Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Biol Pharm Bull ; 43(10): 1595-1599, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32727970

RESUMEN

Calreticulin (CRT) and calnexin (CNX), homologous major chaperones in the endoplasmic reticulum (ER), are known to translocate to the cell surface in response to chemotherapeutic agents, such as mitoxantrone (MIT), and cellular stresses, including apoptosis. Cell surface CRT (ecto-CRT) is relevant to the phagocytic uptake of cancer cells and dying cells, and pre-apoptotic exposure of CRT has been reported to result in enhanced immunogenicity of dying tumor cells, serving as a damage-associated molecular pattern (DAMP). In this study, HT-29 cells were treated with MIT to induce ER stress, and ecto-CRT and cell surface CNX were quantified by flow cytometry in the absence or presence of caspase inhibitors, a calpain inhibitor, or a scavenger of reactive oxygen species. The biphasic (early transient and late sustained) increase of ecto-CRT on HT-29 cells was observed after treatment with MIT. We confirmed that the early increase in ecto-CRT after 4 h of MIT treatment was not related to apoptosis, whereas the increase of ecto-CRT, as well as that of cell-surface CNX, during the later stage of treatment was caspase dependent and related to apoptosis. In addition, our results suggested that the early peak of ecto-CRT was mediated by activation of caspase 8 by ER stress. Thus, the physiological significance of the late increases in cell-surface CRT and/or CNX might be considered an "eat-me signal" for the removal of dead cells by phagocytosis, while the early increase in ecto-CRT caused by ER stress might enhance the immunogenicity of stressed tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Calreticulina/metabolismo , Membrana Celular/metabolismo , Mitoxantrona/farmacología , Neoplasias/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Calnexina/análisis , Calnexina/metabolismo , Calreticulina/análisis , Membrana Celular/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/inmunología , Células HT29 , Humanos , Mitoxantrona/uso terapéutico , Neoplasias/inmunología , Neoplasias/patología , Fagocitosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
2.
Biochem Biophys Res Commun ; 490(4): 1375-1380, 2017 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-28694193

RESUMEN

The initiation of DNA replication is strictly regulated by multiple mechanisms to ensure precise duplication of chromosomes. In higher eukaryotes, activity of the Cdt1 protein is temporally regulated during the cell cycle, and deregulation of Cdt1 induces DNA re-replication. In previous studies, we showed that excess Cdt1 inhibits DNA replication by suppressing progression of replication forks in Xenopus egg extracts. Here, we investigated the functional regions of Cdt1 that are required for the inhibition of DNA replication. We constructed a series of N-terminally or C-terminally deleted mutants of Cdt1 and examined their inhibitory effects on DNA replication in Xenopus egg extracts. Our results showed that the region spanning amino acids (a. a.) 255-620 is required for efficient inhibition of DNA replication, and that, within this region, a. a. 255-289 have a critical role in inhibition. Moreover, one of the Cdt1 mutants, Cdt1 R285A, was compromised with respect to the licensing activity but still inhibited DNA replication. This result suggests that Cdt1 has an unforeseen function in the negative regulation of DNA replication, and that this function is located within a molecular region that is distinct from those required for the licensing activity.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cromatina/química , Replicación del ADN , Proteínas de Unión al ADN/genética , Geminina/genética , Óvulo/química , Proteínas de Xenopus/genética , Xenopus laevis/genética , Animales , Sitios de Unión , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/química , Núcleo Celular/metabolismo , Cromatina/metabolismo , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Geminina/metabolismo , Expresión Génica , Masculino , Mutación , Óvulo/citología , Óvulo/metabolismo , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espermatozoides/química , Espermatozoides/citología , Espermatozoides/metabolismo , Relación Estructura-Actividad , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
3.
Biochem Biophys Res Commun ; 470(2): 405-410, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26773501

RESUMEN

Cdt1 is a protein essential for initiation of DNA replication; it recruits MCM helicase, a core component of the replicative DNA helicase, onto replication origins. In our previous study, we showed that addition of excess Cdt1 inhibits nascent strand elongation during DNA replication in Xenopus egg extracts. In the present study, we investigated the mechanism behind the inhibitory effect of Cdt1. We found that addition of recombinant Cdt1 inhibited nascent DNA synthesis in a reinitiation-independent manner. To identify the mechanism by which Cdt1 inhibits nascent strand elongation, the effect of Cdt1 on loading of Mcm4 and Rpa70 onto chromatin was examined. The results showed that Cdt1 suppressed the excessive Rpa70 binding caused by extensive, aphidicolin-induced DNA unwinding; this unwinding occurs between stalled DNA polymerases and advancing replication forks. These findings suggested that excess Cdt1 suppressed the progression of replication forks.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN/genética , Óvulo/fisiología , Elongación de la Transcripción Genética/fisiología , Animales , Xenopus laevis
4.
J Biol Chem ; 289(16): 11374-11384, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24573676

RESUMEN

The replication fork temporarily stalls when encountering an obstacle on the DNA, and replication resumes after the barrier is removed. Simultaneously, activation of the replication checkpoint delays the progression of S phase and inhibits late origin firing. Camptothecin (CPT), a topoisomerase I (Top1) inhibitor, acts as a DNA replication barrier by inducing the covalent retention of Top1 on DNA. The Timeless-Tipin complex, a component of the replication fork machinery, plays a role in replication checkpoint activation and stabilization of the replication fork. However, the role of the Timeless-Tipin complex in overcoming the CPT-induced replication block remains elusive. Here, we generated viable TIPIN gene knock-out (KO) DT40 cells showing delayed S phase progression and increased cell death. TIPIN KO cells were hypersensitive to CPT. However, homologous recombination and replication checkpoint were activated normally, whereas DNA synthesis activity was markedly decreased in CPT-treated TIPIN KO cells. Proteasome-dependent degradation of chromatin-bound Top1 was induced in TIPIN KO cells upon CPT treatment, and pretreatment with aphidicolin, a DNA polymerase inhibitor, suppressed both CPT sensitivity and Top1 degradation. Taken together, our data indicate that replication forks formed without Tipin may collide at a high rate with Top1 retained on DNA by CPT treatment, leading to CPT hypersensitivity and Top1 degradation in TIPIN KO cells.


Asunto(s)
Proteínas Aviares/metabolismo , Camptotecina/farmacología , Replicación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas Nucleares/metabolismo , Inhibidores de Topoisomerasa I/farmacología , Animales , Proteínas Aviares/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular , Pollos , ADN/biosíntesis , ADN/genética , Replicación del ADN/fisiología , ADN-Topoisomerasas de Tipo I/genética , Técnicas de Silenciamiento del Gen , Proteínas Nucleares/genética , Proteolisis/efectos de los fármacos , Fase S/efectos de los fármacos , Fase S/fisiología
5.
Biochem Biophys Res Commun ; 452(1): 48-52, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25139235

RESUMEN

WRNIP1 (WRN-interacting protein 1) was first identified as a factor that interacts with WRN, the protein that is defective in Werner syndrome (WS). WRNIP1 associates with DNA polymerase η (Polη), but the biological significance of this interaction remains unknown. In this study, we analyzed the functional interaction between WRNIP1 and Polη by generating knockouts of both genes in DT40 chicken cells. Disruption of WRNIP1 in Polη-disrupted (POLH(-/-)) cells suppressed the phenotypes associated with the loss of Polη: sensitivity to ultraviolet light (UV), delayed repair of cyclobutane pyrimidine dimers (CPD), elevated frequency of mutation, elevated levels of UV-induced sister chromatid exchange (SCE), and reduced rate of fork progression after UV irradiation. These results suggest that WRNIP1 functions upstream of Polη in the response to UV irradiation.


Asunto(s)
Proteínas Portadoras/fisiología , Daño del ADN , Proteínas de Unión al ADN/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , ADN/efectos de la radiación , Rayos Ultravioleta , ATPasas Asociadas con Actividades Celulares Diversas , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Intercambio de Cromátides Hermanas
6.
J Biol Chem ; 286(35): 30504-30512, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21757688

RESUMEN

Ordered nucleosome disassembly and reassembly are required for eukaryotic DNA replication. The facilitates chromatin transcription (FACT) complex, a histone chaperone comprising Spt16 and SSRP1, is involved in DNA replication as well as transcription. FACT associates with the MCM helicase, which is involved in DNA replication initiation and elongation. Although the FACT-MCM complex is reported to regulate DNA replication initiation, its functional role in DNA replication elongation remains elusive. To elucidate the functional role of FACT in replication fork progression during DNA elongation in the cells, we generated and analyzed conditional SSRP1 gene knock-out chicken (Gallus gallus) DT40 cells. SSRP1-depleted cells ceased to grow and exhibited a delay in S-phase cell cycle progression, although SSRP1 depletion did not affect the level of chromatin-bound DNA polymerase α or nucleosome reassembly on daughter strands. The tracking length of newly synthesized DNA, but not origin firing, was reduced in SSRP1-depleted cells, suggesting that the S-phase cell cycle delay is mainly due to the inhibition of replication fork progression rather than to defects in the initiation of DNA replication in these cells. We discuss the mechanisms of how FACT promotes replication fork progression in the cells.


Asunto(s)
Cromatina/química , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Chaperonas de Histonas/química , Transcripción Genética , Factores de Elongación Transcripcional/metabolismo , Animales , Ciclo Celular , Pollos , Epigénesis Genética , Citometría de Flujo/métodos , Histonas/química , Humanos , Chaperonas Moleculares/metabolismo , Nucleosomas/metabolismo , Fase S
7.
Biochim Biophys Acta ; 1813(3): 473-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21256165

RESUMEN

Rothmund-Thomson syndrome (RTS) is a rare genetic disorder characterized by premature aging, developmental abnormalities, and a predisposition to cancer. RTS is caused by mutations in the RECQL4 gene, which encodes one of the five human RecQ helicases. To identify the cellular functions of RECQL4, we generated a chicken DT40 cell line in which RECQL4 expression could be turned off by doxycycline (Dox). Upon exposure to Dox, cells stopped growing and underwent apoptosis. The cells could be rescued by expression of the N-terminal region of RECQL4 (amino acids 1-496), which lacks the helicase domain and has sequence similarity to yeast Sld2, which plays an essential function in the initiation of DNA replication in Saccharomyces cerevisiae. Smaller fragments of the N-terminal region of RECQL4 did not rescue the cells from lethality. RECQL4 gene knockout cells complemented with RECQL4 (1-496) showed relatively high sensitivity to DNA damaging agents that induce double strand breaks and cross-links, suggesting that the C-terminal region including the helicase domain of RECQL4 is involved in the repair of certain types of DNA lesions.


Asunto(s)
Supervivencia Celular , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Síndrome Rothmund-Thomson/genética , Animales , Antibacterianos/farmacología , Muerte Celular , Línea Celular , Pollos , ADN Helicasas/metabolismo , Reparación del ADN , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Estructura Terciaria de Proteína , RecQ Helicasas/química , Síndrome Rothmund-Thomson/metabolismo
8.
Biochim Biophys Acta ; 1813(6): 1129-36, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21232560

RESUMEN

The facilitates chromatin transcription (FACT) complex affects nuclear DNA transactions in a chromatin context. Though the involvement of FACT in eukaryotic DNA replication has been revealed, a clear understanding of its biochemical behavior during DNA replication still remains elusive. Here, we analyzed the chromatin-binding dynamics of FACT using Xenopus egg extract cell-free system. We found that FACT has at least two distinct chromatin-binding phases: (1) a rapid chromatin-binding phase at the onset of DNA replication that did not involve origin licensing and (2) a second phase of chromatin binding that initiated after origin licensing. Intriguingly, early-binding FACT dissociated from chromatin when DNA replication was blocked by the addition of Cdc6 in the licensed state before origin firing. Cdc6-induced removal of FACT was blocked by the inhibition of origin licensing with geminin, but not by suppressing the activity of DNA polymerases, CDK, or Cdc7. Furthermore, chromatin transfer experiments revealed that impairing the later binding of FACT severely compromises DNA replication activity. Taken together, we propose that even though FACT has rapid chromatin-binding activity, the binding pattern of FACT on chromatin changes after origin licensing, which may contribute to the establishment of its functional link to the DNA replication machinery.


Asunto(s)
Cromatina/metabolismo , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Factores de Elongación Transcripcional/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Proteínas de Unión al ADN/genética , Células Eucariotas/metabolismo , Femenino , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Humanos , Immunoblotting , Cinética , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/metabolismo , Unión Proteica , Espermatozoides/metabolismo , Factores de Tiempo , Factores de Elongación Transcripcional/genética , Xenopus laevis
9.
Biochem Biophys Res Commun ; 427(3): 682-6, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23036200

RESUMEN

Saccharomyces cerevisiae RecQ helicase (Sgs1) combines with DNA topoisomerase III (Top3) and RecQ-mediated genome instability 1 (Rmi1) to form an evolutionarily conserved complex that is required for processing homologous recombination intermediates and restarting collapsed replication forks. It was previously reported that Rmi1 contributes to sister chromatid cohesion; however, the underlying molecular mechanism has been unclear. In the present study, Rmi1 was found to be enriched at the region close to an early-firing replication origin when replication forks were arrested near their origins in the presence of hydroxyurea. Genetic analyses revealed that Rmi1 promoted sister chromatid cohesion in a process that was distinct from both the cohesion establishment pathway involving Ctf4, Csm3, and Chl1 and the pathway involving the acetylation of Smc3. On the other hand, Rmi1 seemed to function in a pathway involving the Ctf18-RFC complex and Mrc1, which were previously predicted to regulate leading-strand DNA replication.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Recombinación Homóloga , Proteína de Replicación C/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Acetilación , Acetiltransferasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Replicación del ADN , Proteínas de Unión al ADN/genética , Proteínas Nucleares/metabolismo , Conformación Proteica , RecQ Helicasas/metabolismo , Fase S , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal
10.
Genes Cells ; 16(10): 1050-62, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21895891

RESUMEN

The nucleosome, which is composed of DNA wrapped around a histone octamer, is a fundamental unit of chromatin and is duplicated during the eukaryotic DNA replication process. The evolutionarily conserved histone chaperone cell cycle gene 1 (CCG1) interacting factor A/anti-silencing function 1 (CIA/Asf1) is involved in histone transfer and nucleosome reassembly during DNA replication. CIA/Asf1 has been reported to split the histone (H3-H4)(2) tetramer into histone H3-H4 dimer(s) in vitro, raising a possibility that, in DNA replication, CIA/Asf1 is involved in nucleosome disassembly and the promotion of semi-conservative histone H3-H4 dimer deposition onto each daughter strand in vivo. Despite numerous studies on the functional roles of CIA/Asf1, its mechanistic role(s) remains elusive because of lack of biochemical analyses. The biochemical studies described here show that a V94R CIA/Asf1 mutant, which lacks histone (H3-H4)(2) tetramer splitting activity, does not form efficiently a quaternary complex with histones H3-H4 and the minichromosome maintenance 2 (Mcm2) subunit of the Mcm2-7 replicative DNA helicase. Interestingly, the mutant enhances nascent DNA strand synthesis in a cell-free chromosomal DNA replication system using Xenopus egg extracts. These results suggest that CIA/Asf1 in the CIA/Asf1-H3-H4-Mcm2 complex, which is considered to be an intermediate in histone transfer during DNA replication, negatively regulates the progression of the replication fork.


Asunto(s)
Replicación del ADN/fisiología , Chaperonas de Histonas/metabolismo , Nucleosomas/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Chaperonas de Histonas/genética , Histonas/metabolismo , Cinética , Modelos Moleculares , Mutación/genética , Unión Proteica , Xenopus
11.
J Cell Biol ; 179(1): 53-63, 2007 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-17923529

RESUMEN

Bloom's syndrome (BS), which is caused by mutations in the BLM gene, is characterized by a predisposition to a wide variety of cancers. BS cells exhibit elevated frequencies of sister chromatid exchanges (SCEs), interchanges between homologous chromosomes (mitotic chiasmata), and sensitivity to several DNA-damaging agents. To address the mechanism that confers these phenotypes in BS cells, we characterize a series of double and triple mutants with mutations in BLM and in other genes involved in repair pathways. We found that XRCC3 activity generates substrates that cause the elevated SCE in blm cells and that BLM with DNA topoisomerase IIIalpha suppresses the formation of SCE. In addition, XRCC3 activity also generates the ultraviolet (UV)- and methyl methanesulfonate (MMS)-induced mitotic chiasmata. Moreover, disruption of XRCC3 suppresses MMS and UV sensitivity and the MMS- and UV-induced chromosomal aberrations of blm cells, indicating that BLM acts downstream of XRCC3.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Síndrome de Bloom/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/fisiología , Animales , Síndrome de Bloom/genética , Línea Celular , Pollos , Aberraciones Cromosómicas , ADN Helicasas/genética , ADN Helicasas/fisiología , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Predisposición Genética a la Enfermedad , Humanos , Mutación , RecQ Helicasas , Recombinación Genética , Intercambio de Cromátides Hermanas , Rayos Ultravioleta
12.
Nucleic Acids Res ; 38(16): 5409-18, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20421204

RESUMEN

Mcm2-7 is recruited to eukaryotic origins of DNA replication by origin recognition complex, Cdc6 and Cdt1 thereby licensing the origins. Cdc6 is essential for origin licensing during DNA replication and is readily destabilized from chromatin after Mcm2-7 loading. Here, we show that after origin licensing, deregulation of Cdc6 suppresses DNA replication in Xenopus egg extracts without the involvement of ATM/ATR-dependent checkpoint pathways. DNA replication is arrested specifically after chromatin binding of Cdc7, but before Cdk2-dependent pathways and deregulating Cdc6 after this step does not impair activation of origin firing or elongation. Detailed analyses revealed that Cdc6 deregulation leads to strong suppression of Cdc7-mediated hyperphosphorylation of Mcm4 and subsequent chromatin loading of Cdc45, Sld5 and DNA polymerase α. Mcm2 phosphorylation is also repressed although to a lesser extent. Remarkably, Cdc6 itself does not directly inhibit Cdc7 kinase activity towards Mcm2-4-6-7 in purified systems, rather modulates Mcm2-7 phosphorylation on chromatin context. Taken together, we propose that Cdc6 on chromatin acts as a modulator of Cdc7-mediated phosphorylation of Mcm2-7, and thus destabilization of Cdc6 from chromatin after licensing is a key event ensuring proper transition to the initiation of DNA replication.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Replicación del ADN , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas de Xenopus/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Cromatina/enzimología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Componente 4 del Complejo de Mantenimiento de Minicromosoma , Componente 6 del Complejo de Mantenimiento de Minicromosoma , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Proteínas Nucleares/metabolismo , Complejo de Reconocimiento del Origen/metabolismo , Óvulo/enzimología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Xenopus laevis
13.
Biochem Biophys Rep ; 29: 101222, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35146135

RESUMEN

Calreticulin (CRT), a chaperone typically located in the endoplasmic reticulum (ER), is known to translocate to the cell surface in response to anticancer drugs. Cell surface CRT (ecto-CRT) on apoptotic or pre-apoptotic cells serves as an "eat me" signal that can promote phagocytosis. In this study, we observed the biphasic (early transient and late sustained) increase of ecto-CRT on HT-29 cells after treatment with oxaliplatin (L-OHP). To investigate the role of ecto-CRT that accumulates in the early and late phases as "eat me" signals, we examined the phagocytosis of HT-29 cells by macrophage-like cells and dendritic cell (DC) -like cells prepared from THP-1 cells. The results indicated that the early ecto-CRT-expressed cells were phagocytosed by immature DC-like cells, and the late ecto-CRT-expressed cells were phagocytosed primarily by macrophage-like cells, while mature DC-like cells did not respond to the either class of ecto-CRT-expressed cells. Both types of phagocytotic events were inhibited by CRT Blocking Peptide, suggesting that such events depended on the ecto-CRT. Our results suggested that the early increase of ecto-CRT is related to phagocytosis as part of immunogenic cell death (ICD), while the late increase of ecto-CRT is related to the removal of apoptotic cells by macrophages.

14.
Biochem Biophys Res Commun ; 414(2): 298-303, 2011 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-21945441

RESUMEN

Claspin was originally identified as a Check1 (Chk1)-interacting protein. Claspin and Rad17 are reportedly involved in the DNA damage-induced phosphorylation of Chk1, a hallmark of checkpoint activation. To understand the cellular functions of Claspin and the functional relationship between Claspin and Rad17, we generated Claspin(-/-) and Claspin(-/-)/RAD17(-) cells using chicken DT40 cells, which contain an exogenously introduced Claspin that can be suppressed by the addition of doxycycline (Dox). In the presence of Dox, Claspin(-/-) cells ceased growth within 2 days, leading to cell death. In addition, a remarkable reduction in the rate of DNA elongation was observed in Claspin-depleted cells, suggesting that Claspin plays a critical role in DNA replication in the absence of exogenous stress. When cells were exposed to methyl methanesulfonate (MMS), a DNA damaging agent, RAD17(-) cells showed a greater defect in checkpoint activation than Claspin(-/-) cells as monitored by progression of cell cycle and phosphorylation of Chk1. Knocking out RAD17 gene showed almost no additive effects on cell death and DNA elongation rates in Claspin-depleted cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Pollos , Replicación del ADN/genética , Técnicas de Inactivación de Genes , Humanos
15.
Biol Pharm Bull ; 34(8): 1314-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21804224

RESUMEN

Werner interacting protein 1 (WRNIP1) that is highly conserved from Escherichia coli to human was originally identified as a protein that interacts with the Werner syndrome responsible gene product (WRN). Here, human WRNIP1 and WRN are shown to bind to template-primer DNA, and WRNIP1, but not WRN, requires ATP for DNA binding. Under conditions of a limiting amount of WRN, WRNIP1 facilitated binding of WRN to DNA in a dose-dependent manner. However, WRNIP1 did not stimulate the DNA helicase activity of WRN, and WRN displaced pre-bound WRNIP1 from DNA. Functional relationships between WRNIP1 and WRN will be discussed.


Asunto(s)
Proteínas Portadoras/metabolismo , Cartilla de ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , RecQ Helicasas/metabolismo , Síndrome de Werner/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfato/metabolismo , Humanos , Unión Proteica , Síndrome de Werner/metabolismo , Helicasa del Síndrome de Werner
16.
Gene ; 787: 145647, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-33845136

RESUMEN

RecQ4, a member of the RecQ helicase family, is required for the maintenance of genome integrity. RecQ4 has been shown to promote the following two DNA double-strand break (DSB) repair pathways: non-homologous end joining (NHEJ) and homologous recombination (HR). However, its molecular function has not been fully elucidated. In the present study, we aimed to investigate the role of RecQ4 in NHEJ using Xenopus egg extracts. The N-terminal 598 amino acid region of Xenopus RecQ4 (N598), which lacks a central helicase domain and a downstream C-terminal region, was added to the extracts and its effect on the joining of DNA ends was analyzed. We found that N598 inhibited the joining of linearized DNA ends in the extracts. In addition, N598 inhibited DSB-induced chromatin binding of Ku70, which is essential for NHEJ, while the DSB-induced chromatin binding of the HR-associated proteins, replication protein A (RPA) and Rad51, increased upon the addition of N598. These results suggest that RecQ4 possibly influences the choice of the DSB repair pathway by influencing the association of the Ku heterodimer with the DNA ends.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Autoantígeno Ku/metabolismo , RecQ Helicasas/metabolismo , Proteínas de Xenopus/fisiología , Animales , Cromatina , ADN/metabolismo , Autoantígeno Ku/antagonistas & inhibidores , Unión Proteica , RecQ Helicasas/genética , Xenopus laevis
17.
DNA Repair (Amst) ; 8(6): 690-6, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19261547

RESUMEN

Checkpoints are cellular surveillance and signaling pathways that regulate responses to DNA damage and perturbations of DNA replication. Here we show that high levels of sumoylated Rad52 are present in the mec1 sml1 and rad53 sml1 checkpoint mutants exposed to DNA-damaging agents such as methyl methanesulfonate (MMS) or the DNA replication inhibitor hydroxyurea (HU). The kinase-defective mutant rad53-K227A also showed high levels of Rad52 sumoylation. Elevated levels of Rad52 sumoylation occur in checkpoint mutants proceeding S phase being exposed DNA-damaging agent. Interestingly, chromatin immunoprecipitation (ChIP) on chip analyses revealed non-canonical chromosomal localization of Rad52 in the HU-treated rad53-K227A cells arrested in early S phase: Rad52 localization at dormant and early DNA replication origins. However, such unusual localization was not dependent on the sumoylation of Rad52. In addition, we also found that Rad52 could be highly sumoylated in the absence of Rad51. Double mutation of RAD51 and RAD53 exhibited the similar levels of Rad52 sumoylation to RAD53 single mutation. The significance and regulation mechanism of Rad52 sumoylation by checkpoint pathways will be discussed.


Asunto(s)
Replicación del ADN , ADN Bacteriano/genética , Mutación/genética , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2 , Inmunoprecipitación de Cromatina , ADN de Hongos , Citometría de Flujo , Immunoblotting , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
18.
Nature ; 430(7002): 913-7, 2004 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-15286659

RESUMEN

To maintain chromosome stability in eukaryotic cells, replication origins must be licensed by loading mini-chromosome maintenance (MCM2-7) complexes once and only once per cell cycle. This licensing control is achieved through the activities of geminin and cyclin-dependent kinases. Geminin binds tightly to Cdt1, an essential component of the replication licensing system, and prevents the inappropriate reinitiation of replication on an already fired origin. The inhibitory effect of geminin is thought to prevent the interaction between Cdt1 and the MCM helicase. Here we describe the crystal structure of the mouse geminin-Cdt1 complex using tGeminin (residues 79-157, truncated geminin) and tCdt1 (residues 172-368, truncated Cdt1). The amino-terminal region of a coiled-coil dimer of tGeminin interacts with both N-terminal and carboxy-terminal parts of tCdt1. The primary interface relies on the steric complementarity between the tGeminin dimer and the hydrophobic face of the two short N-terminal helices of tCdt1 and, in particular, Pro 181, Ala 182, Tyr 183, Phe 186 and Leu 189. The crystal structure, in conjunction with our biochemical data, indicates that the N-terminal region of tGeminin might be required to anchor tCdt1, and the C-terminal region of tGeminin prevents access of the MCM complex to tCdt1 through steric hindrance.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Animales , Sitios de Unión , Cristalografía por Rayos X , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Dimerización , Geminina , Ratones , Proteína 1 de Mantenimiento de Minicromosoma/metabolismo , Modelos Moleculares , Proteínas Nucleares , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Relación Estructura-Actividad , Xenopus , Proteínas de Xenopus
19.
Bioessays ; 30(3): 208-11, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18293358

RESUMEN

Although initiation of DNA replication is considered to be highly coordinated through multiple protein-DNA and protein-protein interactions, it is poorly understood how particular locations within the eukaryotic chromosome are selected as origins of DNA replication. Here, we discuss recent reports that present structural information on the interaction characteristics of the archaeal orthologues of the eukaryotic origin recognition complex with their cognate binding sequences. Since the archaeal replication system is postulated as a simplified version of the one in eukaryotes, by analogy, these works provide insights into the functions of the eukaryotic initiator proteins.


Asunto(s)
Replicación del ADN , ADN/metabolismo , Complejo de Reconocimiento del Origen , Proteínas Arqueales/metabolismo , Proteínas Bacterianas/metabolismo , Mapeo Cromosómico , Cromosomas , Escherichia coli/metabolismo , Modelos Biológicos , Conformación de Ácido Nucleico , Estructura Terciaria de Proteína , Origen de Réplica
20.
Biochim Biophys Acta ; 1782(2): 75-81, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18078829

RESUMEN

RECQL1 and RECQL5 as well as BLM reportedly interact with TOP3alpha whose defect is lethal for the cell. Therefore in this study, we characterized recql5/recql1/blm triple mutants from DT40 cells to determine whether the triple mutants show a top3alpha disrupted cell-like phenotype. The triple mutants are viable. Moreover, both blm/recql1 and recql5/blm cells, and recql5/recql1/blm cells grew slightly slower than blm cells, that is, triple mutant cells grew almost the same rate as either of the double mutant cells. The blm cells showed sensitivity to methyl methanesulfonate (MMS) and ultraviolet light (UV), about a 10-fold increase in sister chromatid exchange (SCE), and about a 3-fold increase in damage-induced mitotic chiasma compared to wild-type cells. The triple mutants showed the same sensitivity to MMS or UV and the same frequency of damage-induced mitotic chiasma compared to those of blm cells, indicating that unlike BLM, RECQL1 and RECQL5 play a little role in the repair of or tolerance to DNA damages. However, recql5/blm cells showed higher frequency of SCE than blm cells, whereas the RECQL1 gene disruption had no effect on SCE in blm cells and even in recql5/blm cells.


Asunto(s)
Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/fisiología , ADN Helicasas/genética , ADN Helicasas/fisiología , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo I/fisiología , RecQ Helicasas/genética , RecQ Helicasas/fisiología , Adenosina Trifosfatasas/metabolismo , Animales , Células Cultivadas , Pollos , Daño del ADN/fisiología , ADN Helicasas/metabolismo , Metilmetanosulfonato/farmacología , Modelos Biológicos , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , RecQ Helicasas/metabolismo , Intercambio de Cromátides Hermanas/efectos de la radiación , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA