Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Radiat Biol ; 99(5): 760-768, 2023.
Article in English | MEDLINE | ID: mdl-36352506

ABSTRACT

PURPOSE: We studied the effects of computed tomography (CT) scan irradiation on proliferation and differentiation of human embryonic stem cells (hESCs). It was reported that hESC is extremely radiosensitive; exposure of hESC in cultures to 1 Gy of ionizing radiation (IR) results in massive apoptosis of the damaged cells and, thus, they are eliminated from the cultures. However, after recovery the surviving cells proliferate and differentiate normally. We hypothesized that IR-exposed hESC may still have growth rate disadvantage when they proliferate or differentiate in the presence of non-irradiated hESC, as has been shown for mouse hematopoietic stem cells in vivo. MATERIALS AND METHODS: To study such competitive proliferation and differentiation, we obtained cells of H9 hESC line that stably express green fluorescent protein (H9GFP). Irradiated with 50 mGy or 500 mGy H9GFP and non-irradiated H9 cells (or vice versa) were mixed and allowed to grow under pluripotency maintaining conditions or under conditions of directed differentiation into neuronal lineage for several passages. The ratio of H9GFP to H9 cells was measured after every passage or approximately every week. RESULTS: We observed competition of H9 and H9GFP cells; we found that the ratio of H9GFP to H9 cells increased with time in both proliferation and differentiation conditions regardless of irradiation, i.e. the H9GFP cells in general grew faster than H9 cells in the mixtures. However, we did not observe any consistent changes in the relative growth rate of irradiated versus non-irradiated hESC. CONCLUSIONS: We conclude that population of pluripotent hESC is very resilient; while damaged cells are eliminated from colonies, the surviving cells retain their pluripotency, ability to differentiate, and compete with non-irradiated isogenic cells. These findings are consistent with the results of our previous studies, and with the concept that early in pregnancy omnipotent cells injured by IR can be replaced by non-damaged cells with no impact on embryo development.


Subject(s)
Human Embryonic Stem Cells , Humans , Animals , Mice , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/radiation effects , Embryonic Stem Cells/metabolism , Cell Differentiation , Cell Proliferation/radiation effects , Radiation, Ionizing
2.
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
3.
Radiat Res ; 193(5): 460-470, 2020 05.
Article in English | MEDLINE | ID: mdl-32216708

ABSTRACT

Human embryonic brain development is highly sensitive to ionizing radiation. However, detailed information on the mechanisms of this sensitivity is not available due to limited experimental data. In this study, differentiation of human embryonic stem cells (hESCs) to neural lineages was used as a model for early embryonic brain development to assess the effect of exposure to low (17 mGy) and high (572 mGy) doses of radiation on gene expression. Transcriptomes were assessed using RNA sequencing during neural differentiation at three time points in control and irradiated samples. The first time point was when the cells were still pluripotent (day 0), the second time point was during the stage of embryoid body formation (day 6), and the third and final time point was during the stage of neural rosette formation (day 10). Analysis of the transcriptomes revealed neurodifferentiation in both the control and irradiated cells. Low-dose irradiation did not result in changes in gene expression at any of the time points, whereas high-dose irradiation resulted in downregulation of some major neurodifferentiation markers on days 6 and 10. Gene ontology analysis showed that pathways related to nervous system development, neurogenesis and generation of neurons were among the most affected. Expression of such key regulators of neuronal development as NEUROG1, ARX, ASCL1, RFX4 and INSM1 was reduced more than twofold. In conclusion, exposure to a 17 mGy low dose of radiation was well tolerated by hESCs while exposure to 572 mGy significantly affected their genetic reprogramming into neuronal lineages.


Subject(s)
Cell Differentiation/radiation effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/radiation effects , Transcriptome/radiation effects , Human Embryonic Stem Cells/cytology , Humans , Neurogenesis/radiation effects , Time Factors , Tomography, X-Ray Computed/adverse effects
4.
PLoS One ; 14(11): e0224846, 2019.
Article in English | MEDLINE | ID: mdl-31710637

ABSTRACT

Optogenetically engineered human neural progenitors (hNPs) are viewed as promising tools in regenerative neuroscience because they allow the testing of the ability of hNPs to integrate within nervous system of an appropriate host not only structurally, but also functionally based on the responses of their differentiated progenies to light. Here, we transduced H9 embryonic stem cell-derived hNPs with a lentivirus harboring human channelrhodopsin (hChR2) and differentiated them into a forebrain lineage. We extensively characterized the fate and optogenetic functionality of hChR2-hNPs in vitro with electrophysiology and immunocytochemistry. We also explored whether the in vivo phenotype of ChR2-hNPs conforms to in vitro observations by grafting them into the frontal neocortex of rodents and analyzing their survival and neuronal differentiation. Human ChR2-hNPs acquired neuronal phenotypes (TUJ1, MAP2, SMI-312, and synapsin 1 immunoreactivity) in vitro after an average of 70 days of coculturing with CD1 astrocytes and progressively displayed both inhibitory and excitatory neurotransmitter signatures by immunocytochemistry and whole-cell patch clamp recording. Three months after transplantation into motor cortex of naïve or injured mice, 60-70% of hChR2-hNPs at the transplantation site expressed TUJ1 and had neuronal cytologies, whereas 60% of cells also expressed ChR2. Transplant-derived neurons extended axons through major commissural and descending tracts and issued synaptophysin+ terminals in the claustrum, endopiriform area, and corresponding insular and piriform cortices. There was no apparent difference in engraftment, differentiation, or connectivity patterns between injured and sham subjects. Same trends were observed in a second rodent host, i.e. rat, where we employed longer survival times and found that the majority of grafted hChR2-hNPs differentiated into GABAergic neurons that established dense terminal fields and innervated mostly dendritic profiles in host cortical neurons. In physiological experiments, human ChR2+ neurons in culture generated spontaneous action potentials (APs) 100-170 days into differentiation and their firing activity was consistently driven by optical stimulation. Stimulation generated glutamatergic and GABAergic postsynaptic activity in neighboring ChR2- cells, evidence that hChR2-hNP-derived neurons had established functional synaptic connections with other neurons in culture. Light stimulation of hChR2-hNP transplants in vivo generated complicated results, in part because of the variable response of the transplants themselves. Our findings show that we can successfully derive hNPs with optogenetic properties that are fully transferrable to their differentiated neuronal progenies. We also show that these progenies have substantial neurotransmitter plasticity in vitro, whereas in vivo they mostly differentiate into inhibitory GABAergic neurons. Furthermore, neurons derived from hNPs have the capacity of establishing functional synapses with postsynaptic neurons in vitro, but this outcome is technically challenging to explore in vivo. We propose that optogenetically endowed hNPs hold great promise as tools to explore de novo circuit formation in the brain and, in the future, perhaps launch a new generation of neuromodulatory therapies.


Subject(s)
Human Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/cytology , Optogenetics , Animals , Astrocytes/cytology , Astrocytes/radiation effects , Axons/metabolism , Axons/radiation effects , Cell Differentiation/radiation effects , Cell Lineage/radiation effects , Cell Survival/radiation effects , Channelrhodopsins/metabolism , Human Embryonic Stem Cells/radiation effects , Humans , Lentivirus/metabolism , Light , Mice, Nude , Motor Cortex/metabolism , Neural Stem Cells/radiation effects , Neuronal Plasticity/radiation effects , Neurons/radiation effects , Neurotransmitter Agents/metabolism , Phenotype , Photic Stimulation , Rats, Nude , Synaptic Transmission/radiation effects
5.
Int J Mol Sci ; 20(16)2019 Aug 10.
Article in English | MEDLINE | ID: mdl-31405104

ABSTRACT

We studied the effect of radiation from computed tomography (CT) scans on differentiation of human embryonic stem cells (hESCs) into neuronal lineage. hESCs were divided into three radiation exposure groups: 0-dose, low-dose, or high-dose exposure. Low dose was accomplished with a single 15 mGy CT dose index (CTDI) CT scan that approximated the dose for abdominal/pelvic CT examinations in adults while the high dose was achieved with several consecutive CT scans yielding a cumulative dose of 500 mGy CTDI. The neural induction was characterized by immunocytochemistry. Quantitative polymerase chain reaction (qPCR) and Western blots were used to measure expression of the neuronal markers PAX6 and NES and pluripotency marker OCT4. We did not find any visible morphological differences between neural precursors from irradiated and non-irradiated cells. However, quantitative analyses of neuronal markers showed that PAX6 expression was reduced following exposure to the high dose compared to 0-dose controls, while no such decrease in PAX6 expression was observed following exposure to the low dose. Similarly, a statistically significant reduction in expression of NES was observed following high-dose exposure, while after low-dose exposure, a modest but statistically significant reduction in NES expression was only observed on Day 8 of differentiation. Further studies are warranted to elucidate how lower or delayed expression of PAX6 and NES can impact human fetal brain development.


Subject(s)
Human Embryonic Stem Cells/radiation effects , Neural Stem Cells/radiation effects , Neurogenesis/radiation effects , Tomography, X-Ray Computed/adverse effects , Cell Line , Down-Regulation/radiation effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , PAX6 Transcription Factor/genetics , Radiation Dosage , Radiation, Ionizing
6.
Biomaterials ; 199: 63-75, 2019 04.
Article in English | MEDLINE | ID: mdl-30738336

ABSTRACT

Tissue specific extracellular matrices (ECM) provide structural support and enable access to molecular signals and metabolites, which are essential for directing stem cell renewal and differentiation. To mimic this phenomenon in vitro, tissue decellularisation approaches have been developed, resulting in the generation of natural ECM scaffolds that have comparable physical and biochemical properties of the natural tissues and are currently gaining traction in tissue engineering and regenerative therapies due to the ease of standardised production, and constant availability. In this manuscript we report the successful generation of decellularised ECM-derived peptides from neural retina (decel NR) and retinal pigment epithelium (decel RPE), and their impact on differentiation of human pluripotent stem cells (hPSCs) to retinal organoids. We show that culture media supplementation with decel RPE and RPE-conditioned media (CM RPE) significantly increases the generation of rod photoreceptors, whilst addition of decel NR and decel RPE significantly enhances ribbon synapse marker expression and the light responsiveness of retinal organoids. Photoreceptor maturation, formation of correct synapses between retinal cells and recording of robust light responses from hPSC-derived retinal organoids remain unresolved challenges for the field of regenerative medicine. Enhanced rod photoreceptor differentiation, synaptogenesis and light response in response to addition of decellularised matrices from RPE and neural retina as shown herein provide a novel and substantial advance in generation of retinal organoids for drug screening, tissue engineering and regenerative medicine.


Subject(s)
Biomarkers/metabolism , Extracellular Matrix/chemistry , Light , Organoids/cytology , Peptides/pharmacology , Pluripotent Stem Cells/cytology , Retinal Pigment Epithelium/metabolism , Synapses/metabolism , Adult , Animals , Cattle , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/radiation effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/radiation effects , Human Embryonic Stem Cells/ultrastructure , Humans , Organoids/drug effects , Organoids/radiation effects , Organoids/ultrastructure , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/radiation effects , Photoreceptor Cells, Vertebrate/ultrastructure , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/radiation effects , Synapses/drug effects , Synapses/radiation effects
7.
Mol Biol Rep ; 46(1): 1343-1348, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30523518

ABSTRACT

Radiation induces DNA and protein damage and free radical formation, effectively establishing cellular senescence in a variety of models. We demonstrate the effects of two known pleiotropic drugs following gamma radiation damage in neurosphere/cerebral organoid system based on human embryonic stem cells. mTORC1 repression by rapamycin prior to irradiation, or metabolic activation by minocycline after irradiation, partially rescues neuroepithelium integrity, neurite-growing capacity, ventricle formation and extracellular acidification rate as an integral measure of metabolic output. Cerebral organoid model thus provides valid and robust readouts for radiation studies in a complex 3D setting.


Subject(s)
Brain/cytology , Gamma Rays , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/radiation effects , Minocycline/pharmacology , Organoids/cytology , Sirolimus/pharmacology , Cell Line , Gene Expression Regulation/drug effects , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Neurites/drug effects , Neurites/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
8.
Biomaterials ; 143: 65-78, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28763631

ABSTRACT

Multipotent cells have similar basic features of all stem cells but limitation in ability of self-renewal and differentiation compared with pluripotent cells. Here, we have developed an ultra effective, gene- and chemical-free method of generating extra multipotent (xpotent) cells which have differentiation potential more than limited cell types, by the mechanism of ultrasound-directed permeation of environmental transition-guided cellular reprogramming (Entr). Ultrasound stimulus generated a massive number of Entr-mediated xpotent (x/Entr) spheroids from human dermal fibroblasts (HDFs) 6 days after treatment. The emergence of x/Entr was first initiated by the introduction of human embryonic stem cell (ESC) environments into the HDFs to start fast cellular reprogramming including activation of stress-related kinase signaling pathways, subsequent chromatin remodeling, and expression of pluripotent-related genes via transient membrane damage caused by ultrasound-induced cavitation. And then, pluripotent markers were transported into their adjacent HDFs via direct cell-to-cell connections in order to generate xpotent clusters. The features of x/Entr cells were intermediate between pluripotency and multipotency in terms of pluripotency with three germ layer markers, multi-lineage differentiation potential, and no teratoma formation. This physical stimulus-mediated reprogramming strategy was cost-effective, simple, quick, produced significant yields, and was safe, and can therefore provide a new paradigm for clinical application.


Subject(s)
Cell Differentiation , Cellular Reprogramming/radiation effects , Fibroblasts/cytology , Fibroblasts/radiation effects , Adult , Cell Culture Techniques , Cell Line , Cell Self Renewal , Cells, Cultured , Chromatin Assembly and Disassembly/radiation effects , Fibroblasts/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/radiation effects , Humans , Middle Aged , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Spheroids, Cellular/radiation effects , Ultrasonic Waves
9.
Sci Rep ; 7: 43995, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266624

ABSTRACT

We studied the effect of ionizing radiation (IR) on continuous growth of seven hESC lines. Cells were exposed to 0, 0.2, or 1 Gy of X-rays, and the growth rates of cell populations were assessed by measuring areas of the same individual colonies versus time. The population doubling times (DT) of sham-irradiated cells varied from 18.9 to 28.7 hours for different cell lines. All cell lines showed similar reaction to IR, i.e. cell populations dropped within 24-48 hours post IR; after that they recovered and grew with the same rate as the sham-irradiated cells. The relative cell survival (RCS), i.e. the ratio of normalized cell population in the irradiated samples to that of the sham-irradiated ones varied from 0.6 to 0.8 after 0.2 Gy, and from 0.1 to 0.2 after 1 Gy IR for different cell lines. We found that the RCS values of hESC lines correlated directly with their DT, i.e. the faster cells grow the more radiosensitive they are. We also found that DT and RCS values of individual colonies varied significantly within all hESC lines. We believe that the method developed herein can be useful for assessing other cytotoxic insults on cultures of hESC.


Subject(s)
Cell Proliferation/radiation effects , Human Embryonic Stem Cells/physiology , Human Embryonic Stem Cells/radiation effects , Radiation, Ionizing , Cell Survival/radiation effects , Cells, Cultured , Humans
10.
Mol Med Rep ; 15(4): 1952-1962, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259963

ABSTRACT

Despite the well-demonstrated efficacy of stem cell (SC) therapy, this approach has a number of key drawbacks. One important concern is the response of pluripotent SCs to treatment with ionizing radiation (IR), given that SCs used in regenerative medicine will eventually be exposed to IR for diagnostic or treatment­associated purposes. Therefore, the aim of the present study was to examine and compare early IR­induced responses of pluripotent SCs to assess their radioresistance and radiosensitivity. In the present study, 3 cell lines; human embryonic SCs (hESCs), human induced pluripotent SCs (hiPSCs) and primary human dermal fibroblasts (PHDFs); were exposed to IR at doses ranging from 0 to 15 gray (Gy). Double strand breaks (DSBs), and the gene expression of the following DNA repair genes were analyzed: P53; RAD51; BRCA2; PRKDC; and XRCC4. hiPSCs demonstrated greater radioresistance, as fewer DSBs were identified, compared with hESCs. Both pluripotent SC lines exhibited distinct gene expression profiles in the most common DNA repair genes that are involved in homologous recombination, non­homologous end­joining and enhanced DNA damage response following IR exposure. Although hESCs and hiPSCs are equivalent in terms of capacity for pluripotency and differentiation into 3 germ layers, the results of the present study indicate that these 2 types of SCs differ in gene expression following exposure to IR. Consequently, further research is required to determine whether hiPSCs and hESCs are equally safe for application in clinical practice. The present study contributes to a greater understanding of DNA damage response (DDR) mechanisms activated in pluripotent SCs and may aid in the future development of safe SC­based clinical protocols.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , Human Embryonic Stem Cells/radiation effects , Induced Pluripotent Stem Cells/radiation effects , Transcriptome/radiation effects , BRCA2 Protein/genetics , Cells, Cultured , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , Gene Expression/radiation effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Nuclear Proteins/genetics , Rad51 Recombinase/genetics , Radiation, Ionizing
11.
Stem Cell Res Ther ; 8(1): 73, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28327192

ABSTRACT

BACKGROUND: Human induced pluripotent stem cells (hiPSCs) play roles in both disease modelling and regenerative medicine. It is critical that the genomic integrity of the cells remains intact and that the DNA repair systems are fully functional. In this article, we focused on the detection of DNA double-strand breaks (DSBs) by phosphorylated histone H2AX (known as γH2AX) and p53-binding protein 1 (53BP1) in three distinct lines of hiPSCs, their source cells, and one line of human embryonic stem cells (hESCs). METHODS: We measured spontaneously occurring DSBs throughout the process of fibroblast reprogramming and during long-term in vitro culturing. To assess the variations in the functionality of the DNA repair system among the samples, the number of DSBs induced by γ-irradiation and the decrease over time was analysed. The foci number was detected by fluorescence microscopy separately for the G1 and S/G2 cell cycle phases. RESULTS: We demonstrated that fibroblasts contained a low number of non-replication-related DSBs, while this number increased after reprogramming into hiPSCs and then decreased again after long-term in vitro passaging. The artificial induction of DSBs revealed that the repair mechanisms function well in the source cells and hiPSCs at low passages, but fail to recognize a substantial proportion of DSBs at high passages. CONCLUSIONS: Our observations suggest that cellular reprogramming increases the DSB number but that the repair mechanism functions well. However, after prolonged in vitro culturing of hiPSCs, the repair capacity decreases.


Subject(s)
Cellular Senescence/radiation effects , DNA Breaks, Double-Stranded , DNA Repair/genetics , Fibroblasts/radiation effects , Human Embryonic Stem Cells/radiation effects , Induced Pluripotent Stem Cells/radiation effects , Cell Line , Cellular Reprogramming , Cellular Senescence/genetics , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Gamma Rays , Gene Expression , Histones/genetics , Histones/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Phosphorylation/radiation effects , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
12.
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
13.
Adv Mater ; 28(48): 10732-10737, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27787919

ABSTRACT

A new method of spatially controlled gene regulation in 3D-cultured human embryonic stem cells is developed using hollow gold nanoshells (HGNs) and near-infrared (NIR) light. Targeted cell(s) are discriminated from neighboring cell(s) by focusing NIR light emitted from a two-photon microscope. Irradiation of cells that have internalized HGNs releases surface attached siRNAs and leads to concomitant gene downregulation.


Subject(s)
Cell Culture Techniques , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/radiation effects , RNA Interference/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/radiation effects , Down-Regulation , Gold , Human Embryonic Stem Cells/cytology , Humans , Infrared Rays , Microscopy, Fluorescence, Multiphoton , Nanoshells
14.
Int J Mol Sci ; 17(1)2016 Jan 02.
Article in English | MEDLINE | ID: mdl-26729112

ABSTRACT

Chromatin structure affects the extent of DNA damage and repair. Thus, it has been shown that heterochromatin is more protective against DNA double strand breaks (DSB) formation by ionizing radiation (IR); and that DNA DSB repair may proceed differently in hetero- and euchromatin regions. Human embryonic stem cells (hESC) have a more open chromatin structure than differentiated cells. Here, we study the effect of chromatin structure in hESC on initial DSB formation and subsequent DSB repair. DSB were scored by comet assay; and DSB repair was assessed by repair foci formation via 53BP1 antibody staining. We found that in hESC, heterochromatin is confined to distinct regions, while in differentiated cells it is distributed more evenly within the nuclei. The same dose of ionizing radiation produced considerably more DSB in hESC than in differentiated derivatives, normal human fibroblasts; and one cancer cell line. At the same time, the number of DNA repair foci were not statistically different among these cells. We showed that in hESC, DNA repair foci localized almost exclusively outside the heterochromatin regions. We also noticed that exposure to ionizing radiation resulted in an increase in heterochromatin marker H3K9me3 in cancer HT1080 cells, and to a lesser extent in IMR90 normal fibroblasts, but not in hESCs. These results demonstrate the importance of chromatin conformation for DNA protection and DNA damage repair; and indicate the difference of these processes in hESC.


Subject(s)
Chromatin/ultrastructure , DNA Breaks, Double-Stranded/radiation effects , Human Embryonic Stem Cells/radiation effects , Cell Differentiation , Cell Line , Chromatin/radiation effects , Heterochromatin/radiation effects , Heterochromatin/ultrastructure , Human Embryonic Stem Cells/cytology , Humans , Radiation, Ionizing
15.
Oncotarget ; 6(26): 22258-69, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26082437

ABSTRACT

Studies have implicated that a small sub-population of cells within a tumour, termed cancer stem cells (CSCs), have an enhanced capacity for tumour formation in multiple cancers and may be responsible for recurrence of the disease after treatment, including radiation. Although comparisons have been made between CSCs and bulk-tumour, the more important comparison with respect to therapy is between tumour-sustaining CSC versus normal stem cells that maintain the healthy tissue. However, the absence of normal known counterparts for many CSCs has made it difficult to compare the radiation responses of CSCs with the normal stem cells required for post-radiotherapy tissue regeneration and the maintenance of tissue homeostasis. Here we demonstrate that transformed human embryonic stem cells (t-hESCs), showing features of neoplastic progression produce tumours resistant to radiation relative to their normal counterpart upon injection into immune compromised mice. We reveal that t-hESCs have a reduced capacity for radiation induced cell death via apoptosis and exhibit altered cell cycle arrest relative to hESCs in vitro. t-hESCs have an increased expression of BclXL in comparison to their normal counterparts and re-sensitization of t-hESCs to radiation upon addition of BH3-only mimetic ABT737, suggesting that overexpression of BclXL underpins t-hESC radiation insensitivity. Using this novel discovery platform to investigate radiation resistance in human CSCs, our study indicates that chemotherapy targeting Bcl2-family members may prove to be an adjuvant to radiotherapy capable of targeting CSCs.


Subject(s)
Human Embryonic Stem Cells/radiation effects , Neoplasms/radiotherapy , Neoplastic Stem Cells/radiation effects , Animals , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , Heterografts , Human Embryonic Stem Cells/pathology , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Neoplastic Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...