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











Database
Language
Publication year range
1.
Stem Cell Reports ; 15(1): 13-21, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32531193

ABSTRACT

Pluripotency is tightly regulated and is crucial for stem cells and their implementation for regenerative medicine. Non-coding RNAs, especially long non-coding RNAs (lncRNAs) emerged as orchestrators of versatile (patho)-physiological processes on the transcriptional and post-transcriptional level. Cyrano, a well-conserved lncRNA, is highly expressed in stem cells suggesting an important role in pluripotency, which we aimed to investigate in loss-off-function (LOF) experiments. Cyrano was described previously to be essential for the maintenance of mouse embryonic stem cell (ESC) pluripotency. In contrast, using different genetic models, we here found Cyrano to be dispensable in murine and human iPSCs and in human ESCs. RNA sequencing revealed only a moderate influence of Cyrano on the global transcriptome. In line, Cyrano-depleted iPSCs retained the potential to differentiate into the three germ layers. In conclusion, different methods were applied for LOF studies to rule out potential off-target effects. These approaches revealed that Cyrano does not impact pluripotency.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , RNA, Long Noncoding/metabolism , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cell Differentiation/genetics , Cell Self Renewal/genetics , Gene Silencing , Human Embryonic Stem Cells/metabolism , Humans , Mice, Knockout , RNA, Long Noncoding/genetics , Transcriptome/genetics
2.
Cardiovasc Res ; 115(1): 71-82, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29931050

ABSTRACT

Aims: Chronic heart failure is becoming increasingly prevalent and is still associated with a high mortality rate. Myocardial hypertrophy and fibrosis drive cardiac remodelling and heart failure, but they are not sufficiently inhibited by current treatment strategies. Furthermore, despite increasing knowledge on cardiomyocyte intracellular signalling proteins inducing pathological hypertrophy, therapeutic approaches to target these molecules are currently unavailable. In this study, we aimed to establish and test a therapeutic tool to counteract the 22 kDa calcium and integrin binding protein (CIB) 1, which we have previously identified as nodal regulator of pathological cardiac hypertrophy and as activator of the maladaptive calcineurin/NFAT axis. Methods and results: Among three different sequences, we selected a shRNA construct (shCIB1) to specifically down-regulate CIB1 by 50% upon adenoviral overexpression in neonatal rat cardiomyocytes (NRCM), and upon overexpression by an adeno-associated-virus (AAV) 9 vector in mouse hearts. Overexpression of shCIB1 in NRCM markedly reduced cellular growth, improved contractility of bioartificial cardiac tissue and reduced calcineurin/NFAT activation in response to hypertrophic stimulation. In mice, administration of AAV-shCIB1 strongly ameliorated eccentric cardiac hypertrophy and cardiac dysfunction during 2 weeks of pressure overload by transverse aortic constriction (TAC). Ultrastructural and molecular analyses revealed markedly reduced myocardial fibrosis, inhibition of hypertrophy associated gene expression and calcineurin/NFAT as well as ERK MAP kinase activation after TAC in AAV-shCIB1 vs. AAV-shControl treated mice. During long-term exposure to pressure overload for 10 weeks, AAV-shCIB1 treatment maintained its anti-hypertrophic and anti-fibrotic effects, but cardiac function was no longer improved vs. AAV-shControl treatment, most likely resulting from a reduction in myocardial angiogenesis upon downregulation of CIB1. Conclusions: Inhibition of CIB1 by a shRNA-mediated gene therapy potently inhibits pathological cardiac hypertrophy and fibrosis during pressure overload. While cardiac function is initially improved by shCIB1, this cannot be kept up during persisting overload.


Subject(s)
Calcium-Binding Proteins/metabolism , Heart Failure/therapy , Hypertrophy, Left Ventricular/therapy , Myocytes, Cardiac/metabolism , RNA, Small Interfering/metabolism , RNAi Therapeutics , Ventricular Dysfunction, Left/therapy , Ventricular Function, Left , Ventricular Remodeling , Animals , Calcineurin/metabolism , Calcium-Binding Proteins/genetics , Cells, Cultured , Disease Models, Animal , Fibrosis , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , NFATC Transcription Factors/metabolism , Neovascularization, Physiologic , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
3.
Adv Biochem Eng Biotechnol ; 163: 117-146, 2018.
Article in English | MEDLINE | ID: mdl-29218360

ABSTRACT

Engineered cardiac tissue might enable novel therapeutic strategies for the human heart in a number of acquired and congenital diseases. With recent advances in stem cell technologies, namely the availability of pluripotent stem cells, the generation of potentially autologous tissue grafts has become a realistic option. Nevertheless, a number of limitations still have to be addressed before clinical application of engineered cardiac tissue based on human stem cells can be realized. We summarize current progress and pending challenges regarding the optimal cell source, cardiomyogenic lineage specification, purification, safety of genetic cell engineering, and genomic stability. Cardiac cells should be combined with clinical grade scaffold materials for generation of functional myocardial tissue in vitro. Scale-up to clinically relevant dimensions is mandatory, and tissue vascularization is most probably required both for preclinical in vivo testing in suitable large animal models and for clinical application. Graphical Abstract.


Subject(s)
Myocardium , Myocytes, Cardiac , Pluripotent Stem Cells , Tissue Engineering/methods , Animals , Humans , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
4.
J Proteome Res ; 16(3): 1133-1149, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28139119

ABSTRACT

Human pluripotent stem cells (hPSCs), both embryonic (hESCs) and induced (hiPSCs), can be differentiated into derivatives of the three germ layers and are promising tools in regenerative medicine. Cardiovascular diseases are the top-ranking cause of premature death worldwide, and cell replacement therapies based on in vitro differentiated cardiomyocytes might provide a promising perspective to cure patients in the future. The molecular processes during hPSC cardiomyogenesis are far from being fully understood, and we thus have focused here on characterizing the proteome along hESC in vitro differentiation into cardiomyocytes (CMs). Stable isotope labeling of amino acids in cell culture was applied to quantitatively assess the proteome throughout defined stages of hESC cardiomyogenesis. Genetically enriched, >90% pure CM populations were used for shotgun proteomics, leading to the identification and quantitative determination of several thousand proteins. Pathway analysis revealed alterations in energy metabolism during cardiomyogenesis. Enzymes of glycolysis were identified as up-regulated upon differentiation, whereas enzymes involved in oxidative phosphorylation were down-regulated in aggregates on day 20 of differentiation (<10% CMs) and reconstituted on day 35 in >90% pure CMs. A structural protein that attracted our attention was the PDZ and LIM domain containing protein 5 (PDLIM5), which was strongly up-regulated during cardiomyogenesis and for which we detected novel stage-specific isoforms. Notably, expression of the 53 kDa isoforms b and g (corresponding to transcript variants 2 and 7) of PDLIM5 occurred simultaneously to the onset of expression of the early cardiac transcription factor NKX2.5, known to play a key role in cardiac development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Enzymes/metabolism , Gene Expression Regulation , LIM Domain Proteins/metabolism , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Proteomics/methods , Adaptor Proteins, Signal Transducing/chemistry , Cell Differentiation , Cells, Cultured , Energy Metabolism , Glycolysis , Homeobox Protein Nkx-2.5/analysis , Humans , Isotope Labeling , LIM Domain Proteins/chemistry , Metabolic Networks and Pathways , Oxidative Phosphorylation , Protein Isoforms/genetics , Regenerative Medicine
5.
Stem Cell Reports ; 8(2): 305-317, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28089668

ABSTRACT

Subtype-specific human cardiomyocytes (CMs) are valuable for basic and applied research. Induction of cardiomyogenesis and enrichment of nodal-like CMs was described for mouse pluripotent stem cells (mPSCs) in response to 1-ethyl-2-benzimidazolinone (EBIO), a chemical modulator of small-/intermediate-conductance Ca2+-activated potassium channels (SKs 1-4). Investigating EBIO in human pluripotent stem cells (PSCs), we have applied three independent differentiation protocols of low to high cardiomyogenic efficiency. Equivalent to mPSCs, timed EBIO supplementation during hPSC differentiation resulted in dose-dependent enrichment of up to 80% CMs, including an increase in nodal- and atrial-like phenotypes. However, our study revealed extensive EBIO-triggered cell loss favoring cardiac progenitor preservation and, subsequently, CMs with shortened action potentials. Proliferative cells were generally more sensitive to EBIO, presumably via an SK-independent mechanism. Together, EBIO did not promote cardiogenic differentiation of PSCs, opposing previous findings, but triggered lineage-selective survival at a cardiac progenitor stage, which we propose as a pharmacological strategy to modulate CM subtype composition.


Subject(s)
Benzimidazoles/pharmacology , Calcium Channel Agonists/pharmacology , Cell Differentiation/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Biomarkers , Cell Differentiation/genetics , Cell Line , Cell Lineage , Cell Survival/drug effects , Cells, Cultured , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/embryology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism
6.
Stem Cell Reports ; 3(6): 1132-46, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25454631

ABSTRACT

To harness the potential of human pluripotent stem cells (hPSCs), an abundant supply of their progenies is required. Here, hPSC expansion as matrix-independent aggregates in suspension culture was combined with cardiomyogenic differentiation using chemical Wnt pathway modulators. A multiwell screen was scaled up to stirred Erlenmeyer flasks and subsequently to tank bioreactors, applying controlled feeding strategies (batch and cyclic perfusion). Cardiomyogenesis was sensitive to the GSK3 inhibitor CHIR99021 concentration, whereas the aggregate size was no prevailing factor across culture platforms. However, in bioreactors, the pattern of aggregate formation in the expansion phase dominated subsequent differentiation. Global profiling revealed a culture-dependent expression of BMP agonists/antagonists, suggesting their decisive role in cell-fate determination. Furthermore, metallothionein was discovered as a potentially stress-related marker in hPSCs. In 100 ml bioreactors, the production of 40 million predominantly ventricular-like cardiomyocytes (up to 85% purity) was enabled that were directly applicable to bioartificial cardiac tissue formation.


Subject(s)
Batch Cell Culture Techniques , Cell Culture Techniques , Cell Differentiation , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Bioreactors , Culture Media , Culture Media, Conditioned , Humans , Membrane Potentials , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism
7.
Hum Gene Ther Methods ; 25(2): 136-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24483184

ABSTRACT

Human pluripotent stem cells (hPSCs) represent a prime cell source for pharmacological research and regenerative therapies because of their extensive expansion potential and their ability to differentiate into essentially all somatic lineages in vitro. Improved methods to stably introduce multiple transgenes into hPSCs will promote, for example, their preclinical testing by facilitating lineage differentiation and purification in vitro and the subsequent in vivo monitoring of respective progenies after their transplantation into relevant animal models. To date, the establishment of stable transgenic hPSC lines is still laborious and time-consuming. Current limitations include the low transfection efficiency of hPSCs via nonviral methods, the inefficient recovery of genetically engineered clones, and the silencing of transgene expression. Here we describe a fast, electroporation-based method for the generation of multitransgenic hPSC lines by overcoming the need for any preadaptation of conventional hPSC cultures to feeder-free conditions before genetic manipulation. We further show that the selection for a single antibiotic resistance marker encoded on one plasmid allowed for the stable genomic (co-)integration of up to two additional, independent expression plasmids. The method thereby enables the straightforward, nonviral generation of valuable multitransgenic hPSC lines in a single step. Practical applicability of the method is demonstrated for antibiotic-based lineage enrichment in vitro and for sodium iodide symporter transgene-based in situ cell imaging after intramyocardial cell infusion into explanted pig hearts.


Subject(s)
Pluripotent Stem Cells/metabolism , Transgenes/genetics , Animals , Cell Differentiation , Cell Line , Drug Resistance/genetics , Genetic Vectors/metabolism , Heart/diagnostic imaging , Humans , Iodine Radioisotopes/chemistry , Mice , Models, Animal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Plasmids/metabolism , Pluripotent Stem Cells/cytology , Radionuclide Imaging , Rats , Swine , Symporters/genetics , Symporters/metabolism , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
8.
Am J Respir Crit Care Med ; 189(2): 167-82, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24279725

ABSTRACT

RATIONALE: Hereditary pulmonary alveolar proteinosis (hPAP) caused by granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor α-chain (CSF2RA) deficiency is a rare, life-threatening lung disease characterized by accumulation of proteins and phospholipids in the alveolar spaces. The disease is caused by a functional insufficiency of alveolar macrophages, which require GM-CSF signaling for terminal differentiation and effective degradation of alveolar proteins and phospholipids. Therapeutic options are extremely limited, and the pathophysiology underlying the defective protein degradation in hPAP alveolar macrophages remains poorly understood. OBJECTIVES: To further elucidate the cellular mechanisms underlying hPAP and evaluate novel therapeutic strategies, we here investigated the potential of hPAP patient-derived induced pluripotent stem cell (PAP-iPSCs) derived monocytes and macrophages. METHODS: Patient-specific PAP-iPSCs were generated from CD34(+) bone marrow cells of a CSF2RA-deficient patient with PAP. We assessed pluripotency, chromosomal integrity, and genetic correction of established iPSC lines. On hematopoietic differentiation, genetically corrected or noncorrected monocytes and macrophages were investigated in GM-CSF-dependent assays. MEASUREMENTS AND MAIN RESULTS: Although monocytes and macrophages differentiated from noncorrected PAP-iPSCs exhibited distinct defects in GM-CSF-dependent functions, such as perturbed CD11b activation, phagocytic activity, and STAT5 phosphorylation after GM-CSF exposure and lack of GM-CSF uptake, these defects were fully repaired on lentiviral gene transfer of a codon-optimized CSF2RA-cDNA. CONCLUSIONS: These data establish PAP-iPSC-derived monocytes and macrophages as a valid in vitro disease model of CSF2RA-deficient PAP, and introduce gene-corrected iPSC-derived monocytes and macrophages as a potential autologous cell source for innovative therapeutic strategies. Transplantation of such cells to patients with hPAP could serve as a paradigmatic proof for the potential of iPSC-derived cells in clinical gene therapy.


Subject(s)
Genetic Diseases, X-Linked/therapy , Genetic Therapy , Induced Pluripotent Stem Cells , Pulmonary Alveolar Proteinosis/therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/genetics , Child, Preschool , Female , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Humans , Macrophages, Alveolar/metabolism , Models, Biological , Monocytes/metabolism , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL