Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Stem Cells ; 13(1): 65-79, 2020 Mar 30.
Article in English | MEDLINE | ID: mdl-31887845

ABSTRACT

BACKGROUND AND OBJECTIVES: Pluripotent embryonic stem (ES) cells represent a perfect model system for the investigation of early developmental processes. Besides their differentiation into derivatives of the three primary germ layers, they can also be differentiated into derivatives of the 'fourth' germ layer, the neural crest (NC). Due to its multipotency, extensive migration and outstanding capacity to generate a remarkable number of different cell types, the NC plays a key role in early developmental processes. Cardiac neural crest (CNC) cells are a subpopulation of the NC, which are of crucial importance for precise cardiovascular and pharyngeal glands' development. CNC-associated malformations are rare, but always severe and life-threatening. Appropriate cell models could help to unravel underlying pathomechanisms and to develop new therapeutic options for relevant heart malformations. METHODS: Murine ES cells were differentiated according to a mesodermal-lineage promoting protocol. Expression profiles of ES cell-derived progeny at various differentiation stages were investigated on transcript and protein level. RESULTS: Comparative expression profiling of murine ES cell multilineage progeny versus undifferentiated ES cells confirmed differentiation into known cell derivatives of the three primary germ layers and provided evidence that ES cells have the capacity to differentiate into NC/CNC-like cells. Applying the NC/CNC cell-specific marker, 4E9R, an unambiguous identification of ES cell-derived NC/CNC-like cells was achieved. CONCLUSIONS: Our findings will facilitate the establishment of an ES cell-derived CNC cell model for the investigation of molecular pathways during cardiac development in health and disease.

2.
Stem Cell Res ; 25: 278-282, 2017 12.
Article in English | MEDLINE | ID: mdl-29246574

ABSTRACT

The elucidation of molecular mechanisms that restrict the potential of pluripotent stem cells and promote cardiac lineage differentiation is of crucial relevance, since embryonic stem cells (ESCs) hold great potential for cell based heart therapies. The homeodomain transcription factor Shox2 is essential for the development and proper function of the native cardiac pacemaker, the sinoatrial node. This prompted us to develop a cardiac differentiation model using ESC lines isolated from blastocysts of Shox2-deficient mice. The established cell model provides a fundamental basis for the investigation of molecular pathways under physiological and pathophysiological conditions for evaluating novel therapeutic approaches.


Subject(s)
Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Sinoatrial Node/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism
3.
GMS J Med Educ ; 34(2): Doc22, 2017.
Article in English | MEDLINE | ID: mdl-28584870

ABSTRACT

Introduction: The German Council of Science and Humanities as well as a number of medical professional associations support the strengthening of scientific competences by developing longitudinal curricula for teaching scientific competences in the undergraduate medical education. The National Competence Based Catalogue of Learning Objectives for Undergraduate Medical Education (NKLM) has also defined medical scientific skills as learning objectives in addition to the role of the scholar. The development of the Mannheim science curriculum started with a systematic inventory of the teaching of scientific competences in the Mannheim Reformed Curriculum of Medicine (MaReCuM). Methods: The inventory is based on the analysis of module profiles, teaching materials, surveys among experts, and verbatims from memory. Furthermore, science learning objectives were defined and prioritized, thus enabling the contents of the various courses to be assigned to the top three learning objectives. Results: The learning objectives systematic collection of information regarding the current state of research, critical assessment of scientific information and data sources, as well as presentation and discussion of the results of scientific studies are facilitated by various teaching courses from the first to the fifth year of undergraduate training. The review reveals a longitudinal science curriculum that has emerged implicitly. Future efforts must aim at eliminating redundancies and closing gaps; in addition, courses must be more closely aligned with each other, regarding both their contents and their timing, by means of a central coordination unit. Conclusion: The teaching of scientific thinking and working is a central component in the MaReCuM. The inventory and prioritization of science learning objectives form the basis for a structured ongoing development of the curriculum. An essential aspect here is the establishment of a central project team responsible for the planning, coordination, and review of these measures.


Subject(s)
Clinical Competence/standards , Curriculum/standards , Education, Medical, Undergraduate/organization & administration , Equipment and Supplies/standards , Models, Educational , Science/education , Germany , Humans , Longitudinal Studies
4.
Stem Cell Res ; 21: 51-57, 2017 05.
Article in English | MEDLINE | ID: mdl-28390247

ABSTRACT

The homeodomain transcription factor Shox2 controls the development and function of the native cardiac pacemaker, the sinoatrial node (SAN). Moreover, SHOX2 mutations have been associated with cardiac arrhythmias in humans. For detailed examination of Shox2-dependent developmental mechanisms in SAN cells, we established a murine embryonic stem cell (ESC)-based model using Shox2 as a molecular tool. Shox2+/+ and Shox2-/- ESC clones were isolated and differentiated according to five different protocols in order to evaluate the most efficient enrichment of SAN-like cells. Expression analysis of cell subtype-specific marker genes revealed most efficient enrichment after CD166-based cell sorting. Comparative cardiac expression profiles of Shox2+/+ and Shox2-/- ESCs were examined by nCounter technology. Among other genes, we identified Nppb as a novel putative Shox2 target during differentiation in ESCs. Differential expression of Nppb could be confirmed in heart tissue of Shox2-/- embryos. Taken together, we established an ESC-based cardiac differentiation model and successfully purified Shox2+/+ and Shox2-/- SAN-like cells. This now provides an excellent basis for the investigation of molecular mechanisms under physiological and pathophysiological conditions for evaluating novel therapeutic approaches.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Sinoatrial Node/cytology , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , Cell Separation , Female , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism
5.
Stem Cells Int ; 2011: 130970, 2011.
Article in English | MEDLINE | ID: mdl-21941564

ABSTRACT

Transcription factor Lbx1 is known to play a role in the migration of muscle progenitor cells in limb buds and also in neuronal determination processes. In addition, involvement of Lbx1 in cardiac neural crest-related cardiogenesis was postulated. Here, we used mouse embryonic stem (ES) cells which have the capacity to develop into cells of all three primary germ layers. During in vitro differentiation, ES cells recapitulate cellular developmental processes and gene expression patterns of early embryogenesis. Transcript analysis revealed a significant upregulation of Lbx1 at the progenitor cell stage. Immunofluorescence staining confirmed the expression of Lbx1 in skeletal muscle cell progenitors and GABAergic neurons. To verify the presence of Lbx1 in cardiac cells, triple immunocytochemistry of ES cell-derived cardiomyocytes and a quantification assay were performed at different developmental stages. Colabeling of Lbx1 and cardiac specific markers troponin T, α-actinin, GATA4, and Nkx2.5 suggested a potential role in early myocardial development.

6.
Int J Dev Biol ; 54(1): 41-54, 2010.
Article in English | MEDLINE | ID: mdl-19876843

ABSTRACT

We have previously shown that mouse embryonic stem (ES) cells differentiate into insulin-positive cells via multi-lineage progenitors. Here, we used Affymetrix chips and quantitative RT-PCR analysis to determine transcriptional profiles of undifferentiated wildtype (wt) and Pax4 expressing (Pax4+) ES cells and differentiated cells of committed progenitor and advanced stages. From undifferentiated to the committed stage, 237 (wt) and 263 (Pax4+) transcripts were 5- or more-fold up-regulated, whereas from the committed to the advanced stage, 28 (wt) and 5 (Pax4+) transcripts, respectively, were two- or more-fold up-regulated. Transcripts were classified into main subclasses including transcriptional regulation, signalling/growth factors, adhesion/extracellular matrix, membrane/transport, metabolism and organogenesis. Remarkably, endoderm-specific Sox17 and early pancreas-specific Isl1 transcripts were up-regulated at an earlier stage of multi-lineage progenitors, whereas highly up-regulated probe sets and transcripts of genes involved in endoderm, pancreatic, hepatic, angiogenic and neural differentiation were detected at the committed progenitor stage. Pax4+ cells showed specific differences in transcript up-regulation and a lower amount of up-regulated neural-specific transcripts in comparison to wt cells, but no enhanced gene expression complexity. Immunocytochemical analysis of selected proteins involved in endoderm and pancreatic differentiation, such as chromogranin B, transthyretin, Foxa1 and neuronatin revealed co-expression with insulin- or C-peptide-positive cells. The comparison of transcript profiles of ES cells differentiating in vitro with those of the embryonic and adult pancreas in vivo suggested that in vitro differentiated cells resemble an embryonal stage of development, supporting the view that ES-derived pancreatic cells are unable to complete pancreatic differentiation in vitro.


Subject(s)
Cell Differentiation , Cell Lineage , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Insulin-Secreting Cells/metabolism , Pancreas/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Immunoenzyme Techniques , In Vitro Techniques , Mice , Oligonucleotide Array Sequence Analysis , Paired Box Transcription Factors/metabolism , Pancreas/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
PLoS One ; 4(9): e6804, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19727443

ABSTRACT

Embryonic stem (ES) cells have high self-renewal capacity and the potential to differentiate into a large variety of cell types. To investigate gene networks operating in pluripotent ES cells and their derivatives, the "Functional Genomics in Embryonic Stem Cells" consortium (FunGenES) has analyzed the transcriptome of mouse ES cells in eleven diverse settings representing sixty-seven experimental conditions. To better illustrate gene expression profiles in mouse ES cells, we have organized the results in an interactive database with a number of features and tools. Specifically, we have generated clusters of transcripts that behave the same way under the entire spectrum of the sixty-seven experimental conditions; we have assembled genes in groups according to their time of expression during successive days of ES cell differentiation; we have included expression profiles of specific gene classes such as transcription regulatory factors and Expressed Sequence Tags; transcripts have been arranged in "Expression Waves" and juxtaposed to genes with opposite or complementary expression patterns; we have designed search engines to display the expression profile of any transcript during ES cell differentiation; gene expression data have been organized in animated graphs of KEGG signaling and metabolic pathways; and finally, we have incorporated advanced functional annotations for individual genes or gene clusters of interest and links to microarray and genomic resources. The FunGenES database provides a comprehensive resource for studies into the biology of ES cells.


Subject(s)
Databases, Genetic , Genomics , Stem Cells/cytology , Animals , Cell Differentiation , Cell Line , Cluster Analysis , Expressed Sequence Tags , Gene Expression Profiling , Mice , Multigene Family , Oligonucleotide Array Sequence Analysis , Signal Transduction , Software
8.
Biol Chem ; 390(9): 845-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19558327

ABSTRACT

Adult cells have been reprogrammed into induced pluripotent stem (iPS) cells by introducing pluripotency-associated transcription factors. Here, we discuss recent advances and challenges of in vitro reprogramming and future prospects of iPS cells for their use in diagnosis and cell therapy. The generation of patient-specific iPS cells for clinical application requires alternative strategies, because genome-integrating viral vectors may cause insertional mutagenesis. Moreover, when suitable iPS cell lines will be available, efficient and selective differentiation protocols are needed to generate transplantable grafts. Finally, we point to the requirement of a regulatory framework necessary for the commercial use of iPS cells.


Subject(s)
Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Cell Differentiation , Cell- and Tissue-Based Therapy/methods , Humans , Models, Biological , Pluripotent Stem Cells/metabolism
9.
BMC Cell Biol ; 10: 46, 2009 Jun 17.
Article in English | MEDLINE | ID: mdl-19534768

ABSTRACT

BACKGROUND: P53 is a key tumor suppressor protein. In response to DNA damage, p53 accumulates to high levels in differentiated cells and activates target genes that initiate cell cycle arrest and apoptosis. Since stem cells provide the proliferative cell pool within organisms, an efficient DNA damage response is crucial. RESULTS: In proliferating embryonic stem cells, p53 is localized predominantly in the cytoplasm. DNA damage-induced nuclear accumulation of p53 in embryonic stem cells activates transcription of the target genes mdm2, p21, puma and noxa. We observed bi-phasic kinetics for nuclear accumulation of p53 after ionizing radiation. During the first wave of nuclear accumulation, p53 levels were increased and the p53 target genes mdm2, p21 and puma were transcribed. Transcription of noxa correlated with the second wave of nuclear accumulation. Transcriptional activation of p53 target genes resulted in an increased amount of proteins with the exception of p21. While p21 transcripts were efficiently translated in 3T3 cells, we failed to see an increase in p21 protein levels after IR in embryonal stem cells. CONCLUSION: In embryonic stem cells where (anti-proliferative) p53 activity is not necessary, or even unfavorable, p53 is retained in the cytoplasm and prevented from activating its target genes. However, if its activity is beneficial or required, p53 is allowed to accumulate in the nucleus and activates its target genes, even in embryonic stem cells.


Subject(s)
Cell Nucleus/metabolism , DNA Damage , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis Regulatory Proteins , Cell Line , Cell Proliferation , Cell Survival , Cytoplasm/metabolism , Embryonic Stem Cells/radiation effects , Gamma Rays , Homeodomain Proteins/genetics , Mice , Nanog Homeobox Protein , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuro-Oncological Ventral Antigen , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/isolation & purification , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Methods Mol Biol ; 530: 219-50, 2009.
Article in English | MEDLINE | ID: mdl-19266345

ABSTRACT

Pluripotent embryonic stem (ES) cells are characterized by their almost unlimited potential to self-renew and to differentiate into virtually any cell type of the organism. Here we describe basic protocols for the in vitro differentiation of mouse ES cells into cells of the cardiac, neuronal, pancreatic, and hepatic lineage. The protocols include (1) the formation of embryoid bodies (EBs) followed by (2) the spontaneous differentiation of EBs into progenitor cells of the ecto-, endo-, and mesodermal germ layer and (3) the directed differentiation of early progenitors into the respective lineages. Differentiation induction via growth and extracellular matrix factors leads to titin-expressing spontaneously beating cardiac cells, tyrosine hydroxylase-expressing dopaminergic neurons, insulin and c-peptide co-expressing pancreatic islet-like clusters, and albumin-positive hepatic cells, respectively. The differentiated cells show tissue-specific proteins and electrophysiological properties (action potentials and ion channels) in cardiac and neuronal cells, glucose-dependent insulin release in pancreatic cells, or glycogen storage and albumin synthesis in hepatic cells. The protocols presented here provide basic systems to study differentiation processes in vitro and to establish strategies for the use of stem cells in regenerative therapies.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Biomarkers/analysis , Mice
11.
Cells Tissues Organs ; 188(1-2): 103-15, 2008.
Article in English | MEDLINE | ID: mdl-18600024

ABSTRACT

Hematopoietic stem cells maintain the development of all mature blood cells throughout life due to their sustained self-renewal capacity and multilineage differentiation potential. During development into specific cell lineages, the options of stem cells and multipotent progenitor cells become increasingly restricted concomitant with a successive decline in self-renewal potential. Here we describe an Flt3+CD11b+ multipotent progenitor that can be amplified in vitro with a specific combination of cytokines to yield homogeneous populations in high cell numbers. By employing gene expression profiling with DNA microarrays, we studied the transcription factor repertoire of Flt3+CD11b+ progenitors and related it to the transcription factor repertoire of hematopoietic stem cells and embryonic stem cells. We report here on overlapping and nonoverlapping expression patterns of transcription factors in these cells and thus provide novel insights into the dynamic networks of transcriptional regulators in embryonic and adult stem cells. Additionally, the results obtained open the perspective for elucidating lineage and 'stemness' determinants in hematopoiesis.


Subject(s)
Hematopoietic Stem Cells/metabolism , Transcription Factors/genetics , fms-Like Tyrosine Kinase 3/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , CD11b Antigen/metabolism , Cell Line , Cluster Analysis , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phenotype , Transcription Factors/metabolism
12.
Stem Cells ; 26(4): 920-6, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18203677

ABSTRACT

Chromatin architecture in stem cells determines the pattern of gene expression and thereby cell identity and fate. The chromatin-modifying agents trichostatin A (TSA) and 5-Aza-2'-deoxycytidine (AzaC) affect histone acetylation and DNA methylation, respectively, and thereby influence chromatin structure and gene expression. In our previous work, we demonstrated that TSA/AzaC treatment of neurosphere cells induces hematopoietic activity in vivo that is long-term, multilineage, and transplantable. Here, we have analyzed the TSA/AzaC-induced changes in gene expression by global gene expression profiling. TSA/AzaC caused both up- and downregulation of genes, without increasing the total number of expressed genes. Chromosome analysis showed no hot spot of TSA/AzaC impact on a particular chromosome or chromosomal region. Hierarchical cluster analysis revealed common gene expression patterns among neurosphere cells treated with TSA/AzaC, embryonic stem (ES) cells, and hematopoietic stem cells. Furthermore, our analysis identified several stem cell genes and pluripotency-associated genes that are induced by TSA/AzaC in neurosphere cells, including Cd34, Cd133, Oct4, Nanog, Klf4, Bex1, and the Dppa family members Dppa2, 3, 4, and 5. Sox2 and c-Myc are constitutively expressed in neurosphere cells. We propose a model in which TSA/AzaC, by removal of epigenetic inhibition, induces the reactivation of several stem cell and pluripotency-associated genes, and their coordinate expression enlarges the differentiation potential of somatic precursor cells.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Developmental/genetics , Neurons/physiology , Pluripotent Stem Cells/physiology , Animals , Cells, Cultured , Chromatin/metabolism , Gene Expression Regulation, Developmental/drug effects , Humans , Hydroxamic Acids/pharmacology , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/physiology
13.
Lab Chip ; 7(6): 777-85, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17538721

ABSTRACT

We describe a multi-purpose platform for the three-dimensional cultivation of tissues. The device is composed of polymer chips featuring a microstructured area of 1-2 cm(2). The chip is constructed either as a grid of micro-containers measuring 120-300 x 300 x 300 microm (h x l x w), or as an array of round recesses (300 microm diameter, 300 microm deep). The micro-containers may be separately equipped with addressable 3D-micro-electrodes, which allow for electrical stimulation of excitable cells and on-site measurements of electrochemically accessible parameters. The system is applicable for the cultivation of high cell densities of up to 8 x 10(6) cells and, because of the rectangular grid layout, allows the automated microscopical analysis of cultivated cells. More than 1000 micro-containers enable the parallel analysis of different parameters under superfusion/perfusion conditions. Using different polymer chips in combination with various types of bioreactors we demonstrated the principal suitability of the chip-based bioreactor for tissue culture applications. Primary and established cell lines have been successfully cultivated and analysed for functional properties. When cells were cultured in non-perfused chips, over time a considerable degree of apoptosis could be observed indicating the need for an active perfusion. The system presented here has also been applied for the differentiation analysis of pluripotent embryonic stem cells and may be suitable for the analysis of the stem cell niche.


Subject(s)
Bioreactors , Embryonic Stem Cells/cytology , Microfluidics/methods , Pluripotent Stem Cells/cytology , Polymers/chemistry , Tissue Engineering , Animals , Cell Line , Cell Proliferation , Cell Survival , Electrochemistry , Embryonic Stem Cells/physiology , Embryonic Stem Cells/ultrastructure , Equipment Design , Microfluidics/instrumentation , Microscopy, Electron, Scanning , Pluripotent Stem Cells/physiology , Pluripotent Stem Cells/ultrastructure , Rats
14.
Cell Signal ; 19(2): 367-77, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16978842

ABSTRACT

Sphingosylphosphorylcholine (SPC) is the major component of high-density lipoproteins (HDL) in blood plasma. The bioactive lipid acts mainly via G protein coupled receptors (GPCRs). Similar to ligands of other GPCRs, SPC has multiple biological roles including the regulation of proliferation, migration, angiogenesis, wound healing and heart rate. Lysophospholipids and their receptors have also been implicated in cell differentiation. A potential role of SPC in stem cell or tumour cell differentiation has been elusive so far. Here we examined the effect of SPC on the differentiation of mouse embryonic stem (ES) cells and of human NB4 promyelocytic leukemia cells, a well established tumour differentiation model. Our data show that mouse embryonic stem cells and NB4 cells express the relevant GPCRs for SPC. We demonstrate both at the level of morphology and of gene expression that SPC induces neuronal and cardiac differentiation of mouse ES cells. Furthermore, SPC induces differentiation of NB4 cells by a mechanism which is critically dependent on the activity of the MEK-ERK cascade. Thus, the bioactive lipid SPC is a novel differentiation inducing agent both for mouse ES cells, but also of certain human tumour cells.


Subject(s)
Embryonic Stem Cells/physiology , Leukemia, Promyelocytic, Acute/metabolism , Phosphorylcholine/analogs & derivatives , Receptors, G-Protein-Coupled/metabolism , Sphingosine/analogs & derivatives , Actins/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Embryonic Stem Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesions , Humans , Mice , Phosphorylcholine/pharmacology , Sphingosine/pharmacology
15.
Differentiation ; 74(9-10): 499-509, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17177847

ABSTRACT

In mouse blastocysts six facilitative glucose transporter isoforms (GLUT)1-4, 8 and 9 are expressed. We have used the mouse embryonic stem (ES) cell line D3 and spontaneously differentiating embryoid bodies (EB) to investigate GLUT expression and the influence of glucose during differentiation of early embryonic cells. Both ES cells and EBs (2d-20d) expressed GLUT1, 3, and 8, whereas the isoforms 2 and 4 were detectable exclusively in EBs. Differentiation-associated expression of GLUT was analyzed by double staining with stage-specific embryonic antigen (SSEA-1), cytokeratins (CK18, 19), nestin, and desmin. Similar to trophoblast cells in mouse blastocysts the outer cell layer of endoderm-like cells showed a high GLUT3 expression in early EBs. In 20-day-old EBs no GLUT3 protein and only minor GLUT3 mRNA amounts could be detected. A minimal glucose concentration of 5 mM applied during 2 and 8 days of EB culture resulted in up-regulated GLUT4, Oct-4 and SSEA-1 levels and a delay in EB differentiation. We conclude that GLUT expression depends on cellular differentiation and that the expression is modulated by glucose concentration. The developmental and glucose-dependent regulation of GLUT strongly suggests a functional role of glucose and glucose transporters in ES cell differentiation and embryonic development.


Subject(s)
Embryonic Stem Cells/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Gene Expression , Glucose Transport Proteins, Facilitative/analysis , Glucose Transport Proteins, Facilitative/genetics , Immunohistochemistry , Keratins/metabolism , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism
16.
Methods Mol Biol ; 325: 181-205, 2006.
Article in English | MEDLINE | ID: mdl-16761727

ABSTRACT

Embryonic stem (ES) cells, the pluripotent cells of early embryos have been successfully cultured as undifferentiated cells. The cells are characterized by two unique properties, unlimited self-renewal capacity and the ability to differentiate into all cells of the body. Because of the high in vitro differentiation potential, ES cells have been used as model system in cell and developmental biology. Here we present methods that use mouse embryonic stem cells for the in vitro differentiation and characterization of neuronal, cardiac, pancreatic and hepatic cells, derivatives of the ectoderm, mesoderm and endoderm, respectively. In the future, differentiated cells may be also generated from human ES cells by cultivation of early embryos or from reprogrammed cells derived by nuclear transfer. Such cells could represent potential sources for tissue repair of serious human diseases.


Subject(s)
Cell Culture Techniques/methods , Embryo, Mammalian/cytology , Stem Cells/cytology , Animals , Cell Adhesion , Cell Differentiation , Culture Media/pharmacology , Enzyme-Linked Immunosorbent Assay , Liver/metabolism , Mice , Myocytes, Cardiac/cytology , Neurons/metabolism , Pancreas/metabolism
17.
Stem Cells ; 24(9): 2085-97, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16741226

ABSTRACT

The intestinal epithelium has one of the greatest regenerative capacities in the body; however, neither stem nor progenitor cells have been successfully cultivated from the intestine. In this study, we applied an "artificial niche" of mouse embryonic fibroblasts to derive multipotent cells from the intestinal epithelium. Cocultivation of adult mouse and human intestinal epithelium with fibroblast feeder cells led to the generation of a novel type of nestin-positive cells (intestinal epithelium-derived nestin-positive cells [INPs]). Transcriptome analyses demonstrated that mouse embryonic fibroblasts expressed relatively high levels of Wnt/bone morphogenetic protein (BMP) transcripts, and the formation of INPs was specifically associated with an increase in Lef1, Wnt4, Wnt5a, and Wnt/BMP-responsive factors, but a decrease of BMP4 transcript abundance. In vitro, INPs showed a high but finite proliferative capacity and readily differentiated into cells expressing neural, pancreatic, and hepatic transcripts and proteins; however, these derivatives did not show functional properties. In vivo, INPs failed to form chimeras following injection into mouse blastocysts but integrated into hippocampal brain slice cultures in situ. We conclude that the use of embryonic fibroblasts seems to reprogram adult intestinal epithelial cells by modulation of Wnt/BMP signaling to a cell type with a more primitive embryonic-like stage of development that has a high degree of flexibility and plasticity.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Enterocytes/cytology , Fibroblasts/cytology , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Cells, Cultured , Ectoderm/cytology , Endoderm/cytology , Gene Expression Profiling , Humans , Mice , Nestin , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics , Wnt Proteins/genetics
18.
Nat Protoc ; 1(2): 495-507, 2006.
Article in English | MEDLINE | ID: mdl-17406275

ABSTRACT

Here, we describe a basic protocol for the in vitro differentiation of mouse embryonic stem (ES) cells into insulin-producing cells. The three-step protocol comprises (i) the formation of embryoid bodies, (ii) the spontaneous differentiation of embryoid bodies into progenitor cells of ecto-, meso- and endodermal lineages, and (iii) the induction of differentiation of early progenitors into the pancreatic lineage. Differentiated cells can be obtained within approximately 33 d. Differentiation induction by growth and extracellular-matrix factors, including laminin, nicotinamide and insulin, leads to the formation of ES-derived progeny that resembles cells committed to the pancreatic lineage. During differentiation, transcript levels of genes expressed in early pancreatic cells are upregulated. Continued differentiation results in the development of C-peptide/insulin-positive islet-like clusters that release insulin upon glucose stimulation. Differentiated ES cells that overexpress the pancreatic developmental control gene Pax4 develop insulin-secretory granules and reveal functional properties with respect to the pancreas-specific ATP-modulated K+ channel and the normalization of glycemia of streptozotocin-treated diabetic mice.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Animals , Cell Line , Homeodomain Proteins/metabolism , Insulin/metabolism , Mice , Paired Box Transcription Factors/metabolism
19.
FASEB J ; 19(12): 1686-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16116041

ABSTRACT

Mouse embryonic stem (ES) cells were used as an experimental model to study the effects of electromagnetic fields (EMF). ES-derived nestin-positive neural progenitor cells were exposed to extremely low frequency EMF simulating power line magnetic fields at 50 Hz (ELF-EMF) and to radiofrequency EMF simulating the Global System for Mobile Communication (GSM) signals at 1.71 GHz (RF-EMF). Following EMF exposure, cells were analyzed for transcript levels of cell cycle regulatory, apoptosis-related, and neural-specific genes and proteins; changes in proliferation; apoptosis; and cytogenetic effects. Quantitative RT-PCR analysis revealed that ELF-EMF exposure to ES-derived neural cells significantly affected transcript levels of the apoptosis-related bcl-2, bax, and cell cycle regulatory "growth arrest DNA damage inducible" GADD45 genes, whereas mRNA levels of neural-specific genes were not affected. RF-EMF exposure of neural progenitor cells resulted in down-regulation of neural-specific Nurr1 and in up-regulation of bax and GADD45 mRNA levels. Short-term RF-EMF exposure for 6 h, but not for 48 h, resulted in a low and transient increase of DNA double-strand breaks. No effects of ELF- and RF-EMF on mitochondrial function, nuclear apoptosis, cell proliferation, and chromosomal alterations were observed. We may conclude that EMF exposure of ES-derived neural progenitor cells transiently affects the transcript level of genes related to apoptosis and cell cycle control. However, these responses are not associated with detectable changes of cell physiology, suggesting compensatory mechanisms at the translational and posttranslational level.


Subject(s)
Apoptosis , Electromagnetic Fields , Embryo, Mammalian/cytology , Embryo, Nonmammalian , Neurons/radiation effects , Stem Cells/cytology , Stem Cells/radiation effects , Transcription, Genetic/radiation effects , Cell Proliferation , Comet Assay , DNA/chemistry , DNA Damage , Down-Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Neurons/metabolism , Protein Biosynthesis , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Up-Regulation , bcl-2-Associated X Protein/metabolism , GADD45 Proteins
20.
Toxicol Lett ; 149(1-3): 361-9, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15093282

ABSTRACT

Primary cultures or established cell lines of vertebrates are commonly used to analyse the cytotoxic potential of chemical factors, drugs and xenobiotics in vitro. An alternative approach will be provided by permanent lines of pluripotent embryonic stem (ES) cells, which are able to differentiate into specialised somatic cell types in vitro. Here, we demonstrate the capacity of ES cells to generate functional cardiac, neuronal and pancreatic cells. We show that during ES cell differentiation, tissue-specific genes, proteins as well as functional properties are expressed in a developmentally regulated manner recapitulating processes of early embryonic development. We present data that show the use of ES-derived cardiomyocytes and dopaminergic neurons in toxicological studies and the potential of ES-derived pancreatic beta-like cells in future in vitro assays. The application of these differentiation systems to human ES cells opens up new perspectives in basic and applied toxicology.


Subject(s)
Myocardium/cytology , Neurons/drug effects , Pancreas/drug effects , Stem Cells/drug effects , Toxicity Tests , Cell Differentiation/drug effects , Cells, Cultured , Dopamine/physiology , Electrophysiology , Fluorescent Antibody Technique , Humans , Pancreas/cytology , Patch-Clamp Techniques , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...