Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Molecules ; 29(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39339434

ABSTRACT

A gallium nitride (GaN) semiconductor is one of the most promising materials integrated into biomedical devices to play the roles of connecting, monitoring, and manipulating the activity of biological components, due to its excellent photoelectric properties, chemical stability, and biocompatibility. In this work, it was found that the photogenerated free charge carriers of the GaN substrate, as an exogenous stimulus, served to promote neural stem cells (NSCs) to differentiate into neurons. This was observed through the systematic investigation of the effect of the persistent photoconductivity (PPC) of GaN on the differentiation of primary NSCs from the embryonic rat cerebral cortex. NSCs were directly cultured on the GaN surface with and without ultraviolet (UV) irradiation, with a control sample consisting of tissue culture polystyrene (TCPS) in the presence of fetal bovine serum (FBS) medium. Through optical microscopy, the morphology showed a greater number of neurons with the branching structures of axons and dendrites on GaN with UV irradiation. The immunocytochemical results demonstrated that GaN with UV irradiation could promote the NSCs to differentiate into neurons. Western blot analysis showed that GaN with UV irradiation significantly upregulated the expression of two neuron-related markers, ßIII-tubulin (Tuj-1) and microtubule-associated protein 2 (MAP-2), suggesting that neurite formation and the proliferation of NSCs during differentiation were enhanced by GaN with UV irradiation. Finally, the results of the Kelvin probe force microscope (KPFM) experiments showed that the NSCs cultured on GaN with UV irradiation displayed about 50 mV higher potential than those cultured on GaN without irradiation. The increase in cell membrane potential may have been due to the larger number of photogenerated free charges on the GaN surface with UV irradiation. These results could benefit topical research and the application of GaN as a biomedical material integrated into neural interface systems or other bioelectronic devices.


Subject(s)
Cell Differentiation , Gallium , Neural Stem Cells , Semiconductors , Ultraviolet Rays , Gallium/chemistry , Gallium/pharmacology , Animals , Neural Stem Cells/cytology , Neural Stem Cells/radiation effects , Neural Stem Cells/metabolism , Cell Differentiation/radiation effects , Rats , Cells, Cultured , Cell Proliferation , Neurons/cytology , Neurons/radiation effects , Neurons/metabolism
2.
Mol Psychiatry ; 26(1): 322-340, 2021 01.
Article in English | MEDLINE | ID: mdl-31723242

ABSTRACT

Cranial radiotherapy in children has detrimental effects on cognition, mood, and social competence in young cancer survivors. Treatments harnessing hippocampal neurogenesis are currently of great relevance in this context. Lithium, a well-known mood stabilizer, has both neuroprotective, pro-neurogenic as well as antitumor effects, and in the current study we introduced lithium treatment 4 weeks after irradiation. Female mice received a single 4 Gy whole-brain radiation dose on postnatal day (PND) 21 and were randomized to 0.24% Li2CO3 chow or normal chow from PND 49 to 77. Hippocampal neurogenesis was assessed on PND 77, 91, and 105. We found that lithium treatment had a pro-proliferative effect on neural progenitors, but neuronal integration occurred only after it was discontinued. Also, the treatment ameliorated deficits in spatial learning and memory retention observed in irradiated mice. Gene expression profiling and DNA methylation analysis identified two novel factors related to the observed effects, Tppp, associated with microtubule stabilization, and GAD2/65, associated with neuronal signaling. Our results show that lithium treatment reverses irradiation-induced loss of hippocampal neurogenesis and cognitive impairment even when introduced long after the injury. We propose that lithium treatment should be intermittent in order to first make neural progenitors proliferate and then, upon discontinuation, allow them to differentiate. Our findings suggest that pharmacological treatment of cognitive so-called late effects in childhood cancer survivors is possible.


Subject(s)
Cognition/drug effects , Lithium Compounds/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/radiation effects , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Female , Hippocampus/cytology , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects
3.
Bull Exp Biol Med ; 172(2): 228-235, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855083

ABSTRACT

We studied the effect of neural stem cells (NSC) and mesenchymal stem cells (MSC) from mouse adipose tissue on survival, clonogenic activity, and senescence of NSC after exposure to γ-radiation. It was found that survival and clonogenic activity of NSC irradiated in doses of 1 and 2 Gy was enhanced when irradiated cells were co-cultured with non-irradiated NSC and MSC in permeable Transwell inserts. The proportion of senescent NSC (cells with high ß-galactosidase activity) increased with increasing irradiation dose. Co-culturing with non-irradiated NSC in 3 days after irradiation in a dose of 1 Gy led to a decrease in the proportion of senescent cells among irradiated NSC. Factors secreted by NSC and MSC can become the basis for the development of means for prevention and treatment of damage to brain cells resulting from radiation therapy of head and neck cancer.


Subject(s)
Gamma Rays/adverse effects , Mesenchymal Stem Cells/cytology , Neural Stem Cells/radiation effects , Adipose Tissue/cytology , Animals , Apoptosis/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Coculture Techniques , Colony-Forming Units Assay , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/physiology
4.
Biochem Biophys Res Commun ; 523(3): 555-560, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31864707

ABSTRACT

DNA damaging agents, such as ionizing radiation (IR), induce cell cycle arrest, senescence, differentiation, or cell death of stem cells, which may affect tissue homeostasis. The specific response of stem cells upon irradiation seems to vary depending on the cell type and their developmental stages. Drosophila larval brain contains neural stem cells called neuroblasts (NBs) and maintaining an appropriate number of NBs is critical to maintain brain size. Irradiation of larvae at early larval stage results in microcephaly, whereas the DNA damage response of NBs that could explain this small brain size is not clearly understood. We observed that the irradiation of larvae in the second instar retarded brain growth, accompanied by fewer NBs. The IR-induced microcephaly does not seem to result from apoptosis since the irradiated larval brain was not stained with activated Caspase nor was the microcephaly affected by the ectopic expression of the apoptosis inhibitor. When analyzed for the percentage of mitotic cells, irradiated NBs recovered their proliferative potential within 6 h post-irradiation after transient cell cycle arrest. However, IR eventually reduced the proliferation of NBs at later time points and induced the premature differentiation of NBs. In summary, IR-induced microcephaly occurs by NB loss due to premature differentiation, rather than apoptotic cell death.


Subject(s)
Drosophila/radiation effects , Neural Stem Cells/radiation effects , Neurogenesis/radiation effects , Animals , Brain/growth & development , Brain/radiation effects , Drosophila/cytology , Drosophila/growth & development , Larva/cytology , Larva/growth & development , Larva/radiation effects , Microcephaly/etiology , Neural Stem Cells/cytology , Organ Size/radiation effects , Radiation, Ionizing
5.
Biochem Biophys Res Commun ; 531(4): 535-542, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32807492

ABSTRACT

INTRODUCTION: Pre-clinical testing of small molecules for therapeutic development across many pathologies relies on the use of in-vitro and in-vivo models. When designed and implemented well, these models serve to predict the clinical outcome as well as the toxicity of the evaluated therapies. The two-dimensional (2D) reductionist approach where cells are incubated in a mono-layer on hard plastic microtiter plates is relatively inexpensive but not physiologically relevant. In contrast, well developed and applied three dimensional (3D) in vitro models could be employed to bridge the gap between 2D in vitro primary screening and expensive in vivo rodent models by incorporating key features of the tissue microenvironment to explore differentiation, cortical development, cancers and various neuronal dysfunctions. These features include an extracellular matrix, co-culture, tension and perfusion and could replace several hundred rodents in the drug screening validation cascade. METHODS: Human neural progenitor cells from middle brain (ReN VM, Merck Millipore, UK) were expanded as instructed by the supplier (Merck Millipore, UK), and then seeded in 96-well low-attachment plates (Corning, UK) to form multicellular spheroids followed by adding a Matrigel layer to mimic extracellular matrix around neural stem cell niche. ReN VM cells were then differentiated via EGF and bFGF deprivation for 7 days and were imaged at day 7. Radiotherapy was mimicked via gamma-radiation at 2Gy in the absence and presence of selected DYRK1A inhibitors Harmine, INDY and Leucettine 41 (L41). Cell viability was measured by AlamarBlue assay. Immunofluorescence staining was used to assess cell pluripotency marker SOX2 and differentiation marker GFAP. RESULTS: After 7 days of differentiation, neuron early differentiation marker (GFAP, red) started to be expressed among the cells expressing neural stem cell marker SOX2 (green). Radiation treatment caused significant morphology change including the reduced viability of the spheroids. These spheroids also revealed sensitizing potential of DYRK1A inhibitors tested in this study, including Harmine, INDY and L41. DISCUSSION & CONCLUSIONS: Combined with the benefit of greatly reducing the issues associated with in vivo rodent models, including reducing numbers of animals used in a drug screening cascade, cost, ethics, and potential animal welfare burden, we feel the well-developed and applied 3D neural spheroid model presented in this study will provide a crucial tool to evaluate combinatorial therapies, optimal drug concentrations and treatment dosages.


Subject(s)
Drug Evaluation, Preclinical/methods , Neural Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Spheroids, Cellular/drug effects , Cell Line , Collagen , Dioxoles/pharmacology , Drug Combinations , Extracellular Matrix , Gamma Rays , Harmine/pharmacology , Humans , Imidazoles/pharmacology , Laminin , Neural Stem Cells/radiation effects , Neurites/drug effects , Proteoglycans , Radiation-Sensitizing Agents/pharmacology , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/radiation effects , Dyrk Kinases
6.
PLoS Biol ; 15(5): e2001264, 2017 05.
Article in English | MEDLINE | ID: mdl-28489848

ABSTRACT

Stem and differentiated cells frequently differ in their response to DNA damage, which can determine tissue sensitivity. By exploiting insight into the spatial arrangement of subdomains within the adult neural subventricular zone (SVZ) in vivo, we show distinct responses to ionising radiation (IR) between neural stem and progenitor cells. Further, we reveal different DNA damage responses between neonatal and adult neural stem cells (NSCs). Neural progenitors (transit amplifying cells and neuroblasts) but not NSCs (quiescent and activated) undergo apoptosis after 2 Gy IR. This response is cell type- rather than proliferation-dependent and does not appear to be driven by distinctions in DNA damage induction or repair capacity. Moreover, exposure to 2 Gy IR promotes proliferation arrest and differentiation in the adult SVZ. These 3 responses are ataxia telangiectasia mutated (ATM)-dependent and promote quiescent NSC (qNSC) activation, which does not occur in the subdomains that lack progenitors. Neuroblasts arising post-IR derive from activated qNSCs rather than irradiated progenitors, minimising damage compounded by replication or mitosis. We propose that rather than conferring sensitive cell death, apoptosis is a form of rapid cell death that serves to remove damaged progenitors and promote qNSC activation. Significantly, analysis of the neonatal (P5) SVZ reveals that although progenitors remain sensitive to apoptosis, they fail to efficiently arrest proliferation. Consequently, their repopulation occurs rapidly from irradiated progenitors rather than via qNSC activation.


Subject(s)
Apoptosis , DNA Damage , Lateral Ventricles/radiation effects , Neural Stem Cells/radiation effects , Animals , Animals, Newborn , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Differentiation , Cell Proliferation/radiation effects , Mice, Inbred C57BL , X-Rays
7.
Arch Toxicol ; 93(10): 2879-2893, 2019 10.
Article in English | MEDLINE | ID: mdl-31493029

ABSTRACT

Prenatal brain development is a complex and sensitive process, highly susceptible to environmental influences such as pollutants, stress, malnutrition, drugs, tobacco exposure, or ionizing radiation (IR). Disturbances in development may cause life-long disabilities and diseases, such as ADHD, childhood cancers, cognitive problems, depression, anxiety and more severe developmental disabilities. Due to increasing medical imaging, radiation therapy, natural terrestrial radiation, radioactive pollution and long-distance flights, humans are increasingly exposed to IR. However, data on impact of IR on very early human brain development are scarce, particularly in the very first weeks of gestation. Here we investigated the effects of low-dose X-ray IR (1 Gy) in a 3D early brain developmental model derived from human pluripotent stem cells. In this model very early neural stem cells, neuroectodermal progenitor cells (NEP), were exposed to low-dose IR and direct as well as delayed effects were investigated. Expression of 20 different marker genes crucial for normal neural development was determined 48 h and 9 days post IR (pIR). All but one of the analyzed marker genes were reduced 48 h after IR, and all but seven genes normalized their expression by day 9 pIR. Among the seven markers were genes involved in neurodevelopmental and growth abnormalities. Moreover, we could show that stemness of the NEP was reduced after IR. We were thus able to identify a significant impact of radiation in cells surviving low-dose IR, suggesting that low-dose IR could have a negative impact on the early developing human brain, with potential later detrimental effects.


Subject(s)
Gene Expression Regulation/radiation effects , Induced Pluripotent Stem Cells/radiation effects , Neural Stem Cells/radiation effects , Radiation, Ionizing , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Radiation Dosage , Time Factors
8.
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
9.
Sheng Li Xue Bao ; 71(3): 431-438, 2019 Jun 25.
Article in Zh | MEDLINE | ID: mdl-31218334

ABSTRACT

The present study was aimed to investigate the effects and mechanisms of electro-acupuncture (EA) on proliferation and differentiation of neural stem cells in the hippocampus of C57 mice exposed to different doses of X-ray radiation. Thirty-day-old C57BL/6J mice were randomly divided into control, irradiation, and EA groups. The control group was not treated with irradiation. The irradiation groups were exposed to different doses of X-ray (4, 8 or 16 Gy) for 10 min. The EA groups were electro-acupunctured at Baihui, Fengfu and bilateral Shenyu for 3 courses of treatment after X-ray radiation. Immunohistochemistry was used to evaluate proliferation and differentiation of the hippocampal neural stem cell. RT-PCR and Western blot were used to detect mRNA and protein expressions of Notch1 and Mash1 in the hippocampus, respectively. The results showed that, compared with the control group, the numbers of BrdU positive cells (4, 8 Gy subgroup) and BrdU/NeuN double-labeling positive cells (3 dose subgroups) were decreased significantly in the irradiation group, but the above changes could be reversed by EA. Compared with the control group, the number of BrdU/GFAP double-labeling positive cells in each dose subgroup of irradiation group was decreased significantly, while EA could reverse the change of 4 and 8 Gy dose subgroups. In addition, compared with the control group, the expression levels of Notch1 mRNA and protein in hippocampus were up-regulated, and the expression levels of Mash1 mRNA and protein were significantly decreased in each dose subgroup of irradiation group. Compared with irradiation group, the expression levels of Notch1 mRNA and protein in hippocampus of EA group were decreased significantly in each dose subgroup, and the expression levels of Mash1 mRNA and protein were increased significantly in 4 and 8 Gy subgroups. These results suggest that irradiation affects the proliferation and differentiation of neural stem cells in hippocampus of mice, whereas EA may significantly increase the proliferation and differentiation of hippocampal neural stem cells via the regulation of Notch signaling pathway.


Subject(s)
Cell Differentiation , Cell Proliferation , Electroacupuncture , Neural Stem Cells/cytology , X-Rays/adverse effects , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hippocampus/cytology , Hippocampus/radiation effects , Mice, Inbred C57BL , Neural Stem Cells/radiation effects , Random Allocation , Receptor, Notch1/metabolism
10.
Cell Physiol Biochem ; 48(2): 657-669, 2018.
Article in English | MEDLINE | ID: mdl-30025390

ABSTRACT

BACKGROUND/AIMS: Alternative splicing and DNA damage exhibit cross-regulation, with not only DNA damage inducing changes in alternative splicing, but alternative splicing itself possibly modulating the DNA damage response (DDR). Sirt1, a prominent anti-aging player, plays pivotal roles in the DDR. However, few studies have examined alternative splicing with DNA damage in neural stem cells (NSCs) and, in essence, nothing is known about whether SIRT1 regulates alternative splicing. Hence, we investigated the potential involvement of Sirt1-mediated alternative splicing in the NSC DDR. METHODS: Genome-wide alternative splicing profiling was performed upon DNA damage induction and SIRT1 deletion. RESULTS: DNA damage caused genome-wide changes in alternative splicing in adult NSCs and Sirt1 deficiency dramatically altered DDR-related alternative splicing. In particular, extensive alternative splicing changes in DDR-related processes such as cell cycle control and DNA damage repair were observed; these processes were dramatically influenced by Sirt1 deficiency. Phenotypically, Sirt1 deficiency altered the proliferation and DNA repair of adult NSCs, possibly by regulating alternative splicing. CONCLUSION: SIRT1 helps to regulate alternative splicing, which itself affects the DDR of NSCs. Our findings provide novel insight into the mechanisms underlying the DDR in stem cells.


Subject(s)
DNA Repair , Sirtuin 1/genetics , Alternative Splicing/radiation effects , Animals , Cell Cycle Checkpoints , Cell Proliferation , Cells, Cultured , DNA Damage/radiation effects , Lateral Ventricles/cytology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , Radiation, Ionizing , Sirtuin 1/deficiency , Sirtuin 1/metabolism
11.
J Neurooncol ; 138(3): 509-518, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29564746

ABSTRACT

Glioblastoma (GBM) is a highly fatal disease with a 5 year survival rate of less than 22%. One of the most effective treatment regimens to date is the use of radiotherapy which induces lethal DNA double-strand breaks to prevent tumour growth. However, recurrence occurs in the majority of patients and is in-part a result of robust radioresistance mechanisms. In this study, we demonstrate that the multifunctional cytokine, interleukin-6 (IL-6), confers a growth advantage in GBM cells but does not have the same effect on normal neural progenitor cells. Further analysis showed IL-6 can promote radioresistance in GBM cells when exposed to ionising radiation. Ablation of the Ataxia-telangiectasia mutated serine/threonine kinase that is recruited and activated by DNA double-strand breaks reverses the effect of radioresistance and re-sensitised GBM to DNA damage thus leading to increase cell death. Our finding suggests targeting the signaling cascade of DNA damage response is a potential therapeutic approach to circumvent IL-6 from promoting radioresistance in GBM.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cell Proliferation/radiation effects , Central Nervous System Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Interleukin-6/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Death/physiology , Cell Death/radiation effects , Cell Line , Cell Proliferation/physiology , Cell Survival/physiology , Cell Survival/radiation effects , Central Nervous System Neoplasms/metabolism , DNA Damage/radiation effects , Glioblastoma/metabolism , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , RNA, Messenger/metabolism , Radiation Tolerance/physiology , Radiation, Ionizing , Receptors, Interleukin-6/metabolism
12.
Nanomedicine ; 14(7): 2485-2494, 2018 10.
Article in English | MEDLINE | ID: mdl-28552650

ABSTRACT

Carbon-based nanomaterials have shown great promise in regenerative medicine because of their unique electrical, mechanical, and biological properties; however, it is still difficult to engineer 2D pure carbon nanomaterials into a 3D scaffold while maintaining its structural integrity. In the present study, we developed novel carbon nanofibrous scaffolds by annealing electrospun mats at elevated temperature. The resultant scaffold showed a cohesive structure and excellent mechanical flexibility. The graphitic structure generated by annealing renders superior electrical conductivity to the carbon nanofibrous scaffold. By integrating the conductive scaffold with biphasic electrical stimulation, neural stem cell proliferation was promoted associating with upregulated neuronal gene expression level and increased microtubule-associated protein 2 immunofluorescence, demonstrating an improved neuronal differentiation and maturation. The findings suggest that the integration of the conducting carbon nanofibrous scaffold and electrical stimulation may pave a new avenue for neural tissue regeneration.


Subject(s)
Electric Stimulation , Guided Tissue Regeneration/instrumentation , Nanofibers/chemistry , Nerve Regeneration/physiology , Neural Stem Cells/physiology , Tissue Engineering , Tissue Scaffolds , Animals , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , Guided Tissue Regeneration/methods , Mice , Nerve Regeneration/radiation effects , Neural Stem Cells/cytology , Neural Stem Cells/radiation effects
13.
J Cell Sci ; 128(19): 3597-606, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26303202

ABSTRACT

The embryonic neural stem cell compartment is characterised by rapid proliferation from embryonic day (E)11 to E16.5, high endogenous DNA double-strand break (DSB) formation and sensitive activation of apoptosis. Here, we ask whether DSBs arise in the adult neural stem cell compartments, the sub-ventricular zone (SVZ) of the lateral ventricles and the sub-granular zone (SGZ) of the hippocampal dentate gyrus, and whether they activate apoptosis. We used mice with a hypomorphic mutation in DNA ligase IV (Lig4(Y288C)), ataxia telangiectasia mutated (Atm(-/-)) and double mutant Atm(-/-)/Lig4(Y288C) mice. We demonstrate that, although DSBs do not arise at a high frequency in adult neural stem cells, the low numbers of DSBs that persist endogenously in Lig4(Y288C) mice or that are induced by low radiation doses can activate apoptosis. A temporal analysis shows that DSB levels in Lig4(Y288C) mice diminish gradually from the embryo to a steady state level in adult mice. The neonatal SVZ compartment of Lig4(Y288C) mice harbours diminished DSBs compared to its differentiated counterpart, suggesting a process selecting against unfit stem cells. Finally, we reveal high endogenous apoptosis in the developing SVZ of wild-type newborn mice.


Subject(s)
Apoptosis/radiation effects , DNA Breaks, Double-Stranded/radiation effects , Neural Stem Cells/radiation effects , X-Rays , Animals , Apoptosis/genetics , Cells, Cultured , Female , In Situ Nick-End Labeling , Male , Mice
14.
Biochem Biophys Res Commun ; 479(4): 820-826, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27680314

ABSTRACT

Oscillations in Notch signaling are essential for reserving neural progenitors for cellular diversity in developing brains. Thus, steady and prolonged overactivation of Notch signaling is not suitable for generating neurons. To acquire greater temporal control of Notch activity and mimic endogenous oscillating signals, here we adopted a light-inducible transgene system to induce active form of Notch NICD in neural progenitors. Alternating Notch activity saved more progenitors that are prone to produce neurons creating larger number of mixed clones with neurons and progenitors in vitro, compared to groups with no light or continuous light stimulus. Furthermore, more upper layer neurons and astrocytes arose upon intermittent Notch activity, indicating that dynamic Notch activity maintains neural progeny and fine-tune neuron-glia diversity.


Subject(s)
Light , Neurogenesis/physiology , Neurogenesis/radiation effects , Receptor, Notch1/metabolism , Receptor, Notch1/radiation effects , Animals , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/radiation effects , Cell Differentiation/radiation effects , Cell Line , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , Neuroglia/cytology , Neuroglia/metabolism , Neuroglia/radiation effects , Neurons/cytology , Neurons/metabolism , Neurons/radiation effects , Protein Domains , Receptor, Notch1/chemistry , Signal Transduction
15.
Int J Mol Sci ; 17(5)2016 Jun 20.
Article in English | MEDLINE | ID: mdl-27331809

ABSTRACT

Mammalian tissue-specific stem cells and progenitors demonstrate differential DNA damage response. Neural progenitors in dentate gyrus of the hippocampus are known to undergo apoptosis after irradiation. Using a mouse model of hippocampal neuronal development, we characterized the apoptosis sensitivity of the different neural progenitor subpopulations in adult mouse dentate gyrus after irradiation. Two different bromodeoxyuridine incorporation paradigms were used for cell fate mapping. We identified two apoptosis sensitive neural progenitor subpopulations after irradiation. The first represented non-proliferative and non-newborn neuroblasts and immature neurons that expressed doublecortin, calretinin or both. The second consisted of proliferative intermediate neural progenitors. The putative radial glia-like neural stem cells or type-1 cells, regardless of proliferation status, were apoptosis resistant after irradiation. There was no evidence of radiation-induced apoptosis in the absence of the Trp53 (p53) gene but absence of Cdkn1a (p21) did not alter the apoptotic response. Upregulation of nuclear p53 was observed in neuroblasts after irradiation. We conclude that adult hippocampal neural progenitors may demonstrate differential p53-dependent apoptosis sensitivity after irradiation.


Subject(s)
Apoptosis , Hippocampus/radiation effects , Neural Stem Cells/radiation effects , Radiation Tolerance , Animals , Calbindin 2/genetics , Calbindin 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Doublecortin Domain Proteins , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Neuropeptides/genetics , Neuropeptides/metabolism , Radiation, Ionizing , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Radiats Biol Radioecol ; 56(1): 35-43, 2016.
Article in Russian | MEDLINE | ID: mdl-27245003

ABSTRACT

Mesenchymal stem cells (MSC) exist in the brain in addition to the neural stem cells (NSC). The aim of this work was to investigate the sensitivity of mouse brain MSC (MSC(BR)) to sublethal doses of γ-radiation in comparison with the sensitivity of bone marrow MSC (MSC(BM)) and NSC and to study the effects of γ-irradiation at low doses on these cells. Cells were exposed to γ-radiation (137Cs) at the doses of 10 to 200 mGy at a dose rate of 10 mGy/min; higher doses were achieved at the dose rates of 200 and 500 mGy/min (60Co). The survival of cells was assessed by counting living cells after staining with trypan blue in the Goryaev's chamber or using the MTT test for NSC growing as neurospheres. SP fraction was measured using flow cytometry after incubation with rhodamine-123. Exposure to the doses in the range of 10 to 500 mGy stimulated cell proliferation. The maximum decrease in the cells number was seen on the seventh day after irradiation and it was practically the same for the MSC(BR) and MSC(BM). NCS were more radiosensitive than MSC. Exposure to the doses of 100 to 500 mGy stimulated cells proliferation of all SCs except of MSC(BM). It was shown that the size of SP fraction of MSC(BR) was diminished after γ-irradiation at low doses. Thus, the stimulation of cell proliferation after γ-irradiation at low doses is accompanied by the redistribution of distinct cell subpopulations: the decrease in the SP fraction and the increase in the general population of cells were observed.


Subject(s)
Cell Proliferation/radiation effects , Gamma Rays/adverse effects , Mesenchymal Stem Cells/radiation effects , Neural Stem Cells/radiation effects , Animals , Brain/radiation effects , Cell Differentiation/radiation effects , Flow Cytometry , Mice , Radiation Dosage
17.
Stem Cells ; 32(1): 59-69, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24038623

ABSTRACT

Radiation is a common tool in the treatment of brain tumors that induces neurological deficits as a side effect. Some of these deficits appear to be related to the impact of radiation on the neurogenic niches, producing a drastic decrease in the proliferative capacity of these regions. In the adult mammalian brain, the subventricular zone (SVZ) of the lateral ventricles is the main neurogenic niche. Neural stem/precursor cells (NSCs) within the SVZ play an important role in brain repair following injuries. However, the irradiated NSCs' ability to respond to damage has not been previously elucidated. In this study, we evaluated the effects of localized radiation on the SVZ ability to respond to a lysolecithin-induced demyelination of the striatum. We demonstrated that the proliferation rate of the irradiated SVZ was increased after brain damage and that residual NSCs were reactivated. The irradiated SVZ had an expansion of doublecortin positive cells that appeared to migrate from the lateral ventricles toward the demyelinated striatum, where newly generated oligodendrocytes were found. In addition, in the absence of demyelinating damage, remaining cells in the irradiated SVZ appeared to repopulate the neurogenic niche a year post-radiation. These findings support the hypothesis that NSCs are radioresistant and can respond to a brain injury, recovering the neurogenic niche. A more complete understanding of the effects that localized radiation has on the SVZ may lead to improvement of the current protocols used in the radiotherapy of cancer.


Subject(s)
Cerebral Ventricles/radiation effects , Demyelinating Diseases/metabolism , Neural Stem Cells/radiation effects , Animals , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Movement/physiology , Cell Movement/radiation effects , Cell Proliferation , Cerebral Ventricles/metabolism , Cerebral Ventricles/pathology , Demyelinating Diseases/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neural Stem Cells/cytology
18.
Proc Natl Acad Sci U S A ; 109(52): 21522-7, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236175

ABSTRACT

Cranial irradiation is widely used in cancer therapy, but it often causes cognitive defects in cancer survivors. Oxidative stress is considered a major cause of tissue injury from irradiation. However, in an earlier study mice deficient in the antioxidant enzyme extracellular superoxide dismutase (EC-SOD KO) showed reduced sensitivity to radiation-induced defects in hippocampal functions. To further dissect the role of EC-SOD in neurogenesis and in response to irradiation, we generated a bigenic EC-SOD mouse model (OE mice) that expressed high levels of EC-SOD in mature neurons in an otherwise EC-SOD-deficient environment. EC-SOD deficiency was associated with reduced progenitor cell proliferation in the subgranular zone of dentate gyrus in KO and OE mice. However, high levels of EC-SOD in the granule cell layer supported normal maturation of newborn neurons in OE mice. Following irradiation, wild-type mice showed reduced hippocampal neurogenesis, reduced dendritic spine densities, and defects in cognitive functions. OE and KO mice, on the other hand, were largely unaffected, and the mice performed normally in neurocognitive tests. Although the resulting hippocampal-related functions were similar in OE and KO mice following cranial irradiation, molecular analyses suggested that they may be governed by different mechanisms: whereas neurotrophic factors may influence radiation responses in OE mice, dendritic maintenance may be important in the KO environment. Taken together, our data suggest that EC-SOD plays an important role in all stages of hippocampal neurogenesis and its associated cognitive functions, and that high-level EC-SOD may provide protection against irradiation-related defects in hippocampal functions.


Subject(s)
Cognition/radiation effects , Extracellular Space/enzymology , Hippocampus/physiology , Hippocampus/radiation effects , Neurogenesis/radiation effects , Radiation, Ionizing , Superoxide Dismutase/metabolism , Animals , Axons/metabolism , Axons/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Dendrites/metabolism , Dendrites/radiation effects , Memory/radiation effects , Mice , Nerve Growth Factors/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/radiation effects , Time Factors , Transcription Factors/metabolism
19.
Apoptosis ; 19(3): 399-413, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24158598

ABSTRACT

Adult neurons, which are terminally differentiated cells, demonstrate substantial radioresistance. In contrast, human neural stem cells (NSC), which have a significant proliferative capacity, are highly sensitive to ionizing radiation. Cranial irradiation that is widely used for treatment of brain tumors may induce death of NSC and further cause substantial cognitive deficits such as impairing learning and memory. The main goal of our study was to determine a mechanism of NSC radiosensitivity. We observed a constitutive high-level expression of TRAIL-R2 in human NSC. On the other hand, ionizing radiation through generation of reactive oxygen species targeted cell signaling pathways and dramatically changed the pattern of gene expression, including upregulation of TRAIL. A significant increase of endogenous expression and secretion of TRAIL could induce autocrine/paracrine stimulation of the TRAIL-R2-mediated signaling cascade with activation of caspase-3-driven apoptosis. Furthermore, paracrine stimulation could initiate bystander response of non-targeted NSC that is driven by death ligands produced by directly irradiated NSC. Experiments with media transfer from directly irradiated NSC to non-targeted (bystander) NSC confirmed a role of secreted TRAIL for induction of a death signaling cascade in non-targeted NSC. Subsequently, TRAIL production through elimination of bystander TRAIL-R-positive NSC might substantially restrict a final yield of differentiating young neurons. Radiation-induced TRAIL-mediated apoptosis could be partially suppressed by anti-TRAIL antibody added to the cell media. Interestingly, direct gamma-irradiation of SK-N-SH human neuroblastoma cells using clinical doses (2-5 Gy) resulted in low levels of apoptosis in cancer cells that was accompanied however by induction of a strong bystander response in non-targeted NSC. Numerous protective mechanisms were involved in the maintenance of radioresistance of neuroblastoma cells, including constitutive PI3K-AKT over-activation and endogenous synthesis of TGFß1. Specific blockage of these survival pathways was accompanied by a dramatic increase in radiosensitivity of neuroblastoma cells. Intercellular communication between cancer cells and NSC could potentially be involved in amplification of cancer pathology in the brain.


Subject(s)
Apoptosis/radiation effects , Bystander Effect/radiation effects , Neural Stem Cells/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Cell Line, Tumor , Gamma Rays/adverse effects , Humans , Neural Stem Cells/pathology , Neural Stem Cells/radiation effects , Neuroblastoma/pathology , Radiation Tolerance , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Signal Transduction/radiation effects , TNF-Related Apoptosis-Inducing Ligand/genetics
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