Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38870324

RESUMO

PURPOSE: Recently, there have been a few reports regarding the usefulness of a novel saline injection technique using a spiral flow-generating tube. The purpose of this study was to evaluate whether simultaneous saline injection using a spiral flow-generating tube was able to improve hepatic contrast enhancement and lesion conspicuity of metastatic liver tumors. METHODS: We randomized a total of 411 patients with various liver diseases including metastases by total body weight (A, n = 204) and contrast dilution protocol (B, n = 207). Group A received 400 mgI/kg of contrast medium alone without a spiral flow-generating tube; group B received contrast medium 400 mgI/kg simultaneous with injection of a 0.57-ml/kg physiologic saline solution through a spiral flow-generating tube. Abdominal aorta computed tomography (CT) number, hepatic enhancement (ΔHU), percentage of tests demonstrating an enhancement effect of the liver parenchyma exceeding Δ50 HU in 3 measured segments (S2, S6, and S8), and the contrast-to-noise ratio of the metastatic liver tumors were measured. RESULTS: The mean aortic CT number of group B (417.0 HU ± 61.7; P < 0.01) was approximately 10% higher than that of group A (384.6 ± 79.1 HU). The average ΔHU was 59.8 ± 11.4 HU for group A and 61.7 ± 11.7 for group B. The ΔHU for group B was significantly higher than that for group A (P = 0.017). The percentage of tests demonstrating with the enhancement effect of group B was more than 80% in all subgroups; however, that of group A was less than 80% in all subgroups. The contrast-to-noise ratio of group B (7.8 ± 3.3 HU) was significantly higher compared to that of group A (6.5 ± 2.8 HU) (P < 0.05). CONCLUSIONS: Because of the volume effect, injecting a contrast medium diluted with normal saline improved the degree of hepatic and aortic contrast enhancement and achieved better visualization of liver metastases. CLINICAL IMPACT: The use of spiral flow-generating tube may help diagnostic of hepatic and aortic contrast enhancement and liver metastases. IMPORTANCE: The use of a spiral flow-generating tube improved the degree of hepatic and aortic contrast enhancement and achieve better visualization of liver metastases. POINTS: The use of low-concentration syringe formulations is limited by body weight. However, the use of spiral flow-generating tube provides low-concentration contrast medium regardless of body weight.

2.
Biochem Biophys Res Commun ; 668: 42-48, 2023 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-37244033

RESUMO

Camptothecin (CPT) exhibits strong cytotoxicity by inducing DNA double-strand breaks (DSBs) through DNA replication. Unlike radiation-induced DSBs, which have two DNA ends, CPT-induced DSBs are considered to have only one DNA end. However, the differences in cellular responses to one-ended and two-ended DSBs are not well understood. Our previous study showed that proteasome inhibitor treatment suppressed CPT-induced activation of DNA-PK, a factor required for non-homologous end-joining in DSB repair, suggesting that the ubiquitin-proteasome pathway is involved in DNA-PK activation in response to one-ended DSBs. To identify the ubiquitination factors required for DNA-PK activation, we screened an siRNA library against E2 ubiquitin-conjugating enzymes and identified UbcH5c. Knockdown of UbcH5c suppressed DNA-PK activation caused by CPT, but not by the radio-mimetic drug neocarzinostatin. UbcH5c-dependent DNA-PK activation occurred independent of DNA end resection. Furthermore, loss of UbcH5c reduced DNA-PK-dependent chromosomal aberrations and suppressed the activation of cell cycle checkpoint in response to CPT. These results suggest that UbcH5c regulates DNA-PK activation in response to one-ended DSBs caused by replication fork collapse. To our knowledge, this is the first report of a DSB repair-related factor that is differentially involved in the response to one- and two-ended DSBs.


Assuntos
Quebras de DNA de Cadeia Dupla , Proteína Quinase Ativada por DNA , Proteína Quinase Ativada por DNA/metabolismo , Replicação do DNA , DNA/metabolismo , Reparo do DNA por Junção de Extremidades , Reparo do DNA
3.
Nucleic Acids Res ; 46(6): 2932-2944, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29394375

RESUMO

During mild replication stress provoked by low dose aphidicolin (APH) treatment, the key Fanconi anemia protein FANCD2 accumulates on common fragile sites, observed as sister foci, and protects genome stability. To gain further insights into FANCD2 function and its regulatory mechanisms, we examined the genome-wide chromatin localization of FANCD2 in this setting by ChIP-seq analysis. We found that FANCD2 mostly accumulates in the central regions of a set of large transcribed genes that were extensively overlapped with known CFS. Consistent with previous studies, we found that this FANCD2 retention is R-loop-dependent. However, FANCD2 monoubiquitination and RPA foci formation were still induced in cells depleted of R-loops. Interestingly, we detected increased Proximal Ligation Assay dots between FANCD2 and R-loops following APH treatment, which was suppressed by transcriptional inhibition. Collectively, our data suggested that R-loops are required to retain FANCD2 in chromatin at the middle intronic region of large genes, while the replication stress-induced upstream events leading to the FA pathway activation are not triggered by R-loops.


Assuntos
Cromatina/genética , Sítios Frágeis do Cromossomo/genética , Replicação do DNA/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Instabilidade Genômica/genética , Afidicolina/farmacologia , Linhagem Celular Tumoral , Cromatina/metabolismo , DNA/química , DNA/genética , DNA/metabolismo , Dano ao DNA , Reparo do DNA , Replicação do DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Conformação de Ácido Nucleico , Transdução de Sinais/genética , Ubiquitinação/efeitos dos fármacos
4.
Exp Cell Res ; 362(2): 450-460, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29233683

RESUMO

X-ray repair cross-complementing group 4 (XRCC4), a repair protein for DNA double-strand breaks, is cleaved by caspases during apoptosis. In this study, we examined the role of XRCC4 in apoptosis. Cell lines, derived from XRCC4-deficient M10 mouse lymphoma cells and stably expressing wild-type XRCC4 or caspase-resistant XRCC4, were established and treated with staurosporine (STS) to induce apoptosis. In STS-induced apoptosis, expression of wild-type, but not caspase-resistant, XRCC4 in XRCC4-deficient cells enhanced oligonucleosomal DNA fragmentation and the appearance of TUNEL-positive cells by promoting nuclear translocation of caspase-activated DNase (CAD), a major nuclease for oligonucleosomal DNA fragmentation. CAD activity is reportedly regulated by the ratio of two inhibitor of CAD (ICAD) splice variants, ICAD-L and ICAD-S mRNA, which, respectively, produce proteins with and without the ability to transport CAD into the nucleus. The XRCC4-dependent promotion of nuclear import of CAD in STS-treated cells was associated with reduction of ICAD-S mRNA and protein, and enhancement of phosphorylation and nuclear import of serine/arginine-rich splicing factor (SRSF) 1. These XRCC4-dependent, apoptosis-enhancing effects were canceled by depletion of SRSF1 or SR protein kinase (SRPK) 1. In addition, overexpression of SRSF1 in XRCC4-deficient cells restored the normal level of apoptosis, suggesting that SRSF1 functions downstream of XRCC4 in activating CAD. This XRCC4-dependent, SRPK1/SRSF1-mediated regulatory mechanism was conserved in apoptosis in Jurkat human leukemia cells triggered by STS, and by two widely used anti-cancer agents, Paclitaxel and Vincristine. These data imply that the level of XRCC4 expression could be used to predict the effects of apoptosis-inducing drugs in cancer treatment.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinases/genética , Fatores de Processamento de Serina-Arginina/genética , Animais , Núcleo Celular/genética , Fragmentação do DNA/efeitos dos fármacos , Reparo do DNA/genética , Desoxirribonucleases/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Células Jurkat , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Paclitaxel/farmacologia , Transdução de Sinais/efeitos dos fármacos , Estaurosporina/farmacologia , Vincristina/farmacologia
5.
J Biol Chem ; 288(19): 13269-77, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23536184

RESUMO

BACKGROUND: It is unclear how DNA-damaging agents target cancer cells over normal somatic cells. RESULTS: Arf/p53-dependent down-regulation of H2AX enables normal cells to survive after DNA damage. CONCLUSION: Transformed cells, which harbor mutations in either Arf or p53, are more sensitive to DNA-damaging agents. SIGNIFICANCE: Cellular transformation renders cells more susceptible to some DNA-damaging agents. Anti-cancer drugs generally target cancer cells rather than normal somatic cells. However, the factors that determine this differential sensitivity are poorly understood. Here we show that Arf/p53-dependent down-regulation of H2AX induced the selective survival of normal cells after drug treatment, resulting in the preferential targeting of cancer cells. Treatment with camptothecin, a topoisomerase I inhibitor, caused normal cells to down-regulate H2AX and become quiescent, a process mediated by both Arf and p53. In contrast, transformed cells that harbor mutations in either Arf or p53 do not down-regulate H2AX and are more sensitive to drugs unless they have developed drug resistance. Such transformation-associated changes in H2AX expression rendered cancer cells more susceptible to drug-induced damage (by two orders of magnitude). Thus, the expression of H2AX and γH2AX (phosphorylated form of H2AX at Ser-139) is a critical factor that determines drug sensitivity and should be considered when administering chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regulação para Baixo , Histonas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Camptotecina/farmacologia , Forma Celular , Células Cultivadas , Senescência Celular , Cisplatino/farmacologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Dano ao DNA , Replicação do DNA/efeitos dos fármacos , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Técnicas de Silenciamento de Genes , Histonas/genética , Humanos , Hidroxiureia/farmacologia , Camundongos , Camundongos Knockout , Mutação , Fenantrenos/farmacologia , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases , Proteína Supressora de Tumor p53/genética
6.
Genes Cells ; 17(12): 962-70, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23157317

RESUMO

FANCJ, also called BACH1/BRIP1, is a 5'-3' DEAH helicase, whose mutations are known as a risk factor for Fanconi anemia and also breast and ovarian cancer. FANCJ is thought to contribute to DNA double-strand break (DSB) repair and S-phase checkpoint through binding to multiple partner proteins, such as BRCA1 and TopBP1, but its molecular regulation remains unclear. We focused on DNA damage-induced phosphorylation of FANCJ and found that reagents that cause DSB or replication fork stalling induce FANCJ hyperphosphorylation. In particular, camptothecin (CPT) induced rapid and efficient FANCJ hyperphosphorylation that was largely dependent on TopBP1 and ATM-Rad3 related (ATR) kinase. Furthermore, DNA end resection that exposes single-strand DNA at the DSB site was required for hyperphosphorylation. Interestingly, upon CPT treatment, a dramatic increase in the FANCJ-TopBP1 complex was observed, and this increase was not alleviated even when ATR-dependent hyperphosphorylation was suppressed. These results suggest that FANCJ function may be modulated by hyperphosphorylation in a DNA end resection- and ATR-dependent manner and by FANCJ-TopBP1 complex formation in response to replication-coupled DSBs.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Replicação do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Camptotecina/toxicidade , Quebras de DNA de Cadeia Simples , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases , Células HeLa , Humanos , Fosforilação/efeitos dos fármacos
7.
DNA Repair (Amst) ; 113: 103318, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35325630

RESUMO

DNA-damaging anti-cancer drugs are used clinically to induce cell death by causing DNA strand breaks or DNA replication stress. Camptothecin (CPT) and cisplatin are commonly used anti-cancer drugs, and their combined use enhances the anti-tumour effects. However, the mechanism underlying this enhanced effect has not been well studied. In this study, we analysed the combined effect of CPT and cisplatin or ultraviolet (UV) and found that CPT suppresses transcription recovery after UV damage and induces the disappearance of the Cockayne syndrome group B (CSB) protein, a transcription-coupled nucleotide excision repair (TC-NER) factor. This CPT-induced disappearance of CSB expression was suppressed by proteasome and transcription inhibitors. Moreover, CSB ubiquitination was detected after CPT treatment in a transcription-dependent manner, suggesting that the transcription stress caused by CPT induces CSB ubiquitination, resulting in CSB undetectability. However, Cockayne syndrome group A (CSA) and CUL4A were not involved in the CPT-induced CSB undetectability, suggesting that CSB ubiquitination caused by CPT is regulated differently from the UV response. However, cisplatin or UV sensitivity was enhanced by CPT even in CSB- or CSA-knockout cells. Furthermore, the excessive CSB expression, which suppressed CSB ubiquitination, did not cancel the combined effect of CPT. These results suggest that CPT blocks the repair of cisplatin or UV-induced DNA damage regardless of TC-NER status. CPT possibly compromised the alternative repair pathways other than TC-NER, leading to the suppression of transcription recovery and enhancement of cell killing.

8.
J Biol Chem ; 285(20): 15201-15208, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20304914

RESUMO

Camptothecin (CPT) is a topoisomerase I inhibitor, derivatives of which are being used for cancer chemotherapy. CPT-induced DNA double-strand breaks (DSBs) are considered a major cause of its tumoricidal activity, and it has been shown that CPT induces DNA damage signaling through the phosphatidylinositol 3-kinase-related kinases, including ATM (ataxia telangiectasia mutated), ATR (ATM and Rad3-related), and DNA-PK (DNA-dependent protein kinase). In addition, CPT causes DNA strand breaks mediated by transcription, although the downstream signaling events are less well characterized. In this study, we show that CPT-induced activation of ATM requires transcription. Mechanistically, transcription inhibition suppressed CPT-dependent activation of ATM and blocked recruitment of the DNA damage mediator p53-binding protein 1 (53BP1) to DNA damage sites, whereas ATM inhibition abrogated CPT-induced G(1)/S and S phase checkpoints. Functional inactivation of ATM resulted in DNA replication-dependent hyperactivation of DNA-PK in CPT-treated cells and dramatic CPT hypersensitivity. On the other hand, simultaneous inhibition of ATM and DNA-PK partially restored CPT resistance, suggesting that activation of DNA-PK is proapoptotic in the absence of ATM. Correspondingly, comet assay and cell cycle synchronization experiments suggested that transcription collapse occurring as the result of CPT treatment are converted to frank double-strand breaks when ATM-deficient cells bypass the G(1)/S checkpoint. Thus, ATM suppresses DNA-PK-dependent cell death in response to topoisomerase poisons, a finding with potential clinical implications.


Assuntos
Camptotecina/farmacologia , Proteínas de Ciclo Celular/metabolismo , Morte Celular/fisiologia , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Inibidores da Topoisomerase I , Transcrição Gênica , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Citometria de Fluxo , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Supressoras de Tumor/genética
9.
Genes Cells ; 15(4): 327-37, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20298437

RESUMO

Developmental pluripotency associated 4 (DPPA4) is one of the uncharacterized genes that is highly expressed in embryonic stem (ES) cells. DPPA4 is associated with active chromatin and involved in the pluripotency of mouse ES cells. However, the biological function of DPPA4 remains poorly understood. In this study, we performed fluorescence recovery after photobleaching (FRAP) analysis to examine the dynamics of DPPA4 in ES cells. FRAP analysis showed that the mobility of DPPA4 is similar to that of histone H1. In addition, biochemical analysis with purified proteins and immunoprecipitation analysis showed that DPPA4 directly binds to both DNA and core histone H3. The analysis using truncated proteins indicated that DPPA4 is associated with DNA via the N-terminal region and histone H3 via the C-terminal region. In vitro assembled chromatin showed resistance to micrococcal nuclease (MNase) digestion in the presence of DPPA4. Moreover, MNase assay and FRAP analysis with the truncated proteins implies that DPPA4 binding to both DNA and histone H3 is necessary for the chromatin structure resistant to MNase and for the proper localization of DPPA4 in ES cell nuclei. These results suggest that DPPA4 modulates the chromatin structure in association with DNA and histone H3 in ES cells.


Assuntos
Cromatina/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Histonas/metabolismo , Animais , DNA/genética , DNA/metabolismo , Histonas/química , Histonas/genética , Camundongos
10.
Mech Ageing Dev ; 196: 111492, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33862037

RESUMO

Tropomyosin (Tpm) 1 and 2 are important in the epithelial mesenchymal transition of lens epithelial cells; however, the effect of Tpm1 depletion during aging remains obscure. We analyzed the age-related changes in the crystalline lens of Tpm1- conditional knockout mice (Tpm1-CKO). Floxed alleles of Tpm1 were conditionally deleted in the lens, using Pax6-cre transgenic mice. Lenses of embryonic day (ED) 14, postnatal 1-, 11-, and 48-week-old Tpm1-CKO and wild type mice were dissected to prepare paraffin sections, which subsequently underwent histological and immunohistochemical analysis. Tpm1 and α smooth muscle actin (αSMA) mRNA expression were assessed using RT-PCR. The homozygous Tpm1-CKO (Tpm1-/-) lenses displayed a dramatic reduction in Tpm1 transcript, with no change to αSMA mRNA expression. Tpm1-/- mice had small lenses with disorganized, vesiculated fiber cells, and loss of epithelial cells. The lenses of Tpm1-/- mice had abnormal and disordered lens fiber cells with cortical and peri-nuclear liquefaction. Expression of filamentous-actin was reduced in the equator region of lenses derived from ED14, 1-, 11-, and 48-week-old Tpm1-/- mice. Therefore, Tpm1 plays an integral role in mediating the integrity and fate of lens fiber differentiation and lens homeostasis during aging. Age-related Tpm1 dysregulation or deficiency may induce cataract formation.


Assuntos
Actinas/metabolismo , Envelhecimento/fisiologia , Catarata , Senescência Celular/fisiologia , Tropomiosina/genética , Animais , Catarata/metabolismo , Catarata/patologia , Catarata/fisiopatologia , Diferenciação Celular , Transição Epitelial-Mesenquimal/fisiologia , Perfilação da Expressão Gênica , Imuno-Histoquímica , Cristalino/metabolismo , Cristalino/patologia , Camundongos , Camundongos Knockout , RNA Mensageiro
11.
Oncogenesis ; 9(6): 60, 2020 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-32541651

RESUMO

The nucleus of mammalian cells is compartmentalized by nuclear bodies such as nuclear speckles, however, involvement of nuclear bodies, especially nuclear speckles, in DNA repair has not been actively investigated. Here, our focused screen for nuclear speckle factors involved in homologous recombination (HR), which is a faithful DNA double-strand break (DSB) repair mechanism, identified transcription-related nuclear speckle factors as potential HR regulators. Among the top hits, we provide evidence showing that USP42, which is a hitherto unidentified nuclear speckles protein, promotes HR by facilitating BRCA1 recruitment to DSB sites and DNA-end resection. We further showed that USP42 localization to nuclear speckles is required for efficient HR. Furthermore, we established that USP42 interacts with DHX9, which possesses DNA-RNA helicase activity, and is required for efficient resolution of DSB-induced R-loop. In conclusion, our data propose a model in which USP42 facilitates BRCA1 loading to DSB sites, resolution of DSB-induced R-loop and preferential DSB repair by HR, indicating the importance of nuclear speckle-mediated regulation of DSB repair.

12.
Sci Rep ; 7(1): 13808, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-29061988

RESUMO

Accumulating evidence indicates that transcription is closely related to DNA damage formation and that the loss of RNA biogenesis factors causes genome instability. However, whether such factors are involved in DNA damage responses remains unclear. We focus here on the RNA helicase Aquarius (AQR), a known R-loop processing factor, and show that its depletion in human cells results in the accumulation of DNA damage during S phase, mediated by R-loop formation. We investigated the involvement of Aquarius in DNA damage responses and found that AQR knockdown decreased DNA damage-induced foci formation of Rad51 and replication protein A, suggesting that Aquarius contributes to homologous recombination (HR)-mediated repair of DNA double-strand breaks (DSBs). Interestingly, the protein level of CtIP, a DSB processing factor, was decreased in AQR-knockdown cells. Exogenous expression of Aquarius partially restored CtIP protein level; however, CtIP overproduction did not rescue defective HR in AQR-knockdown cells. In accordance with these data, Aquarius depletion sensitized cells to genotoxic agents. We propose that Aquarius contributes to the maintenance of genomic stability via regulation of HR by CtIP-dependent and -independent pathways.


Assuntos
Proteínas de Transporte/metabolismo , Quebras de DNA de Cadeia Dupla , Instabilidade Genômica , Neoplasias/genética , Proteínas Nucleares/metabolismo , RNA Helicases/metabolismo , Reparo de DNA por Recombinação , Proteínas de Transporte/genética , Endodesoxirribonucleases , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Nucleares/genética , RNA Helicases/antagonistas & inibidores , RNA Helicases/genética , Células Tumorais Cultivadas
13.
Genes Genet Syst ; 90(4): 187-94, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26616758

RESUMO

Camptothecin (CPT) inhibits DNA topoisomerase I (Top1) through a non-catalytic mechanism that stabilizes the Top1-DNA cleavage complex (Top1cc) and blocks the DNA re-ligation step, resulting in the accumulation in the genome of DNA single-strand breaks (SSBs), which are converted to secondary strand breaks when they collide with the DNA replication and RNA transcription machinery. DNA strand breaks mediated by replication, which have one DNA end, are distinct in repair from the DNA double-strand breaks (DSBs) that have two ends and are caused by ionizing radiation and other agents. In contrast to two-ended DSBs, such one-ended DSBs are preferentially repaired through the homologous recombination pathway. Conversely, the repair of one-ended DSBs by the non-homologous end-joining pathway is harmful for cells and leads to cell death. The choice of repair pathway has a crucial impact on cell fate and influences the efficacy of anticancer drugs such as CPT derivatives. In addition to replication-mediated one-ended DSBs, transcription also generates DNA strand breaks upon collision with the Top1cc. Some reports suggest that transcription-mediated DNA strand breaks correlate with neurodegenerative diseases. However, the details of the repair mechanisms of, and cellular responses to, transcription-mediated DNA strand breaks still remain unclear. In this review, combining our recent results and those of previous reports, we introduce and discuss the responses to CPT-induced DNA damage mediated by DNA replication and RNA transcription.


Assuntos
Camptotecina/farmacologia , Quebras de DNA de Cadeia Simples , Reparo do DNA , Replicação do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , Humanos , Transcrição Gênica/efeitos dos fármacos
14.
Nat Commun ; 7: 11117, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-27030108

RESUMO

Temporal regulation of microtubule dynamics is essential for proper progression of mitosis and control of microtubule plus-end tracking proteins by phosphorylation is an essential component of this regulation. Here we show that Aurora B and CDK1 phosphorylate microtubule end-binding protein 2 (EB2) at multiple sites within the amino terminus and a cluster of serine/threonine residues in the linker connecting the calponin homology and end-binding homology domains. EB2 phosphorylation, which is strictly associated with mitotic entry and progression, reduces the binding affinity of EB2 for microtubules. Expression of non-phosphorylatable EB2 induces stable kinetochore microtubule dynamics and delays formation of bipolar metaphase plates in a microtubule binding-dependent manner, and leads to aneuploidy even in unperturbed mitosis. We propose that Aurora B and CDK1 temporally regulate the binding affinity of EB2 for microtubules, thereby ensuring kinetochore microtubule dynamics, proper mitotic progression and genome stability.


Assuntos
Aurora Quinase B/fisiologia , Quinases Ciclina-Dependentes/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Mitose/fisiologia , Aurora Quinase B/análise , Aurora Quinase B/metabolismo , Sítios de Ligação , Proteína Quinase CDC2 , Linhagem Celular , Quinases Ciclina-Dependentes/análise , Quinases Ciclina-Dependentes/metabolismo , Instabilidade Genômica , Humanos , Cinetocoros/metabolismo , Cinetocoros/ultraestrutura , Proteínas Associadas aos Microtúbulos/análise , Proteínas Associadas aos Microtúbulos/química , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Mitose/genética , Fosforilação
15.
Genes Genet Syst ; 87(4): 265-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23229313

RESUMO

Topoisomerase 1 (Top1)-DNA cleavage complexes induced by camptothecin (CPT) cause DNA strand breaks during DNA replication or transcription. Although the cellular responses to replication-mediated DNA double-strand breaks have been well studied, the responses to transcription-mediated DNA strand breaks have not. Here, we show that poly (ADP-ribose) polymerase (PARP) and cockayne syndrome group B protein (CSB) modulate the CPT-induced formation of discrete p53-binding protein 1 (53BP1) nuclear foci at sites of transcription-mediated DNA strand breaks. Inhibition of PARP activity enhanced the formation of these foci, while knockdown of essential components of the base excision repair (BER) pathway did not. These findings suggest that PARP suppresses transcription-mediated 53BP1 foci formation, but that this does not occur through the BER pathway. In addition, knockdown of CSB, one of the key factors of transcription-coupled repair, slowed the kinetics of 53BP1 foci formation. These data suggest that PARP and CSB modulate the formation of 53BP1 foci during the processing of transcription-mediated DNA strand breaks.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Transcrição Gênica , Camptotecina/toxicidade , Linhagem Celular Tumoral , Reparo do DNA , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Proteínas de Ligação a Poli-ADP-Ribose , RNA Interferente Pequeno , Inibidores da Topoisomerase I/toxicidade , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
16.
DNA Repair (Amst) ; 11(3): 247-58, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22188649

RESUMO

5-Fluorouracil (5-FU) has long been a mainstay antimetabolite chemotherapeutic drug for the treatment of major solid tumors, particularly colorectal cancer. 5-FU is processed intracellularly to yield active metabolites that compromise RNA and DNA metabolism. However, the mechanisms responsible for its cytotoxicity are not fully understood. From the phenotypic analysis of mutant chicken B lymphoma DT40 cells, we found that homologous recombinational repair (HRR), involving Rad54 and BRCA2, and the ATR-Chk1 signaling pathway, involving Rad9 and Rad17, significantly contribute to 5-FU tolerance. 5-FU induced γH2AX nuclear foci, which were colocalized with the key HRR factor Rad51, but not with DNA double-strand breaks (DSBs), in a dose-dependent manner as cells accumulated in the S phase. Inhibition of Chk1 kinase by UCN-01 increased 5-FU-induced γH2AX and enhanced 5-FU cytotoxicity not only in wild-type cells but also in Rad54- or BRCA2-deficient cells, suggesting that HRR and Chk1 kinase have non-overlapping roles in 5-FU tolerance. 5-FU-induced Chk1 phosphorylation was significantly impaired in Rad9- or Rad17-deficient cells, and severe γH2AX nuclear foci and DSBs were formed, which was followed by apoptosis. Finally, inhibition of Chk1 kinase by UCN-01 increased 5-FU-induced γH2AX nuclear foci and enhanced 5-FU cytotoxicity in Rad9- or Rad17-deficient cells. These results suggest that Rad9- and Rad17-independent activation of the ATR-Chk1 signaling pathway also significantly contributes to 5-FU tolerance.


Assuntos
Núcleo Celular/metabolismo , Citoproteção/efeitos dos fármacos , Fluoruracila/farmacologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Reparo de DNA por Recombinação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Quinase 1 do Ponto de Checagem , Galinhas , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Histonas/metabolismo , Linfoma de Células B/enzimologia , Linfoma de Células B/patologia , Mutação/genética , Fosforilação/efeitos dos fármacos , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia , Fatores de Tempo
17.
DNA Repair (Amst) ; 9(1): 76-82, 2010 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-19959400

RESUMO

The ubiquitin-proteasome pathway plays an important role in DNA damage signaling and repair by facilitating the recruitment and activation of DNA repair factors and signaling proteins at sites of damaged chromatin. Proteasome activity is generally not thought to be required for activation of apical signaling kinases including the PI3K-related kinases (PIKKs) ATM, ATR, and DNA-PK that orchestrate downstream signaling cascades in response to diverse genotoxic stimuli. In a previous work, we showed that inhibition of the proteasome by MG-132 suppressed 53BP1 (p53 binding protein1) phosphorylation as well as RPA2 (replication protein A2) phosphorylation in response to the topoisomerase I (TopI) poison camptothecin (CPT). To address the mechanism of proteasome-dependent RPA2 phosphorylation, we investigated the effects of proteasome inhibitors on the upstream PIKKs. MG-132 sharply suppressed CPT-induced DNA-PKcs autophosphorylation, a marker of the activation, whereas the phosphorylation of ATM and ATR substrates was only slightly suppressed by MG-132, suggesting that DNA-PK among the PIKKs is specifically regulated by the proteasome in response to CPT. On the other hand, MG-132 did not suppress DNA-PK activation in response to UV or IR. MG-132 blocked the interaction between DNA-PKcs and Ku heterodimer enhanced by CPT, and hydroxyurea pre-treatment completely abolished CPT-induced DNA-PKcs autophosphorylation, indicating a requirement for ongoing DNA replication. CPT-induced TopI degradation occurred independent of DNA-PK activation, suggesting that DNA-PK activation does not require degradation of trapped TopI complexes. The combined results suggest that CPT-dependent replication fork collapse activates DNA-PK signaling through a proteasome dependent, TopI degradation-independent pathway. The implications of DNA-PK activation in the context of TopI poison-based therapies are discussed.


Assuntos
Camptotecina/farmacologia , Proteína Quinase Ativada por DNA/metabolismo , Leupeptinas/farmacologia , Inibidores de Proteases/farmacologia , Linhagem Celular Tumoral , Quinases Ciclina-Dependentes/metabolismo , Replicação do DNA/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Inibidores de Proteassoma , Proteína de Replicação A/metabolismo
18.
J Biol Chem ; 284(12): 8083-92, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19112176

RESUMO

TDP-43 (43-kDa TAR DNA-binding domain protein) is a major constituent of ubiquitin-positive cytoplasmic aggregates present in neurons of patients with fronto-temporal lobular dementia and amyotrophic lateral sclerosis (ALS). The pathologic significance of TDP-43 aggregation is not known; however, dominant mutations in TDP-43 cause a subset of ALS cases, suggesting that misfolding and/or altered trafficking of TDP-43 is relevant to the disease process. Here, we show that the presenilin-binding protein ubiquilin 1 (UBQLN) plays a role in TDP-43 aggregation. TDP-43 interacted with UBQLN both in yeast and in vitro, and the carboxyl-terminal ubiquitin-associated domain of UBQLN was both necessary and sufficient for binding to polyubiquitylated forms of TDP-43. Overexpression of UBQLN recruited TDP-43 to detergent-resistant cytoplasmic aggregates that colocalized with the autophagosomal marker, LC3. UBQLN-dependent aggregation required the UBQLN UBA domain, was mediated by non-overlapping regions of TDP-43, and was abrogated by a mutation in UBQLN previously linked to Alzheimer disease. Four ALS-associated alleles of TDP-43 also coaggregated with UBQLN, and the extent of aggregation correlated with in vitro UBQLN binding affinity. Our findings suggest that UBQLN is a polyubiquitin-TDP-43 cochaperone that mediates the autophagosomal delivery and/or proteasome targeting of TDP-43 aggregates.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Chaperonas Moleculares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Proteínas Adaptadoras de Transdução de Sinal , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/genética , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Chaperonas Moleculares/genética , Mutação , Complexo de Endopeptidases do Proteassoma/genética , Estrutura Terciária de Proteína/genética , Ubiquitinação/genética
19.
J Biol Chem ; 283(20): 13549-55, 2008 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-18337245

RESUMO

The DNA damage surveillance network orchestrates cellular responses to DNA damage through the recruitment of DNA damage-signaling molecules to DNA damage sites and the concomitant activation of protein phosphorylation cascades controlled by the ATM (ataxia-telangiectasia-mutated) and ATR (ATM-Rad3-related) kinases. Activation of ATM/ATR triggers cell cycle checkpoint activation and adaptive responses to DNA damage. Recent studies suggest that protein ubiquitylation or degradation plays an important role in the DNA damage response. In this study, we examined the potential role of the proteasome in checkpoint activation and ATM/ATR signaling in response to UV light-induced DNA damage. HeLa cells treated with the proteasome inhibitor MG-132 showed delayed phosphorylation of ATM substrates in response to UV light. UV light-induced phosphorylation of 53BP1, as well as its recruitment to DNA damage foci, was strongly suppressed by proteasome inhibition, whereas the recruitment of upstream regulators of 53BP1, including MDC1 and H2AX, was unaffected. The ubiquitin-protein isopeptide ligase RNF8 was critical for 53BP1 focus targeting and phosphorylation in ionizing radiation-damaged cells, whereas UV light-induced 53BP1 phosphorylation and targeting exhibited partial dependence on RNF8 and the ubiquitin-conjugating enzyme UBC13. Suppression of RNF8 or UBC13 also led to subtle defects in UV light-induced G2/M checkpoint activation. These findings are consistent with a model in which RNF8 ubiquitylation pathways are essential for 53BP1 regulation in response to ionizing radiation, whereas RNF8-independent pathways contribute to 53BP1 targeting and phosphorylation in response to UV light and potentially other forms of DNA replication stress.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular , Replicação do DNA , Histonas/química , Humanos , Leupeptinas/farmacologia , Modelos Biológicos , Proteínas Nucleares/química , Fosforilação , Inibidores de Proteassoma , Transdução de Sinais , Transativadores/química , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina/química , Ubiquitina-Proteína Ligases , Raios Ultravioleta
20.
Genes Cells ; 11(3): 237-46, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16483312

RESUMO

Replication protein A2 (RPA2), a component of the RPA heterotrimer, is hyperphosphorylated and forms nuclear foci in response to camptothecin (CPT) that directly induces replication-mediated DNA double-strand breaks (DSBs). Ataxia-telangiectasia mutated and Rad3-related kinase (ATR) and DNA-dependent protein kinase (DNA-PK) are activated by CPT, and RPA2 is hyperphosphorylated in a DNA-PK-dependent manner. To distinguish the roles of phosphatidylinositol 3-kinase-related protein kinases including DNA-PK, ataxia-telangiectasia mutated (ATM), and ATR, in the response to replication-mediated DSBs, we analyzed RPA2 focus formation and hyperphosphorylation during exposure to CPT. ATR knock-down with siRNA suppressed CPT-induced RPA2 hyperphosphorylation and focus formation. CPT-induced RPA2 focus formation was normally observed in DNA-PK- or ATM-deficient cells. Comparison between CPT and hydroxyurea (HU) indirectly inducing DSBs showed that RPA2 hyperphosphorylation is DNA-PK-dependent in CPT-treated cells and DNA-PK-independent in HU-treated cells. Although RPA2 foci rapidly formed in response to HU and CPT, the RPA2 hyperphosphorylation in HU-treated cells occurred later than in the CPT-treated cells, indicating that the DNA-PK dependency of RPA2 hyperphosphorylation is likely to be related to the mode of DSB induction. These results suggest that DNA-PK is responsible for the RPA2 hyperphosphorylation following ATR-dependent RPA2 focus formation in response to replication-mediated DSBs directly induced by CPT.


Assuntos
Dano ao DNA , Replicação do DNA , DNA/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína de Replicação A/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Camptotecina/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Hidroxiureia/farmacologia , Rim/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/farmacologia , Proteína de Replicação A/genética , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa