metabolism , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/genetics
15.
Free Radic Biol Med ; 51(2): 432-43, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21570463

ABSTRACT

Ascorbic acid (AA) increases cardiomyogenesis of embryonic stem (ES) cells. Herein we show that treatment of mouse ES cells with AA enhanced cardiac differentiation accompanied by an upregulation of the NADPH oxidase isoforms NOX2 and NOX4, phosphorylation of endothelial nitric oxide synthase (eNOS), and cyclic GMP (cGMP) formation, indicating that reactive oxygen species (ROS) as well as nitric oxide (NO) may be involved in cardiomyogenesis. In whole mount embryoid bodies as well as isolated Flk-1-positive (Flk-1(+)) cardiovascular progenitor cells ROS elevation by AA was observed in early stages of differentiation (Days 4-7), and absent at Day 10. In contrast NO generation following incubation with AA was absent at Day 4 and increased at Days 7 and 10. AA-mediated cardiomyogenesis was blunted by the NADPH oxidase inhibitors diphenylen iodonium (DPI) and apocynin, the free radical scavengers N-(2-mercaptopropionyl)-glycine (NMPG) and ebselen, and the NOS inhibitor L-NAME. Downregulation of NOX4 by short hairpin RNA (shRNA) resulted in significant inhibition of cardiomyogenesis and abolished the stimulation of MHC-ß and MLC2v gene expression observed on AA treatment. Our data demonstrate that AA stimulates cardiomyocyte differentiation from ES cells by signaling pathways that involve ROS generated at early stages and NO at late stages of cardiomyogenesis.


Subject(s)
Ascorbic Acid/pharmacology , Embryonic Stem Cells/drug effects , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Base Sequence , Blotting, Western , Cell Differentiation , Cell Lineage , DNA Primers , Embryonic Stem Cells/cytology , Mice , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
J Cell Sci ; 124(Pt 11): 1819-30, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21540297

ABSTRACT

VEGF-, phosphoinositide 3-kinase (PI3K)- and protein kinase C (PKC)-regulated signaling in cardiac and vascular differentiation was investigated in mouse ES cells and in ES cell-derived Flk-1⁺ cardiovascular progenitor cells. Inhibition of PI3K by wortmannin and LY294002, disruption of PI3K catalytic subunits p110α and p110δ using short hairpin RNA (shRNA), or inhibition of p110α with compound 15e and of p110δ with IC-87114 impaired cardiac and vascular differentiation. By contrast, TGX-221, an inhibitor of p110ß, and shRNA knockdown of p110ß were without significant effects. Antagonists of the PKC family, i.e. bisindolylmaleimide-1 (BIM-1), GÖ 6976 (targeting PKCα/ßII) and rottlerin (targeting PKCδ) abolished vasculogenesis, but not cardiomyogenesis. Inhibition of Akt blunted cardiac as well as vascular differentiation. VEGF induced phosphorylation of PKCα/ßII and PKCδ but not PKCζ. This was abolished by PI3K inhibitors and the VEGFR-2 antagonist SU5614. Furthermore, phosphorylation of Akt and phosphoinositide-dependent kinase-1 (PDK1) was blunted upon inhibition of PI3K, but not upon inhibition of PKC by BIM-1, suggesting that activation of Akt and PDK1 by VEGF required PI3K but not PKC. In summary, we demonstrate that PI3K catalytic subunits p110α and p110δ are central to cardiovasculogenesis of ES cells. Akt downstream of PI3K is involved in both cardiomyogenesis and vasculogenesis, whereas PKC is involved only in vasculogenesis.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Embryoid Bodies/cytology , Muscle Development , Myocardium/cytology , Neovascularization, Physiologic , Protein Kinase C/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Catalytic Domain , Cell Differentiation , Cell Line , Chromones/pharmacology , Class Ia Phosphatidylinositol 3-Kinase/genetics , Embryoid Bodies/drug effects , Enzyme Activation , Mice , Morpholines/pharmacology , Myocardium/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction
17.
Antioxid Redox Signal ; 13(12): 1813-27, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20722506

ABSTRACT

Thalidomide [α-(N-phthalimido)-glutarimide] exerts antiangiogenic properties and causes cardiac malformations in embryos. Herein the effects of thalidomide on cardiovascular differentiation were investigated in mouse embryonic stem (ES) cell-derived embryoid bodies. Thalidomide inhibited the formation of capillary-like blood vessels and decreased tumor-induced angiogenesis in confrontation cultures of embryoid bodies and multicellular prostate tumor spheroids, but stimulated cardiomyogenesis of ES cells. The number of CD31- and CD144-positive endothelial cells was not impaired, suggesting that thalidomide acted on vascular tube formation and cell migration rather than endothelial differentiation. Thalidomide increased reactive oxygen species generation, which was abolished by the NADPH oxidase inhibitor VAS2870 and the complex I respiratory chain inhibitor rotenone. Conversely, thalidomide decreased nitric oxide (NO) generation and endothelial NO synthase activity. VAS2870 abrogated thalidomide stimulation of cardiomyogenesis, whereas inhibition of vasculogenesis persisted. In NOX-1 and NOX-4 shRNA gene-inactivated ES cells, cardiomyogenesis was severely impaired and thalidomide failed to stimulate cardiac cell commitment. The NO donor S-nitrosopenicillamine reversed the antiangiogenic effect of thalidomide and increased capillary structure formation, whereas scavenging NO by 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide and inhibition of endothelial NO synthase by N(G)-nitro-l-arginine methyl ester decreased cardiovascular differentiation. Our data demonstrate that thalidomide causes an imbalance of reactive oxygen species/NO generation, thus stimulating cardiomyogenesis and impairing vascular sprout formation.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Thalidomide/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Transformed , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Flow Cytometry , Humans , Immunohistochemistry , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Cardiovasc Res ; 86(1): 37-44, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19965960

ABSTRACT

AIMS: It is well established that leukotrienes (LTs), products of the 5-lipoxygenase (5-LO) pathway, participate in inflammatory tissue reactions and immune responses. In the present study, the impact of the 5-LO pathway on vasculogenesis of mouse embryonic stem (ES) cells was investigated. METHODS AND RESULTS: Immunohistochemistry studies demonstrated that 5-LO(+) cells first appeared at day 6 of embryoid body (EB) formation from ES cells. 5-LO(+)/CD68(+) as well as 5-LO(+)/CD45(+) cells were prominent at day 10 of EB differentiation. Real-time PCR and western blot analysis revealed all constituents of the 5-LO pathway. High performance liquid chromatography analyses indicated the synthesis of LTB(4) and LTD(4) in conformity with induction of the 5-LO pathway. Furthermore, Flk-1(+)/CD105(+) cells displayed calcium- (Ca(2+)) transients in response to LTB(4), whereas CD11b(+) cells responded to LTD(4). Treatment of EBs with LTB(4) and LTD(4) resulted in phosphorylation of the mitogen-activated protein kinase ERK1/2. Pharmacological inhibition of the 5-LO pathway and stable shRNA targeting of 5-LO-activating protein decreased capillary cell areas positive for PECAM-1. CONCLUSION: Our data demonstrate that the 5-LO pathway emerges early during ES cell differentiation into cells of the myeloid lineage and that LTs play an until now unrecognized role in vascular development of ES cells.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Carrier Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Membrane Proteins/metabolism , Neovascularization, Pathologic/metabolism , 5-Lipoxygenase-Activating Proteins , Animals , Arachidonate 5-Lipoxygenase/genetics , Carrier Proteins/genetics , Cell Differentiation/physiology , Cell Lineage/physiology , Cells, Cultured , Embryonic Stem Cells/drug effects , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Leukotriene B4/pharmacology , Leukotriene D4/pharmacology , Leukotrienes/metabolism , Lymphocytes/cytology , Lymphocytes/physiology , Macrophages/cytology , Macrophages/physiology , Membrane Proteins/genetics , Mice , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Phosphorylation/drug effects , Phosphorylation/physiology , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Leukotriene/genetics , Receptors, Leukotriene/metabolism , Receptors, Leukotriene B4/genetics , Receptors, Leukotriene B4/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
J Cell Biochem ; 109(2): 434-46, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19950199

ABSTRACT

UNLABELLED: ABC transporters like P-glycoprotein (P-gp/ABCB1) are membrane proteins responsible for the transport of toxic compounds out of non-malignant cells and tumor tissue. AIM: To investigate the effect of glycolysis and the tissue redox state on P-gp expression in multicellular tumor spheroids derived from prostate adenocarcinoma cells (DU-145), glioma cells (Gli36), and the human cervix carcinoma cell line KB-3-1 transfected with a P-gp-EGFP fusion gene that allows monitoring of P-gp expression in living cells. During cell culture of DU-145, Gli36, and KB-3-1 tumor spheroids P-gp expression was observed as well as increased lactate and decreased pyruvate levels and expression of glycolytic enzymes. Inhibition of glycolysis for 24 h by either iodoacetate (IA) or 2-deoxy-D-glucose (2-DDG) downregulated P-gp expression which was reversed upon coincubation with the radical scavenger ebselen as shown by semi-quantitative immunohistochemisty in DU-145 and Gli36 tumor spheroids, and by EGFP fluorescence in KB-3-1 tumor spheroids. Consequently endogenous ROS generation in DU-145 tumor spheroids was increased in the presence of either IA or 2-DDG, which was abolished upon coincubation with ebselen. Exogenous addition of pyruvate significantly reduced ROS generation, increased P-gp expression as well as efflux of the P-gp substrate doxorubicin. Doxorubicin transport was significantly blunted by 2-DDG and IA, indicating that inhibition of glycolysis reversed the multidrug resistance phenotype. In summary our data demonstrate that P-gp expression in tumor spheroids is closely related to the glycolytic metabolism of tumor cells and can be downregulated by glycolysis inhibitors via mechanisms that involve changes in the cellular redox state.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Biological Transport, Active/drug effects , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism , Spheroids, Cellular/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm , Glycolysis , Humans , Lactic Acid/metabolism , Male , Oxidation-Reduction , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyruvic Acid/pharmacology
20.
Stem Cells Dev ; 19(5): 731-43, 2010 May.
Article in English | MEDLINE | ID: mdl-19788349

ABSTRACT

Electromagnetic fields (EMFs) are used to treat bone diseases. Herein, the effects of static EMFs on chondroosteogenesis and vasculogenesis of embryonic stem (ES) cells and bone mineralization of mouse fetuses were investigated. Treatment of differentiating ES cells with static EMFs (0.4-2 mT) stimulated vasculogenesis and chondro-osteogenesis and increased reactive oxygen species (ROS), which was abolished by the free radical scavengers trolox, 1,10-phenanthroline (phen), and the NAD(P)H oxidase inhibitor diphenylen iodonium (DPI). In contrast, EMFs of 10 mT field strength exerted inhibitory effects on vasculogenesis and chondro-osteogenesis despite robust ROS generation. EMFs of 1 mT and 10 mT increased and decreased vascular endothelial growth factor (VEGF) expression, respectively, which was abolished by DPI and radical scavengers. EMFs activated extracellular-regulated kinase 1/2 (ERK1/2), p38, and c-jun N-terminal kinase (JNK), which was sensitive to DPI treatment. The increase in VEGF by EMFs was inhibited by the ERK1/2 inhibitor U0126 but not by SB203580 and SP600125, which are p38 and JNK inhibitors, respectively, suggesting VEGF regulation by ERK1/2. Chondroosteogenesis and vasculogenesis of ES cells was blunted by trolox, DPI, and the VEGF receptor-2 (flk-1) antagonist SU5614. In mouse fetuses 1 mT EMFs increased and 10 mT EMFs decreased bone mineralization, which was abolished in the presence of trolox. Hence, EMFs induced chondro-osteogenesis and vasculogenesis in ES cells and bone mineralization of mouse fetuses by a ROS-dependent up-regulation of VEGF expression.


Subject(s)
Electromagnetic Fields , Embryonic Stem Cells/physiology , Neovascularization, Physiologic , Osteogenesis , Reactive Oxygen Species/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Calcification, Physiologic , Embryonic Stem Cells/metabolism , Fetus , Mice , Mitogen-Activated Protein Kinases/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...