Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article in English | MEDLINE | ID: mdl-33495326

ABSTRACT

Adaptation to different forms of environmental stress is crucial for maintaining essential cellular functions and survival. The nucleolus plays a decisive role as a signaling hub for coordinating cellular responses to various extrinsic and intrinsic cues. p53 levels are normally kept low in unstressed cells, mainly due to E3 ubiquitin ligase MDM2-mediated degradation. Under stress, nucleophosmin (NPM) relocates from the nucleolus to the nucleoplasm and binds MDM2, thereby preventing degradation of p53 and allowing cell-cycle arrest and DNA repair. Here, we demonstrate that the mammalian sirtuin SIRT7 is an essential component for the regulation of p53 stability during stress responses induced by ultraviolet (UV) irradiation. The catalytic activity of SIRT7 is substantially increased upon UV irradiation through ataxia telangiectasia mutated and Rad3 related (ATR)-mediated phosphorylation, which promotes efficient deacetylation of the SIRT7 target NPM. Deacetylation is required for stress-dependent relocation of NPM into the nucleoplasm and MDM2 binding, thereby preventing ubiquitination and degradation of p53. In the absence of SIRT7, stress-dependent stabilization of p53 is abrogated, both in vitro and in vivo, impairing cellular stress responses. The study uncovers an essential SIRT7-dependent mechanism for stabilization of the tumor suppressor p53 in response to genotoxic stress.


Subject(s)
DNA Damage , Nuclear Proteins/metabolism , Sirtuins/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Acetylation/radiation effects , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Catalysis/radiation effects , Cell Line, Tumor , Cell Nucleolus/metabolism , Cell Nucleolus/radiation effects , Humans , Lysine/metabolism , Mice , Mice, Inbred C57BL , Nucleophosmin , Phosphorylation/radiation effects , Protein Stability/radiation effects , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription, Genetic/radiation effects , Ubiquitination/radiation effects
2.
Nucleic Acids Res ; 48(18): 10342-10352, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32894284

ABSTRACT

Ribosomal DNA (rDNA) consists of highly repeated sequences that are prone to incurring damage. Delays or failure of rDNA double-strand break (DSB) repair are deleterious, and can lead to rDNA transcriptional arrest, chromosomal translocations, genomic losses, and cell death. Here, we show that the zinc-finger transcription factor GLI1, a terminal effector of the Hedgehog (Hh) pathway, is required for the repair of rDNA DSBs. We found that GLI1 is activated in triple-negative breast cancer cells in response to ionizing radiation (IR) and localizes to rDNA sequences in response to both global DSBs generated by IR and site-specific DSBs in rDNA. Inhibiting GLI1 interferes with rDNA DSB repair and impacts RNA polymerase I activity and cell viability. Our findings tie Hh signaling to rDNA repair and this heretofore unknown function may be critically important in proliferating cancer cells.


Subject(s)
DNA, Ribosomal/genetics , Hedgehog Proteins/genetics , RNA Polymerase I/genetics , Triple Negative Breast Neoplasms/radiotherapy , Zinc Finger Protein GLI1/genetics , Cell Cycle Proteins/genetics , Cell Nucleolus/genetics , Cell Nucleolus/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA, Ribosomal/radiation effects , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Humans , RNA Polymerase I/radiation effects , Radiation, Ionizing , Ribosomes/genetics , Ribosomes/radiation effects , Signal Transduction/radiation effects , Transcription, Genetic/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
3.
Oncol Rep ; 42(6): 2293-2302, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31578593

ABSTRACT

Ribosomes are important cellular components that maintain cellular homeostasis through overall protein synthesis. The nucleolus is a prominent subnuclear structure that contains ribosomal DNA (rDNA) encoding ribosomal RNA (rRNA), an essential component of ribosomes. Despite the significant role of the rDNA­rRNA­ribosome axis in cellular homeostasis, the stability of rDNA in the context of the DNA damage response has not been fully investigated. In the present study, the number and morphological changes of nucleolin, a marker of the nucleolus, were examined following ionizing radiation (IR) in order to investigate the impact of DNA damage on nucleolar stability. An increase in the number of nucleoli per cell was found in HCT116 and U2OS cells following IR. Interestingly, the IR­dependent increase in nucleolar fragmentation was enhanced by p53 deficiency. In addition, the morphological analysis revealed several distinct types of nucleolar fragmentation following IR. The pattern of nucleolar morphology differed between HCT116 and U2OS cells, and the p53 deficiency altered the pattern of nucleolar morphology. Finally, a significant decrease in rRNA synthesis was observed in HCT116 p53­/­ cells following IR, suggesting that severe nucleolar fragmentation downregulates rRNA transcription. The findings of the present study suggest that p53 plays a key role in protecting the transcriptional activity of rDNA in response to DNA damage.


Subject(s)
Bone Neoplasms/genetics , Cell Nucleolus/metabolism , Colorectal Neoplasms/genetics , Osteosarcoma/genetics , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Tumor Suppressor Protein p53/deficiency , Apoptosis , Bone Neoplasms/pathology , Cell Nucleolus/genetics , Cell Nucleolus/radiation effects , Colorectal Neoplasms/pathology , DNA Damage , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Humans , Osteosarcoma/pathology , Phosphoproteins/genetics , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA-Binding Proteins/genetics , Radiation, Ionizing , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Nucleolin
4.
Nucleic Acids Res ; 46(19): 10119-10131, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30165463

ABSTRACT

The RING finger protein TRAIP protects genome integrity and its mutation causes Seckel syndrome. TRAIP encodes a nucleolar protein that migrates to UV-induced DNA lesions via a direct interaction with the DNA replication clamp PCNA. Thus far, mechanistically how UV mobilizes TRAIP from the nucleoli remains unknown. We found that PCNA binding is dispensable for the nucleolus-nucleoplasm shuttling of TRAIP following cell exposure to UV irradiation, and that its redistribution did not rely on the master DNA damage kinases ATM and ATR. Interestingly, I-PpoI-induced ribosomal DNA damage led to TRAIP exclusion from the nucleoli, raising the possibility that active ribosomal DNA transcription may underlie TRAIP retention in the nuclear sub-compartments. Accordingly, chemical inhibition of RNA polymerase I activity led to TRAIP diffusion into the nucleoplasm, and was coupled with marked reduction of DNA/RNA hybrids in the nucleoli, suggesting that TRAIP may be sequestered via binding to nucleic acid structures in the nucleoli. Consistently, cell pre-treatment with DNase/RNase effectively released TRAIP from the nucleoli. Taken together, our study defines a bipartite mechanism that drives TRAIP trafficking in response to UV damage, and highlights the nucleolus as a stress sensor that contributes to orchestrating DNA damage responses.


Subject(s)
Cell Nucleolus/metabolism , DNA, Ribosomal/genetics , RNA Polymerase I/genetics , Transcription, Genetic , Ubiquitin-Protein Ligases/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Nucleolus/radiation effects , Cell Nucleolus/ultrastructure , DNA Damage , DNA, Ribosomal/metabolism , Deoxyribonucleases/chemistry , Dwarfism/genetics , Dwarfism/metabolism , Dwarfism/pathology , Facies , Gene Expression Regulation , HeLa Cells , Humans , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Naphthyridines/pharmacology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoblasts/radiation effects , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Transport , RNA Polymerase I/antagonists & inhibitors , RNA Polymerase I/metabolism , Ribonucleases/chemistry , Ribosomes/genetics , Ribosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ultraviolet Rays
5.
J Neurochem ; 141(5): 750-765, 2017 06.
Article in English | MEDLINE | ID: mdl-28345128

ABSTRACT

The retina is highly sensitive to oxidative stress because of its high consumption of oxygen associated with the phototransductional processes. Recent findings have suggested that oxidative stress is involved in the pathology of age-related macular degeneration, a progressive degeneration of the central retina. A well-known environmental risk factor is light exposure, as excessive and continuous light exposure can damage photoreceptors. Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a transcriptional factor that controls antioxidative responses and phase 2 enzymes. Thus, we hypothesized that RS9, a specific activator of Nrf2, decreases light-induced retinal cell death in vivo and in vitro. Nrf2 was detected in the nucleus of the 661W cells exposed to RS9 and also after light exposure, and the Nrf2-antioxidant response element binding was increased in 661W cells after exposure to RS9. Consequentially, the expression of the phase 2 enzyme's mRNAs of Ho-1, Nqo-1, and Gclm genes was increased in 661W cells after exposure to RS9. Furthermore, RS9 decreased the light-induced death of 661W cells (2500 lux, 24 h), and also reduced the functional damages and the histological degeneration of the nuclei in the outer nuclear layer or the retina in the in vivo studies (8000 lux, 3 h). Heme oxygenase-1 was increased after light exposure, and Nrf2 was translocated into the nucleus after light exposure in vivo. Silencing of Ho-1 reduced the protective effects of RS9 against light-induced death of 661W cells. These findings indicate that RS9 has therapeutic potential for retinal diseases that are aggravated by light exposure.


Subject(s)
Cell Death/drug effects , Ependymoglial Cells/drug effects , Light/adverse effects , Photoreceptor Cells/drug effects , Triterpenes/pharmacology , Animals , Cell Death/radiation effects , Cell Line, Transformed , Cell Nucleolus/drug effects , Cell Nucleolus/radiation effects , Cytosol/drug effects , Cytosol/radiation effects , Ependymoglial Cells/cytology , Ependymoglial Cells/radiation effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , In Vitro Techniques , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NF-E2 Transcription Factor/genetics , NF-E2 Transcription Factor/metabolism , Photoreceptor Cells/radiation effects , Protein Biosynthesis/drug effects , Protein Biosynthesis/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Retina/cytology , Retinal Degeneration/etiology , Retinal Degeneration/prevention & control , Time Factors , Triterpenes/chemistry
6.
Oncotarget ; 7(50): 83241-83257, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27829214

ABSTRACT

PICT-1 is an essential ribosome biogenesis factor whose loss induces p53 accumulation and apoptosis. Here, we show that DNA damage changes PICT-1 localization and decreases PICT-1 protein levels via the proteasome pathway. Two important phosphatidylinositol 3-kinase-like kinases (PIKKs), ataxia-telangiectasia mutated (ATM) and the Ku70 subunit of DNA-dependent protein kinase (DNA-PK), co-localize and interact with PICT-1 in the nucleolus. Computational prediction of phosphorylation sites and detection using an anti-phospho-substrate antibody suggest that PICT-1 might be a substrate of PIKKs. PICT-1 S233 and T289 were identified as the key phosphorylation sites in this pathway, as mutating both to alanine abolished UVB-induced increase of PICT-1 phosporylation. Inhibition of PIKKs or ATM (with wortmannin and KU55933, respectively) prevented the agglomeration and degradation of PICT-1, suggesting that ATM is a key regulator of PICT-1. PICT-1(S233A, T289A) demonstrated marked resistance to DNA damage-induced agglomeration and loss of PICT-1. Phosphomimetic PICT-1 (S233D, T289D) showed a different nuclear distribution and was more rapidly degraded after DNA damage than wild-type PICT-1. Furthermore, both phosphorylation and degradation of PICT-1 released RPL11 from the nucleolus to the nucleoplasm, increased binding of RPL11 to MDM2, and promoted p53 accumulation and apoptosis in an ATM-dependent manner after DNA damage. These data indicate that PICT-1 is a major nucleolar sensor of the DNA damage repair response and an important upstream regulator of p53 via the RPL11-MDM2-p53 pathway.


Subject(s)
Apoptosis , Cell Nucleolus/metabolism , DNA Damage , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Active Transport, Cell Nucleus , Apoptosis/drug effects , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins/metabolism , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Nucleolus/drug effects , Cell Nucleolus/pathology , Cell Nucleolus/radiation effects , DNA Repair , HEK293 Cells , Humans , Mitomycin/pharmacology , Multienzyme Complexes , Mutation , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Signal Transduction , Time Factors , Tumor Suppressor Proteins/genetics , Ultraviolet Rays
7.
Redox Biol ; 8: 333-40, 2016 08.
Article in English | MEDLINE | ID: mdl-26966892

ABSTRACT

Radioadaptive response (RAR) is an important phenomenon induced by low dose radiation. However, the molecular mechanism of RAR is obscure. In this study, we focused on the possible role of heme oxygenase 1 (HO-1) in RAR. Consistent with previous studies, priming dose of X-ray radiation (1-10cGy) induced significant RAR in normal human skin fibroblasts (AG 1522 cells). Transcription and translation of HO-1 was up-regulated more than two fold by a priming dose of radiation (5cGy). Zinc protoporphyrin Ⅸ, a specific competitive inhibitor of HO-1, efficiently inhibited RAR whereas hemin, an inducer of HO-1, could mimic priming dose of X-rays to induce RAR. Knocking down of HO-1 by transfection of HO-1 siRNA significantly attenuated RAR. Furthermore, the expression of HO-1 gene was modulated by the nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which translocated from cytoplasm to nucleus after priming dose radiation and enhance the antioxidant level of cells.


Subject(s)
Antioxidants/metabolism , Fibroblasts/metabolism , Heme Oxygenase-1/genetics , NF-E2-Related Factor 2/genetics , Cell Line , Cell Nucleolus/metabolism , Cell Nucleolus/radiation effects , Cytoplasm/radiation effects , Dose-Response Relationship, Radiation , Fibroblasts/radiation effects , Gene Expression Regulation/radiation effects , Heme Oxygenase-1/antagonists & inhibitors , Hemin/genetics , Hemin/metabolism , Humans , NF-E2-Related Factor 2/metabolism , Protoporphyrins/genetics , Protoporphyrins/metabolism , RNA, Small Interfering/genetics , X-Rays
8.
Nucleus ; 6(4): 301-13, 2015.
Article in English | MEDLINE | ID: mdl-26208041

ABSTRACT

The nucleolus is a well-organized site of ribosomal gene transcription. Moreover, many DNA repair pathway proteins, including ATM, ATR kinases, MRE11, PARP1 and Ku70/80, localize to the nucleolus (Moore et al., 2011 ). We analyzed the consequences of DNA damage in nucleoli following ultraviolet A (UVA), C (UVC), or γ-irradiation in order to test whether and how radiation-mediated genome injury affects local motion and morphology of nucleoli. Because exposure to radiation sources can induce changes in the pattern of UBF1-positive nucleolar regions, we visualized nucleoli in living cells by GFP-UBF1 expression for subsequent morphological analyses and local motion studies. UVA radiation, but not 5 Gy of γ-rays, induced apoptosis as analyzed by an advanced computational method. In non-apoptotic cells, we observed that γ-radiation caused nucleolar re-positioning over time and changed several morphological parameters, including the size of the nucleolus and the area of individual UBF1-positive foci. Radiation-induced nucleoli re-arrangement was observed particularly in G2 phase of the cell cycle, indicating repair of ribosomal genes in G2 phase and implying that nucleoli are less stable, thus sensitive to radiation, in G2 phase.


Subject(s)
Cell Cycle/radiation effects , G2 Phase/radiation effects , Gamma Rays/adverse effects , Animals , Apoptosis/radiation effects , Cell Line , Cell Line, Tumor , Cell Nucleolus/radiation effects , Computational Biology , DNA Damage/radiation effects , Mice , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , Transcription, Genetic , Ultraviolet Rays
9.
Folia Biol (Praha) ; 60 Suppl 1: 76-84, 2014.
Article in English | MEDLINE | ID: mdl-25369346

ABSTRACT

Every day, genomes are affected by genotoxic factors that create multiple DNA lesions. Several DNA repair systems have evolved to counteract the deleterious effects of DNA damage. These systems include a set of DNA repair mechanisms, damage tolerance processes, and activation of cell-cycle checkpoints. This study describes selected confocal microscopy techniques that investigate DNA damage-related nuclear events after UVA- and γ-irradiation and compare the DNA damage response (DDR) induced by the two experimental approaches. In both cases, we observed induction of the nucleotide excision repair (NER) pathway and formation of localized double-strand breaks (DSBs). This was confirmed by analysis of cyclobutane pyrimidine dimers (CPDs) in the DNA lesions and by increased levels of γH2AX and 53BP1 proteins in the irradiated genome. DNA damage by UVA-lasers was potentiated by either BrdU or Hoechst 33342 pre-sensitization and compared to non-photosensitized cells. DSBs were also induced without BrdU or Hoechst 33342 pre-treatment. Interestingly, no cyclobutane pyrimidine dimers (CPDs) were detected after 405 nm UVA laser micro-irradiation in non-photosensitized cells. The effects of UVA and γ-irradiation were also studied by silver staining of nucleolar organizer regions (AgNORs). This experimental approach revealed changes in the morphology of nucleoli after genome injury. Additionally, to precisely characterize DDR in locally induced DNA lesions, we analysed the kinetics of the 53BP1 protein involved in DDR by fluorescence recovery after photobleaching (FRAP).


Subject(s)
Cell Nucleolus/radiation effects , DNA Damage , Gamma Rays , Microscopy/methods , Ultraviolet Rays , Animals , Antigens, Nuclear , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Kinetics , Luminescent Proteins/metabolism , Mice , Pyrimidine Dimers/metabolism , Tumor Suppressor p53-Binding Protein 1 , Red Fluorescent Protein
10.
Radiats Biol Radioecol ; 53(1): 55-62, 2013.
Article in Russian | MEDLINE | ID: mdl-23700835

ABSTRACT

The effects of a single exposure of rats to the whole-body roentgen irradiation at the doses of 3.5 Gy and 4.5 Gy on the activity of creatine kinase, purine nucleoside phosphorylase, alanine aminotransferase, aspartate aminotransferase, as well as on the state of the nuclear-nucleolar apparatus in rat hepatocytes on the 6th and 13th days after radiation exposure have been studied. Irradiation at the above doses induced changes in the levels of enzymatic activity of different values and different directions within the same time periods, as well as oscillating changes in this type of enzymatic activity over time. This demonstrates various radiosensitivity and adaptation abilities of these enzymatic activities. The changes in the enzymatic activity significantly correspond to the changes in the morphometric indices of nuclear-nucleolar apparatus of hepatocytes, as well as the distribution of hepatocytes within the ploidy classes: in particular, stabilization of the enzymatic activity on the 13th day after irradiation correlates with the increased transcriptional activity, which is detectable through the increased number of nucleoli per nucleus and the expanded space of a hepatocyte nucleus. The compensation mechanisms are likely to be targeted at the changes in the functional activity of surviving hepatocytes, rather than at the replacement of the damaged cells by the new ones.


Subject(s)
Cell Nucleolus , Hepatocytes , Liver , Radiation, Ionizing , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Cell Nucleolus/enzymology , Cell Nucleolus/radiation effects , Creatine Kinase/metabolism , Hepatocytes/enzymology , Hepatocytes/radiation effects , Liver/enzymology , Liver/radiation effects , Male , Ploidies , Purine-Nucleoside Phosphorylase/metabolism , Rats , Whole-Body Irradiation
11.
Radiats Biol Radioecol ; 53(6): 549-55, 2013.
Article in Russian | MEDLINE | ID: mdl-25486735

ABSTRACT

In the current study the authors investigated the connection between the proliferation rate of stimulated hu- man peripheral blood lymphocytes with some other characteristics of the population: expression of immunological markers, spontaneous genome damage, radiosensitivity. The population's proliferation index (PI) was taken as a measure of the rate. It was calculated using the composition of a cell population, which was cyto- kinesis-blocked with a cytochalasin B. If the genotoxic action is absent, the PI does not correlate with the spontaneous frequency of cells with micronuclei or with cell radiosensitivity, but is tightly linked with immunological indexes. It has been determined that after stimulation the level of marker-positive cells (CD25, CD69 and Ki67) is closely related to PI and is greater in the populations with lower proliferation rates. Irradiation of a cell culture 48 h after stimulation at a dose of 1 Gy leads to a correlation between PI and radiosensitivity, measured directly after the irradiation and in the same time frame as the PI measured in the non-irradiated population. The irradiated population's PI is not connected with the level of marker expression.


Subject(s)
Cell Nucleolus/metabolism , Cell Proliferation/radiation effects , Genome, Human/radiation effects , Lymphocytes/radiation effects , Cell Cycle/radiation effects , Cell Nucleolus/radiation effects , Chromosome Aberrations/radiation effects , Cytogenetics , DNA Damage/radiation effects , Dose-Response Relationship, Radiation , Humans , Lymphocytes/cytology , Radiation Tolerance
12.
Mol Biol Cell ; 23(21): 4323-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22933572

ABSTRACT

The AP-1 transcription factor c-Jun has been shown to be essential for stress-induced apoptosis in several models. However, the molecular mechanisms underlying the proapoptotic activity of c-Jun are poorly understood. We identify the apoptosis-antagonizing transcription factor (AATF) as a novel nucleolar stress sensor, which is required as a cofactor for c-Jun-mediated apoptosis. Overexpression or down-regulation of AATF expression levels led to a respective increase or decrease in the amount of activated and phosphorylated c-Jun with a proportional alteration in the induction levels of the proapoptotic c-Jun target genes FasL and TNF-α. Accordingly, AATF promoted commitment of ultraviolet (UV)-irradiated cells to c-Jun-dependent apoptosis. Whereas AATF overexpression potentiated UV-induced apoptosis in wild-type cells, c-Jun-deficient mouse embryonic fibroblasts were resistant to AATF-mediated apoptosis induction. Furthermore, AATF mutants defective in c-Jun binding were also defective in inducing AP-1 activity and c-Jun-mediated apoptosis. UV irradiation induced a translocation of AATF from the nucleolus to the nucleus, thereby enabling its physical association to c-Jun. Analysis of AATF deletion mutants revealed that the AATF domains required for compartmentalization, c-Jun binding, and enhancement of c-Jun transcriptional activity were all also required to induce c-Jun-dependent apoptosis. These results identify AATF as a nucleolar-confined c-Jun cofactor whose expression levels and spatial distribution determine the stress-induced activity of c-Jun and the levels of c-Jun-mediated apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cell Nucleolus/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/chemistry , Cell Nucleolus/radiation effects , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , HEK293 Cells , Humans , Mice , Mice, Knockout , Nuclear Proteins/chemistry , Protein Binding/radiation effects , Protein Structure, Tertiary , Protein Transport/radiation effects , Repressor Proteins/chemistry , Ultraviolet Rays
13.
Cell Death Dis ; 3: e283, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22419112

ABSTRACT

Cellular senescence-inhibited gene (CSIG) protein, a nucleolar protein with a ribosomal L1 domain in its N-terminus, can exert non-ribosomal functions to regulate biological processes, such as cellular senescence. Here, we describe a previously unknown function for CSIG: promotion of apoptosis in response to ultraviolet (UV) irradiation-induced CSIG upregulation. We identified p33ING1 as a binding partner that interacts with CSIG. After UV irradiation, p33ING1 increases its protein expression, translocates into the nucleolus and binds CSIG. p33ING1 requires its nucleolar targeting sequence region to interact with CSIG and enhance CSIG protein stability, which is essential for activation of downstream effectors, Bcl-2-associated X protein, to promote apoptosis. Thus, our data imply that p33ING1-CSIG axis functions as a novel pro-apoptotic regulator in response to DNA damage.


Subject(s)
Apoptosis/radiation effects , Cell Nucleolus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Pregnancy Proteins/metabolism , Ribosomal Proteins/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism , Apoptosis/genetics , Binding Sites , Cell Nucleolus/genetics , Cell Nucleolus/radiation effects , DNA Damage/genetics , DNA Damage/radiation effects , Gene Expression/radiation effects , HEK293 Cells , HeLa Cells , Humans , Inhibitor of Growth Protein 1 , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Pregnancy Proteins/genetics , Protein Binding , Protein Transport/genetics , Protein Transport/radiation effects , Ribosomal Proteins/genetics , Signal Transduction/radiation effects , Transfection , Tumor Suppressor Proteins/genetics , Ultraviolet Rays , Up-Regulation/radiation effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
14.
J Toxicol Sci ; 36(5): 515-21, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22008527

ABSTRACT

To date, biological risk assessment studies of chemicals that induce DNA lesions have been primarily based on the action of DNA polymerases during replication. However, DNA lesions interfere not only with replication but also with transcription. Therefore, detecting the damaging effects of DNA lesions during transcription might be important for estimating the safety of chemical mutagens and carcinogens. However, methods to address these effects have not been developed. Here, we report a simple, non-isotopic method for determining the toxicity of chemical agents by visualizing transcription in a mammalian cell system. The method is based on the measurement of the incorporation of bromouridine (as the uridine analogue) into the nascent RNA during RNA synthesis inhibition (RSI) induced by the stalling of RNA polymerases at DNA lesions on the transcribed DNA strand, which triggers transcription-coupled nucleotide excision repair (TC-NER). When we tested chemical agents (camptothecin, etoposide, 4-nitroquinoline-1-oxide, mitomycin C, methyl methanesulfonate, and cisplatin) in HeLa cells by the method, RSI indicative of genomic toxicity was observed in the nucleoli of the tested cells. This procedure provides the following advantages: 1) it uses common, affordable mammalian cells (HeLa cells, WI38VA13 cells, human dermal fibroblasts, or Chinese hamster ovary cells) rather than genetically modified microorganisms; 2) it can be completed within approximately 8 hr after the cells are prepared because RNA polymerase responses during TC-NER are faster than other DNA damage responses (replication, recombination, and apoptosis); and 3) it is safe because it uses non-radioactive bromouridine and antibodies to detect RNA synthesis on undamaged transcribed DNA strands.


Subject(s)
DNA Damage , Mutagenicity Tests/methods , Mutagens/toxicity , RNA/biosynthesis , Transcription, Genetic , Animals , Bromouracil/analogs & derivatives , CHO Cells , Cell Culture Techniques , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Cell Nucleolus/radiation effects , Cell Nucleolus/ultrastructure , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Cell Nucleus/ultrastructure , Cricetinae , DNA-Directed RNA Polymerases/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , RNA/chemistry , Risk Assessment , Ultraviolet Rays , Uridine/analogs & derivatives , Uridine/chemistry
15.
Mol Cell Proteomics ; 10(10): M111.009241, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21778410

ABSTRACT

The nucleolus is a nuclear organelle that coordinates rRNA transcription and ribosome subunit biogenesis. Recent proteomic analyses have shown that the nucleolus contains proteins involved in cell cycle control, DNA processing and DNA damage response and repair, in addition to the many proteins connected with ribosome subunit production. Here we study the dynamics of nucleolar protein responses in cells exposed to stress and DNA damage caused by ionizing and ultraviolet (UV) radiation in diploid human fibroblasts. We show using a combination of imaging and quantitative proteomics methods that nucleolar substructure and the nucleolar proteome undergo selective reorganization in response to UV damage. The proteomic responses to UV include alterations of functional protein complexes such as the SSU processome and exosome, and paraspeckle proteins, involving both decreases and increases in steady state protein ratios, respectively. Several nonhomologous end-joining proteins (NHEJ), such as Ku70/80, display similar fast responses to UV. In contrast, nucleolar proteomic responses to IR are both temporally and spatially distinct from those caused by UV, and more limited in terms of magnitude. With the exception of the NHEJ and paraspeckle proteins, where IR induces rapid and transient changes within 15 min of the damage, IR does not alter the ratios of most other functional nucleolar protein complexes. The rapid transient decrease of NHEJ proteins in the nucleolus indicates that it may reflect a response to DNA damage. Our results underline that the nucleolus is a specific stress response organelle that responds to different damage and stress agents in a unique, damage-specific manner.


Subject(s)
Cell Nucleolus/metabolism , DNA Damage , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Proteome/analysis , Antigens, Nuclear/analysis , Antigens, Nuclear/metabolism , Cell Nucleolus/radiation effects , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Exosomes/metabolism , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Isotope Labeling , Ku Autoantigen , Microscopy, Electron, Transmission , Nuclear Proteins/genetics , Proteome/genetics , Proteome/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Radiation, Ionizing , Stress, Physiological , Transcription, Genetic , Ultraviolet Rays
16.
FEBS Lett ; 584(22): 4559-64, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20974138

ABSTRACT

hCINAP is an atypical nucleoplasmic enzyme, combining structural features of adenylate kinases and ATPases, which exhibits dual enzymatic activity. It interacts with the Cajal Body marker coilin and its level of expression and enzymatic activity influence Cajal Body numbers. Here we show that upon specific transcriptional inhibition of RNA pol.II, hCINAP segregates in perinuclear caps identified as Dark Nucleolar Caps (DNCs). These are distinct from perinucleolar caps where coilin and fibrillarin (both Cajal Body components) accumulate. In DNCs, hCINAP co-localizes with Paraspeckle Protein (PSP1) and also co-segregates with PSP1, and not coilin, in nuclear and nucleolar foci upon UV irradiation.


Subject(s)
Adenosine Triphosphatases/metabolism , Adenylate Kinase/metabolism , Cell Nucleolus/metabolism , RNA Polymerase II/metabolism , Active Transport, Cell Nucleus/radiation effects , Cell Nucleolus/radiation effects , HeLa Cells , Humans , Ultraviolet Rays
17.
Neuro Endocrinol Lett ; 31(2): 208-14, 2010.
Article in English | MEDLINE | ID: mdl-20424591

ABSTRACT

OBJECTIVES: We aimed to investigate the effects of weak extremely low frequency electromagnetic fields (ELF-EMFs) on the nucleus size, the silver staining nucleolar organizer regions (AgNORs), the frequency of micro nucleated peripheral blood lymphocytes (MPBLs) and the micro nucleated polychromatic erythrocytes (MPCEs). METHODS: One hundred and twenty Swiss albino mice were equally divided into 6 groups. The study groups were exposed to 1, 2, 3, 4 and 5 microT 50 Hz-EMFs for 40 days. Micronucleus number (MN) per PBL was determined.. RESULTS: ELF-EMF exposure caused a nonlinear decline of nucleus area. A sharp drop occurred in AgNOR area of 1 microT group, and following it gained an insignificantly higher level than that of the control group. The field did not change mean AgNOR numbers per nucleus of the groups. Relative AgNOR area had the highest level in 1 microT-exposure group, and the level was quite similar to that of the 5 microT-exposure group. The remaining groups had significantly lower values quite similar to that of the control level. The field exposure at any intensity did not affect significantly the frequency of either MPBLs or MPCEs. The number of MN per PBL in the 4 and 5 microT-exposure groups were significantly higher than those of the lower intensity exposure groups. The males in 4 microT-exposure group displayed the highest MN number per PBL, whereas values changed in a nonlinear manner. CONCLUSIONS: The results of the present study suggest that

Subject(s)
Cell Nucleolus/radiation effects , Electromagnetic Fields , Erythroblasts/radiation effects , Lymphocytes/radiation effects , Nucleolus Organizer Region/radiation effects , Animals , Leukocyte Count , Mice , Mice, Inbred Strains , Nucleolus Organizer Region/metabolism , Time Factors
18.
Radiats Biol Radioecol ; 50(1): 42-51, 2010.
Article in Russian | MEDLINE | ID: mdl-20297680

ABSTRACT

Transposition and mutual approaching of pericentromeric loci 1q12 of homological chromosomes from the nuclear membrane towards the nuclear centre as well as activation of the chromosomal nucleolus-forming regions (NFR) are observed in human mesenchymal stem cells (hMSCs) as an initial stages of the adaptive response (AR) after exposure to low doses of X-radiation (10 cGy). All these reactions are also induced after addition of cultivation medium from irradiated cells to intact bystander-cells and this phenomenon called bystander effect (BE). Recently the same AR and BE induction results were obtained for human G0-lymphocytes. All these data indicate the existence of universal reaction of homological chromosome loci transposition which was revealed during AR development in differentiated (lymphocytes) and non-differentiated (hMSCs) and also it shows possibility of radiational BE development in suspension and monolayer cell cultures upon addition of stress-signalization factors in incubation medium. We suppose that these factors are extracellular genome DNA fragments apoptotic cells.


Subject(s)
Bystander Effect , Mesenchymal Stem Cells/radiation effects , Cell Nucleolus/radiation effects , Dose-Response Relationship, Radiation , Humans , Mesenchymal Stem Cells/physiology , Nucleolus Organizer Region/radiation effects , X-Rays
20.
Radiats Biol Radioecol ; 49(5): 528-37, 2009.
Article in Russian | MEDLINE | ID: mdl-19947515

ABSTRACT

Recently we found that transposition of homologous chromosomes 1q12 loci towards the nuclear centre and activation of the chromosomal nucleolus-forming regions (NFR) are observed in human lymphocytes after exposure to low doses of X-radiation (10 cGy). These cell reactions were studied for human breast cancer stem cell cultures. There are two cell types in cell culture from single donor: with two (type 1) and three (type 2) loci of 1q12. It was shown that an adaptive response induced by X-ray irradiation is developed only in cells of the type 1 but not in type 2 ones after 3 and 10 cGy doses. We observed a considerable death of cell type 2 after low-dose exposure. Activation of the NFR in breast cancer stem cells after irradiation was not found. In this paper we discuss features of studied cancer stem cells lines and their responses to low doses of ionizing radiation.


Subject(s)
Adenocarcinoma/ultrastructure , Breast Neoplasms/ultrastructure , Neoplastic Stem Cells/radiation effects , Nucleolus Organizer Region/radiation effects , Radiation Tolerance , Cell Nucleolus/radiation effects , Dose-Response Relationship, Radiation , Female , Humans , Neoplastic Stem Cells/ultrastructure , Tumor Cells, Cultured , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...