Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Front Cardiovasc Med ; 11: 1200786, 2024.
Article in English | MEDLINE | ID: mdl-38450366

ABSTRACT

Background: Cardiac arrhythmias have markedly increased in recent decades, highlighting the urgent need for appropriate test systems to evaluate the efficacy and safety of new pharmaceuticals and the potential side effects of established drugs. Methods: The Microelectrode Array (MEA) system may be a suitable option, as it provides both real-time and non-invasive monitoring of cellular networks of spontaneously active cells. However, there is currently no commercially available cell source to apply this technology in the context of the cardiac conduction system (CCS). In response to this problem, our group has previously developed a protocol for the generation of pure functional cardiac pacemaker cells from mouse embryonic stem cells (ESCs). In addition, we compared the hanging drop method, which was previously utilized, with spherical plate-derived embryoid bodies (EBs) and the pacemaker cells that are differentiated from these. Results: We described the application of these pacemaker cells on the MEA platform, which required a number of crucial optimization steps in terms of coating, dissociation, and cell density. As a result, we were able to generate a monolayer of pure pacemaker cells on an MEA surface that is viable and electromechanically active for weeks. Furthermore, we introduced spherical plates as a convenient and scalable method to be applied for the production of induced sinoatrial bodies. Conclusion: We provide a tool to transfer modeling and analysis of cardiac rhythm diseases to the cell culture dish. Our system allows answering CCS-related queries within a cellular network, both under baseline conditions and post-drug exposure in a reliable and affordable manner. Ultimately, our approach may provide valuable guidance not only for cardiac pacemaker cells but also for the generation of an MEA test platform using other sensitive non-proliferating cell types.

2.
Cell Physiol Biochem ; 53(2): 337-354, 2019.
Article in English | MEDLINE | ID: mdl-31373783

ABSTRACT

BACKGROUND/AIMS: The availability of truly maturated cardiomyocytic subtypes is a major prerequisite for cardiovascular cell replacement therapies. Pluripotent stem cells provide a suitable source for the development of new strategies to overcome enormous hurdles such as yield, purity and safety of in vitro generated cells. METHODS: To address these issues, we have refined existing forward programming protocols by combining forced exogenous overexpression of the early cardiovascular transcription factor Nkx2.5 with a αMHC-promoter-based antibiotic selection step. Additionally, we applied small molecules such as ascorbic acid to enhance cardiomyogenic differentiation efficiency. Subsequently, we evaluated the cell fate of the resulting cardiomyocytes on the mRNA as well as protein levels. The latter was performed using high-resolution confocal microscopy. Furthermore, we examined the response of the cells` beating activities to pharmacological substance administration. RESULTS: Our results reveal an apparent influence of Nkx2.5 on the cell fate of ESC-derived cardiomyocytes. Resulting single cells exhibit characteristics of early ventricular cardiomyocytes, such as sarcomeric marker expression, spontaneous beating frequency, and distinct L-type calcium channel occurrence. CONCLUSION: Therefore, we demonstrate cardiovascular subtype forward programming of ESCs using a combination of transcription factors along with small molecule administration. However, our findings also underline current assumptions, that a terminal maturation of PSC derived cardiomyocytes in vitro is still an unsolved problem which urgently needs to be addressed in the field.


Subject(s)
Cellular Reprogramming , Embryonic Stem Cells/metabolism , Homeobox Protein Nkx-2.5/metabolism , Myocytes, Cardiac/metabolism , Animals , Ascorbic Acid/pharmacology , Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Homeobox Protein Nkx-2.5/antagonists & inhibitors , Homeobox Protein Nkx-2.5/genetics , Mice , Microscopy, Confocal , Myocytes, Cardiac/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Verapamil/pharmacology
3.
J Breath Res ; 12(2): 026014, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29231842

ABSTRACT

Bacterial and cell cultures are known to emit a large number of volatile organic compounds (VOCs). Conventional biochemical methods are often destructive, time-consuming and expensive. In contrast, VOC analysis of headspace over cultures may offer a non-destructive alternative for the monitoring of cell proliferation and metabolism. VOC profiles from cultures of murine pluripotent stem cells and fibroblasts were assessed every 24 h for 3 days. Pure cell media were measured as parallel controls. VOC analysis was highly standardized with respect to time of measurement and phases of cell growth. Cultures were grown in custom-made inert boxes. In order to determine the effects of fresh media supply on VOC emissions, both cell types were cultured with and without daily media exchange. VOCs from headspace were preconcentrated by means of needle trap micro-extraction and analysed by gas chromatography-mass spectrometry (GC-MS). Murine pluripotent stem cells emitted increasing concentrations of thiirane and methyl-methoxy-hydroxy-methyl-amine (MMHA). Substance concentration correlated with cell numbers. Murine fibroblasts did not emit thiirane or MMHA. Concentrations of aldehydes, especially benzaldehyde, were lower in both cell cultures than in pure media samples. Daily media exchange resulted in higher cell numbers, but had no major effects on VOC concentrations emitted from the cells. Investigation and monitoring of volatile substances such as thiirane and MMHA may enhance the understanding of stem cell properties and lead to a destruction-free characterization of pluripotent stem cells.


Subject(s)
Smell , Stem Cells/cytology , Volatile Organic Compounds/analysis , Aldehydes/analysis , Amines/analysis , Animals , Breath Tests , Cell Count , Cell Proliferation , Gas Chromatography-Mass Spectrometry , Limit of Detection , Mice , Sulfides/analysis
4.
Nat Commun ; 8(1): 1258, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29097735

ABSTRACT

AMPK is a conserved serine/threonine kinase whose activity maintains cellular energy homeostasis. Eukaryotic AMPK exists as αßγ complexes, whose regulatory γ subunit confers energy sensor function by binding adenine nucleotides. Humans bearing activating mutations in the γ2 subunit exhibit a phenotype including unexplained slowing of heart rate (bradycardia). Here, we show that γ2 AMPK activation downregulates fundamental sinoatrial cell pacemaker mechanisms to lower heart rate, including sarcolemmal hyperpolarization-activated current (I f) and ryanodine receptor-derived diastolic local subsarcolemmal Ca2+ release. In contrast, loss of γ2 AMPK induces a reciprocal phenotype of increased heart rate, and prevents the adaptive intrinsic bradycardia of endurance training. Our results reveal that in mammals, for which heart rate is a key determinant of cardiac energy demand, AMPK functions in an organ-specific manner to maintain cardiac energy homeostasis and determines cardiac physiological adaptation to exercise by modulating intrinsic sinoatrial cell behavior.


Subject(s)
AMP-Activated Protein Kinases/genetics , Bradycardia/genetics , Calcium/metabolism , Heart Rate/genetics , Sarcolemma/metabolism , Sinoatrial Node/metabolism , Adult , Animals , Bradycardia/metabolism , Electrocardiography, Ambulatory , Exercise , Heart/diagnostic imaging , Humans , Magnetic Resonance Imaging, Cine , Magnetic Resonance Spectroscopy , Mice , Microscopy, Electron, Transmission , Mutation , Myocardium/metabolism , Myocardium/pathology , Myocardium/ultrastructure , Physical Conditioning, Animal , Physical Endurance , Ryanodine Receptor Calcium Release Channel/metabolism , Sinoatrial Node/pathology
5.
Adv Drug Deliv Rev ; 120: 142-167, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28916499

ABSTRACT

Adult cardiomyocytes (CMs) possess a highly restricted intrinsic regenerative potential - a major barrier to the effective treatment of a range of chronic degenerative cardiac disorders characterized by cellular loss and/or irreversible dysfunction and which underlies the majority of deaths in developed countries. Both stem cell programming and direct cell reprogramming hold promise as novel, potentially curative approaches to address this therapeutic challenge. The advent of induced pluripotent stem cells (iPSCs) has introduced a second pluripotent stem cell source besides embryonic stem cells (ESCs), enabling even autologous cardiomyocyte production. In addition, the recent achievement of directly reprogramming somatic cells into cardiomyocytes is likely to become of great importance. In either case, different clinical scenarios will require the generation of highly pure, specific cardiac cellular-subtypes. In this review, we discuss these themes as related to the cardiovascular stem cell and programming field, including a focus on the emergent topic of pacemaker cell generation for the development of biological pacemakers and in vitro drug testing.


Subject(s)
Cellular Reprogramming/physiology , Myocytes, Cardiac/physiology , Animals , Cardiovascular Diseases/therapy , Guided Tissue Regeneration , Humans , Induced Pluripotent Stem Cells/physiology
6.
Microvasc Res ; 112: 87-92, 2017 07.
Article in English | MEDLINE | ID: mdl-28363496

ABSTRACT

OBJECTIVES: Toll-like-receptor (TLR) mediated immune response has been shown to regulate myocardial damage following cardiac ischemia-reperfusion (IR). It has not been described conclusively so far whether migration of therapeutically applied progenitor cells following an IR event depends on TLR-signaling. METHODS: In vivo migratory capacity murine c-kit+ cells following IR injury was quantified by intravital fluorescence microscopy, utilizing the mouse cremaster muscle model and analyzing early (rolling) and late (adhesion) c-kit+ cell interaction with the local endothelium. The role of TLR-2 and TLR-4, as well as MyD88 and TRIF was analyzed by applying specific knock-out models. RESULTS: A sequence of 15min ischemia followed by 15min of reperfusion induced firm endothelial c-kit+ cell adhesion (5.6±1.3cells/mm2 in Control vs. 30.2±10.1cells/mm2 in IR, p<0.05). Knock-out of TLR-2 and TLR-4 diminished both IR induced early c-kit+ cell-endothelial cell interactions (67.6±2.3% c-kit+ cell rolling in IR vs. 46.3±4.8% c-kit+ cell rolling in IR-TLR-2-ko vs. 45.3±4.8% c-kit+ cell rolling in IR-TLR-4-ko, p<0.05) as well as firm endothelial c-kit+ cell adhesion (30.2±10.1cells/mm2 in IR vs. 16.3±3.9cells/mm2 in IR-TLR-2-ko vs. 14.5±4.4cells/mm2 in IR-TLR-4-ko, p<0.05). Adaptor protein knock-out resulted in a significantly decreased firm endothelial c-kit+ cell adhesion only in MyD88 knock-out but not in TRIF knock-out (9.2±2.2cells/mm2 in IR-MyD88-ko vs. 30.1±9.9cells/mm2 in IR-WT, p<0.05). CONCLUSION: Artificially applied c-kit+ cells interact with the target organ endothelium following IR injury. This interaction seems to depend on TLR-MyD88 signaling. Therapeutic blockade of TLR signaling for anti-inflammatory purposes might interfere with regenerative cell-based therapy protocols.


Subject(s)
Abdominal Muscles/blood supply , Cell Movement , Proto-Oncogene Proteins c-kit/metabolism , Regeneration , Reperfusion Injury/surgery , Stem Cell Transplantation , Stem Cells/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Abdominal Muscles/pathology , Abdominal Muscles/physiopathology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Adhesion , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Genetic Predisposition to Disease , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phenotype , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Time Factors , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
7.
Nanomedicine ; 12(8): 2353-2364, 2016 11.
Article in English | MEDLINE | ID: mdl-27389150

ABSTRACT

Genetic modulation of angiogenesis is a powerful tool for the treatment of multiple disorders. Here, we describe a strategy to produce modified endothelial cells, which can be efficiently magnetically guided. First, we defined optimal transfection conditions with both plasmid and microRNA, using a polyethyleneimine/magnetic nanoparticle-based vector (PEI/MNP), previously designed in our group. Further, two approaches were assessed in vitro: direct vector guidance and magnetic targeting of transfected cells. Due to its higher efficiency, including simulated dynamic conditions, production of miR/PEI/MNP-modified magnetically responsive cells was selected for further detailed investigation. In particular, we have studied internalization of transfection complexes, functional capacities and intercellular communication of engineered cells and delivery of therapeutic miR. Moreover, we demonstrated that 104 miRNA/PEI/MNP-modified magnetically responsive cells loaded with 0.37pg iron/cell are detectable with MRI. Taken together, our in vitro findings show that PEI/MNP is highly promising as a multifunctional tool for magnetically guided angiogenesis regulation.


Subject(s)
Magnetics , MicroRNAs , Plasmids , DNA , Endothelial Cells , Nanotechnology/methods , Neovascularization, Pathologic , Polyethyleneimine , Transfection
8.
BMC Bioinformatics ; 17: 21, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26738481

ABSTRACT

BACKGROUND: Technical advances in Next Generation Sequencing (NGS) provide a means to acquire deeper insights into cellular functions. The lack of standardized and automated methodologies poses a challenge for the analysis and interpretation of RNA sequencing data. We critically compare and evaluate state-of-the-art bioinformatics approaches and present a workflow that integrates the best performing data analysis, data evaluation and annotation methods in a Transparent, Reproducible and Automated PipeLINE (TRAPLINE) for RNA sequencing data processing (suitable for Illumina, SOLiD and Solexa). RESULTS: Comparative transcriptomics analyses with TRAPLINE result in a set of differentially expressed genes, their corresponding protein-protein interactions, splice variants, promoter activity, predicted miRNA-target interactions and files for single nucleotide polymorphism (SNP) calling. The obtained results are combined into a single file for downstream analysis such as network construction. We demonstrate the value of the proposed pipeline by characterizing the transcriptome of our recently described stem cell derived antibiotic selected cardiac bodies ('aCaBs'). CONCLUSION: TRAPLINE supports NGS-based research by providing a workflow that requires no bioinformatics skills, decreases the processing time of the analysis and works in the cloud. The pipeline is implemented in the biomedical research platform Galaxy and is freely accessible via www.sbi.uni-rostock.de/RNAseqTRAPLINE or the specific Galaxy manual page (https://usegalaxy.org/u/mwolfien/p/trapline---manual).


Subject(s)
Computational Biology/standards , High-Throughput Nucleotide Sequencing/standards , Sequence Analysis, RNA/standards , Computational Biology/methods , High-Throughput Nucleotide Sequencing/methods , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Annotation , Polymorphism, Single Nucleotide , Protein Interaction Domains and Motifs , Sequence Alignment , Sequence Analysis, RNA/methods , Transcriptome
9.
J Vis Exp ; (96): e52465, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25742394

ABSTRACT

Treatment of the "sick sinus syndrome" is based on artificial pacemakers. These bear hazards such as battery failure and infections. Moreover, they lack hormone responsiveness and the overall procedure is cost-intensive. "Biological pacemakers" generated from PSCs may become an alternative, yet the typical content of pacemaker cells in Embryoid Bodies (EBs) is extremely low. The described protocol combines "forward programming" of murine PSCs via the sinus node inducer TBX3 with Myh6-promoter based antibiotic selection. This yields cardiomyocyte aggregates consistent of >80% physiologically functional pacemaker cells. These "induced-sinoatrial-bodies" ("iSABs") are spontaneously contracting at yet unreached frequencies (400-500 bpm) corresponding to nodal cells isolated from mouse hearts and are able to pace murine myocardium ex vivo. Using the described protocol highly pure sinus nodal single cells can be generated which e.g. can be used for in vitro drug testing. Furthermore, the iSABs generated according to this protocol may become a crucial step towards heart tissue engineering.


Subject(s)
Pluripotent Stem Cells/physiology , Sinoatrial Node/physiology , T-Box Domain Proteins/genetics , Animals , Cell Aggregation/physiology , Cell Differentiation/physiology , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Sinoatrial Node/cytology , Sinoatrial Node/metabolism , Stem Cell Transplantation/methods , T-Box Domain Proteins/biosynthesis , Transfection
10.
Pharmacogenet Genomics ; 25(1): 8-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25379722

ABSTRACT

BACKGROUND: The efficacy of statins, which are used commonly in primary and secondary prevention of cardiovascular diseases, shows a wide range of interindividual variability. Genetic variants of OATP1B1, a hepatic uptake transporter, can modify access of statins to its therapeutic target, thereby potentially altering drug efficacy. We studied the impact of genetic variants of OATP1B1 on the lipid-lowering efficacy of statins in a population-based setting. MATERIALS AND METHODS: The basis of the analysis was the Study of Health in Pomerania, a cohort of 2732 men and women aged 20-81 years. Included in the statistical analysis to evaluate the impact of OATP1B1 on therapeutic efficacy of statins were 214 individuals diagnosed with dyslipidaemia during initial recruitment and receiving statins during the 5-year follow-up. RESULTS: Analysing the impact of the OATP1B1 genotype, we observed a trend for lower statin-induced total cholesterol reduction in carriers of the SLCO1B1 512C variant. Restricting the analysis to patients receiving simvastatin, pravastatin, lovastatin and fluvastatin indicated a statistically significant association of the OATP1B1 genotype on lipid parameters at the 5-year follow-up. No such effect was observed for atorvastatin. Calculation of achievement of treatment goals according to the NCEP-ATPIII guidelines showed a lower rate of successful treatment when harbouring the mutant allele for patients taking simvastatin (46.7 vs. 73.9%). A similar trend was observed for pravastatin (34.4 vs. 70.4%). CONCLUSION: Genetic variants of OATP1B1 leading to impaired hepatic uptake of statins translated into reduced drug efficacy in a population-based cohort.


Subject(s)
Coronary Disease/genetics , Genetic Association Studies , Lipid Metabolism/genetics , Organic Anion Transporters/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Pharmacological , Coronary Disease/blood , Coronary Disease/drug therapy , Coronary Disease/pathology , Fatty Acids, Monounsaturated/administration & dosage , Female , Fluvastatin , Genotype , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Indoles/administration & dosage , Lipid Metabolism/drug effects , Liver-Specific Organic Anion Transporter 1 , Lovastatin/administration & dosage , Lovastatin/genetics , Male , Middle Aged , Pravastatin/administration & dosage , Pravastatin/genetics , Risk Assessment , Simvastatin/administration & dosage
11.
Am J Physiol Renal Physiol ; 307(9): F1041-51, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25209865

ABSTRACT

Renal tubular handling of urate is realized by a network of uptake and efflux transporters, including members of drug transporter families such as solute carrier proteins and ATP-binding cassette transporters. Solute carrier family 2, member 9 (SLC2A9), is one key factor of this so called "urate transportosome." The aim of the present study was to understand the transcriptional regulation of SLC2A9 and to test whether identified factors might contribute to a coordinated transcriptional regulation of the transporters involved in urate handling. In silico analysis and cell-based reporter gene assays identified a hepatocyte nuclear factor (HNF)4α-binding site in the promoter of SLC2A9 isoform 1, whose activity was enhanced by transient HNF4α overexpression, whereas mutation of the binding site diminished activation. HNF4α overexpression induced endogenous SLC2A9 expression in vitro. The in vivo role of HNF4α in the modulation of renal SLC2A9 gene expression was supported by findings of quantitative real-time RT-PCR analyses and chromatin immunoprecipitation assays. Indeed, mRNA expression of SLC2A9 and HNF4α in human kidney samples was significantly correlated. We also showed that in renal clear cell carcinoma, downregulation of HNF4α mRNA and protein expression was associated with a significant decline in expression of the transporter. Taken together, our data suggest that nuclear receptor family member HNF4α contributes to the transcriptional regulation of SLC2A9 isoform 1. Since HNF4α has previously been assumed to be a modulator of several urate transporters, our findings support the notion that there could be a transcriptional network providing synchronized regulation of the functional network of the urate transportosome.


Subject(s)
Glucose Transport Proteins, Facilitative/biosynthesis , Hepatocyte Nuclear Factor 4/physiology , Organic Anion Transporters/biosynthesis , Binding Sites/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/physiopathology , Cell Dedifferentiation , Gene Expression Regulation , Glucose Transport Proteins, Facilitative/genetics , HeLa Cells , Humans , Organic Anion Transporters/genetics , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Transcription, Genetic
12.
Eur J Clin Invest ; 44(9): 833-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25047264

ABSTRACT

BACKGROUND: In primary hyperparathyroidism (PHPT), the increased levels of parathyroid hormone (PTH) result in mobilisation of bone-marrow-derived cells (BMCs) into peripheral blood. However, the fate of these cells is still unknown. MATERIALS AND METHODS: In this study, we sought to investigate cells with typical surface markers of BMCs within parathyroid adenomas (PA) of patients with primary hyperparathyroidism. We therefore investigated PA and normal parathyroid glands (NPG) of 15 patients with PHPT by immunohistochemistry and PCR. RESULTS: mRNA levels for CD31, CD34 and CD45 were significantly increased in PA compared to NPG. Immunohistochemical staining for CD31 and CD34 revealed a significantly higher vessel density in PA. Furthermore, scattered single cells expressing CD31, CD34 or CD45 were significantly augmented compared to normal parathyroid glands and directly correlated with vessel density. mRNA levels of SDF-1 was increased whereas its major inhibitor dipeptidylpeptidase IV (DPP IV) is decreased in PA, suggesting that the SDF-1 axis plays a role in the migration of BMCs into PA. CONCLUSION: These data indicate a possible role of BMCs in the pathophysiology of PA of patients with PHPT.


Subject(s)
Adenoma/pathology , Hyperparathyroidism, Primary/pathology , Parathyroid Neoplasms/pathology , Adult , Aged , Antigens, CD/metabolism , Bone Marrow Cells , Female , Humans , Immunohistochemistry , Male , Middle Aged , Parathyroid Glands/metabolism , Prospective Studies
13.
Stem Cell Reports ; 2(5): 592-605, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24936448

ABSTRACT

Therapeutic approaches for "sick sinus syndrome" rely on electrical pacemakers, which lack hormone responsiveness and bear hazards such as infection and battery failure. These issues may be overcome via "biological pacemakers" derived from pluripotent stem cells (PSCs). Here, we show that forward programming of PSCs with the nodal cell inducer TBX3 plus an additional Myh6-promoter-based antibiotic selection leads to cardiomyocyte aggregates consisting of >80% physiologically and pharmacologically functional pacemaker cells. These induced sinoatrial bodies (iSABs) exhibited highly increased beating rates (300-400 bpm), coming close to those found in mouse hearts, and were able to robustly pace myocardium ex vivo. Our study introduces iSABs as highly pure, functional nodal tissue that is derived from PSCs and may be important for future cell therapies and drug testing in vitro.


Subject(s)
Cellular Reprogramming , Pluripotent Stem Cells/cytology , Sinoatrial Node/physiology , Animals , Biological Clocks , Calcium/metabolism , Cell Differentiation , Cell Line , Coculture Techniques , In Vitro Techniques , Mice , Models, Biological , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Patch-Clamp Techniques , Pluripotent Stem Cells/metabolism , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology , Sick Sinus Syndrome/veterinary , Sinoatrial Node/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
14.
PLoS One ; 9(5): e94882, 2014.
Article in English | MEDLINE | ID: mdl-24788344

ABSTRACT

Heritability estimates for body mass index (BMI) variation are high. For mothers and their offspring higher BMI correlations have been described than for fathers. Variation(s) in the exclusively maternally inherited mitochondrial DNA (mtDNA) might contribute to this parental effect. Thirty-two to 40 mtDNA single nucleotide polymorphisms (SNPs) were available from genome-wide association study SNP arrays (Affymetrix 6.0). For discovery, we analyzed association in a case-control (CC) sample of 1,158 extremely obese children and adolescents and 435 lean adult controls. For independent confirmation, 7,014 population-based adults were analyzed as CC sample of n = 1,697 obese cases (BMI ≥ 30 kg/m2) and n = 2,373 normal weight and lean controls (BMI<25 kg/m2). SNPs were analyzed as single SNPs and haplogroups determined by HaploGrep. Fisher's two-sided exact test was used for association testing. Moreover, the D-loop was re-sequenced (Sanger) in 192 extremely obese children and adolescents and 192 lean adult controls. Association testing of detected variants was performed using Fisher's two-sided exact test. For discovery, nominal association with obesity was found for the frequent allele G of m.8994G/A (rs28358887, p = 0.002) located in ATP6. Haplogroup W was nominally overrepresented in the controls (p = 0.039). These findings could not be confirmed independently. For two of the 252 identified D-loop variants nominal association was detected (m.16292C/T, p = 0.007, m.16189T/C, p = 0.048). Only eight controls carried the m.16292T allele, five of whom belonged to haplogroup W that was initially enriched among these controls. m.16189T/C might create an uninterrupted poly-C tract located near a regulatory element involved in replication of mtDNA. Though follow-up of some D-loop variants still is conceivable, our hypothesis of a contribution of variation in the exclusively maternally inherited mtDNA to the observed larger correlations for BMI between mothers and their offspring could not be substantiated by the findings of the present study.


Subject(s)
DNA, Mitochondrial/genetics , Genome-Wide Association Study , Obesity/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Aged , Case-Control Studies , Female , Haplotypes/genetics , Humans , Male , Middle Aged , Sequence Analysis, DNA , Young Adult
15.
Basic Res Cardiol ; 108(6): 388, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24065117

ABSTRACT

Endogenous circulation of bone marrow-derived cells (BMCs) was observed in patients with dilated cardiomyopathy (DCM) who showed cardiac upregulation of Vascular Cell Adhesion Protein-1 (VCAM-1). However, the underlying pathophysiology is currently unknown. Thus, we aimed to analyze circulation, migration and G-CSF-based mobilization of BMCs in a murine model of virus-induced DCM. Mice with coxsackievirus B3 (CVB3) induced DCM and healthy controls were analyzed regarding their myocardial homing factors by PCR. To determine cardiac VCAM-1 expression ELISA and immunohistochemistry were applied. Flow cytometry was performed to analyze BMCs. Cardiac diameters and function were evaluated by echocardiography before and 4 weeks after G-CSF treatment. In murine CVB3-induced DCM an increase of BMCs in peripheral blood and a decrease of BMCs in bone marrow was observed. We found an enhanced migration of Very Late Antigen-4 (VLA-4⁺) BMCs to the diseased heart overexpressing VCAM-1 and higher numbers of CD45⁻CD34⁻Sca-1⁺ and CD45⁻CD34⁻c-kit⁺ cells. Mobilization of BMCs by G-CSF boosted migration along the VCAM-1/VLA-4 axis and reduced apoptosis of cardiomyocytes. Significant improvement of cardiac function was detected by echocardiography in G-CSF-treated mice. Blocking VCAM-1 by a neutralizing antibody reduced the G-CSF-dependent effects on stem cell migration and cardiac function. This is the first study showing that in virus-induced DCM VCAM-1/VLA-4 interaction is crucial for recruitment of circulating BMCs leading to beneficial anti-apoptotic effects resulting in improved cardiac function after G-CSF-induced mobilization.


Subject(s)
Bone Marrow Cells/cytology , Cardiomyopathy, Dilated/physiopathology , Cell Movement/physiology , Integrin alpha4beta1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/virology , Coxsackievirus Infections , Disease Models, Animal , Echocardiography , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Immunohistochemistry , Mice , Real-Time Polymerase Chain Reaction , Stem Cells/cytology
16.
Basic Res Cardiol ; 108(1): 312, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23184392

ABSTRACT

Common cardiovascular progenitor cells are characterized and induced by expression of the transcription factor MesP1. To characterize this population we used a 3.4-kb promoter fragment previously described by our group. This served to isolate MesP1-positive cells from differentiating ES stem cells via magnetic cell sorting based on a truncated CD4 surface marker. As this proximal promoter fragment omits a distal non-cardiovasculogenic enhancer region, we were able to achieve a synchronized fraction of highly enriched cardiovascular progenitors. These led to about 90% of cells representing the three cardiovascular lineages: cardiomyocytes, endothelial cells and smooth muscle cells as evident from protein and mRNA analyses. In addition, electrophysiological and pharmacological parameters of the cardiomyocytic fraction show that almost all correspond to the multipotent early/intermediate cardiomyocyte subtype at day 18 of differentiation. Further differentiation of these cells was not impaired as evident from strong and synchronous beating at later stages. Our work contributes to the understanding of the earliest cardiovasculogenic events and may become an important prerequisite for cell therapy, tissue engineering and pharmacological testing in the culture dish using pluripotent stem cell-derived as well as directly reprogrammed cardiovascular cell types. Likewise, these cells provide an ideal source for large-scale transcriptome and proteome analyses.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cardiovascular System/cytology , Multipotent Stem Cells/cytology , Promoter Regions, Genetic , Animals , Cell Differentiation , Cell Separation , Endothelial Cells/cytology , Mice , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology
17.
BMC Cancer ; 13: 617, 2013 Dec 31.
Article in English | MEDLINE | ID: mdl-24380367

ABSTRACT

BACKGROUND: Resistance of the highly aggressive glioblastoma multiforme (GBM) to drug therapy is a major clinical problem resulting in a poor patient's prognosis. Beside promoter methylation of the O6-methylguanine-DNA-methyltransferase (MGMT) gene the efflux transporters ABCB1 and ABCG2 have been suggested as pivotal factors contributing to drug resistance, but the methylation of ABCB1 and ABCG2 has not been assessed before in GBM. METHODS: Therefore, we evaluated the proportion and prognostic significance of promoter methylation of MGMT, ABCB1 and ABCG2 in 64 GBM patient samples using pyrosequencing technology. Further, the single nucleotide polymorphisms MGMT C-56 T (rs16906252), ABCB1 C3435T (rs1045642) and ABCG2 C421A (rs2231142) were determined using the restriction fragment length polymorphism method (RFLP). To study a correlation between promoter methylation and gene expression, we analyzed MGMT, ABCB1 and ABCG2 expression in 20 glioblastoma and 7 non-neoplastic brain samples. RESULTS: Despite a significantly increased MGMT and ABCB1 promoter methylation in GBM tissue, multivariate regression analysis revealed no significant association between overall survival of glioblastoma patients and MGMT or ABCB1 promoter methylation. However, a significant negative correlation between promoter methylation and expression could be identified for MGMT but not for ABCB1 and ABCG2. Furthermore, MGMT promoter methylation was significantly associated with the genotypes of the MGMT C-56 T polymorphism showing a higher methylation level in the T allele bearing GBM. CONCLUSIONS: In summary, the data of this study confirm the previous published relation of MGMT promoter methylation and gene expression, but argue for no pivotal role of MGMT, ABCB1 and ABCG2 promoter methylation in GBM patients' survival.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Brain Neoplasms/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Glioblastoma/genetics , Neoplasm Proteins/genetics , Tumor Suppressor Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Adult , Aged , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/therapy , DNA Methylation , Female , Gene Expression , Glioblastoma/mortality , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Recurrence
18.
Pharmacogenet Genomics ; 21(11): 694-700, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21934639

ABSTRACT

BACKGROUND: CYP2C19 is a polymorphic enzyme that plays a pivotal role in the metabolism of 10% of clinically used drugs worldwide. The CYP2C19*3 allele is characterized by a premature stop codon that leads to a truncated nonfunctional protein and consequently a poor metabolizer phenotype. Aminoglycoside antibiotics have been shown to induce readthrough of premature stop codons and partial restoration of protein function. We investigated the ability of the aminoglycosides gentamicin and G418 to induce readthrough of CYP2C19*3 premature stop codon in human cells. METHODS: A CYP2C19*3 expression model in HeLa cells was used in all experiments. CYP2C19-EGFP expression was assessed by flow cytometry, immunoblotting, and fluorescence microscopy, whereas CYP2C19 enzymatic activity was quantified by hydroxylation of 3-cyano-7-ethoxycoumarin. RESULTS: G418 and gentamicin promoted readthrough of the CYP2C19*3 premature stop codon in a concentration-dependent manner. Flow cytometry revealed a maximum 23% protein restoration with the highest aminoglycoside concentrations tested, namely 300 mg/ml G418 and 1000 mg/ml gentamicin. At these concentrations, G418 was more effective than gentamicin in restoring CYP2C19 expression in immunoblotting and fluorescence microscopy assays, as well as in restoring CYP2C19 enzymatic activity. CONCLUSION: This is the first demonstration of readthrough of a stop codon in a pharmacogenetic target of clinical relevance, namely CYP2C19*3. The experimental models may be adapted to explore readthrough of stop codons in other genes of pharmacogenetic interest.


Subject(s)
Aminoglycosides/pharmacology , Aryl Hydrocarbon Hydroxylases/genetics , Codon, Nonsense/genetics , Gene Expression Regulation/drug effects , Biological Assay , Cytochrome P-450 CYP2C19 , Flow Cytometry , HeLa Cells , Humans , Models, Genetic
19.
Drug Metab Dispos ; 39(1): 132-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20930123

ABSTRACT

Pancreatic adenocarcinoma is one of the malignancies that is highly resistant to therapy and among the leading causes of cancer-related death. Several factors may influence pancreatic cancer resistance, and expression of ATP-binding cassette transport proteins is one of the major mechanisms of drug resistance. Members of this family's C-branch, also referred to as multidrug resistance-associated proteins (MRPs), might be of particular interest because they are able to efflux nucleoside analogs used in the treatment of pancreatic cancer. Expression of MRP1, MRP3, MRP4, and MRP5 in human pancreas and pancreatic carcinoma has been reported. However, contributions of MRPs to chemoresistance of pancreatic cancer are not fully understood. MRP5 mRNA expression in pancreatic adenocarcinoma cell lines correlated significantly with cellular sensitivity to 5-fluorouracil (5-FU) (r = 0.738, p < 0.05). Long-term treatment with 5-FU increased expression of MRP5 by 2.4-fold and was associated with significant drug resistance [IC(50) values for control and 5-fluorouracil (5-FU)-resistant Patu-T cell lines were 11.3 ± 5.3 and 33.2 ± 6.9 µM, respectively (p < 0.05)]. Consequently, overexpression of MRP5 in Colo-357 cells resulted in significantly reduced accumulation of 5-FU related radioactivity and 5-FU cytotoxicity. Knockdown of MRP5 significantly increased cellular cytotoxicity of 5-FU to Patu-02 cells and enhanced accumulation of radioactivity related to 5-FU and its metabolites. Our results suggest that MRP5 is expressed and functionally active and contributes to variable sensitivities of pancreatic adenocarcinoma cell lines to 5-FU. Further investigations using models that resemble human pancreas tumors are necessary to prove a causative relation between expression and activity of MRP5 and tumor resistance to 5-FU.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Fluorouracil/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Adenocarcinoma , Cell Line, Tumor , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Gene Knockdown Techniques , Humans , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , RNA/analysis , RNA Interference , Tumor Cells, Cultured
20.
PLoS Genet ; 6(4): e1000916, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421936

ABSTRACT

Meta-analyses of population-based genome-wide association studies (GWAS) in adults have recently led to the detection of new genetic loci for obesity. Here we aimed to discover additional obesity loci in extremely obese children and adolescents. We also investigated if these results generalize by estimating the effects of these obesity loci in adults and in population-based samples including both children and adults. We jointly analysed two GWAS of 2,258 individuals and followed-up the best, according to lowest p-values, 44 single nucleotide polymorphisms (SNP) from 21 genomic regions in 3,141 individuals. After this DISCOVERY step, we explored if the findings derived from the extremely obese children and adolescents (10 SNPs from 5 genomic regions) generalized to (i) the population level and (ii) to adults by genotyping another 31,182 individuals (GENERALIZATION step). Apart from previously identified FTO, MC4R, and TMEM18, we detected two new loci for obesity: one in SDCCAG8 (serologically defined colon cancer antigen 8 gene; p = 1.85x10(-8) in the DISCOVERY step) and one between TNKS (tankyrase, TRF1-interacting ankyrin-related ADP-ribose polymerase gene) and MSRA (methionine sulfoxide reductase A gene; p = 4.84x10(-7)), the latter finding being limited to children and adolescents as demonstrated in the GENERALIZATION step. The odds ratios for early-onset obesity were estimated at approximately 1.10 per risk allele for both loci. Interestingly, the TNKS/MSRA locus has recently been found to be associated with adult waist circumference. In summary, we have completed a meta-analysis of two GWAS which both focus on extremely obese children and adolescents and replicated our findings in a large followed-up data set. We observed that genetic variants in or near FTO, MC4R, TMEM18, SDCCAG8, and TNKS/MSRA were robustly associated with early-onset obesity. We conclude that the currently known major common variants related to obesity overlap to a substantial degree between children and adults.


Subject(s)
Body Weight/genetics , Genetic Loci , Genome, Human , Obesity/genetics , Adolescent , Adult , Age of Onset , Alleles , Body Mass Index , Child , France/epidemiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Germany/epidemiology , Humans , Obesity/epidemiology , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...