Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Genesis ; 59(12): e23457, 2021 12.
Article in English | MEDLINE | ID: mdl-34687271

ABSTRACT

The Cre-loxP system has been widely used for specific DNA recombination which induces gene inactivation or expression. Recently, photoactivatable-Cre (PA-Cre) proteins have been developed as a tool for spatiotemporal control of the enzymatic activity of Cre recombinase. Here, we generated transgenic mice bearing a PA-Cre gene and systematically investigated the conditions of photoactivation for the PA-Cre in embryonic stem cells (ESCs) derived from the transgenic mice and in a simple mathematical model. Cre-mediated DNA recombination was induced in 16% of the PA-Cre ESCs by 6 hr continuous illumination. We show that repetitive pulsed illumination efficiently induced DNA recombination with low light energy as efficient as continuous illumination in the ESCs (96 ± 15% of continuous illumination when pulse cycle was 2 s), which was also supported by a minimal mathematical model. DNA recombination by the PA-Cre was also successfully induced in the transgenic mouse pre-implantation embryos under the developed conditions. These results suggest that strategies based on repetitive pulsed illumination are efficient for the activation of photoactivatable Cre and, possibly other photo-switchable proteins.


Subject(s)
Embryonic Stem Cells/radiation effects , Genetic Engineering , Integrases/genetics , Recombination, Genetic/radiation effects , Animals , Blastocyst/radiation effects , Embryonic Stem Cells/metabolism , Integrases/radiation effects , Light , Mice , Mice, Transgenic , Promoter Regions, Genetic/radiation effects
2.
Cancer Res ; 81(13): 3706-3716, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33941615

ABSTRACT

Fanconi anemia is an inherited genome instability syndrome characterized by interstrand cross-link hypersensitivity, congenital defects, bone marrow failure, and cancer predisposition. Although DNA repair mediated by Fanconi anemia genes has been extensively studied, how inactivation of these genes leads to specific cellular phenotypic consequences associated with Fanconi anemia is not well understood. Here we report that Fanconi anemia stem cells in the C. elegans germline and in murine embryos display marked nonhomologous end joining (NHEJ)-dependent radiation resistance, leading to survival of progeny cells carrying genetic lesions. In contrast, DNA cross-linking does not induce generational genomic instability in Fanconi anemia stem cells, as widely accepted, but rather drives NHEJ-dependent apoptosis in both species. These findings suggest that Fanconi anemia is a stem cell disease reflecting inappropriate NHEJ, which is mutagenic and carcinogenic as a result of DNA misrepair, while marrow failure represents hematopoietic stem cell apoptosis. SIGNIFICANCE: This study finds that Fanconi anemia stem cells preferentially activate error-prone NHEJ-dependent DNA repair to survive irradiation, thereby conferring generational genomic instability that is instrumental in carcinogenesis.


Subject(s)
Cesium Radioisotopes/adverse effects , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Embryonic Stem Cells/pathology , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/pathology , Genomic Instability , Animals , Apoptosis , Caenorhabditis elegans , DNA Repair , Embryonic Stem Cells/radiation effects , Fanconi Anemia/genetics , Fanconi Anemia/radiotherapy , Fanconi Anemia Complementation Group Proteins/genetics , Mice
3.
Signal Transduct Target Ther ; 6(1): 129, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33785736

ABSTRACT

Maintenance of genetic stability via proper DNA repair in stem and progenitor cells is essential for the tissue repair and regeneration, while preventing cell transformation after damage. Loss of PUMA dramatically increases the survival of mice after exposure to a lethal dose of ionizing radiation (IR), while without promoting tumorigenesis in the long-term survivors. This finding suggests that PUMA (p53 upregulated modulator of apoptosis) may have a function other than regulates apoptosis. Here, we identify a novel role of PUMA in regulation of DNA repair in embryonic or induced pluripotent stem cells (PSCs) and immortalized hematopoietic progenitor cells (HPCs) after IR. We found that PUMA-deficient PSCs and HPCs exhibited a significant higher double-strand break (DSB) DNA repair activity via Rad51-mediated homologous recombination (HR). This is because PUMA can be associated with early mitotic inhibitor 1 (EMI1) and Rad51 in the cytoplasm to facilitate EMI1-mediated cytoplasmic Rad51 ubiquitination and degradation, thereby inhibiting Rad51 nuclear translocation and HR DNA repair. Our data demonstrate that PUMA acts as a repressor for DSB DNA repair and thus offers a new rationale for therapeutic targeting of PUMA in regenerative cells in the context of DNA damage.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Proteins/genetics , Rad51 Recombinase/genetics , Tumor Suppressor Proteins/genetics , Animals , Carcinogenesis/radiation effects , Cell Line, Tumor , Cytoplasm/genetics , Cytoplasm/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Embryonic Stem Cells/pathology , Embryonic Stem Cells/radiation effects , Gene Expression Regulation, Developmental/radiation effects , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Mice , Radiation, Ionizing , Recombinational DNA Repair/radiation effects , Regeneration/genetics , Ubiquitination/genetics
4.
J Photochem Photobiol B ; 188: 77-86, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30232055

ABSTRACT

It is known that visible light, including sunlight and laboratory lighting, adversely affect the development of embryos in vitro. In with article we present a technology for the synthesis of composite screens, capable to photoconvert UV and a part of the blue spectrum into red light with the maximum ~630 nm. It is established that the application of such transformed light with an evident red component raises the chances of embryos to survive and protects embryonic stem cells. To create photoconversion screens, the CdZn/Se quantum dots were obtained, the average size being about 7 nm. When the quantum dots are excited by electromagnetic waves of the UV and blue spectral range, photoluminescence is observed. The average photon energy for photoluminescence is of the order of 2 eV. On the basis of CdZn/Se quantum dots and methylphenylsiloxane polymer, light-transforming composite screens were made. In case of the light-transforming composite screen, the UV component disappeared from the energy spectrum, and the intensity of the blue region of the spectrum was reduced. On the contrary, in the red region (λmax = 630 nm) one can see a little more than two-fold increase of intensity. It is shown that when exposed to 2-cell embryos by transformed light, the proportion of normally developing embryos increases by 20%, the number of dead embryos decreases twice, and number of dead and apoptotic cells was lower in blastocysts, what's decreased by 70%, as compared to the control group. When blastocysts are transferred to the feeder substrate, colonies of embryonic stem cells are formed. Cells obtained from blastocysts irradiated with transformed visible light are in a normal state in 90% of cases and did not change expression levels, biochemistry and morphology for at least 20 passages. It is assumed that the data obtained can be used for the design of systems of efficient cultivation of embryonic cells for tissue engineering and cell therapy.


Subject(s)
Embryo, Mammalian/radiation effects , Light , Animals , Cell Differentiation/radiation effects , Embryonic Development/radiation effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Gene Expression Regulation/radiation effects , Mice , Polymers/chemistry , Quantum Dots/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
5.
J Assist Reprod Genet ; 35(6): 1061-1069, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29546598

ABSTRACT

PURPOSE: To study the outcomes of mouse preimplantation embryos irradiated with low doses of X-rays (≤ 1 Gy) and investigate apoptosis and pluripotency of the irradiated embryos. METHODS: Mouse embryos at the 2-cell stage were collected for in vitro culture. After reaching the 8-cell stage, embryos were irradiated with various low doses of X-rays (0-1 Gy). Blastocysts with a normal appearance were transferred into a pseudopregnant uterus. The developmental rate to blastocysts and the survival rate following embryo transfer were examined. Expression levels of p21, Smad2, Foxo1, Cdx2, Oct4, and Nanog genes were measured by RT-PCR. Apoptotic cells in mouse blastocysts were examined immunofluorescently by staining for cleaved caspase-3. RESULTS: More than 90% of non-irradiated and low-dose X-ray-irradiated preimplantation embryos developed to morphologically normal blastocysts that could be implanted and survive in the uterus. However, embryos irradiated with X-rays had more apoptotic cells in a dose-dependent manner. Expression of p21, Smad2, and Foxo1 genes in X-ray-irradiated embryos was increased significantly, while expression of Cdx2, Oct4, and Nanog genes was maintained in comparison with non-irradiated embryos. CONCLUSIONS: Although irradiated embryos contained apoptotic cells, the low doses of irradiation did not disturb development of 8-cell stage embryos to blastocysts or their survival in utero. The underlying mechanisms might involve anti-apoptotic systems, including the Smad-p21 pathway, and preservation of pluripotency.


Subject(s)
Blastocyst/cytology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryonic Development/radiation effects , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/radiation effects , Pluripotent Stem Cells/cytology , Smad Proteins/metabolism , Animals , Blastocyst/metabolism , Blastocyst/radiation effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Dose-Response Relationship, Radiation , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Female , Mice , Mice, Inbred ICR , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/radiation effects , Smad Proteins/genetics , X-Rays
6.
Stem Cell Res ; 23: 98-104, 2017 08.
Article in English | MEDLINE | ID: mdl-28715716

ABSTRACT

Early mammalian embryonic cells must maintain a particularly robust DNA repair system, as mutations at this developmental point have detrimental consequences for the organism. How the repair system can be tuned to fulfill such elevated requirements is largely unknown, but it may involve transcriptional regulation. Ronin (Thap11) is a transcriptional regulator responsible for vital programs in pluripotent cells. Here, we report that this protein also modulates the DNA damage response of such cells. We show that conditional Ronin knockout sensitizes embryonic stem cells (ESCs) to UV-C-induced DNA damage in association with Atr pathway activation and G2/M arrest. Ronin binds to and regulates the genes encoding several DNA repair factors, including Gtf2h4 and Rad18, providing a potential mechanism for this phenotype. Our results suggest that the unique DNA repair requirements of the early embryo are not met by a static system, but rather via highly regulated processes.


Subject(s)
DNA Damage , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Cycle Checkpoints/radiation effects , DNA Damage/genetics , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , G2 Phase/radiation effects , Mice, Knockout , Mitosis/radiation effects , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/radiation effects , Radiation, Ionizing , Ultraviolet Rays
7.
Stem Cell Reports ; 7(6): 1013-1022, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27974220

ABSTRACT

Dynamic spatiotemporal modification of chromatin around DNA damage is vital for efficient DNA repair. Normal stem cells exhibit an attenuated DNA damage response (DDR), inefficient DNA repair, and high radiosensitivity. The impact of unique chromatin characteristics of stem cells in DDR regulation is not yet recognized. We demonstrate that murine embryonic stem cells (ES) display constitutively elevated acetylation of histone H3 lysine 9 (H3K9ac) and low H3K9 tri-methylation (H3K9me3). DNA damage-induced local deacetylation of H3K9 was abrogated in ES along with the subsequent H3K9me3. Depletion of H3K9ac in ES by suppression of monocytic leukemia zinc finger protein (MOZ) acetyltransferase improved ATM activation, DNA repair, diminished irradiation-induced apoptosis, and enhanced clonogenic survival. Simultaneous suppression of the H3K9 methyltransferase Suv39h1 abrogated the radioprotective effect of MOZ inhibition, suggesting that high H3K9ac promoted by MOZ in ES cells obstructs local upregulation of H3K9me3 and contributes to muted DDR and increased radiosensitivity.


Subject(s)
Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Histones/metabolism , Lysine/metabolism , Radiation Tolerance , Radiation, Ionizing , Acetylation , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Breaks/radiation effects , DNA Repair/radiation effects , Down-Regulation/radiation effects , Histone Acetyltransferases/metabolism , Methylation , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , Radiation Tolerance/radiation effects , Up-Regulation/genetics , Up-Regulation/radiation effects
8.
Exp Dermatol ; 25 Suppl 3: 41-4, 2016 08.
Article in English | MEDLINE | ID: mdl-27539901

ABSTRACT

The aryl hydrocarbon receptor (AHR) mediates melanocyte activation and skin tanning. We hypothesized that the AHR also mediates melanoblast-to-melanocyte maturation. In a cloned cell line, NCCmelb4, derived from mouse neural crest cells, we investigated AHR expression in melanoblasts stimulated by UV irradiation and AHR agonists. We irradiated the cells with UV, ranging from 280 to 380 nm in 10-nm increments, using a multiwavelength irradiation spectral apparatus. Tyrosinase expression significantly increased with bimodal peaks at 310 and 360 nm. Although melanoblast activation peaked 48 hours after irradiation, the most suitable irradiation interval was 24 hours. AHR expression significantly increased at 360 nm, but not at 310 nm. The AHR agonist, VAF347, and water-soluble tobacco smoke extract induced melanoblast maturation and AHR activation. The culture supernatant derived from the NS47 fibroblast cell line also induced melanoblast maturation and AHR activation. These findings suggest that UV and environmental stimulation of melanoblast-to-melanocyte maturation are enhanced via the AHR pathway.


Subject(s)
Melanocytes/cytology , Melanocytes/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Melanocytes/radiation effects , Mice , Monophenol Monooxygenase/genetics , Neural Crest/cytology , Neural Crest/metabolism , Neural Crest/radiation effects , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/genetics , Smoke/adverse effects , Nicotiana , Ultraviolet Rays/adverse effects
9.
Radiat Res ; 186(1): 17-26, 2016 07.
Article in English | MEDLINE | ID: mdl-27332952

ABSTRACT

Pluripotent stem cells (PSCs) hold great promise in regenerative medicine, disease modeling, functional genomics, toxicological studies and cell-based therapeutics due to their unique characteristics of self-renewal and pluripotency. Novel methods for generation of pluripotent stem cells and their differentiation to the specialized cell types such as neuronal cells, myocardial cells, hepatocytes and beta cells of the pancreas and many other cells of the body are constantly being refined. Pluripotent stem cell derived differentiated cells, including neuronal cells or cardiac cells, are ideal for stem cell transplantation as autologous or allogeneic cells from healthy donors due to their minimal risk of rejection. Radiation-induced DNA damage, ultraviolet light, genotoxic stress and other intrinsic and extrinsic factors triggers a series of biochemical reactions known as DNA damage response. To maintain genomic stability and avoid transmission of mutations into progenitors cells, stem cells have robust DNA damage response signaling, a contrast to somatic cells. Stem cell transplantation may protect against radiation-induced late effects. In particular, this review focuses on differential DNA damage response between stem cells and derived differentiated cells and the possible pathways that determine such differences.


Subject(s)
DNA Damage , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/radiation effects , Animals , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/radiation effects
10.
PLoS One ; 11(3): e0150923, 2016.
Article in English | MEDLINE | ID: mdl-26950212

ABSTRACT

Exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) can enhance hippocampal neurogenesis in adult mice. However, little is focused on the effects of ELF-EMFs on embryonic neurogenesis. Here, we studied the potential effects of ELF-EMFs on embryonic neural stem cells (eNSCs). We exposed eNSCs to ELF-EMF (50 Hz, 1 mT) for 1, 2, and 3 days with 4 hours per day. We found that eNSC proliferation and maintenance were significantly enhanced after ELF-EMF exposure in proliferation medium. ELF-EMF exposure increased the ratio of differentiated neurons and promoted the neurite outgrowth of eNSC-derived neurons without influencing astrocyes differentiation and the cell apoptosis. In addition, the expression of the proneural genes, NeuroD and Ngn1, which are crucial for neuronal differentiation and neurite outgrowth, was increased after ELF-EMF exposure. Moreover, the expression of transient receptor potential canonical 1 (TRPC1) was significantly up-regulated accompanied by increased the peak amplitude of intracellular calcium level induced by ELF-EMF. Furthermore, silencing TRPC1 expression eliminated the up-regulation of the proneural genes and the promotion of neuronal differentiation and neurite outgrowth induced by ELF-EMF. These results suggest that ELF-EMF exposure promotes the neuronal differentiation and neurite outgrowth of eNSCs via up-regulation the expression of TRPC1 and proneural genes (NeuroD and Ngn1). These findings also provide new insights in understanding the effects of ELF-EMF exposure on embryonic brain development.


Subject(s)
Cell Differentiation/radiation effects , Electromagnetic Fields , Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Neurites/radiation effects , TRPC Cation Channels/genetics , Up-Regulation/radiation effects , Animals , Brain/embryology , Brain/radiation effects , Cell Proliferation/radiation effects , Embryonic Stem Cells/radiation effects , Mice , Mice, Inbred BALB C , Neural Stem Cells/radiation effects , Neurites/metabolism , RNA, Small Interfering/genetics , TRPC Cation Channels/deficiency
11.
Int J Mol Sci ; 16(7): 14737-48, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26133243

ABSTRACT

The biological effects of low-dose ionizing radiation (LDIR) exposure in humans are not comprehensively understood, generating a high degree of controversy in published literature. The earliest stages of human development are known to be among the most sensitive to stress exposures, especially genotoxic stresses. However, the risks stemming from exposure to LDIR, particularly within the clinical diagnostic relevant dose range, have not been directly evaluated in human embryonic stem cells (hESCs). Here, we describe the dynamics of the whole genome transcriptional responses of different hESC lines to both LDIR and, as a reference, high-dose IR (HDIR). We found that even doses as low as 0.05 Gy could trigger statistically significant transient changes in a rather limited subset of genes in all hESCs lines examined. Gene expression signatures of hESCs exposed to IR appear to be highly dose-, time-, and cell line-dependent. We identified 50 genes constituting consensus gene expression signature as an early response to HDIR across all lines of hESC examined. We observed substantial differences in biological pathways affected by either LDIR or HDIR in hESCs, suggesting that the molecular mechanisms underpinning the responses of hESC may fundamentally differ depending on radiation doses.


Subject(s)
Embryonic Stem Cells/radiation effects , Radiation, Ionizing , Transcriptome/radiation effects , Cell Line , Dose-Response Relationship, Radiation , Embryonic Stem Cells/metabolism , Genome, Human , Humans
12.
Int J Radiat Biol ; 91(6): 495-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25786477

ABSTRACT

PURPOSE: Following in utero exposure to low dose radiation (10-200 mGy), we recently observed a linear induction of DNA double-strand breaks (DSB) and activation of apoptosis in the embryonic neuronal stem/progenitor cell compartment. No significant induction of DSB or apoptosis was observed following exposure to magnetic fields (MF). In the present study, we exploited this in vivo system to examine whether exposure to MF before and after exposure to 100 mGy X-rays impacts upon DSB repair rates. MATERIALS AND METHODS: 53BP1 foci were quantified following combined exposure to radiation and MF in the embryonic neuronal stem/progenitor cell compartment. Embryos were exposed in utero to 50 Hz MF at 300 µT for 3 h before and up to 9 h after exposure to 100 mGy X-rays. Controls included embryos exposed to MF or X-rays alone plus sham exposures. RESULTS: Exposure to MF before and after 100 mGy X-rays did not impact upon the rate of DSB repair in the embryonic neuronal stem cell compartment compared to repair rates following radiation exposure alone. CONCLUSIONS: We conclude that in this sensitive system MF do not exert any significant level of DNA damage and do not impede the repair of X-ray induced damage.


Subject(s)
Brain/metabolism , Brain/radiation effects , DNA Breaks, Double-Stranded , DNA Repair/radiation effects , Magnetic Fields/adverse effects , Animals , Brain/embryology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Female , Lateral Ventricles/embryology , Lateral Ventricles/metabolism , Lateral Ventricles/radiation effects , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , Pregnancy
13.
PLoS One ; 10(2): e0117437, 2015.
Article in English | MEDLINE | ID: mdl-25706293

ABSTRACT

Rapidly-frozen hydrated (cryopreserved) specimens combined with cryo-scanning x-ray fluorescence microscopy provide an ideal approach for investigating elemental distributions in biological cells and tissues. However, because cryopreservation does not deactivate potentially infectious agents associated with Risk Group 2 biological materials, one must be concerned with contamination of expensive and complicated cryogenic x-ray microscopes when working with such materials. We employed ultraviolet germicidal irradiation to decontaminate previously cryopreserved cells under liquid nitrogen, and then investigated its effects on elemental distributions under both frozen hydrated and freeze dried states with x-ray fluorescence microscopy. We show that the contents and distributions of most biologically important elements remain nearly unchanged when compared with non-ultraviolet-irradiated counterparts, even after multiple cycles of ultraviolet germicidal irradiation and cryogenic x-ray imaging. This provides a potential pathway for rendering Risk Group 2 biological materials safe for handling in multiuser cryogenic x-ray microscopes without affecting the fidelity of the results.


Subject(s)
Cryopreservation , Embryonic Stem Cells/radiation effects , Fibroblasts/radiation effects , Ultraviolet Rays/adverse effects , Animals , Electron Probe Microanalysis/methods , Embryonic Stem Cells/chemistry , Fibroblasts/chemistry , Mice , Microscopy, Fluorescence
14.
Cell Death Dis ; 6: e1588, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25569105

ABSTRACT

Prolonged culture of embryonic stem cells (ESCs) leads them to adopt embryonal carcinoma cell features, creating enormous dangers for their further application. The mechanism involved in ESC stability has not, however, been extensively studied. We previously reported that SMAD family member 3 (Smad3) has an important role in maintaining mouse ESC stability, as depletion of Smad3 results in cancer cell-like properties in ESCs and Smad3-/- ESCs are prone to grow large, malignant teratomas. To understand how Smad3 contributes to ESC stability, we performed microarray analysis to compare the transcriptome of wild-type and Smad3-/- ESCs. We found that Rif1 (RAP1-associated protein 1), a factor important for genomic stability, is significantly upregulated in Smad3-/- ESCs. The expression level of Rif1 needs to be tightly controlled in ESCs, as a low level of Rif1 is associated with ESC differentiation, but a high level of Rif1 is linked to ESC transformation. In ESCs, Oct4 activates Rif1, whereas Smad3 represses its expression. Oct4 recruits Smad3 to bind to Rif1 promoter, but Smad3 joining facilitates the loading of a polycomb complex that generates a repressive epigenetic modification on Rif1 promoter, and thus maintains the expression of Rif1 at a proper level in ESCs. Interestingly, Rif1 short hairpin RNA (shRNA)-transduced Smad3-/- ESCs showed less malignant properties than the control shRNA-transduced Smad3-/- ESCs, suggesting a critical role of Rif1 in maintaining the stability of ESCs during proliferation.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Smad3 Protein/metabolism , Telomere-Binding Proteins/metabolism , Animals , Cell Differentiation/radiation effects , Cell Movement/radiation effects , Cell Proliferation/radiation effects , DNA Repair/radiation effects , Embryonic Stem Cells/radiation effects , Gene Expression Profiling , Gene Expression Regulation, Developmental/radiation effects , Gene Knockdown Techniques , Histones/metabolism , Lysine/metabolism , Methylation/radiation effects , Mice , Models, Biological , Promoter Regions, Genetic/genetics , Protein Binding/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Smad3 Protein/deficiency , Smad3 Protein/genetics , Telomere-Binding Proteins/genetics , Teratoma/pathology , Ultraviolet Rays
15.
Sci Rep ; 5: 7749, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25582954

ABSTRACT

Terahertz (THz) radiation was proposed recently for use in various applications, including medical imaging and security scanners. However, there are concerns regarding the possible biological effects of non-ionising electromagnetic radiation in the THz range on cells. Human embryonic stem cells (hESCs) are extremely sensitive to environmental stimuli, and we therefore utilised this cell model to investigate the non-thermal effects of THz irradiation. We studied DNA damage and transcriptome responses in hESCs exposed to narrow-band THz radiation (2.3 THz) under strict temperature control. The transcription of approximately 1% of genes was subtly increased following THz irradiation. Functional annotation enrichment analysis of differentially expressed genes revealed 15 functional classes, which were mostly related to mitochondria. Terahertz irradiation did not induce the formation of γH2AX foci or structural chromosomal aberrations in hESCs. We did not observe any effect on the mitotic index or morphology of the hESCs following THz exposure.


Subject(s)
DNA Damage/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Genome, Human , Terahertz Radiation , Transcription, Genetic/radiation effects , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Cell Shape/radiation effects , Chromosome Aberrations , Cluster Analysis , Cyclin B1/metabolism , Cytogenetic Analysis , DNA Breaks, Double-Stranded/radiation effects , G1 Phase/radiation effects , Histones/metabolism , Humans , Indoles/metabolism , Mitotic Index , Molecular Sequence Annotation , Phosphorylation/radiation effects
16.
Toxicol Sci ; 144(1): 138-50, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25516496

ABSTRACT

Because of high exposure to systemic noxae, vascular endothelial cells (EC) have to ensure distinct damage defense and regenerative mechanisms to guarantee vascular health. For meaningful toxicological drug assessments employing embryonic stem cell (ESC)-based in vitro models, functional competence of differentiated progeny and detailed knowledge regarding damage defense mechanisms are essential. Here, mouse ESCs (mESC) were differentiated into functionally competent vascular cells (EC and smooth muscle cells [SMC]). mESC, EC, and SMC were comparatively analyzed regarding DNA repair and DNA damage response (DDR). Differentiation was accompanied by both congruent and unique alterations in repair and DDR characteristics. EC and SMC shared the downregulation of genes involved cell cycle regulation and repair of DNA double-strand breaks (DSBs) and mismatches, whereas genes associated with nucleotide excision repair (NER), apoptosis, and autophagy were upregulated when compared with mESC. Expression of genes involved in base excision repair (BER) was particularly low in SMC. IR-induced formation of DSBs, as detected by nuclear γH2AX foci formation, was most efficient in SMC, the repair of DSBs was fastest in EC. Together with substantial differences in IR-induced phosphorylation of p53, Chk1, and Kap1, the data demonstrate complex alterations in DDR capacity going along with the loss of pluripotency and gain of EC- and SMC-specific functions. Notably, IR exposure of early vascular progenitors did not impair differentiation into functionally competent EC and SMC. Summarizing, mESC-based vascular differentiation models are informative to study the impact of environmental stressors on differentiation and function of vascular cells.


Subject(s)
Cell Differentiation/radiation effects , Embryonic Stem Cells/radiation effects , Endothelial Progenitor Cells/radiation effects , Muscle, Smooth, Vascular/radiation effects , Myocytes, Smooth Muscle/radiation effects , Pluripotent Stem Cells/radiation effects , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Autophagy/radiation effects , Biomarkers/metabolism , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA Breaks, Double-Stranded , DNA Repair , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Gene Expression Regulation , Histones/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/radiation effects , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , RNA, Messenger/metabolism , Time Factors
17.
Int J Mol Sci ; 17(1)2015 Dec 31.
Article in English | MEDLINE | ID: mdl-26729107

ABSTRACT

Exposure to ionizing radiation (IR) is inevitable to humans in real-life scenarios; the hazards of IR primarily stem from its mutagenic, carcinogenic, and cell killing ability. For many decades, extensive research has been conducted on the human cell responses to IR delivered at a low dose/low dose (LD) rate. These studies have shown that the molecular-, cellular-, and tissue-level responses are different after low doses of IR (LDIR) compared to those observed after a short-term high-dose IR exposure (HDIR). With the advent of high-throughput technologies in the late 1990s, such as DNA microarrays, changes in gene expression have also been found to be ubiquitous after LDIR. Very limited subset of genes has been shown to be consistently up-regulated by LDIR, including CDKN1A. Further research on the biological effects and mechanisms induced by IR in human cells demonstrated that the molecular and cellular processes, including transcriptional alterations, activated by LDIR are often related to protective responses and, sometimes, hormesis. Following LDIR, some distinct responses were observed, these included bystander effects, and adaptive responses. Changes in gene expression, not only at the level of mRNA, but also miRNA, have been found to crucially underlie these effects having implications for radiation protection purposes.


Subject(s)
Gene Expression Regulation/radiation effects , Blood Cells/metabolism , Blood Cells/radiation effects , Dose-Response Relationship, Radiation , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/radiation effects , Oligonucleotide Array Sequence Analysis , Radiation, Ionizing , Skin/cytology , Skin/metabolism , Skin/radiation effects
18.
PLoS One ; 9(11): e110472, 2014.
Article in English | MEDLINE | ID: mdl-25415264

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are a class of small non-coding single-stranded RNA molecules that inhibit gene expression at post-transcriptional level. Gadd45g (growth arrest and DNA-damage-inducible 45 gamma) is a stress-response protein, which has been implicated in several biological processes, including DNA repair, the cell cycle and cell differentiation. RESULTS: In this work, we found that miR-383 is a negative regulator of Gadd45g. Forced expression of miR-383 decreased the expression of Gadd45g through binding to the 3' untranslated region (3'-UTR), whereas inhibition of miR-383 increased Gadd45g expression. The presence of miR-383 increased the cellular sensitivity to DNA damage in breast cancer cells, which was rescued by ectopic expression of Gadd45g without the 3'-UTR. miR-383 also regulates the expression of Gadd45g in embryonic stem (ES) cells, but not their apoptosis under genotoxic stress. miR-383 was further showed to negatively regulate ES cell differentiation via targeting Gadd45g, which subsequently modulates the pluripotency-associated genes. Taken together, our study demonstrates that miR-383 is a negative regulator of Gadd45g in both tumor cells and ES cells, however, has distinct function in regulating cell apoptosis. miR-383 may be used as antineoplastic agents in cancer chemotherapy. CONCLUSION: We demonstrate for the first time that miR-383 can specifically regulates the expression of Gadd45g by directly targeting to the 3-UTR region of Gadd45g mRNA, a regulatory process conserved in human tumor cells and mouse embryonic stem cells. These two compotents can be potentially used as antineoplastic agents in cancer chemotherapy.


Subject(s)
Apoptosis/physiology , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/physiology , 3' Untranslated Regions , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Differentiation , Cisplatin/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , MCF-7 Cells , MicroRNAs/metabolism , Protein Binding , Ultraviolet Rays
19.
Cell Death Dis ; 5: e1508, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25375373

ABSTRACT

Human embryonic stem cells (hESC) and induced pluripotent stem cells (hiPSC) are characterised by an unusual and tightly regulated cell cycle that has been shown to be important for the maintenance of a pluripotent phenotype. Cyclin-dependant kinase 1 (CDK1) is a key player in cell cycle regulation and particularly mitosis; however, its role has not been studied previously in hESC and hiPSC. To investigate the impacts of CDK1 downregulation, we performed RNA interference studies which in addition to expected mitotic deficiencies revealed a large range of additional phenotypes related to maintenance of pluripotency, ability to repair double strand breaks (DSBs) and commitment to apoptosis. Downregulation of CDK1 led to the loss of typical pluripotent stem cell morphology, downregulation of pluripotency markers and upregulation of a large number of differentiation markers. In addition, human pluripotent stem cells with reduced CDK1 expression accumulated a higher number of DSBs were unable to activate CHK2 expression and could not maintain G2/M arrest upon exposure to ionising radiation. CDK1 downregulation led to the accumulation of cells with abnormal numbers of mitotic organelles, multiple chromosomal abnormalities and polyploidy. Furthermore, such cells demonstrated an inability to execute apoptosis under normal culture conditions, despite a significant increase in the expression of active PARP1, resulting in tolerance and very likely further propagation of genomic instabilities and ensuing of differentiation process. On the contrary, apoptosis but not differentiation, was the preferred route for such cells when they were subjected to ionising radiation. Together these data suggest that CDK1 regulates multiple events in human pluripotent stem cells ranging from regulation of mitosis, G2/M checkpoint maintenance, execution of apoptosis, maintenance of pluripotency and genomic stability.


Subject(s)
Cyclin-Dependent Kinases/genetics , DNA Repair , Embryonic Stem Cells/metabolism , Genomic Instability/radiation effects , Induced Pluripotent Stem Cells/metabolism , Apoptosis/radiation effects , Biomarkers/metabolism , CDC2 Protein Kinase , Cell Differentiation/radiation effects , Cell Line , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , DNA Breaks, Double-Stranded , Embryonic Stem Cells/cytology , Embryonic Stem Cells/radiation effects , G2 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/radiation effects , Gamma Rays , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/radiation effects , Mitosis/radiation effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Polyploidy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
20.
Apoptosis ; 19(12): 1736-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25273222

ABSTRACT

Ionizing radiation alone or in combination with chemotherapy is the main treatment modality for brain tumors including glioblastoma. Adult neurons and astrocytes demonstrate substantial radioresistance; in contrast, human neural stem cells (NSC) are highly sensitive to radiation via induction of apoptosis. Irradiation of tumor cells has the potential risk of affecting the viability and function of NSC. In this study, we have evaluated the effects of irradiated glioblastoma cells on viability, proliferation and differentiation potential of non-irradiated (bystander) NSC through radiation-induced signaling cascades. Using media transfer experiments, we demonstrated significant effects of the U87MG glioblastoma secretome after gamma-irradiation on apoptosis in non-irradiated NSC. Addition of anti-TRAIL antibody to the transferred media partially suppressed apoptosis in NSC. Furthermore, we observed a dramatic increase in the production and secretion of IL8, TGFß1 and IL6 by irradiated glioblastoma cells, which could promote glioblastoma cell survival and modify the effects of death factors in bystander NSC. While differentiation of NSC into neurons and astrocytes occurred efficiently with the corresponding differentiation media, pretreatment of NSC for 8 h with medium from irradiated glioblastoma cells selectively suppressed the differentiation of NSC into neurons, but not into astrocytes. Exogenous IL8 and TGFß1 increased NSC/NPC survival, but also suppressed neuronal differentiation. On the other hand, IL6 was known to positively affect survival and differentiation of astrocyte progenitors. We established a U87MG neurosphere culture that was substantially enriched by SOX2(+) and CD133(+) glioma stem-like cells (GSC). Gamma-irradiation up-regulated apoptotic death in GSC via the FasL/Fas pathway. Media transfer experiments from irradiated GSC to non-targeted NSC again demonstrated induction of apoptosis and suppression of neuronal differentiation of NSC. In summary, intercellular communication between glioblastoma cells and bystander NSC/NPC could be involved in the amplification of cancer pathology in the brain.


Subject(s)
Apoptosis/radiation effects , Cell Communication/radiation effects , Cell Differentiation/radiation effects , Embryonic Stem Cells/radiation effects , Glioblastoma/metabolism , Neural Stem Cells/radiation effects , Signal Transduction/radiation effects , Cell Survival/radiation effects , Cytokines/metabolism , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Fas Ligand Protein/metabolism , Gamma Rays , Glioblastoma/pathology , Humans , Ligands , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Receptors, Death Domain/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...