Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 2155, 2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33846325

RESUMO

Cryptochromes (CRYs) are photoreceptors or components of the molecular clock in various evolutionary lineages, and they are commonly regulated by polyubiquitination and proteolysis. Multiple E3 ubiquitin ligases regulate CRYs in animal models, and previous genetics study also suggest existence of multiple E3 ubiquitin ligases for plant CRYs. However, only one E3 ligase, Cul4COP1/SPAs, has been reported for plant CRYs so far. Here we show that Cul3LRBs is the second E3 ligase of CRY2 in Arabidopsis. We demonstrate the blue light-specific and CRY-dependent activity of LRBs (Light-Response Bric-a-Brack/Tramtrack/Broad 1, 2 & 3) in blue-light regulation of hypocotyl elongation. LRBs physically interact with photoexcited and phosphorylated CRY2, at the CCE domain of CRY2, to facilitate polyubiquitination and degradation of CRY2 in response to blue light. We propose that Cul4COP1/SPAs and Cul3LRBs E3 ligases interact with CRY2 via different structure elements to regulate the abundance of CRY2 photoreceptor under different light conditions, facilitating optimal photoresponses of plants grown in nature.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Criptocromos/metabolismo , Fotorreceptores de Plantas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Arabidopsis/efeitos da radiação , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Criptocromos/química , Criptocromos/genética , Células HEK293 , Humanos , Luz , Modelos Biológicos , Mutação/genética , Fosforilação/efeitos da radiação , Poliubiquitina/metabolismo , Ligação Proteica/efeitos da radiação , Proteólise/efeitos da radiação , Plântula/efeitos da radiação , Ubiquitinação/efeitos da radiação
2.
J Biol Chem ; 296: 100570, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33753168

RESUMO

Nonmelanoma skin cancers occur primarily in individuals over the age of 60 and are characterized by an abundance of ultraviolet (UV) signature mutations in keratinocyte DNA. Though geriatric skin removes UV photoproducts from DNA less efficiently than young adult skin, it is not known whether the utilization of other prosurvival but potentially mutagenic DNA damage tolerance systems such as translesion synthesis (TLS) is altered in older individuals. Using monoubiquitination of the replicative DNA polymerase clamp protein PCNA (proliferating cell nuclear antigen) as a biochemical marker of TLS pathway activation, we find that UVB exposure of the skin of individuals over the age of 65 results in a higher level of PCNA monoubiquitination than in the skin of young adults. Furthermore, based on previous reports showing a role for deficient insulin-like growth factor-1 (IGF-1) signaling in altered UVB DNA damage responses in geriatric human skin, we find that both pharmacological inhibition of the IGF-1 receptor (IGF-1R) and deprivation of IGF-1 potentiate UVB-induced PCNA monoubiquitination in both human skin ex vivo and keratinocytes in vitro. Interestingly, though the TLS DNA polymerase Pol eta can accurately replicate the major photoproducts induced in DNA by UV radiation, we find that it fails to accumulate on chromatin in the absence of IGF-1R signaling and that this phenotype is correlated with increased mutagenesis in keratinocytes in vitro. Thus, altered IGF-1/IGF-1R signaling in geriatric skin may predispose epidermal keratinocytes to carry out a more mutagenic form of DNA synthesis following UVB exposure.


Assuntos
Envelhecimento/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Pele/metabolismo , Pele/efeitos da radiação , Ubiquitinação/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Idoso , Envelhecimento/efeitos da radiação , Dano ao DNA , Reparo do DNA/efeitos da radiação , Feminino , Humanos , Masculino , Transdução de Sinais/efeitos da radiação , Pele/citologia
3.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33495326

RESUMO

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.


Assuntos
Dano ao DNA , Proteínas Nucleares/metabolismo , Sirtuínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta , Acetilação/efeitos da radiação , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Catálise/efeitos da radiação , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Nucléolo Celular/efeitos da radiação , Humanos , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nucleofosmina , Fosforilação/efeitos da radiação , Estabilidade Proteica/efeitos da radiação , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transcrição Gênica/efeitos da radiação , Ubiquitinação/efeitos da radiação
4.
Int J Mol Sci ; 21(19)2020 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-32992650

RESUMO

A tight junction (TJ) makes a physical barrier in the epidermal cells of skin. Ultraviolet (UV) light may disrupt the TJ barrier, but the mechanism has not been well clarified. Weak UVB (5 mJ/cm2) caused mislocalization of claudin-1 (CLDN1), a component of the TJ strand, and disruption of TJ barrier in human keratinocyte-derived HaCaT cells. The UVB-induced mislocalization of CLDN1 was inhibited by monodansylcadaverine (MDC), a clathrin-dependent endocytosis inhibitor, suggesting that UVB enhances the internalization of CLDN1. Transepidermal electrical resistance and paracellular flux of lucifer yellow, a fluorescent hydrophilic marker, were rescued by MDC. UVB changed neither the total nor phosphorylation levels of CLDN1, but it increased both mono-ubiquitination and tyrosine nitration levels of CLDN1. Fluorescence measurements revealed that UVB increased intracellular free Ca2+, nitric oxide (NO), and peroxynitrite contents, which were inhibited by Opsin2 (OPN2) siRNA, suggesting that OPN2 functions as a UVB sensor. The effects of UVB were inhibited by an antagonist of transient receptor potential type vanilloid 1 (TRPV1) and Ca2+ chelator. Both NO donor and peroxynitrite donor induced the mislocalization of CLDN1 and disruption of TJ barrier, which were rescued by a NO synthase (NOS) inhibitor and a peroxynitrite scavenger. Weak UVB irradiation induced the disruption of TJ barrier mediated by mislocalization of CLDN1 in HaCaT cells. The OPN2/TRPV1/NOS signaling pathway may be a novel target for preventing destruction of the TJ barrier by UVB irradiation.


Assuntos
Claudina-1/metabolismo , Queratinócitos/metabolismo , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/biossíntese , Transdução de Sinais/efeitos da radiação , Raios Ultravioleta , Cadaverina/análogos & derivados , Cadaverina/farmacologia , Sobrevivência Celular/efeitos da radiação , Endocitose/efeitos dos fármacos , Células HaCaT , Humanos , Óxido Nítrico Sintase/metabolismo , Fosforilação/efeitos da radiação , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/efeitos da radiação , Ubiquitinação/efeitos da radiação
5.
DNA Repair (Amst) ; 95: 102959, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32927239

RESUMO

UV is a significant environmental agent that damages DNA. Translesion synthesis (TLS) is a DNA damage tolerance pathway that utilizes specialized DNA polymerases to replicate through the damaged DNA, often leading to mutagenesis. In eukaryotic cells, genomic DNA is organized into chromatin that is composed of nucleosomes. To date, if and/or how TLS is regulated by a specific nucleosome feature has been undocumented. We found that mutations of multiple histone H4 residues mostly or entirely embedded in the nucleosomal LRS (loss of ribosomal DNA-silencing) domain attenuate UV mutagenesis in Saccharomyces cerevisiae. The attenuation is not caused by an alteration of ubiquitination or sumoylation of PCNA (proliferating cell nuclear antigen), the modifications well-known to regulate TLS. Also, the attenuation is not caused by decreased chromatin accessibility, or by alterations of methylation of histone H3 K79, which is at the center of the LRS surface. The attenuation may result from compromised TLS by both DNA polymerases ζ and η, in which Rad6 and Rad5 are but Rad18 is not implicated. We propose that a feature of the LRS is recognized or accessed by the TLS machineries either during/after a nucleosome is disassembled in front of a lesion-stalled replication fork, or during/before a nucleosome is reassembled behind a lesion-stalled replication fork.


Assuntos
Histonas/química , Histonas/genética , Mutagênese/genética , Mutagênese/efeitos da radiação , Mutação , Antígeno Nuclear de Célula em Proliferação/metabolismo , Raios Ultravioleta/efeitos adversos , Modelos Moleculares , Domínios Proteicos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos da radiação , Sumoilação/genética , Sumoilação/efeitos da radiação , Ubiquitinação/genética , Ubiquitinação/efeitos da radiação
6.
Nucleic Acids Res ; 48(16): 9181-9194, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32789493

RESUMO

The NAD+-dependent deacetylase and mono-ADP-ribosyl transferase SIRT6 stabilizes the genome by promoting DNA double strand break repair, thereby acting as a tumor suppressor. However, whether SIRT6 regulates nucleotide excision repair (NER) remains unknown. Here, we showed that SIRT6 was recruited to sites of UV-induced DNA damage and stimulated the repair of UV-induced DNA damage. Mechanistic studies further indicated that SIRT6 interacted with DDB2, the major sensor initiating global genome NER (GG-NER), and that the interaction was enhanced upon UV irradiation. SIRT6 deacetylated DDB2 at two lysine residues, K35 and K77, upon UV stress and then promoted DDB2 ubiquitination and segregation from chromatin, thereby facilitating downstream signaling. In addition, we characterized several SIRT6 mutations derived from melanoma patients. These SIRT6 mutants ablated the stimulatory effect of SIRT6 on NER and destabilized the genome due to (i) partial loss of enzymatic activity (P27S or H50Y), (ii) a nonsense mutation (R150*) or (iii) high turnover rates (G134W). Overall, we demonstrate that SIRT6 promotes NER by deacetylating DDB2, thereby preventing the onset of melanomagenesis.


Assuntos
Carcinogênese/genética , Dano ao DNA/genética , Proteínas de Ligação a DNA/genética , Melanoma/genética , Sirtuínas/genética , Carcinogênese/efeitos da radiação , Cromatina/genética , Cromatina/efeitos da radiação , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Células HeLa , Humanos , Melanoma/patologia , Mutação/efeitos da radiação , Ubiquitinação/efeitos da radiação , Raios Ultravioleta/efeitos adversos
7.
J Photochem Photobiol B ; 210: 111977, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32738749

RESUMO

Widely used white light-emitting diodes (LEDs) currently deliver higher levels of blue light than conventional domestic light sources. The high intensity of the blue component is the main source of concern regarding possible health risks of LED to chronic light toxicity to the retina. Therefore, we analyzed retinal injury and genome-wide changes in gene expression induced by white LED light with different correlated color temperatures (CCTs) in a mouse model. Balb/c mice (10 weeks old) were exposed to LED light with CCTs of 2954, 5624, and 7378 K, at different illuminance levels (250, 500, 1000, and 3000 lx) and for different exposure times (7, 14, and 28 days). Hematoxylin and eosin staining revealed that exposure to 7378 K light at 250 lx for 28 days resulted in a significant reduction of outer nuclear layer (ONL) nuclei, whereas 2954 K light at <3000 lx led to only a mild reduction in the number of ONL nuclei. In addition, 5624 and 7378 K light at 3000 lx resulted in a significant increase in TUNEL-positive apoptotic nuclei, which was not found at an illuminance of 1000 lx. Genome-wide expression analyses showed that, compared to a control group, there were 121 upregulated differentially expressed genes (DEGs) and 458 downregulated DEGs found in the 7378 K group, and 59 upregulated and only 4 downregulated DEGs in the 2954 K group. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that the DEGs were involved in 341 GO terms and 16 related pathways for the 7378 K group and in 12 GO terms and 7 related pathways for the 2954 K group. Signal pathways related to ubiquitin potentially played an important role in light-induced retinal degeneration. Furthermore, retinal immunohistochemistry (IHC) indicated downregulation of ubiquitin and autophagy function caused by 7378 K light. Taken together, these results indicate that retinal injury in the mice induced by white LED light occurred in a CCT-dependent manner, and that light with a higher CCT was more likely to reduce ONL nuclei; however, the apoptosis pathway may not be the only mechanism involved. Based on genome-wide expression analyses and retinal IHC, the ubiquitin-mediated proteolysis signal pathway may have participated in the induction retinal degeneration.


Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Luz , Retina/efeitos da radiação , Animais , Autofagia/efeitos da radiação , Núcleo Celular/metabolismo , Análise por Conglomerados , Cor , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Retina/metabolismo , Retina/patologia , Temperatura , Ubiquitinação/efeitos da radiação
8.
Oncogene ; 39(18): 3710-3725, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32157210

RESUMO

Radioresistance becomes the major obstacle to reduce tumor recurrence and improve prognosis in the treatment of esophageal squamous cell carcinoma (ESCC). Thus new strategies for radioresistant ESCC are urgently needed. Herein, we reported that tribbles pseudokinase 3 (TRIB3) serves as a key regulator of radioresistance in ESCC. TRIB3 is overexpressed in ESCC tissues and cell lines. High expression of TRIB3 significantly correlates with poor radiotherapy response and prognosis in ESCC patients. Upregulation of TRIB3 in ESCC cells conferred radioresistance in vitro and in vivo by interacting with TAZ thus impeding ß-TrCP-mediated TAZ ubiquitination and degradation. Conversely, silencing TRIB3 sensitized ESCC cells to ionizing radiation. More importantly, TRIB3 was significantly correlated with TAZ activation in ESCC biopsies, and patients with high expression of both TRIB3 and TAZ suffered the worst radiotherapy response and survival. Our study uncovers the critical mechanism of ESCC resistance to radiotherapy, and provides a new pharmacological opportunity for developing a mechanism-based strategy to eliminate radioresistant ESCC in clinical practice.


Assuntos
Proteínas de Ciclo Celular/genética , Carcinoma de Células Escamosas do Esôfago/radioterapia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tolerância a Radiação/genética , Proteínas Repressoras/genética , Transativadores/genética , Animais , Intervalo Livre de Doença , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Células HEK293 , Xenoenxertos , Humanos , Masculino , Camundongos , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/radioterapia , Prognóstico , Ligação Proteica/genética , Proteínas Serina-Treonina Quinases/genética , Proteólise/efeitos da radiação , Radiossensibilizantes/farmacologia , Transdução de Sinais/genética , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Ubiquitinação/efeitos da radiação
9.
Cell Biochem Funct ; 38(3): 283-289, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31943290

RESUMO

Based on central dogma of genetics, protein is the embodiment and executor of genetic function, post-translational modifications (PTMs) of protein are particularly important and involved in almost all aspects of cell biology and pathogenesis. Studies have shown that ionizing radiation (IR) alters gene expression much more profoundly and a broad variety of cell-process pathways, lots of proteins are modified and activated. Our understanding of the protein in response to ionizing radiation is steadily increasing. Among the various biological processes known to induce radioresistance, PTMs have attracted marked attention in recent years. The present review summarizes the latest knowledge about how PTMs response to ionizing radiation and pathway analysis were conducted. The data provided insights into biological effects of IR and contributing to the development of novel IR-based strategies.


Assuntos
Processamento de Proteína Pós-Traducional/efeitos da radiação , Proteínas/efeitos da radiação , Radiação Ionizante , Motivos de Aminoácidos , Dano ao DNA/efeitos da radiação , Genoma Humano/efeitos da radiação , Glicosilação/efeitos da radiação , Humanos , Metilação/efeitos da radiação , Neoplasias/radioterapia , Fosforilação/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Ubiquitinação/efeitos da radiação
10.
Nucleic Acids Res ; 47(8): 4039-4053, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30722038

RESUMO

FBXW7, a classic tumor suppressor, is a substrate recognition subunit of the Skp1-cullin-F-box (SCF) ubiquitin ligase that targets oncoproteins for ubiquitination and degradation. We recently found that FBXW7 is recruited to DNA damage sites to facilitate nonhomologous end-joining (NHEJ). The detailed underlying molecular mechanism, however, remains elusive. Here we report that the WD40 domain of FBXW7, which is responsible for substrate binding and frequently mutated in human cancers, binds to poly(ADP-ribose) (PAR) immediately following DNA damage and mediates rapid recruitment of FBXW7 to DNA damage sites, whereas ATM-mediated FBXW7 phosphorylation promotes its retention at DNA damage sites. Cancer-associated arginine mutations in the WD40 domain (R465H, R479Q and R505C) abolish both FBXW7 interaction with PAR and recruitment to DNA damage sites, causing inhibition of XRCC4 polyubiquitination and NHEJ. Furthermore, inhibition or silencing of poly(ADP-ribose) polymerase 1 (PARP1) inhibits PAR-mediated recruitment of FBXW7 to the DNA damage sites. Taken together, our study demonstrates that the WD40 domain of FBXW7 is a novel PAR-binding motif that facilitates early recruitment of FBXW7 to DNA damage sites for subsequent NHEJ repair. Abrogation of this ability seen in cancer-derived FBXW7 mutations provides a molecular mechanism for defective DNA repair, eventually leading to genome instability.


Assuntos
Reparo do DNA por Junção de Extremidades , Proteína 7 com Repetições F-Box-WD/genética , Poli(ADP-Ribose) Polimerase-1/genética , Poli Adenosina Difosfato Ribose/metabolismo , Fator de Células-Tronco/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína 7 com Repetições F-Box-WD/química , Proteína 7 com Repetições F-Box-WD/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Fibroblastos/ultraestrutura , Raios gama , Células HCT116 , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efeitos da radiação , Células Secretoras de Insulina/ultraestrutura , Modelos Moleculares , Mutação , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli Adenosina Difosfato Ribose/química , Ligação Proteica , Domínios Proteicos , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Fator de Células-Tronco/metabolismo , Ubiquitinação/efeitos da radiação
11.
Oncogene ; 38(4): 549-563, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30158672

RESUMO

Ionizing radiation (IR) is a conventional cancer therapeutic, to which cancer cells develop radioresistance with exposure. The residual cancer cells after radiation treatment also have increased metastatic potential. The mechanisms by which cancer cells develop radioresistance and gain metastatic potential are still unknown. In this study acute IR exposure induced cancer cell senescence and apoptosis, but after long-term IR exposure, cancer cells exhibited radioresistance. The proliferation of radioresistant cells was retarded, and most cells were arrested in G0/G1 phase. The radioresistant cells simultaneously showed resistance to further IR-induced apoptosis, premature senescence, and epithelial to mesenchymal transformation (EMT). Acute IR exposure steadily elevated CDC6 protein levels due to the attenuation of ubiquitination, while CDC6 overexpression was observed in the radioresistant cells because the insufficiency of CDC6 phosphorylation blocked protein translocation from nucleus to cytoplasm, resulting in subcellular protein accumulation when the cells were arrested in G0/G1 phase. CDC6 ectopic overexpression in CNE2 cells resulted in apoptosis resistance, G0/G1 cell cycle arrest, premature senescence, and EMT, similar to the characteristics of radioresistant CNE2-R cells. Targeting CDC6 with siRNA promoted IR-induced senescence, sensitized cancer cells to IR-induced apoptosis, and reversed EMT. Furthermore, CDC6 depletion synergistically repressed the growth of CNE2-R xenografts when combined with IR. The study describes for the first time cell models for IR-induced senescence, apoptosis resistance, and EMT, three major mechanisms by which radioresistance develops. CDC6 is a novel radioresistance switch regulating senescence, apoptosis, and EMT. These studies suggest that CDC6highKI67low represents a new diagnostic marker of radiosensitivity, and CDC6 represents a new therapeutic target for cancer radiosensitization.


Assuntos
Antígenos CD/fisiologia , Antígenos de Diferenciação de Linfócitos T/fisiologia , Apoptose/efeitos da radiação , Carcinoma/patologia , Senescência Celular/fisiologia , Transição Epitelial-Mesenquimal/efeitos da radiação , Neoplasias Nasofaríngeas/patologia , Proteínas de Neoplasias/fisiologia , Processamento de Proteína Pós-Traducional/efeitos da radiação , Tolerância a Radiação/fisiologia , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/biossíntese , Antígenos de Diferenciação de Linfócitos T/genética , Carcinoma/radioterapia , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Antígeno Ki-67/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Nasofaríngeas/radioterapia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Fosforilação/efeitos da radiação , Estabilidade Proteica , Transporte Proteico/efeitos da radiação , Interferência de RNA , RNA Interferente Pequeno/genética , Ubiquitinação/efeitos da radiação , Raios X
12.
Int Arch Occup Environ Health ; 91(8): 937-950, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29971594

RESUMO

PURPOSE: Radon is a risk factor for lung cancer and uranium miners are more exposed than the general population. A genome-wide interaction analysis was carried out to identify genomic loci, genes or gene sets that modify the susceptibility to lung cancer given occupational exposure to the radioactive gas radon. METHODS: Samples from 28 studies provided by the International Lung Cancer Consortium were pooled with samples of former uranium miners collected by the German Federal Office of Radiation Protection. In total, 15,077 cases and 13,522 controls, all of European ancestries, comprising 463 uranium miners were compared. The DNA of all participants was genotyped with the OncoArray. We fitted single-marker and in multi-marker models and performed an exploratory gene-set analysis to detect cumulative enrichment of significance in sets of genes. RESULTS: We discovered a genome-wide significant interaction of the marker rs12440014 within the gene CHRNB4 (OR = 0.26, 95% CI 0.11-0.60, p = 0.0386 corrected for multiple testing). At least suggestive significant interaction of linkage disequilibrium blocks was observed at the chromosomal regions 18q21.23 (p = 1.2 × 10-6), 5q23.2 (p = 2.5 × 10-6), 1q21.3 (p = 3.2 × 10-6), 10p13 (p = 1.3 × 10-5) and 12p12.1 (p = 7.1 × 10-5). Genes belonging to the Gene Ontology term "DNA dealkylation involved in DNA repair" (GO:0006307; p = 0.0139) or the gene family HGNC:476 "microRNAs" (p = 0.0159) were enriched with LD-blockwise significance. CONCLUSION: The well-established association of the genomic region 15q25 to lung cancer might be influenced by exposure to radon among uranium miners. Furthermore, lung cancer susceptibility is related to the functional capability of DNA damage signaling via ubiquitination processes and repair of radiation-induced double-strand breaks by the single-strand annealing mechanism.


Assuntos
Carcinógenos Ambientais/toxicidade , Neoplasias Pulmonares/genética , Neoplasias Induzidas por Radiação/genética , Proteínas do Tecido Nervoso/genética , Doenças Profissionais/genética , Radônio/toxicidade , Receptores Nicotínicos/genética , Estudos de Casos e Controles , Dano ao DNA/efeitos da radiação , Feminino , Marcadores Genéticos/efeitos da radiação , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Mineração , Exposição Ocupacional/efeitos adversos , Fatores de Risco , Ubiquitinação/efeitos da radiação , Urânio
13.
J Biol Chem ; 293(2): 588-598, 2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29167269

RESUMO

Cells have evolved sophisticated mechanisms to maintain genomic integrity in response to DNA damage. Ionizing radiation (IR)-induced DNA damage results in the formation of IR-induced foci (iRIF) in the nucleus. The iRIF formation is part of the DNA damage response (DDR), which is an essential signaling cascade that must be strictly regulated because either the loss of or an augmented DDR leads to loss of genome integrity. Accordingly, negative regulation of the DDR is as critical as its activation. In this study, we have identified ring finger protein 126 (RNF126) as a negative regulator of the DDR from a screen of iRIF containing 53BP1. RNF126 overexpression abolishes not only the formation of 53BP1 iRIF but also of RNF168, FK2, RAP80, and BRCA1. However, the iRIF formation of γH2AX, MDC1, and RNF8 is maintained, indicating that RNF126 acts between RNF8 and RNF168 during the DDR. In addition, RNF126 overexpression consistently results in the loss of RNF168-mediated H2A monoubiquitination at lysine 13/15 and inhibition of the non-homologous end joining capability. Taken together, our findings reveal that RNF126 is a novel factor involved in the negative regulation of DDR, which is important for sustaining genomic integrity.


Assuntos
Radiação Ionizante , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA/efeitos da radiação , Células HeLa , Histonas/metabolismo , Histonas/efeitos da radiação , Humanos , Imunoprecipitação , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/efeitos da radiação
14.
J Neuropathol Exp Neurol ; 76(7): 578-584, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28535250

RESUMO

Interferon-stimulated gene 15 (ISG15), an antagonist of the ubiquitin pathway, is elevated in cells and brain tissues obtained from ataxia telangiectasia (A-T) patients. Previous studies reveal that an elevated ISG15 pathway inhibits ubiquitin-dependent protein degradation, leading to activation of basal autophagy as a compensatory mechanism for protein turnover in A-T cells. Also, genotoxic stress (ultraviolet [UV] radiation) deregulates autophagy and induces aberrant degradation of ubiquitylated proteins in A-T cells. In the current study, we show that, as in A-T cells, ISG15 protein expression is elevated in cerebellums and various other tissues obtained from Atm-compromised mice in an Atm-allele-dependent manner (Atm+/+ < Atm+/- < Atm-/-). Notably, in cerebellums, the brain part primarily affected in A-T, levels of ISG15 were significantly greater (3-fold higher) than cerebrums obtained from the same set of mice. Moreover, as in A-T cell culture, UV induces aberrant degradation of ubiquitylated proteins and autophagy in Atm-deficient, but not in Atm-proficient, cerebellar brain slices grown in culture. Thus, the ex vivo organotypic A-T mouse brain culture model mimics that of an A-T human cell culture model and could be useful for studying the role of ISG15-dependent proteinopathy in cerebellar neurodegeneration, a hallmark of A-T in humans.


Assuntos
Ataxia Telangiectasia/genética , Ataxia Telangiectasia/patologia , Cerebelo/metabolismo , Citocinas/metabolismo , Mutação/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Proteínas Mutadas de Ataxia Telangiectasia/genética , Autofagia/genética , Autofagia/efeitos da radiação , Cerebelo/efeitos da radiação , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/efeitos da radiação , Genótipo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Técnicas de Cultura de Órgãos , Ubiquitinação/genética , Ubiquitinação/efeitos da radiação , Ubiquitinas/metabolismo , Raios Ultravioleta
15.
Development ; 144(10): 1831-1840, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28420710

RESUMO

The phytochrome-mediated regulation of photomorphogenesis under red and far-red light conditions involves both positively and negatively acting factors. The positively acting factors (e.g. HY5/HFR1/LAF1 and others) are degraded in the dark to prevent photomorphogenesis. By contrast, the negatively acting factors (e.g. phytochrome-interacting factors or PIFs) are degraded in response to light to promote photomorphogenesis. Here, we show that the negatively acting factor PIF1 is also degraded in the dark by direct heterodimerization with the positively acting factor HFR1. Conversely, PIF1 also promotes the degradation of HFR1 in darkness. PIF1 enhances the poly-ubiquitylation of HFR1 by COP1 in vivo and in vitro In addition, the reciprocal co-degradation of PIF1 and HFR1 is dependent on the 26S proteasome pathway in vivo Genetic evidence shows that the hfr1 mutant partially suppresses the constitutive photomorphogenic phenotypes of cop1-6 pif1 and of the quadruple mutant pifq both in the dark and in far-red light conditions. Taken together, these data uncover a co-degradation mechanism between PIFs and HFR1 that underlies photomorphogenic development in Arabidopsis thaliana.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Luz , Proteínas Nucleares/metabolismo , Desenvolvimento Vegetal , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Arabidopsis/crescimento & desenvolvimento , Arabidopsis/metabolismo , Arabidopsis/efeitos da radiação , Desenvolvimento Vegetal/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Ubiquitinação/efeitos da radiação
16.
Oncotarget ; 8(22): 36423-36437, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28430587

RESUMO

Deficiency of Parkin is a major cause of early-onset Parkinson's disease (PD). Notably, PD patients also exhibit a significantly higher risk in melanoma and other skin tumors, while the mechanism remains largely unknown. In this study, we show that depletion of Parkin causes compromised cell viability and genome stability after ultraviolet (UV) radiation. We demonstrate that Parkin promotes efficient Rad18-dependent proliferating cell nuclear antigen (PCNA) monoubiquitination by facilitating the formation of Replication protein A (RPA)-coated ssDNA upon UV radiation. Furthermore, Parkin is found to physically interact with NBS1 (Nijmegen breakage syndrome 1), and to be required for optimal recruitment of NBS1 and DNA polymerase eta (Polη) to UV-induced damage sites. Consequently, depletion of Parkin leads to increased UV-induced mutagenesis. These findings unveil an important role of Parkin in protecting genome stability through positively regulating translesion DNA synthesis (TLS) upon UV damage, providing a novel mechanistic link between Parkin deficiency and predisposition to skin cancers in PD patients.


Assuntos
Reparo do DNA , Replicação do DNA/efeitos da radiação , Ubiquitina-Proteína Ligases/metabolismo , Raios Ultravioleta , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Técnicas de Inativação de Genes , Instabilidade Genômica , Humanos , Camundongos , Mutagênese/efeitos da radiação , Mutação , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ligação Proteica , Tolerância a Radiação/genética , Proteína de Replicação A/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/efeitos da radiação
17.
Dev Cell ; 41(1): 47-58.e4, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28399399

RESUMO

The plant hormones brassinosteroids (BRs) participate in light-mediated regulation of plant growth, although the underlying mechanisms are far from being fully understood. In addition, the function of the core transcription factor in the BR signaling pathway, BRI1-EMS-SUPPRESSOR 1 (BES1), largely depends on its phosphorylation status and its protein stability, but the regulation of BES1 is not well understood. Here, we report that SINA of Arabidopsis thaliana (SINATs) specifically interact with dephosphorylated BES1 and mediate its ubiquitination and degradation. Our genetic data demonstrated that SINATs inhibit BR signaling in a BES1-dependent manner. Interestingly, we found that the protein levels of SINATs were decreased in the dark and increased in the light, which changed BES1 protein levels accordingly. Thus, our study not only uncovered a new mechanism of BES1 degradation but also provides significant insights into how light conditionally regulates plant growth through controlling accumulation of different forms of BES1.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Arabidopsis/efeitos da radiação , Brassinosteroides/farmacologia , Luz , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Arabidopsis/efeitos dos fármacos , Arabidopsis/crescimento & desenvolvimento , Proteínas de Arabidopsis/química , Proteínas de Ligação a DNA , Técnicas de Silenciamento de Genes , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/efeitos da radiação , Estabilidade Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos da radiação , Proteólise/efeitos dos fármacos , Proteólise/efeitos da radiação , Domínios RING Finger , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/efeitos da radiação
18.
Mol Vis ; 23: 52-59, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28331281

RESUMO

PURPOSE: Blue light is a high-energy emitting light with a short wavelength in the visible light spectrum. Blue light induces photoreceptor apoptosis and causes age-related macular degeneration or retinitis pigmentosa. In the present study, we investigated the roles of endoplasmic reticulum (ER) stress induced by blue light-emitting diode (LED) light exposure in murine photoreceptor cells. METHODS: The murine photoreceptor cell line was incubated and exposed to blue LED light (464 nm blue LED light, 450 lx, 3 to 24 h). The expression of the factors involved in the unfolded protein response pathway was examined using quantitative real-time reverse transcription (RT)-PCR and immunoblot analysis. The aggregation of short-wavelength opsin (S-opsin) in the murine photoreceptor cells was observed with immunostaining. The effect of S-opsin knockdown on ATF4 expression in the murine photoreceptor cell line was also investigated. RESULTS: Exposure to blue LED light increased the bip, atf4, and grp94 mRNA levels, induced the expression of ATF4 protein, and increased the levels of ubiquitinated proteins. Exposure to blue LED light in combination with ER stress inducers (tunicamycin and dithiothreitol) induced the aggregation of S-opsin. S-opsin mRNA knockdown prevented the induction of ATF4 expression in response to exposure to blue LED light. CONCLUSIONS: These findings indicate that the aggregation of S-opsin induced by exposure to blue LED light causes ER stress, and ATF4 activation in particular.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Luz , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Opsinas de Bastonetes/metabolismo , Animais , Linhagem Celular , Ditiotreitol/farmacologia , Técnicas de Silenciamento de Genes , Camundongos Endogâmicos C57BL , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Poliubiquitina/metabolismo , Agregados Proteicos/efeitos dos fármacos , Agregados Proteicos/efeitos da radiação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Tunicamicina/farmacologia , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/efeitos da radiação , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos da radiação
19.
Cell Chem Biol ; 24(4): 443-457.e6, 2017 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-28330605

RESUMO

Ubiquitin (Ub) signaling is a diverse group of processes controlled by covalent attachment of small protein Ub and polyUb chains to a range of cellular protein targets. The best documented Ub signaling pathway is the one that delivers polyUb proteins to the 26S proteasome for degradation. However, studies of molecular interactions involved in this process have been hampered by the transient and hydrophobic nature of these interactions and the lack of tools to study them. Here, we develop Ub-phototrap (UbPT), a synthetic Ub variant containing a photoactivatable crosslinking side chain. Enzymatic polymerization into chains of defined lengths and linkage types provided a set of reagents that led to identification of Rpn1 as a third proteasome ubiquitin-associating subunit that coordinates docking of substrate shuttles, unloading of substrates, and anchoring of polyUb conjugates. Our work demonstrates the value of UbPT, and we expect that its future uses will help define and investigate the ubiquitin interactome.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Sítios de Ligação , Reagentes de Ligações Cruzadas/química , Simulação de Acoplamento Molecular , Ressonância Magnética Nuclear Biomolecular , Poliubiquitina/química , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/genética , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina/química , Ubiquitina/genética , Ubiquitinação/efeitos da radiação , Raios Ultravioleta
20.
Mol Cell Biol ; 37(4)2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27895153

RESUMO

Repair of damaged DNA is critical for maintenance of genetic information. In eukaryotes, DNA double-strand breaks (DSBs) are recognized by the Ku70-Ku80 heterodimer, which then recruits proteins that mediate repair by nonhomologous end joining (NHEJ). Prolonged retention of Ku70/80 at DSBs prevents completion of repair, however, with ubiquitylation of Ku80 having been implicated in Ku70/80 dissociation from DNA. Here, we identify RNF126 as a ubiquitin ligase that is recruited to DSBs and ubiquitylates Ku80, with UBE2D3 serving as an E2 enzyme. Knockdown of RNF126 prevented Ku70/80 dissociation from DSBs and inhibited break repair. Attenuation of Ku80 ubiquitylation by replacement of ubiquitylation site lysines with arginine residues delayed Ku70/80 release from chromatin after DSB induction by genotoxic insults. Together, our data indicate that RNF126 is a novel regulator of NHEJ that promotes completion of DNA repair by ubiquitylating Ku80 and releasing Ku70/80 from damaged DNA.


Assuntos
Reparo do DNA por Junção de Extremidades , Autoantígeno Ku/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Cromatina/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Células HEK293 , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Células NIH 3T3 , Multimerização Proteica/efeitos da radiação , Proteólise/efeitos da radiação , Radiação Ionizante , Ubiquitinação/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...