Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Res Sq ; 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37886443

ABSTRACT

Brain tumor patients are commonly treated with radiotherapy, but the efficacy of the treatment is limited by its toxicity, particularly the risk of radionecrosis. We used human cerebral organoids to investigate the mechanisms and nature of postirradiation brain image changes commonly linked to necrosis. Irradiation of cerebral organoids lead to increased formation of ZO1+/AQP1+/CLN3+-choroid plexus (CP) structures. Increased CP formation was triggered by radiation via the NOTCH/WNT signaling pathways and associated with delayed growth and neural stem cell differentiation, but not necrosis. The effect was more pronounced in immature than in mature organoids, reflecting the clinically-observed increased radiosensitivity of the pediatric brain. Protons were more effective than X-rays at the same dose, as also observed in clinical treatments. We conclude that radiation-induced brain image-changes can be attributed to aberrant CP formation, providing a new cellular mechanism and strategy for possible countermeasures.

2.
Bioengineering (Basel) ; 9(5)2022 May 20.
Article in English | MEDLINE | ID: mdl-35621498

ABSTRACT

Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.

3.
Cells ; 10(10)2021 09 30.
Article in English | MEDLINE | ID: mdl-34685588

ABSTRACT

The heart tissue is a potential target of various noxae contributing to the onset of cardiovascular diseases. However, underlying pathophysiological mechanisms are largely unknown. Human stem cell-derived models are promising, but a major concern is cell immaturity when estimating risks for adults. In this study, 3D aggregates of human embryonic stem cell-derived cardiomyocytes were cultivated for 300 days and characterized regarding degree of maturity, structure, and cell composition. Furthermore, effects of ionizing radiation (X-rays, 0.1-2 Gy) on matured aggregates were investigated, representing one of the noxae that are challenging to assess. Video-based functional analyses were correlated to changes in the proteome after irradiation. Cardiomyocytes reached maximum maturity after 100 days in cultivation, judged by α-actinin lengths, and displayed typical multinucleation and branching. At this time, aggregates contained all major cardiac cell types, proven by the patch-clamp technique. Matured and X-ray-irradiated aggregates revealed a subtle increase in beat rates and a more arrhythmic sequence of cellular depolarisation and repolarisation compared to non-irradiated sham controls. The proteome analysis provides first insights into signaling mechanisms contributing to cardiotoxicity. Here, we propose an in vitro model suitable to screen various noxae to target adult cardiotoxicity by preserving all the benefits of a 3D tissue culture.


Subject(s)
Cell Differentiation/drug effects , Human Embryonic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Noxae/pharmacology , X-Rays , Adult , Cardiotoxicity/drug therapy , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Noxae/metabolism
4.
Nucleic Acids Res ; 49(13): 7457-7475, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34165573

ABSTRACT

Using human embryonic, adult and cancer stem cells/stem cell-like cells (SCs), we demonstrate that DNA replication speed differs in SCs and their differentiated counterparts. While SCs decelerate DNA replication, differentiated cells synthesize DNA faster and accumulate DNA damage. Notably, both replication phenotypes depend on p53 and polymerase iota (POLι). By exploring protein interactions and newly synthesized DNA, we show that SCs promote complex formation of p53 and POLι at replication sites. Intriguingly, in SCs the translocase ZRANB3 is recruited to POLι and required for slow-down of DNA replication. The known role of ZRANB3 in fork reversal suggests that the p53-POLι complex mediates slow but safe bypass of replication barriers in SCs. In differentiated cells, POLι localizes more transiently to sites of DNA synthesis and no longer interacts with p53 facilitating fast POLι-dependent DNA replication. In this alternative scenario, POLι associates with the p53 target p21, which antagonizes PCNA poly-ubiquitination and, thereby potentially disfavors the recruitment of translocases. Altogether, we provide evidence for diametrically opposed DNA replication phenotypes in SCs and their differentiated counterparts putting DNA replication-based strategies in the spotlight for the creation of therapeutic opportunities targeting SCs.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase/metabolism , Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Differentiation/genetics , Cells, Cultured , DNA Helicases/metabolism , Embryonic Stem Cells/metabolism , Humans , Neoplastic Stem Cells/metabolism , Stress, Physiological/genetics , DNA Polymerase iota
5.
Methods Mol Biol ; 2269: 25-33, 2021.
Article in English | MEDLINE | ID: mdl-33687669

ABSTRACT

In an increasingly geriatric population, in which elderly people frequently face chronic diseases and degenerative conditions, cell therapies as part of novel regenerative medicine approaches are of great interest. Even though today's cell therapies mostly rely on adult stem cells like the mesenchymal stem cells or primary somatic cells, pluripotent stem cells represent an enormously versatile cell model to explore possible new avenues in the field of regenerative medicine due to their capacity to grow indefinitely and to differentiate into the desired cell types. The discovery of reprogramming somatic cells into induced pluripotent stem cells augmented the pool of applicable cell entities so that researchers nowadays can resort to embryonic stem cells, but also to a plethora of patient- and disease-specific induced pluripotent stem cells. The ease of targeted genome engineering is an additional benefit that allows using pluripotent stem cells for disease modeling, drug discovery, and the development of cell therapies. However, the task is still demanding as the generation of subpopulations and a sufficient cell maturation for some cell entities have yet to be achieved. Likewise, even though for some applications the cells of interest can be produced in the large-scale dimensions and purity that are required for clinical purposes, proper integration, and function in the host tissue remain challenging. Nonetheless, the immense progress that has been made over the last decades warrants the prominent role of pluripotent stem cells in regenerative medicine as in vitro models to broaden our knowledge of disease onset/progression and treatment as well as in vivo as a substitution of damaged/aged tissue.


Subject(s)
Cell- and Tissue-Based Therapy , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Regenerative Medicine , Animals , Humans , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology
6.
Neurotoxicology ; 79: 40-47, 2020 07.
Article in English | MEDLINE | ID: mdl-32320710

ABSTRACT

Ionizing radiation (IR) is increasingly used for diagnostics and therapy of severe brain diseases. However, IR also has adverse effects on the healthy brain tissue, particularly on the neuronal network. This is true for adults but even more pronounced in the developing brain of unborn and pediatric patients. Epidemiological studies on children receiving radiotherapy showed an increased risk for cognitive decline ranging from mild deficits in academic functioning to severe late effects in intellectual ability and language as a consequence of altered neuronal development and connectivity. To provide a comprehensive approach for the analysis of radiation-induced alterations in human neuronal functionality, we developed an in vitro assay by combining microelectrode array (MEA) analyses and human embryonic stem cell (hESC) derived three-dimensional neurospheres (NS). In our proof of principle study, we irradiated hESC with 1 Gy X-rays and let them spontaneously differentiate into neurons within NS. After the onset of neuronal activity, we recorded and analyzed the activity pattern of the developing neuronal networks. The network activity in NS derived from irradiated hESC was significantly reduced, whereas no differences in molecular endpoints such as cell proliferation and transcript or protein expression analyses were found. Thus, the combination of MEA analysis with a 3D model for neuronal functionality revealed radiation sequela that otherwise would not have been detected. We therefore strongly suggest combining traditional biomolecular methods with the new functional assay presented in this work to improve the risk assessment for IR-induced effects on the developing brain.


Subject(s)
Human Embryonic Stem Cells/radiation effects , Nerve Net/radiation effects , Neural Stem Cells/radiation effects , Neurogenesis/radiation effects , Action Potentials/drug effects , Cell Culture Techniques/instrumentation , Cell Proliferation/radiation effects , Cells, Cultured , Gene Expression Regulation, Developmental/radiation effects , Human Embryonic Stem Cells/metabolism , Humans , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Nerve Net/metabolism , Neural Stem Cells/metabolism , Phenotype , Proof of Concept Study , Spheroids, Cellular
7.
Stem Cell Res ; 29: 115-124, 2018 05.
Article in English | MEDLINE | ID: mdl-29655161

ABSTRACT

Human embryonic stem cells (hESCs) differentiated into cardiomyocytes (CM) often develop into complex 3D structures that are composed of various cardiac cell types. Conventional methods to study the electrophysiology of cardiac cells are patch clamp and microelectrode array (MEAs) analyses. However, these methods are not suitable to investigate the contractile features of 3D cardiac clusters that detach from the surface of the culture dishes during differentiation. To overcome this problem, we developed a video-based motion detection software relying on the optical flow by Farnebäck that we call cBRA (cardiac beat rate analyzer). The beating characteristics of the differentiated cardiac clusters were calculated based on the local displacement between two subsequent images. Two differentiation protocols, which profoundly differ in the morphology of cardiac clusters generated and in the expression of cardiac markers, were used and the resulting CM were characterized. Despite these differences, beat rates and beating variabilities could be reliably determined using cBRA. Likewise, stimulation of ß-adrenoreceptors by isoproterenol could easily be identified in the hESC-derived CM. Since even subtle changes in the beating features are detectable, this method is suitable for high throughput cardiotoxicity screenings.


Subject(s)
Human Embryonic Stem Cells/cytology , Imaging, Three-Dimensional/methods , Myocytes, Cardiac/cytology , Cell Differentiation/physiology , Human Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/metabolism , Video Recording
8.
Stem Cells Dev ; 26(5): 341-352, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27937745

ABSTRACT

Exposure of the embryo to ionizing radiation (IR) is detrimental as it can cause genotoxic stress leading to immediate and latent consequences such as functional defects, malformations, or cancer. Human embryonic stem (hES) cells can mimic the preimplantation embryo and help to assess the biological effects of IR during early development. In this study, we describe the alterations H9 hES cells exhibit after X-ray irradiation in respect to cell cycle progression, apoptosis, genomic stability, stem cell signaling, and their capacity to differentiate into definitive endoderm. Early postirradiation, hES cells responded with an arrest in G2/M phase, elevated apoptosis, and increased chromosomal aberrations. Significant downregulation of stem cell signaling markers of the TGF beta-, Wnt-, and Hedgehog pathways was observed. Most prominent were alterations in the expression of activin receptors. However, hES cells responded differently depending on the culture conditions chosen for maintenance. Enzymatically passaged cells were less sensitive to IR than mechanically passaged ones showing fewer apoptotic cells and fewer changes in the stem cell signaling 24 h after irradiation, but displayed higher levels of chromosomal aberrations. Even though many of the observed changes were transient, surviving hES cells, which were differentiated 4 days postirradiation, showed a lower efficiency to form definitive endoderm than their mock-irradiated counterparts. This was demonstrated by lower expression levels of SOX17 and microRNA miR-375. In conclusion, hES cells are a suitable tool for the IR risk assessment during early human development. However, careful choice of the culture methods and a vigorous monitoring of the stem cell quality are mandatory for the use of these cells. Exposure to IR influences the stem cell properties of hES cells even when immediate radiation effects are overcome. This warrants consideration in the risk assessment of radiation effects during the earliest stages of human development.


Subject(s)
Activin Receptors/metabolism , Cell Differentiation/radiation effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/radiation effects , Radiation, Ionizing , Apoptosis/radiation effects , Biomarkers/metabolism , Cell Cycle/radiation effects , Cell Line , Cell Shape/radiation effects , Cell Survival/radiation effects , Chromosome Aberrations , Endoderm/metabolism , Endoderm/radiation effects , Gene Expression Regulation/radiation effects , Human Embryonic Stem Cells/metabolism , Humans , Karyotyping , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/radiation effects
9.
Stem Cells Dev ; 25(2): 178-88, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26506910

ABSTRACT

Little is known about the effects of ionizing radiation on the earliest stages of embryonic development although it is well recognized that ionizing radiation is a natural part of our environment and further exposure may occur due to medical applications. The current study addresses this issue using D3 mouse embryonic stem cells as a model system. Cells were irradiated with either X-rays or carbon ions representing sparsely and densely ionizing radiation and their effect on the differentiation of D3 cells into spontaneously contracting cardiomyocytes through embryoid body (EB) formation was measured. This study is the first to demonstrate that ionizing radiation impairs the formation of beating cardiomyocytes with carbon ions being more detrimental than X-rays. However, after prolonged culture time, the number of beating EBs derived from carbon ion irradiated cells almost reached control levels indicating that the surviving cells are still capable of developing along the cardiac lineage although with considerable delay. Reduced EB size, failure to downregulate pluripotency markers, and impaired expression of cardiac markers were identified as the cause of compromised cardiomyocyte formation. Dysregulation of cardiac differentiation was accompanied by alterations in the expression of endodermal and ectodermal markers that were more severe after carbon ion irradiation than after exposure to X-rays. In conclusion, our data show that carbon ion irradiation profoundly affects differentiation and thus may pose a higher risk to the early embryo than X-rays.


Subject(s)
Embryoid Bodies/cytology , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Radiation, Ionizing , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cell Survival , Cells, Cultured , Mice
10.
Invest Ophthalmol Vis Sci ; 56(8): 4350-63, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26176872

ABSTRACT

PURPOSE: Aqueous tear deficiency due to lacrimal gland insufficiency is one of the major causes of dry eye disease. In severe cases, such as Sjogren's syndrome, Stevens-Johnson syndrome, or ocular cicatricial pemphigoid, therapy with artificial tears is often insufficient to relieve severe discomfort, prevent progressive ocular surface disease, or enable visual rehabilitation by corneal transplantation. Cell or organ generation from stem cells, resulting in tear-like secretion, presents an option as a suitable alternative treatment. To obtain deeper insights into lacrimal gland stem cells we analyzed murine lacrimal glands for markers of pluripotency, self-renewal, and differentiation. METHODS: A special, patented technique with mechanical and enzymatic digestion was used to generate high numbers of cells in vitro from murine lacrimal glands. These presumptive "murine lacrimal gland stem cells" ("mLGSCs") can be propagated as monolayer cultures over multiple passages. By means of RT-PCR, Western blot, and immunohistochemistry, markers of pluripotency and differentiation were demonstrated. Hanging drop culture was used to build organoid bodies from mLGSCs to investigate their spontaneous differentiation in three-dimensional culture with histology, immunohistochemistry, and transmission electron microscopy methods. RESULTS: Isolated mLGSCs were cultured over more than 65 passages. Murine lacrimal gland stem cells expressed markers of pluripotency such as Nanog, Sox2, Kruppel-like factor 4 (Klf4), as well as early-lineage markers of all three germ layers. Three-dimensional culture of these cells revealed their ability to differentiate into various cell types. CONCLUSIONS: Our results suggest that mLGSCs were isolated and cultured successfully. These cells have the ability to differentiate into all three germ layers. The results provide further insights into lacrimal gland stem cell physiology for engineering of a lacrimal gland construct to treat severe cases of tear deficiency in the future.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Dry Eye Syndromes/therapy , Lacrimal Apparatus/ultrastructure , Stem Cells/ultrastructure , Tears/metabolism , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Immunohistochemistry , Kruppel-Like Factor 4 , Lacrimal Apparatus/metabolism , Mice , Microscopy, Electron, Transmission
11.
Methods Mol Biol ; 1213: 3-21, 2014.
Article in English | MEDLINE | ID: mdl-25173369

ABSTRACT

Cell therapy as a replacement for diseased or destroyed endogenous cells is a major component of regenerative medicine. Various types of stem cells are or will be used in clinical settings as autologous or allogeneic products. In this chapter, the progress that has been made to translate basic stem cell research into pharmaceutical manufacturing processes will be reviewed. Even if in public perception, embryonic stem (ES) cells and more recently induced pluripotent stem (iPS) cells dominate the field of regenerative medicine and will be discussed in great detail, it is the adult stem cells that are used for decades as therapeutics. Hence, these cells will be compared to ES and iPS cells. Finally, special emphasis will be placed on the scientific, technical, and economic challenges of developing stem cell-based in vitro model systems and cell therapies that can be commercialized.


Subject(s)
Cell- and Tissue-Based Therapy , Regenerative Medicine , Stem Cells , Animals , Cell Differentiation , Humans , Stem Cells/cytology
12.
Cell Reprogram ; 15(1): 68-79, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23256541

ABSTRACT

Pluripotency is characterized by specific transcription factors such as OCT4, NANOG, and SOX2, but also by pluripotency-associated microRNAs (miRs). Somatic cells can be reprogrammed by forced expression of these factors leading to induced pluripotent stem cells (iPSCs) with characteristics similar to embryonic stem cells (ESCs). However, current reprogramming strategies are commonly based on viral delivery of the pluripotency-associated factors, which affects the integrity of the genome and impedes the use of such cells in any clinical application. In an effort to establish nonviral, nonintegrating reprogramming strategies, we examined the influence of hypoxia on the expression of pluripotency-associated factors and the ESC-specific miR-302 cluster in primary and immortalized mesenchymal stromal cells (MSCs). The combination of hypoxia and fibroblast growth factor 2 (FGF2) treatments led to the induction of OCT4 and NANOG in an immortalized cell line L87 and primary MSCs, accompanied with increased doubling rates and decreased senescence. Most importantly, the endogenous ECS-specific cluster miR-302 was induced upon hypoxic culture and FGF2 supplementation. Hypoxia also improved reprogramming of MSCs via episomal expression of pluripotency factors. Thus, our data illustrate that hypoxia in combination with FGF2 supplementation efficiently facilitates reprogramming of MSCs.


Subject(s)
Cell Dedifferentiation , Embryonic Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/biosynthesis , Multigene Family , Pluripotent Stem Cells/metabolism , Cell Hypoxia/drug effects , Cell Line, Transformed , Embryonic Stem Cells/cytology , Fibroblast Growth Factor 2/pharmacology , Homeodomain Proteins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology
13.
Curr Diab Rep ; 12(5): 490-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22753002

ABSTRACT

Diabetes mellitus type 1 (T1DM) and type 2 (T2DM) are common diseases. To date, it is widely accepted that all forms of DM lead to the loss of beta cells. Therefore, to avoid the debilitating comorbidities when glycemic control cannot be fully achieved, some would argue that beta cell replacement is the only way to cure the disease. Due to organ donor shortage, other cell sources for beta cell replacement strategies have to be employed. Pluripotent stem cells, including embryonic stem (ES) and induced pluripotent stem (iPS) cells offer a valuable alternative to provide the necessary cells to substitute organ transplants but also to serve as a model to study the onset and progression of the disease, resulting in better treatment regimens. This review will summarize recent progress in the establishment of pluripotent stem cells, their differentiation into the pancreatic lineage with a focus on two-dimensional (2D) and three-dimensional (3D) differentiation settings, the special role of iPS cells in the analysis of genetic predispositions to diabetes, and techniques that help to move current approaches to clinical applications. Particular attention, however, is also given to the long-term challenges that have to be addressed before ES or iPS cell-based therapies will become a broadly accepted treatment option.


Subject(s)
Diabetes Mellitus/therapy , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pluripotent Stem Cells/metabolism
14.
Cells Tissues Organs ; 195(6): 507-23, 2012.
Article in English | MEDLINE | ID: mdl-22123608

ABSTRACT

Embryonic stem (ES) cells offer a valuable source for generating insulin-producing cells. However, current differentiation protocols often result in heterogeneous cell populations of various developmental stages. Here we show the activin A-induced differentiation of mouse ES cells carrying a homologous dsRed-IRES-puromycin knock-in within the Sox17 locus into the endoderm lineage. Sox17-expressing cells were selected by fluorescence-assisted cell sorting (FACS) and characterized at the transcript and protein level. Treatment of ES cells with high concentrations of activin A for 10 days resulted in up to 19% Sox17-positive cells selected by FACS. Isolated Sox17-positive cells were characterized by defini- tive endoderm-specific Sox17/Cxcr4/Foxa2 transcripts, but lacked pluripotency-associated Oct4 mRNA and protein. The Sox17-expressing cells showed downregulation of extraembryonic endoderm (Sox7, Afp, Sdf1)-, mesoderm (Foxf1, Meox1)- and ectoderm (Pax6, NeuroD6)-specific transcripts. The presence of Hnf4α, Hes1 and Pdx1 mRNA demonstrated the expression of primitive gut/foregut cell-specific markers. Ngn3, Nkx6.1 and Nkx2.2 transcripts in Sox17-positive cells were determined as properties of pancreatic endocrine progenitors. Immunocytochemistry of activin A-induced Sox17-positive embryoid bodies revealed coexpression of Cxcr4 and Foxa2. Moreover, the histochemical demonstration of E-cadherin-, Cxcr4-, Sox9-, Hnf1ß- and Ngn3-positive epithelial-like structures underlined the potential of Sox17-positive cells to further differentiate into the pancreatic lineage. By reducing the heterogeneity of the ES cell progeny, Sox17-expressing cells are a suitable model to evaluate the effects of growth and differentiation factors and of culture conditions to delineate the differentiation process for the generation of pancreatic cells in vitro.


Subject(s)
Cell Separation/methods , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/metabolism , HMGB Proteins/metabolism , SOXF Transcription Factors/metabolism , Activins/pharmacology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Embryonic Stem Cells/drug effects , Endoderm/drug effects , Epithelium/drug effects , Epithelium/embryology , Epithelium/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/drug effects , Homeobox Protein Nkx-2.2 , Luminescent Proteins/metabolism , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
15.
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
16.
Int J Dev Biol ; 53(4): 569-78, 2009.
Article in English | MEDLINE | ID: mdl-19378248

ABSTRACT

Embryonic stem (ES) cells which constitutively express the Pdx-1, Ngn-3, NeuroD1, Nkx2.2, and Nkx6.1 transcription factors were engineered by means of lentiviral vectors, following a multi-step infection procedure to successively generate ES cell lines expressing one, two, and three factors, respectively. Each ES cell line was allowed to differentiate into nestin+/Isl-1+ endocrine precursors, then into more mature pancreatic cells, and subsequently analysed for expression of Glc, Ins, and Sst, markers of alpha, beta and delta cells, respectively. Each ES cell line generated displayed a unique pattern of gene expression. The ES cell line expressing NeuroD1 displayed vastly elevated levels of Glc, Ins-1, Ins-2 and Sst, and showed an increase in Pdx-1, Pax-4, Nkx6.1, Isl-1, Glut-2 and GK transcript levels. Furthermore, immunofluorescence analysis revealed that differentiation of NeuroD1-expressing ES cells in nestin+/Isl-1+ multilineage progenitors, followed by the formation of C-peptide+/insulin+ clusters, was accelerated. Together, these results indicate that stable expression of NeuroD1 in ES cells facilitates differentiation into endocrine and insulin-producing cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endocrine System/metabolism , Insulin-Secreting Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Endocrine System/cytology , Endocrine System/embryology , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.2 , Insulin/biosynthesis , Insulin-Secreting Cells/cytology , Mice , Transcription Factors/metabolism
17.
Stem Cell Rev Rep ; 5(2): 159-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19263252

ABSTRACT

The differentiation of murine and human embryonic stem (ES) cells into pancreatic cell types has been shown by several methods including spontaneous differentiation, formation of multi-lineage progenitors, lineage selection or transgene expression. However, these strategies led to a mixture of cells of all three primary germ layers and only a low percentage of definitive endoderm cells giving rise to pancreas, liver, lung and intestine. To reproducibly generate functional insulin-producing cells, ES cells have to be differentiated via definitive endoderm and pancreatic endocrine progenitors recapitulating the in vivo development. Activin A, a member of the transforming growth factor beta superfamily, has been shown to induce definitive endoderm cells dependent on concentration, culture conditions and time of application. Moreover, serum components or contamination by feeder cells as well as differentiation and proliferation factors are critical for successful generation of activin A-induced ES cells into endoderm and pancreatic cells. The review presents an overview on those factors that influence activin A activity on endoderm and endocrine progenitor cells and determines the role of signaling factors in the differentiation process into the pancreatic lineage.


Subject(s)
Activins/pharmacology , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Insulin-Secreting Cells/drug effects , Pancreas/drug effects , Animals , Humans , Pancreas/cytology
18.
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
19.
Trends Immunol ; 29(2): 68-74, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18182324

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that results from the destruction of insulin-producing pancreatic islet cells owing to the aggressive effector function of autoreactive T cells. In addition to lifetime supply of exogenous insulin, whole-pancreas or islet transplantation is presently the only alternative therapy for severely ill patients. Here, we discuss the current status of the development of cell-based therapies that are based on essentially two options, i.e. replacement of islet cells by islet-like cells derived from embryonic or adult stem cells, and re-establishment of immunological tolerance to islet self-antigens through regulatory T cells and/or tolerance-promoting monocyte-derived cells. A combination of both approaches will be required to turn cell-based therapy of T1D into clinical success.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Insulin-Secreting Cells/metabolism , Islets of Langerhans Transplantation , Monocytes/immunology , Stem Cells/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/immunology , Autoimmunity , Cell Differentiation , Diabetes Mellitus, Type 1/physiopathology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Self Tolerance , Stem Cell Transplantation , Stem Cells/metabolism , T-Lymphocytes, Regulatory/metabolism
20.
J Clin Invest ; 117(9): 2713-22, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17717605

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide, accounting for an estimated 600,000 deaths annually. Aberrant methylation, consisting of DNA hypomethylation and/or promoter gene CpG hypermethylation, is implicated in the development of a variety of solid tumors, including HCC. We analyzed the global levels of DNA methylation as well as the methylation status of 105 putative tumor suppressor genes and found that the extent of genome-wide hypomethylation and CpG hypermethylation correlates with biological features and clinical outcome of HCC patients. We identified activation of Ras and downstream Ras effectors (ERK, AKT, and RAL) due to epigenetic silencing of inhibitors of the Ras pathway in all HCC. Further, selective inactivation of SPRY1 and -2, DAB2, and SOCS4 and -5 genes and inhibitors of angiogenesis (BNIP3, BNIP3L, IGFBP3, and EGLN2) was associated with poor prognosis. Importantly, several epigenetically silenced putative tumor suppressor genes found in HCC were also inactivated in the nontumorous liver. Our results assign both therapeutic and chemopreventive significance to methylation patterns in human HCC and open the possibility of using molecular targets, including those identified in this study, to effectively inhibit HCC development and progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , DNA Methylation , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Disease Progression , Enzyme Inhibitors/pharmacology , Female , Genome, Human/genetics , Humans , Male , Middle Aged , Prognosis , Protein Binding , Signal Transduction/drug effects , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...