Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.658
1.
J Biophotonics ; 17(5): e202300369, 2024 May.
Article En | MEDLINE | ID: mdl-38332564

5-Aminolevulinic acid (5-ALA) photodynamic therapy (PDT) is a treatment for actinic keratosis (AK) and has been studied as a treatment for noninvasive cutaneous squamous cell carcinoma (cSCC). PDT induces apoptosis and necrosis in AKs and cSCC. 5-ALA blue light PDT may modulate gene expression and pathways in surviving cells. In this study, differential gene expression and pathway analysis of cSCC and human dermal fibroblasts were compared before and after 5-ALA blue light PDT using RNA sequencing. No genes were differentially expressed after correcting for multiple testing (false discovery rate < 0.05). As a result, transcription factor, gene enrichment, and pathway analysis were performed with genes identified before multiple testing (p < 0.05). Pathways associated with proliferation and carcinogenesis were downregulated. These findings using 5-ALA blue light PDT are similar to previously published studies using methyl-aminolevulinic and red light protocols, indicating that surviving residual cells may undergo changes consistent with a less aggressive cancerous phenotype.


Aminolevulinic Acid , Carcinoma, Squamous Cell , Cell Proliferation , Down-Regulation , Photochemotherapy , Skin Neoplasms , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Humans , Skin Neoplasms/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/drug effects , Down-Regulation/drug effects , Cell Division/drug effects , Cell Division/radiation effects , Light , Gene Expression Regulation, Neoplastic/drug effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Blue Light
2.
Sci Rep ; 11(1): 20464, 2021 10 14.
Article En | MEDLINE | ID: mdl-34650158

In this study we demonstrated that exposure of Escherichia coli (E. coli) to terahertz (THz) radiation resulted in a change in the activities of the tdcABCDEFGR and matA-F genes (signs of cell aggregation), gene yjjQ (signs of suppression of cell motility), dicABCF, FtsZ, and minCDE genes (signs of suppression of cell division), sfmACDHF genes (signs of adhesin synthesis), yjbEFGH and gfcA genes (signs of cell envelope stabilization). Moreover, THz radiation induced E. coli csg operon genes of amyloid biosynthesis. Electron microscopy revealed that the irradiated bacteria underwent increased aggregation; 20% of them formed bundle-like structures consisting of two to four pili clumped together. This could be the result of changes in the adhesive properties of the pili. We also found aberrations in cell wall structure in the middle part of the bacterial cell; these aberrations impaired the cell at the initial stages of division and resulted in accumulation of long rod-like cells. Overall, THz radiation was shown to have adverse effects on bacterial populations resulting in cells with abnormal morphology.


Cell Aggregation/radiation effects , Cell Division/radiation effects , Escherichia coli/radiation effects , Terahertz Radiation , Cell Wall/radiation effects , Escherichia coli/cytology , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/radiation effects , Microscopy, Electron , Operon/genetics
3.
PLoS One ; 16(8): e0248381, 2021.
Article En | MEDLINE | ID: mdl-34339441

Biological phenomena induced by terahertz (THz) irradiation are described in recent reports, but underlying mechanisms, structural and dynamical change of specific molecules are still unclear. In this paper, we performed time-lapse morphological analysis of human cells and found that THz irradiation halts cell division at cytokinesis. At the end of cytokinesis, the contractile ring, which consists of filamentous actin (F-actin), needs to disappear; however, it remained for 1 hour under THz irradiation. Induction of the functional structures of F-actin was also observed in interphase cells. Similar phenomena were also observed under chemical treatment (jasplakinolide), indicating that THz irradiation assists actin polymerization. We previously reported that THz irradiation enhances the polymerization of purified actin in vitro; our current work shows that it increases cytoplasmic F-actin in vivo. Thus, we identified one of the key biomechanisms affected by THz waves.


Actins/radiation effects , Cell Division/radiation effects , Terahertz Radiation , Actins/metabolism , Cytokinesis/radiation effects , HeLa Cells/radiation effects , Humans , Interphase/radiation effects , Microscopy, Fluorescence , Single-Cell Analysis
4.
Cell Mol Biol (Noisy-le-grand) ; 66(7): 51-55, 2020 Oct 31.
Article En | MEDLINE | ID: mdl-33287922

Ciprofloxacin hydrochloride and Norfloxacin are second-generation fluoroquinolone antibiotic against bacterial DNA gyrase, which reduces DNA strain throughout replication. As DNA gyrase is essential through DNA replication, subsequent DNA synthesis and cell division are inhibited. Direct photolysis of fluoroquinolones was studied by using UV irradiation in the presence or absence of other substances that generate free radicals. This study aimed to assess the effect of Ultraviolet B (UVB) irradiation in removing ciprofloxacin and norfloxacin by using a simulating model of wastewater contained urea at pH 4. A known concentration of ciprofloxacin and norfloxacin were prepared in an appropriate aqueous solution in presence or absence 0.2M urea and adjusted at pH 4. The dis-solved drugs were irradiated with UVB-lamp in a dark place for 60 minutes. The percent of removal and the rate of elimination (k) of each drug were calculated. The direct photolysis effect of UVB irradiation was observed with ciprofloxacin which amounted to 24.4% removal compared with12.4% removal of norfloxacin after 60 minutes of irradiation. The effect of UVB irradiation was enhanced by urea to reach 38.9% and 15% for ciprofloxacin and norfloxacin. The calculated k of ciprofloxacin has amounted to three folds of that of norfloxacin. Direct photolysis of ciprofloxacin and norfloxacin can be achieved simply by using a simulation model of 0.2 M urea and UVB irradiation at pH 4. UVB is highly effective in removing ciprofloxacin compared with norfloxacin by 2-3 folds.


Cell Division/drug effects , Ciprofloxacin/pharmacology , DNA Replication/drug effects , DNA, Bacterial/drug effects , Norfloxacin/pharmacology , Ultraviolet Rays , Urea/chemistry , Cell Division/radiation effects , Ciprofloxacin/radiation effects , Culture Media , DNA Replication/radiation effects , DNA, Bacterial/radiation effects , Norfloxacin/radiation effects , Regression Analysis
5.
Radiat Res ; 194(4): 379-389, 2020 10 02.
Article En | MEDLINE | ID: mdl-32936901

The effect of low-dose-rate exposure to ionizing radiation on cancer risk is a major issue associated with radiation protection. Tissue stem cells are regarded as one of the targets of radiation-induced carcinogenesis. However, it is hypothesized that the effect of radiation may be reduced if damaged stem cells are eliminated via stem cell competition between damaged and intact stem cells. This would be particularly effective under very low-dose-rate conditions, in which only a few stem cells in a stem cell pool may be affected by radiation. Following this hypothesis, we constructed a simple mathematical model to discuss the influence of stem cell competition attenuating the accumulation of damaged cells under very low-dose-rate conditions. In this model, a constant number of cells were introduced into a cell pool, and the numbers of intact and damaged cells were calculated via transition and turnover events. A transition event emulates radiation dose, whereby an intact cell is changed into a damaged cell with a given probability. On the other hand, a turnover event expresses cell competition, where reproduction and elimination of cells occur depending on the properties of cells. Under very low-dose-rate conditions, this model showed that radiation damage to the stem cell pool was strongly suppressed when the damaged cells were less reproductive and tended to be eliminated compared to the intact cells. Furthermore, the size of the stem cell pool was positively correlated with reduction in radiation damage.


Computer Simulation , Models, Biological , Stem Cells/radiation effects , Cell Division/radiation effects , Dose-Response Relationship, Radiation
6.
J Cell Mol Med ; 24(17): 10027-10041, 2020 09.
Article En | MEDLINE | ID: mdl-32790210

Both SIRT1 and UVA radiation are involved in cellular damage processes such as apoptosis, senescence and ageing. MicroRNAs (miRNAs) have been reported to be closely related to UV radiation, as well as to SIRT1. In this study, we investigated the connections among SIRT1, UVA and miRNA in human skin primary fibroblasts. Our results showed that UVA altered the protein level of SIRT1 in a time point-dependent manner. Using miRNA microarray, bioinformatics analysis, we found that knocking down SIRT1 could cause up-regulation of miR-27a-5p and the latter could down-regulate SMAD2, and these results were verified by qRT-PCR or Western blot. Furthermore, UVA radiation (5 J/cm2 ), knocking down SIRT1 or overexpression of miR-27a-5p led to increased expression of MMP1, and decreased expressions of COL1 and BCL2. We also found additive impacts on MMP1, COL1 and BCL2 under the combination of UVA radiation + Sirtinol (SIRT1 inhibitor), or UVA radiation + miR-27a-5p mimic. SIRT1 activator resveratrol could reverse damage changes caused by UVA radiation. Besides, absent of SIRT1 or overexpression of miR-27a-5p increased cell apoptosis and induced cell arrest in G2/M phase. Taken together, these results demonstrated that UVA could influence a novel SIRT1-miR-27a-5p-SMAD2-MMP1/COL1/BCL2 axis in skin primary fibroblasts, and may provide potential therapeutic targets for UVA-induced skin damage.


Fibroblasts/metabolism , Fibroblasts/radiation effects , Proteins/metabolism , Signal Transduction/radiation effects , Skin/metabolism , Skin/radiation effects , Ultraviolet Rays/adverse effects , Adolescent , Adult , Apoptosis/radiation effects , Cell Cycle Checkpoints/radiation effects , Cell Division/radiation effects , Cells, Cultured , Down-Regulation/radiation effects , G2 Phase/radiation effects , Humans , Up-Regulation/radiation effects , Young Adult
7.
Nat Commun ; 11(1): 2262, 2020 05 08.
Article En | MEDLINE | ID: mdl-32385264

Cell division can perturb the metabolic performance of industrial microbes. The C period of cell division starts from the initiation to the termination of DNA replication, whereas the D period is the bacterial division process. Here, we first shorten the C and D periods of E. coli by controlling the expression of the ribonucleotide reductase NrdAB and division proteins FtsZA through blue light and near-infrared light activation, respectively. It increases the specific surface area to 3.7 µm-1 and acetoin titer to 67.2 g·L-1. Next, we prolong the C and D periods of E. coli by regulating the expression of the ribonucleotide reductase NrdA and division protein inhibitor SulA through blue light activation-repression and near-infrared (NIR) light activation, respectively. It improves the cell volume to 52.6 µm3 and poly(lactate-co-3-hydroxybutyrate) titer to 14.31 g·L-1. Thus, the optogenetic-based cell division regulation strategy can improve the efficiency of microbial cell factories.


Cell Division/radiation effects , Escherichia coli/cytology , Escherichia coli/radiation effects , Light , Acetoin/metabolism , Bioreactors/microbiology , Escherichia coli/genetics , Escherichia coli/ultrastructure , Genes, Bacterial , Polyesters/metabolism
8.
Exp Cell Res ; 389(1): 111887, 2020 04 01.
Article En | MEDLINE | ID: mdl-32027865

The cell fusion is a widespread process, which takes place in many systems in vivo and in vitro. Fusion of cells is frequently related to tetraploidy, which can be found within natural physiological conditions, e.g., placentation, and in pathophysiological conditions, such as cancer and early pregnancy failure in humans. Here we investigate the mechanism of tetraploidization with help of femtosecond laser-induced mouse blastomere fusion by the means of Hoechst staining, GFP, BODIPY dyes and fluorescent species generated intracellularly by a femtosecond laser. We establish diffusive mixing of cytosol, whereas the large components of a cytoplasm (organelles, cytoskeleton) are poorly diffusible and are not completely mixed after cell fusion and a subsequent division. We show that mechanisms which are responsible for the formation of a common metaphase plate triggered tetraploidization in fused mouse embryos and could be a significant factor in polyploidy formation in vivo. Thus, our results suggest that microtubules play a critical role in tetraploidization.


Blastomeres/physiology , Blastomeres/radiation effects , Lasers , Tetraploidy , Animals , Blastomeres/cytology , Cell Division/radiation effects , Cell Fusion/methods , Embryo, Mammalian/cytology , Embryo, Mammalian/radiation effects , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Metaphase/physiology , Metaphase/radiation effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
9.
Nucleic Acids Res ; 48(6): e33, 2020 04 06.
Article En | MEDLINE | ID: mdl-31989175

Light-regulated modules offer unprecedented new ways to control cellular behaviour with precise spatial and temporal resolution. Among a variety of bacterial light-switchable gene expression systems, single-component systems consisting of single transcription factors would be more useful due to the advantages of speed, simplicity, and versatility. In the present study, we developed a single-component light-activated bacterial gene expression system (eLightOn) based on a novel LOV domain from Rhodobacter sphaeroides (RsLOV). The eLightOn system showed significant improvements over the existing single-component bacterial light-activated expression systems, with benefits including a high ON/OFF ratio of >500-fold, a high activation level, fast activation kinetics, and/or good adaptability. Additionally, the induction characteristics, including regulatory windows, activation kinetics and light sensitivities, were highly tunable by altering the expression level of LexRO. We demonstrated the usefulness of the eLightOn system in regulating cell division and swimming by controlling the expression of the FtsZ and CheZ genes, respectively, as well as constructing synthetic Boolean logic gates using light and arabinose as the two inputs. Taken together, our data indicate that the eLightOn system is a robust and highly tunable tool for quantitative and spatiotemporal control of bacterial gene expression.


Gene Expression Regulation, Bacterial/radiation effects , Light , Rhodobacter sphaeroides/cytology , Rhodobacter sphaeroides/radiation effects , Bacterial Proteins/metabolism , Cell Division/radiation effects , Kinetics , Logic , Transcription Factors/metabolism
10.
J Bacteriol ; 202(2)2020 01 02.
Article En | MEDLINE | ID: mdl-31548275

All organisms regulate cell cycle progression by coordinating cell division with DNA replication status. In eukaryotes, DNA damage or problems with replication fork progression induce the DNA damage response (DDR), causing cyclin-dependent kinases to remain active, preventing further cell cycle progression until replication and repair are complete. In bacteria, cell division is coordinated with chromosome segregation, preventing cell division ring formation over the nucleoid in a process termed nucleoid occlusion. In addition to nucleoid occlusion, bacteria induce the SOS response after replication forks encounter DNA damage or impediments that slow or block their progression. During SOS induction, Escherichia coli expresses a cytoplasmic protein, SulA, that inhibits cell division by directly binding FtsZ. After the SOS response is turned off, SulA is degraded by Lon protease, allowing for cell division to resume. Recently, it has become clear that SulA is restricted to bacteria closely related to E. coli and that most bacteria enforce the DNA damage checkpoint by expressing a small integral membrane protein. Resumption of cell division is then mediated by membrane-bound proteases that cleave the cell division inhibitor. Further, many bacterial cells have mechanisms to inhibit cell division that are regulated independently from the canonical LexA-mediated SOS response. In this review, we discuss several pathways used by bacteria to prevent cell division from occurring when genome instability is detected or before the chromosome has been fully replicated and segregated.


Cell Division/drug effects , Cell Division/radiation effects , DNA Replication/drug effects , DNA Replication/radiation effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bleomycin/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Division/genetics , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Replication/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/radiation effects , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Protease La/genetics , Protease La/metabolism , Radiation, Ionizing , SOS Response, Genetics/drug effects , SOS Response, Genetics/genetics , SOS Response, Genetics/radiation effects
11.
Microb Ecol ; 79(1): 64-72, 2020 Jan.
Article En | MEDLINE | ID: mdl-31147731

Temporal programs synchronised with the daily cycle are of adaptive importance for organisms exposed to periodic fluctuations. This study deepens into several aspects of the exogenous and endogenous nature of microbial grazers. We investigated the diel rhythms of cell division and feeding activity of four marine protists under different light regimes. In particular, we tested if the feeding cycle of protistan grazers could be mediated by a light-aided enhancement of prey digestion, and also explored the consequences of cell division on diel feeding rhythms. Cell division occurred at night for the heterotrophic dinoflagellates Gyrodinium dominans and Oxyrrhis marina. In contrast, the mixotrophic dinoflagellate Karlodinium armiger and the ciliate Strombidium sp. mostly divided during the day. Additionally, a significant diurnal feeding rhythm was observed in all species. When exposed to continuous darkness, nearly all species maintained the cell division rhythm, but lost the feeding cycle within several hours/days (with the exception of O. marina that kept the rhythm for 9.5 days). Additional feeding experiments under continuous light also showed the same pattern. We conclude that the feeding rhythms of protistan grazers are generally regulated not by cell division nor by the enhancement of digestion by light. Our study, moreover, indicates that the cell division cycle is under endogenous control, whereas an external trigger is required to maintain the feeding rhythm, at least for most of the species studied here.


Ciliophora/physiology , Dinoflagellida/physiology , Cell Division/radiation effects , Ciliophora/radiation effects , Dinoflagellida/radiation effects , Heterotrophic Processes , Light
12.
Int J Radiat Biol ; 96(2): 172-178, 2020 02.
Article En | MEDLINE | ID: mdl-31633435

Purpose: Accumulated damage in neural stem cells (NSCs) during brain tumor radiotherapy causes cognitive dysfunction to the patients. Carbon-ion radiotherapy can reduce undesired irradiation of normal tissues more efficiently than conventional photon radiotherapy. This study elucidates the responses of NSCs to carbon-ion radiation.Methods: Human NSCs and glioblastoma A-172 cells were irradiated with carbon-ion radiation and γ-rays, which have different linear-energy-transfer (LET) values of 108 and 0.2 keV/µm, respectively. After irradiation, growth rates were measured, apoptotic cells were detected by flow cytometry, and DNA synthesizing cells were immunocytochemically visualized.Results: Growth rates of NSCs and A-172 cells were decreased after irradiation. The percentages of apoptotic cells were remarkably increased in NSCs but not in A-172 cells. In contrast, the fractions of DNA synthesizing A-172 cells were decreased in a dose-dependent manner. These results indicate that apoptosis induction and DNA synthesis inhibition contribute to the growth inhibition of NSCs and glioblastoma cells, respectively. In addition, high-LET carbon ions induced more profound effects than low-LET γ-rays.Conclusions: Apoptosis is an important clinical target to protect NSCs during brain tumor radiotherapy using carbon-ion radiation as well as conventional X-rays.


Apoptosis/radiation effects , Brain Neoplasms/radiotherapy , Gamma Rays , Glioblastoma/radiotherapy , Heavy Ion Radiotherapy/methods , Neural Stem Cells/radiation effects , Biomarkers/metabolism , Carbon , Cell Division/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , DNA/radiation effects , DNA Damage , Dose-Response Relationship, Radiation , Humans , Immunohistochemistry , Ions , Linear Energy Transfer , Nestin/metabolism , Photons , SOXB1 Transcription Factors/metabolism
13.
Sci Rep ; 9(1): 17604, 2019 11 26.
Article En | MEDLINE | ID: mdl-31772211

The factors and signals driving T cell activation and polarisation during immune responses have been studied mainly at the level of cells and chemical mediators. Here we describe a physical driver of these processes in the form of physiological-strength electric fields (EFs). EFs are generated at sites where epithelium is disrupted (e.g. wounded skin/bronchial epithelia) and where T cells frequently are present. Using live-cell imaging, we show human primary T cells migrate directionally to the cathode in low strength (50/150 mV/mm) EFs. Strikingly, we show for the first time that EFs significantly downregulate T cell activation following stimulation with antigen-activated APCs or anti-CD3/CD28 antibodies, as demonstrated by decreased IL-2 secretion and proliferation. These EF-induced functional changes were accompanied by a significant dampening of CD4+ T cell polarisation. Expression of critical markers of the Th17 lineage, RORγt and IL-17, and the Th17 polarisation mediator phospho-STAT3 were reduced significantly, while STAT1, ERK and c-Jun phosphorylation were comparatively unaffected suggesting STAT3 modulation by EFs as one mechanism driving effects. Overall, we identify electrical signals as important contributors to the co-ordination and regulation of human T cell functions, paving the way for a new research area into effects of naturally occurring and clinically-applied EFs in conditions where control of T cell activity is paramount.


Electromagnetic Fields , Lymphocyte Activation/radiation effects , T-Lymphocyte Subsets/radiation effects , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/radiation effects , Cell Division/radiation effects , Cell Movement , Cell Polarity/radiation effects , Cells, Cultured , Cytokines/biosynthesis , Electrodes , Endotoxins/pharmacology , Humans , Interleukin-2/biosynthesis , Lymphocyte Activation/drug effects , Phosphorylation , Protein Processing, Post-Translational , STAT3 Transcription Factor/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Th17 Cells/radiation effects
14.
Plant Signal Behav ; 14(12): 1675471, 2019.
Article En | MEDLINE | ID: mdl-31595819

Studies on UV-B-induced plant photomorphogenesis mainly focus on Arabidopsis shoots (hypocotyl, leaf, petiole, and stem) but less on roots. In the present research, the low-level UV-B (0.2 W·m-2) induced a decrease in the number of root cells in the meristem zone and an inhibition of the cell length in the maturation zone of roots in Arabidopsis thaliana L.Heynh (Col-0). UV-B-induced root growth inhibition was recovered by the addition of GA3 to culture media. GA3 played an important role in UV-B-induced inhibition of root growth. The cop1-4 mutant with more meristem cell and longer mature cells exhibited longer root length under low-level UV-B. COP1 acted as a positive regulator of root growth under UV-B, through regulation of cell division and elongation. The sto mutant exhibited a shorter root length under UV-B with similar cell length but fewer meristem cells compared with wild type (Col-0). STO only regulated cell division, but cell expansion was not affected. UV-B radiation also inhibited the root growth of uvr8 mutant, and the degree of inhibition was greater than for wild type (Ler). UV-B inhibited the growth of Arabidopsis root, possibly because it changes the GA signal and inhibited cell division and cell elongation, which be related to COP1 and STO genes.


Arabidopsis Proteins/metabolism , Arabidopsis/growth & development , Arabidopsis/radiation effects , Gibberellins/metabolism , Plant Roots/growth & development , Plant Roots/radiation effects , Ultraviolet Rays , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Cell Division/drug effects , Cell Division/radiation effects , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Plant/drug effects , Gene Expression Regulation, Plant/radiation effects , Plant Growth Regulators/pharmacology , Plant Roots/cytology , Plant Roots/drug effects , Seedlings/drug effects , Seedlings/growth & development , Seedlings/radiation effects , Ubiquitin-Protein Ligases/metabolism
15.
Sci Rep ; 9(1): 10568, 2019 07 22.
Article En | MEDLINE | ID: mdl-31332255

Charged-particle microbeams (CPMs) provide a unique opportunity to investigate the effects of ionizing radiation on living biological specimens with a precise control of the delivered dose, i.e. the number of particles per cell. We describe a methodology to manipulate and micro-irradiate early stage C. elegans embryos at a specific phase of the cell division and with a controlled dose using a CPM. To validate this approach, we observe the radiation-induced damage, such as reduced cell mobility, incomplete cell division and the appearance of chromatin bridges during embryo development, in different strains expressing GFP-tagged proteins in situ after irradiation. In addition, as the dosimetry of such experiments cannot be extrapolated from random irradiations of cell populations, realistic three-dimensional models of 2 cell-stage embryo were imported into the Geant4 Monte-Carlo simulation toolkit. Using this method, we investigate the energy deposit in various chromatin condensation states during the cell division phases. The experimental approach coupled to Monte-Carlo simulations provides a way to selectively irradiate a single cell in a rapidly dividing multicellular model with a reproducible dose. This method opens the way to dose-effect investigations following targeted irradiation.


Caenorhabditis elegans/radiation effects , Embryo, Nonmammalian/radiation effects , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/ultrastructure , Cell Division/radiation effects , Chromatin/radiation effects , Chromosomes/radiation effects , Embryo, Nonmammalian/ultrastructure , Embryonic Development/radiation effects , Microscopy, Confocal/methods , Monte Carlo Method , Radiometry
16.
JCI Insight ; 4(13)2019 07 11.
Article En | MEDLINE | ID: mdl-31292293

Bone provides supportive microenvironments for hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) and is a frequent site of metastasis. While incidences of bone metastases increase with age, the properties of the bone marrow microenvironment that regulate dormancy and reactivation of disseminated tumor cells (DTCs) remain poorly understood. Here, we elucidate the age-associated changes in the bone secretome that trigger proliferation of HSCs, MSCs, and DTCs in the aging bone marrow microenvironment. Remarkably, a bone-specific mechanism involving expansion of pericytes and induction of quiescence-promoting secretome rendered this proliferative microenvironment resistant to radiation and chemotherapy. This bone-specific expansion of pericytes was triggered by an increase in PDGF signaling via remodeling of specialized type H blood vessels in response to therapy. The decline in bone marrow pericytes upon aging provides an explanation for loss of quiescence and expansion of cancer cells in the aged bone marrow microenvironment. Manipulation of blood flow - specifically, reduced blood flow - inhibited pericyte expansion, regulated endothelial PDGF-B expression, and rendered bone metastatic cancer cells susceptible to radiation and chemotherapy. Thus, our study provides a framework to recognize bone marrow vascular niches in age-associated increases in metastasis and to target angiocrine signals in therapeutic strategies to manage bone metastasis.


Aging/pathology , Bone Marrow/pathology , Bone Neoplasms/therapy , Tumor Microenvironment/physiology , Adrenergic alpha-1 Receptor Antagonists/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Bone Marrow/blood supply , Bone Marrow/drug effects , Bone Marrow/radiation effects , Bone Neoplasms/blood supply , Bone Neoplasms/secondary , Cell Division/drug effects , Cell Division/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Drug Resistance, Neoplasm/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/radiation effects , Mice , Pericytes/drug effects , Pericytes/pathology , Pericytes/radiation effects , Prazosin/administration & dosage , Radiation Tolerance/physiology , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Whole-Body Irradiation , Xenograft Model Antitumor Assays
17.
Sci Rep ; 9(1): 8255, 2019 06 04.
Article En | MEDLINE | ID: mdl-31164689

Using data generated with cells exposed to ionizing-radiation (IR) in G2-phase of the cell cycle, we describe dose-dependent interactions between ATM, ATR and DNA-PKcs revealing unknown mechanistic underpinnings for two key facets of the DNA damage response: DSB end-resection and G2-checkpoint activation. At low IR-doses that induce low DSB-numbers in the genome, ATM and ATR regulate epistatically the G2-checkpoint, with ATR at the output-node, interfacing with the cell-cycle predominantly through Chk1. Strikingly, at low IR-doses, ATM and ATR epistatically regulate also resection, and inhibition of either activity fully suppresses resection. At high IR-doses that induce high DSB-numbers in the genome, the tight ATM/ATR coupling relaxes and independent outputs to G2-checkpoint and resection occur. Consequently, both kinases must be inhibited to fully suppress checkpoint activation and resection. DNA-PKcs integrates to the ATM/ATR module by regulating resection at all IR-doses, with defects in DNA-PKcs causing hyper-resection and G2-checkpoint hyper-activation. Notably, hyper-resection is absent from other c-NHEJ mutants. Thus, DNA-PKcs specifically regulates resection and adjusts the activation of the ATM/ATR module. We propose that selected DSBs are shepherd by DNA-PKcs from c-NHEJ to resection-dependent pathways for processing under the regulatory supervision of the ATM/ATR module.


Ataxia Telangiectasia Mutated Proteins/genetics , DNA-Activated Protein Kinase/genetics , G2 Phase Cell Cycle Checkpoints/radiation effects , Radiation, Ionizing , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Division/genetics , Cell Division/radiation effects , Checkpoint Kinase 1/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/genetics , DNA Damage/radiation effects , DNA End-Joining Repair/radiation effects , DNA-Binding Proteins/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Phosphorylation/radiation effects , Signal Transduction/radiation effects
18.
FASEB J ; 33(9): 10165-10176, 2019 09.
Article En | MEDLINE | ID: mdl-31207192

Gastrointestinal toxicity limits the clinical application of abdominal and pelvic radiotherapy and currently has no effective treatment. Intestinal leucine-rich-repeat-containing GPCR 5 (Lgr5)-positive stem cell depletion and loss of proliferative ability due to radiation may be the primary factors causing intestinal injury following radiation. Here, we report the critical role of ß-arrestin1 (ßarr1) in radiation-induced intestinal injury. Intestinal ßarr1 was highly expressed in radiation enteritis and in a radiation model. ßarr1 knockout (KO) or knockdown mice exhibited increased proliferation in intestinal Lgr5+ stem cell, crypt reproduction, and survival following radiation. Unexpectedly, the beneficial effects of ßarr1 deficiency on intestinal stem cells in response to radiation were compromised when the endoplasmic reticulum stress-related protein kinase RNA-like ER kinase (PERK)/eukaryotic initiation factor-2α (eIF2α) pathway was inhibited, and this result was further supported in vitro. Furthermore, we found that ßarr1 knockdown with small interfering RNA significantly enhanced intestinal Lgr5+ stem cell proliferation after radiation via directly targeting PERK. ßarr1 offers a promising target for mitigating radiation-induced intestinal injury.-Liu, Z., Jiang, J., He, Q., Liu, Z., Yang, Z., Xu, J., Huang, Z., Wu, B. ß-Arrestin1-mediated decrease in endoplasmic reticulum stress impairs intestinal stem cell proliferation following radiation.


Endoplasmic Reticulum Stress/physiology , Enteritis/pathology , Intestines/radiation effects , Radiation Injuries, Experimental/pathology , Radiation Injuries/pathology , Stem Cells/radiation effects , beta-Arrestin 1/physiology , eIF-2 Kinase/physiology , Aged , Animals , Cell Division/radiation effects , Colony-Forming Units Assay , Enteritis/etiology , Enteritis/physiopathology , Eukaryotic Initiation Factor-2/physiology , Female , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Radiation Chimera , Radiation Injuries/physiopathology , Radiation Injuries, Experimental/physiopathology , Radiotherapy/adverse effects , Receptors, G-Protein-Coupled/analysis , Regeneration , Signal Transduction/physiology , Stem Cells/pathology , beta-Arrestin 1/deficiency , beta-Arrestin 1/genetics
19.
Mol Cell ; 75(1): 117-130.e6, 2019 07 11.
Article En | MEDLINE | ID: mdl-31101499

Telomeres are essential for genome stability. Oxidative stress caused by excess reactive oxygen species (ROS) accelerates telomere shortening. Although telomeres are hypersensitive to ROS-mediated 8-oxoguanine (8-oxoG) formation, the biological effect of this common lesion at telomeres is poorly understood because ROS have pleiotropic effects. Here we developed a chemoptogenetic tool that selectively produces 8-oxoG only at telomeres. Acute telomeric 8-oxoG formation increased telomere fragility in cells lacking OGG1, the enzyme that removes 8-oxoG, but did not compromise cell survival. However, chronic telomeric 8-oxoG induction over time shortens telomeres and impairs cell growth. Accumulation of telomeric 8-oxoG in chronically exposed OGG1-deficient cells triggers replication stress, as evidenced by mitotic DNA synthesis at telomeres, and significantly increases telomere losses. These losses generate chromosome fusions, leading to chromatin bridges and micronucleus formation upon cell division. By confining base damage to the telomeres, we show that telomeric 8-oxoG accumulation directly drives telomere crisis.


Chromosome Aberrations/radiation effects , DNA Glycosylases/genetics , DNA Repair/radiation effects , Genomic Instability/radiation effects , Guanine/analogs & derivatives , Telomere/radiation effects , Cell Division/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , DNA Damage , DNA Glycosylases/deficiency , DNA Replication/radiation effects , Gene Expression , Guanine/agonists , Guanine/biosynthesis , HeLa Cells , Humans , Light/adverse effects , Micronuclei, Chromosome-Defective/radiation effects , Optogenetics , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/radiation effects , Oxidative Stress/radiation effects , Singlet Oxygen/agonists , Singlet Oxygen/metabolism , Telomere/metabolism , Telomere Homeostasis/radiation effects
20.
Neuroreport ; 30(8): 538-543, 2019 05 22.
Article En | MEDLINE | ID: mdl-30950935

This study assessed the effects of combined low-dose neutron and γ-ray irradiation on hippocampal neurogenesis and hippocampal-dependent memory. Neural progenitor cell division and survival were evaluated in brain sections and whole hippocampal preparations following head irradiation at a dose of 0.34 Gy for neutron radiation and 0.36 Gy for γ-ray radiation. Hippocampal-dependent memory formation was tested in a contextual fear conditioning task following irradiation at doses of 0.4 Gy for neutron radiation and 0.42 Gy for γ-ray radiation. Cell division was suppressed consistently along the entire dorsoventral axis of the hippocampus 24 h after the irradiation, but quiescent stem cells remained unaffected. The control and irradiated mice showed no differences in terms of exploratory behavior or anxiety 6 weeks after the irradiation. The ability to form hippocampus-dependent memory was also unaffected. The data may be indicative of a negligible effect of the low-dose of fast neutron irradiation and the neurogenesis suppression on animal behavior at 6 weeks after irradiation.


Conditioning, Classical/radiation effects , Electromagnetic Radiation , Hippocampus/radiation effects , Neurogenesis/radiation effects , Animals , Cell Division/radiation effects , Male , Mice, Inbred C57BL , Neural Stem Cells/radiation effects
...