Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Biol Chem ; 288(7): 4649-58, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23283980

RESUMEN

DNA-protein cross-links (DPCs) are formed when cells are exposed to various DNA-damaging agents. Because DPCs are extremely large, steric hindrance conferred by DPCs is likely to affect many aspects of DNA transactions. In DNA replication, DPCs are first encountered by the replicative helicase that moves at the head of the replisome. However, little is known about how replicative helicases respond to covalently immobilized protein roadblocks. In the present study we elucidated the effect of DPCs on the DNA unwinding reaction of hexameric replicative helicases in vitro using defined DPC substrates. DPCs on the translocating strand but not on the nontranslocating strand impeded the progression of the helicases including the phage T7 gene 4 protein, simian virus 40 large T antigen, Escherichia coli DnaB protein, and human minichromosome maintenance Mcm467 subcomplex. The impediment varied with the size of the cross-linked proteins, with a threshold size for clearance of 5.0-14.1 kDa. These results indicate that the central channel of the dynamically translocating hexameric ring helicases can accommodate only small proteins and that all of the helicases tested use the steric exclusion mechanism to unwind duplex DNA. These results further suggest that DPCs on the translocating and nontranslocating strands constitute helicase and polymerase blocks, respectively. The helicases stalled by DPC had limited stability and dissociated from DNA with a half-life of 15-36 min. The implications of the results are discussed in relation to the distinct stabilities of replisomes that encounter tight but reversible DNA-protein complexes and irreversible DPC roadblocks.


Asunto(s)
ADN Helicasas/química , ADN Helicasas/fisiología , ADN/química , Animales , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/farmacología , Daño del ADN , AdnB Helicasas/metabolismo , Escherichia coli/metabolismo , Humanos , Proteínas de Dominio MADS/metabolismo , Modelos Genéticos , Plasticidad Neuronal , Oligonucleótidos/genética , Unión Proteica , Transporte de Proteínas , Sinapsis/metabolismo , Factores de Tiempo , Xenopus
2.
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
3.
Genes Genet Syst ; 94(3): 139, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31353333

RESUMEN

On page 126, on the bottom line of the left column, the sentence opening "Conversion of these two conserved basic amino acids in either Mcm4, 6 or 7 in S. cerevisiae to aspartic acid did not affect cell growth," should be changed to "Conversion of these two conserved basic amino acids in either Mcm4, 6 or 7 in S. cerevisiae to alanine did not affect cell growth,".   On page 127, in the Fig. 3 legend, the second sentence "Two conserved basic amino acids in the amino-terminal region of Mcm4, 6 and 7 proteins were mutated to aspartic acid in S. cerevisiae (Froelich et al., 2014)." should be changed to "Two conserved basic amino acids in the amino-terminal region of Mcm4, 6 and 7 proteins were mutated to alanine in S. cerevisiae (Froelich et al., 2014); the suffix D indicates double-alanine mutation".

4.
Genes Genet Syst ; 94(3): 123-132, 2019 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-31092751

RESUMEN

Cellular aging is characterized by the loss of DNA replication capability and is mainly brought about by various changes in chromatin structure. Here, we examined changes in MCM2-7 proteins, which act as a replicative DNA helicase, during aging of human WI38 fibroblasts at the single-cell level. We used nuclear accumulation of p21 as a marker of senescent cells, and examined changes in MCM2-7 by western blot analysis. First, we found that senescent cells are enriched for cells with a DNA content higher than 4N. Second, the levels of MCM2, MCM3, MCM4 and MCM6 proteins decreased in senescent cells. Third, cytoplasmic localization of MCM2 and MCM7 was observed in senescent cells, from an analysis of MCM2-7 except for MCM5. Consistent with this finding, fragmented MCM2 was predominant in these cells. These age-dependent changes in MCM2-7, a protein complex that directly affects cellular DNA replication, may play a critical role in cellular senescence.


Asunto(s)
Senescencia Celular/genética , Replicación del ADN/genética , Componente 2 del Complejo de Mantenimiento de Minicromosoma/genética , Complejos Multiproteicos/genética , Proteínas de Ciclo Celular/genética , Regulación de la Expresión Génica/genética , Humanos , Componente 2 del Complejo de Mantenimiento de Minicromosoma/química , Componente 3 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 4 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 6 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 7 del Complejo de Mantenimiento de Minicromosoma/genética , Complejos Multiproteicos/química , Análisis de la Célula Individual , Quinasas p21 Activadas/genética
5.
Cancer Sci ; 99(5): 863-71, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18294277

RESUMEN

When a replicative DNA polymerase encounters a lesion on the template strand and stalls, it is replaced with another polymerase(s) with low processivity that bypasses the lesion to continue DNA synthesis. This phenomenon is known as translesion replication or replicative bypass. Failing this, the cell is increasingly likely to undergo apoptosis. In this study, we found that proteasome inhibitors prevent translesion replication in human cancer cells but not in normal cells. Three proteasome inhibitors, MG-132, lactacystin, and MG-262, inhibited UV-induced translesion replication in a wide range of cancer cell lines, including HeLa, HGC-27, MCF-7, HepG2, WiDr, a malignant melanoma, an acute lymphoblastic leukemia, and a multiple myeloma cell line; irrespective of cell origin, histological type, or p53 status. In contrast, these inhibitors had little or no influence on normal fibroblasts (NB1RGB and TIG-1) or a normal liver mesenchymal (LI90) cell line. Among the DNA-damaging antineoplastic agents, cisplatin caused a UV-type translesion reaction; the proteasome inhibitors delayed cisplatin-induced translesion replication in cancer cell lines but had only a weak effect on normal cell lines. Therefore, translesion replication would be an effective target of proteasome inhibitors for cancer chemotherapy by which cancer cells can be efficiently sensitized to DNA-damaging antineoplastic agents, such as cisplatin.


Asunto(s)
Inhibidores de Cisteína Proteinasa/farmacología , Daño del ADN , Replicación del ADN/efectos de los fármacos , Neoplasias/genética , Inhibidores de Proteasoma , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Ácidos Borónicos/farmacología , Cafeína/farmacología , Células Cultivadas , Cisplatino/farmacología , Reparación del ADN , Células HeLa , Humanos , Leupeptinas/farmacología , Neoplasias/enzimología , Células Tumorales Cultivadas , Rayos Ultravioleta
6.
FEBS J ; 275(4): 727-38, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18190532

RESUMEN

The molecular dissection of human MCM2, a constituent of MCM2-7 licensing factor complex, was performed to identify the region responsible for its biochemical activities. Partial digestion with trypsin dissected the MCM2 protein into a central region (148-676) containing ATPase motifs and a C-terminal region (677-895). These two fragments, along with three other fragments (148-441, 442-676 and 442-895), were produced using the wheat germ cell-free system and were examined for their ability to inhibit MCM4/6/7 helicase activity. Two fragments (442-895 and 677-895) containing the C-terminus were partly inhibitory to the activity. Further dissection revealed that one fragment (713-895) has strong inhibitory activity. The inhibitory activity of the smaller fragments derived from the C-terminal region correlated with their ability to inhibit SV40 T antigen helicase activity and also with their ability to bind to ssDNA, which has been shown by gel mobility shift analysis. These results strongly suggest that the MCM2 fragments derived from the C-terminal region inhibit DNA helicase activity through their ability to bind to ssDNA. In contrast, two fragments (148-441 and 442-676) from the central region were mainly responsible for the interaction between MCM2 and MCM4, and this was revealed by a pulldown analysis using MCM4 protein beads. Finally, only complete MCM2, not the smaller fragments, could disassemble the MCM4/6/7 hexamer into the MCM2/4/6/7 tetramer.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/metabolismo , Secuencias de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , ADN Helicasas/metabolismo , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dimerización , Ensayo de Cambio de Movilidad Electroforética , Humanos , 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/química , Proteínas Nucleares/genética , Fragmentos de Péptidos/química , Unión Proteica , Tripsina/metabolismo
7.
Genes Genet Syst ; 93(4): 125-133, 2018 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-30369561

RESUMEN

Recently published structural and functional analyses of the CMG complex have provided insight into the mechanism of its DNA helicase function and into the distinct roles of its central six component proteins MCM2-MCM7 (MCM2-7). To activate CMG helicase, the two protein kinases CDK and DDK, as well as MCM10, are required. In addition to the initiation of DNA replication, MCM function must be regulated at the DNA replication steps of elongation and termination. Polyubiquitylation of MCM7 is involved in terminating MCM function. Reinitiation of DNA replication in a single cell cycle, which is prevented mainly by CDK, is understood at the molecular level. MCM2-7 gene expression is regulated during cellular aging and the cell cycle, and the expression depends on oxygen concentration. These regulatory processes have been described recently. Genomic structural alteration, which is an essential element in cancer progression, is mainly generated by disruptions of DNA replication fork structures. A point mutation in MCM4 that disturbs MCM2-7 function results in genomic instability, leading to the generation of cancer cells. In this review, I focus on the following points: 1) function of the MCM2-7 complex, 2) activation of MCM2-7 helicase, 3) regulation of MCM2-7 function, 4) MCM2-7 expression, and 5) the role of MCM mutation in cancer progression.


Asunto(s)
Proteínas de Mantenimiento de Minicromosoma/metabolismo , Animales , Carcinogénesis , Senescencia Celular , Replicación del ADN , Humanos , Proteínas de Mantenimiento de Minicromosoma/genética
8.
J Biochem ; 164(6): 449-460, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30184107

RESUMEN

The amino-terminal region of eukaryotic MCM4 is characteristic of the presence of a number of phosphorylation sites for CDK and DDK, suggesting that the region plays regulatory roles in the MCM2-7 helicase function. However, the roles are not fully understood. We analyzed the role of the amino-terminal region of human MCM4 by using MCM4/6/7 helicase as a model for MCM2-7 helicase. First we found that deletion of 35 amino acids at the amino-terminal end resulted in inhibition of DNA helicase activity of the MCM4/6/7 complex. Conversion of arginine at amino acid no. 10 and 11 to alanine had similar effect to the deletion mutant of Δ1-35, suggesting that these arginine play a role in the DNA helicase activity. The data suggest that expression of these mutant MCM4 in HeLa cells perturbed the progression of the S phase. Substitution of six CDK phosphorylation sites (3, 7, 19, 32, 54 and 110) in the amino-terminal region by phospho-mimetic glutamic acids affected the hexamer formation of the MCM4/6/7 complex. MCM4 phosphorylation by CDK may play a role in DNA replication licensing system, and the present results suggest that the phosphorylation interferes MCM function by lowering stability of MCM complex.


Asunto(s)
Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Componente 4 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 6 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Procesamiento Proteico-Postraduccional , Fase S , Sustitución de Aminoácidos , Arginina/química , Estabilidad de Enzimas , Eliminación de Gen , Células HeLa , Humanos , Componente 4 del Complejo de Mantenimiento de Minicromosoma/química , Componente 4 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 6 del Complejo de Mantenimiento de Minicromosoma/química , Componente 6 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 7 del Complejo de Mantenimiento de Minicromosoma/química , Componente 7 del Complejo de Mantenimiento de Minicromosoma/genética , Mutagénesis Sitio-Dirigida , Oligopéptidos/química , Oligopéptidos/genética , Oligopéptidos/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación , Mutación Puntual , Multimerización de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
9.
J Biochem ; 163(5): 371-380, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29346578

RESUMEN

It has been reported that daidzein and equol stimulate DNA replication and proliferation of MCF-7 cells. However, their molecular mechanisms of action are still unclear. We examined the effects of daidzein and equol on DNA replication of MCF-7 cells, focusing on MCM2-7 proteins, which function as the replicative helicase. In the presence of either 1 µM of daidzein or equol, the number of cells in S-phase, which was determined by detecting bromodeoxyuridine incorporated into replicated DNA, almost doubled. The total amounts of MCM7 protein and chromatin-bound MCM7 protein increased in the presence of daidzein. The data suggest that phytoestrogens facilitate cell cycle progression in G1-phase by increasing the level of MCM proteins. In the presence of phytoestrogens, phosphorylation of Rb and levels of MCM2, 3 and 7 mRNA increased, suggesting that stimulation of MCM2-7 transcription is involved in the cell cycle progression. Under the same conditions, double-stranded DNA breakage in logarithmically growing MCF-7 cells, which was detected using anti-γ-H2AX antibodies, did not increase in the presence of equol.


Asunto(s)
Replicación del ADN/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , Equol/farmacología , Isoflavonas/farmacología , Proteínas de Ciclo Celular/metabolismo , ADN de Neoplasias/metabolismo , Humanos , Células MCF-7 , Componente 2 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 3 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 4 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 6 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/metabolismo
10.
J Biochem ; 161(3): 259-268, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27794528

RESUMEN

An MCM4 mutation detected in human cancer cells from endometrium was characterized. The mutation of G486D is located within MCM-box and the glycine at 486 in human MCM4 is conserved in Saccharomyces cerevisiae MCM4 and Sulfolobus solfataricus MCM. This MCM4 mutation affected human MCM4/6/7 complex formation, since the complex containing the mutant MCM4 protein is unstable and the mutant MCM4 protein is tend to be degraded. It is likely that the MCM4 mutation affects the interaction with MCM7 to destabilize the MCM4/6/7 complex. Cells with abnormal nuclear morphology were detected when the mutant MCM4 was expressed in HeLa cells, suggesting that DNA replication was perturbed in the presence of the mutant MCM4. Role of the conserved amino acid in MCM4 function is discussed.


Asunto(s)
Componente 4 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 4 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 6 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Proteínas Mutantes/metabolismo , Mutación , Células HeLa , Humanos , Componente 4 del Complejo de Mantenimiento de Minicromosoma/química , Componente 6 del Complejo de Mantenimiento de Minicromosoma/química , Componente 6 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 7 del Complejo de Mantenimiento de Minicromosoma/química , Componente 7 del Complejo de Mantenimiento de Minicromosoma/genética , Complejos Multiproteicos/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , Estabilidad Proteica
11.
FEBS J ; 273(6): 1224-39, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16519687

RESUMEN

MCM4, a subunit of a putative replicative helicase, is phosphorylated during the cell cycle, at least in part by cyclin-dependent kinases (CDK), which play a central role in the regulation of DNA replication. However, detailed characterization of the phosphorylation of MCM4 remains to be performed. We examined the phosphorylation of human MCM4 at Ser3, Thr7, Thr19, Ser32, Ser54, Ser88 and Thr110 using anti-phosphoMCM4 sera. Western blot analysis of HeLa cells indicated that phosphorylation of MCM4 at these seven sites can be classified into two groups: (a) phosphorylation that is greatly enhanced in the G2 and M phases (Thr7, Thr19, Ser32, Ser54, Ser88 and Thr110), and (b) phosphorylation that is firmly detected during interphase (Ser3). We present data indicating that phosphorylation at Thr7, Thr19, Ser32, Ser88 and Thr110 in the M phase requires CDK1, using a temperature-sensitive mutant of mouse CDK1, and phosphorylation at sites 3 and 32 during interphase requires CDK2, using a dominant-negative mutant of human CDK2. Based on these results and those from in vitro phosphorylation of MCM4 with CDK2/cyclin A, we discuss the kinases responsible for MCM4 phosphorylation. Phosphorylated MCM4 detected using anti-phospho sera exhibited different affinities for chromatin. Studies on the nuclear localization of chromatin-bound MCM4 phosphorylated at sites 3 and 32 suggested that they are not generally colocalized with replicating DNA. Unexpectedly, MCM4 phosphorylated at site 32 was enriched in the nucleolus through the cell cycle. These results suggest that phosphorylation of MCM4 has several distinct and site-specific roles in the function of MCM during the mammalian cell cycle.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Cromatina/metabolismo , Quinasa 2 Dependiente de la Ciclina/fisiología , ADN Helicasas/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Ciclina A/fisiología , Células HeLa , Humanos , Mamíferos/metabolismo , Ratones , Componente 4 del Complejo de Mantenimiento de Minicromosoma , Componente 6 del Complejo de Mantenimiento de Minicromosoma , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Mitosis , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Fosforilación , Homología de Secuencia
12.
J Biochem ; 157(6): 561-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25661590

RESUMEN

A number of gene mutations are detected in cells derived from human cancer tissues, but roles of these mutations in cancer cell development are largely unknown. We examined G364R mutation of MCM4 detected in human skin cancer cells. Formation of MCM4/6/7 complex is not affected by the mutation. Consistent with this notion, the binding to MCM6 is comparable between the mutant MCM4 and wild-type MCM4. Nuclear localization of this mutant MCM4 expressed in HeLa cells supports this conclusion. Purified MCM4/6/7 complex containing the G364R MCM4 exhibited similar levels of single-stranded DNA binding and ATPase activities to the complex containing wild-type MCM4. However, the mutant complex showed only 30-50% of DNA helicase activity of the wild-type complex. When G364R MCM4 was expressed in HeLa cells, it was fractionated into nuclease-sensitive chromatin fraction, similar to wild-type MCM4. These results suggest that this mutation does not affect assembly of MCM2-7 complex on replication origins but it interferes some step at function of MCM2-7 helicase. Thus, this mutation may contribute to cancer cell development by disturbing DNA replication.


Asunto(s)
ADN Helicasas/metabolismo , Componente 4 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 6 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Neoplasias Cutáneas/genética , Línea Celular Tumoral , Humanos , Componente 4 del Complejo de Mantenimiento de Minicromosoma/química , Componente 4 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Mutagénesis Sitio-Dirigida , Unión Proteica , Neoplasias Cutáneas/patología
14.
FEBS J ; 281(4): 1057-67, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24299456

RESUMEN

It has been reported that minichromosome maintenance protein-binding protein (MCM-BP) functions in the formation of the pre-replication complex, unloading of minichromosome maintenance (MCM)2-7 from chromatin in late S phase, and formation of the cohesion complex by interacting with MCM3-7 proteins, suggesting that MCM-BP functions in several different reactions during the cell cycle. Here, we examined the interaction of human MCM-BP with MCM2-7 and structural maintenance of chromosome 3 in synchronized HeLa cells by immunoprecipitation. The results show that MCM-BP mainly interacts with MCM7 in the Triton-soluble fraction from S phase and G(2) phase cells, and it also interacts with structural maintenance of chromosome 3 in the fraction from G(2) phase cells. In vitro studies show that MCM-BP disassembles MCM2-7 bound to DNA with a fork-like structure by interacting with MCM3, MCM5, and MCM7. These results suggest that MCM-BP functions in disassembling MCM2-7 on chromatin during S phase and G2 phase by interacting with MCM3, MCM5, and MCM7.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Proteínas Nucleares/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Cromosomas Humanos Par 3/metabolismo , Células HeLa , Humanos , Componente 2 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 3 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Unión Proteica
15.
J Biochem ; 154(4): 363-72, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23864661

RESUMEN

Cyclin-dependent kinase (CDK) that plays a central role in preventing re-replication of DNA phosphorylates several replication proteins to inactivate them. MCM4 in MCM2-7 and RPA2 in RPA are phosphorylated with CDK in vivo. There are inversed correlations between the phosphorylation of these proteins and their chromatin binding. Here, we examined in vitro phosphorylation of human replication proteins of MCM2-7, RPA, TRESLIN, CDC45 and RECQL4 with CDK2/cyclinE, CDK2/cyclinA, CDK1/cyclinB, CHK1, CHK2 and CDC7/DBF4 kinases. MCM4, RPA2, TRESLIN and RECQL4 were phosphorylated with CDKs. Effect of the phosphorylation by CDK2/cyclinA on DNA-binding abilities of MCM2-7 and RPA was examined by gel-shift analysis. The phosphorylation of RPA did not affect its DNA-binding ability but that of MCM4 inhibited the ability of MCM2-7. Change of six amino acids of serine and threonine to alanines in the amino-terminal region of MCM4 rendered the mutant MCM2-7 insensitive to the inhibition with CDK. These biochemical data suggest that phosphorylation of MCM4 at these sites by CDK plays a direct role in dislodging MCM2-7 from chromatin and/or preventing re-loading of the complex to chromatin.


Asunto(s)
Quinasas Ciclina-Dependientes/química , Replicación del ADN/efectos de los fármacos , ADN/química , Proteínas de Mantenimiento de Minicromosoma/química , Ciclo Celular , Ciclina A/química , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/química , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , ADN/metabolismo , Células HeLa , Humanos , Componente 4 del Complejo de Mantenimiento de Minicromosoma/química , Componente 4 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 4 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Proteínas de Mantenimiento de Minicromosoma/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Datos de Secuencia Molecular , Mutación , Fosforilación , Proteínas Recombinantes/química
16.
J Biochem ; 153(4): 381-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23364835

RESUMEN

CDC45, which plays a role in eukaryotic DNA replication, is a member of the CMG (CDC45/MCM2-7/GINS) complex that is thought to function as a replicative DNA helicase. However, the biochemical properties of CDC45 are not fully understood. We systematically examined the interactions of human CDC45 with MCM2-7, GINS and other replication proteins by immunoprecipitation. We found that CDC45 can directly interact with all MCM2-7 proteins; with PSF2, PSF3 and SLD5 in GINS subunits; and with replication protein A2 (RPA2), AND-1 and topoisomerase 2-binding protein 1. These results are consistent with the notion that CDC45 plays a role in progression of DNA replication forks. Experiments using antibodies against CDC45 show that the level of CDC45 recovered from the Triton-insoluble chromatin-containing fraction is peaked at middle of S phase in synchronized HeLa cells. However, incubation of the Triton-insoluble fraction with nucleases resulted in recovery of less than half the amount of CDC45 in the nuclease-sensitive fraction; this result is in contrast with RPA1 and proliferating cell nuclear antigen distribution. These results indicate that a considerable portion of CDC45 localizes in a region other than the DNA replication forks in nuclei or it localizes on the replication forks but it is not fractionated with the fork proteins owing to its tight association with presumably nuclear scaffolds.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/inmunología , Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Inmunoprecipitación/métodos , Insectos , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Mapas de Interacción de Proteínas , Proteína de Replicación A/metabolismo , Fase S
17.
J Biochem ; 152(2): 191-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22668557

RESUMEN

It has been reported that a point mutation of minichromosome maintenance (MCM)4 causes mammary carcinoma, and it deregulates DNA replication to produce abnormal chromosome structures. To understand the effect of this mutation at level of MCM2-7 interaction, we examined the effect of the same mutation of human MCM4 on the complex formation with MCM6 and MCM7 in insect cells. Human MCM4/6/7 complexes containing the mutated MCM4 were formed, but the hexameric complex formation was not evident in comparison with those containing wild-type MCM4. In binary expression of MCM4 and MCM6, decreased levels of MCM6 were recovered with the mutated MCM4, compared with wild-type MCM4. These results suggest that this mutation of MCM4 perturbs proper interaction with MCM6 to affect complex formation of MCM4/6/7 that is a core structure of MCM2-7 complex. Consistent with this notion, nuclear localization and MCM complex formation of forcedly expressed MCM4 in human cells are affected by this mutation. Thus, the defect of this mutant MCM4 in interacting with MCM6 may generate a decreased level of chromatin binding of MCM2-7 complex.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mutación Puntual , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Células HeLa , Humanos , Ratones , Componente 4 del Complejo de Mantenimiento de Minicromosoma , Componente 6 del Complejo de Mantenimiento de Minicromosoma , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Complejos Multiproteicos
18.
J Biochem ; 151(2): 129-37, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22023799

RESUMEN

The antibiotic heliquinomycin inhibited cellular DNA replication at IC(50) of 2.5 µM without affecting level of chromatin-bound MCM4 and without activating the DNA replication stress checkpoint system, suggesting that heliquinomycin perturbs DNA replication mainly by inhibiting the activity of replicative DNA helicase that unwinds DNA duplex at replication forks. Among the DNA helicases involved in DNA replication, DNA helicase B was inhibited by heliquinomycin at IC(50) of 4.3 µM and RECQL4 helicase at IC(50) of 14 µM; these values are higher than that of MCM4/6/7 helicase (2.5 µM). These results suggest that heliquinomycin mainly targets actions of the replicative DNA helicases. Gel-retardation experiment indicates that heliquinomycin binds to single-stranded DNA. The single-stranded DNA-binding ability of MCM4/6/7 was affected in the presence of heliquinomycin. The data suggest that heliquinomycin inhibits the DNA helicase activity of MCM4/6/7 complex by stabilizing its interaction with single-stranded DNA.


Asunto(s)
Antibacterianos/metabolismo , ADN Helicasas/antagonistas & inhibidores , ADN de Cadena Simple/metabolismo , Antibacterianos/química , Benzoquinonas/química , Benzoquinonas/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , ADN Helicasas/química , ADN Helicasas/metabolismo , Replicación del ADN/efectos de los fármacos , ADN de Cadena Simple/química , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Células HeLa , Humanos , Componente 4 del Complejo de Mantenimiento de Minicromosoma , Componente 6 del Complejo de Mantenimiento de Minicromosoma , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Compuestos de Espiro/química , Compuestos de Espiro/metabolismo
19.
J Nucleic Acids ; 2010: 750296, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21151666

RESUMEN

When a replicative DNA polymerase stalls upon encountering a lesion on the template strand, it is relieved by other low-processivity polymerase(s), which insert nucleotide(s) opposite the lesion, extend by a few nucleotides, and dissociate from the 3'-OH. The replicative polymerase then resumes DNA synthesis. This process, termed translesion replication (TLS) or replicative bypass, may involve at least five different polymerases in mammals, although the participating polymerases and their roles have not been entirely characterized. Using siRNAs originally designed and an alkaline sucrose density gradient sedimentation technique, we verified the involvement of several polymerases in ultraviolet (UV) light-induced TLS in HeLa cells. First, siRNAs to Rev3 or Rev7 largely abolished UV-TLS, suggesting that these 2 gene products, which comprise Polζ, play a main role in mutagenic TLS. Second, Rev1-targeted siRNA also abrogated UV-TLS, indicating that Rev1 is also indispensable to mutagenic TLS. Third, Polη-targeted siRNA also prevented TLS to a greater extent than our expectations. Forth, although siRNA to Polι had no detectable effect, that to Polκ delayed UV-TLS. To our knowledge, this is the first study reporting apparent evidence for the participation of Polκ in UV-TLS.

20.
J Biochem ; 148(5): 539-47, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20679368

RESUMEN

RPA, which consisted of three subunits (RPA1, 2 and 3), plays essential roles in DNA transactions. At the DNA replication forks, RPA binds to single-stranded DNA region to stabilize the structure and to assemble other replication proteins. Interactions between RPA and several replication proteins have been reported but the analysis is not comprehensive. We systematically performed the qualitative analysis to identify RPA interaction partners to understand the protein-protein interaction at the replication forks. We expressed in insect cells the three subunits of human RPA, together with one replication protein, which is present at the forks under normal conditions and/or under the replication stress conditions, to examine the interaction. Among 30 proteins examined in total, it was found that at least 14 proteins interacted with RPA. RPA interacted with MCM3-7, MCM-BP and CDC45 proteins among the proteins that play roles in the initiation and the elongation of the DNA replication. RPA bound with TIPIN, CLASPIN and RAD17, which are involved in the DNA replication checkpoint functions. RPA also bound with cyclin-dependent kinases and an amino-terminal fragment of Rb protein that negatively regulates DNA replication. These results suggest that RPA interacts with the specific proteins among those that play roles in the regulation of the replication fork progression.


Asunto(s)
Replicación del ADN , Proteína de Replicación A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Componente 3 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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA