Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Genes (Basel) ; 13(2)2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-35205311

RESUMEN

The ubiquitin ligase CRL4Cdt2 plays a vital role in preserving genomic integrity by regulating essential proteins during S phase and after DNA damage. Deregulation of CRL4Cdt2 during the cell cycle can cause DNA re-replication, which correlates with malignant transformation and tumor growth. CRL4Cdt2 regulates a broad spectrum of cell cycle substrates for ubiquitination and proteolysis, including Cdc10-dependent transcript 1 or Chromatin licensing and DNA replication factor 1 (Cdt1), histone H4K20 mono-methyltransferase (Set8) and cyclin-dependent kinase inhibitor 1 (p21), which regulate DNA replication. However, the mechanism it operates via its substrate receptor, Cdc10-dependent transcript 2 (Cdt2), is not fully understood. This review describes the essential features of the N-terminal and C-terminal parts of Cdt2 that regulate CRL4 ubiquitination activity, including the substrate recognition domain, intrinsically disordered region (IDR), phosphorylation sites, the PCNA-interacting protein-box (PIP) box motif and the DNA binding domain. Drugs targeting these specific domains of Cdt2 could have potential for the treatment of cancer.


Asunto(s)
Ubiquitina-Proteína Ligasas , Ubiquitina , Proteínas de Ciclo Celular/genética , ADN/genética , ADN/metabolismo , Antígeno Nuclear de Célula en Proliferación/química , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/genética
2.
Trends Cell Biol ; 30(4): 290-302, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32044173

RESUMEN

The cullin-RING E3 ubiquitin ligase CRL4Cdt2 has emerged as a master regulator of genome stability, which targets key cell cycle proteins for proteolysis during S phase and after DNA damage. Recent advances shed light on how it couples ubiquitination to DNA synthesis, offering a new paradigm for substrate recognition: Cdt2 binds directly onto proliferating cell nuclear antigen (PCNA) loaded on DNA, which serves as a landing pad for the independent recruitment of the ubiquitin ligase and its substrates. Cyclin-dependent kinases (CDKs) and the ataxia telangiectasia and Rad3-related (ATR) kinase ensure accurate spatiotemporal regulation of CRL4Cdt2 under normal conditions and upon DNA damage. Deregulation of Cdt2 is evident in malignancies and was recently highlighted as a major target of oncogenic viruses, supporting the therapeutic targeting of the ligase as a promising anticancer strategy.


Asunto(s)
Inestabilidad Genómica , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Humanos , Modelos Biológicos , Proteolisis , Especificidad por Sustrato
3.
J Biochem ; 165(6): 505-516, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30649446

RESUMEN

The Cullin-RING ubiquitin ligase CRL4Cdt2 maintains genome integrity by mediating the cell cycle- and DNA damage-dependent degradation of proteins such as Cdt1, p21 and Set8. Human Cdt2 has two regions, a conserved N-terminal seven WD40 repeat region and a less conserved C-terminal region. Here, we showed that the N-terminal region is sufficient for complex formation with CRL4, but the C-terminal region is required for the full ubiquitin ligase activity. UV irradiation-induced polyubiquitination and degradation of Cdt1 were impaired in Cdt2 (N-terminus only)-expressing cells. Deletion and mutation analysis identified a domain in the C-terminal region that increased ubiquitination activity and displayed DNA-binding activity. The identified domain mediated binding to double-stranded DNA and showed higher affinity binding to single-stranded DNA. As the ligase activity of CRL4Cdt2 depends on proliferating cell nuclear antigen (PCNA) loading onto DNA, the present results suggest that the DNA-binding domain facilitates the CRL4Cdt2-mediated recognition and ubiquitination of substrates bound to PCNA on chromatin.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN/biosíntesis , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Sitios de Unión , Células Cultivadas , ADN/química , Humanos
4.
Sci Signal ; 11(525)2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636388

RESUMEN

The symbiotic nitrogen-fixing bacterium Bradyrhizobium japonicum is critical to the agro-industrial production of soybean because it enables the production of high yields of soybeans with little use of nitrogenous fertilizers. The FixL and FixJ two-component system (TCS) of this bacterium ensures that nitrogen fixation is only stimulated under conditions of low oxygen. When it is not bound to oxygen, the histidine kinase FixL undergoes autophosphorylation and transfers phosphate from adenosine triphosphate (ATP) to the response regulator FixJ, which, in turn, stimulates the expression of genes required for nitrogen fixation. We purified full-length B. japonicum FixL and FixJ proteins and defined their structures individually and in complex using small-angle x-ray scattering, crystallographic, and in silico modeling techniques. Comparison of active and inactive forms of FixL suggests that intramolecular signal transduction is driven by local changes in the sensor domain and in the coiled-coil region connecting the sensor and histidine kinase domains. We also found that FixJ exhibits conformational plasticity not only in the monomeric state but also in tetrameric complexes with FixL during phosphotransfer. This structural characterization of a complete TCS contributes both a mechanistic and evolutionary understanding to TCS signal relay, specifically in the context of the control of nitrogen fixation in root nodules.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hemoproteínas/metabolismo , Histidina Quinasa/metabolismo , Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bradyrhizobium/genética , Bradyrhizobium/metabolismo , Cristalografía por Rayos X , Regulación Bacteriana de la Expresión Génica , Hemoproteínas/química , Hemoproteínas/genética , Histidina Quinasa/química , Histidina Quinasa/genética , Modelos Moleculares , Fijación del Nitrógeno/genética , Fosforilación , Unión Proteica , Dominios Proteicos , Transducción de Señal/genética
6.
Genes Cells ; 23(3): 200-213, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29424068

RESUMEN

CRL4Cdt2 ubiquitin ligase plays an important role maintaining genome integrity during the cell cycle. A recent report suggested that Cdk1 negatively regulates CRL4Cdt2 activity through phosphorylation of its receptor, Cdt2, but the involvement of phosphorylation remains unclear. To address this, we mutated all CDK consensus phosphorylation sites located in the C-terminal half region of Cdt2 (Cdt2-18A) and examined the effect on substrate degradation. We show that both cyclinA/Cdk2 and cyclinB/Cdk1 phosphorylated Cdt2 in vitro and that phosphorylation was reduced by the 18A mutation both in vitro and in vivo. The 18A mutation increased the affinity of Cdt2 to PCNA, and a high amount of Cdt2-18A was colocalized with PCNA foci during S phase in comparison with Cdt2-WT. Poly-ubiquitination activity to Cdt1 was concomitantly enhanced in cells expressing Cdt2-18A. Other CRL4Cdt2 substrates, Set8 and thymine DNA glycosylase, begin to accumulate around late S phase to G2 phase, but the accumulation was prevented in Cdt2-18A cells. Furthermore, mitotic degradation of Cdt1 after UV irradiation was induced in these cells. Our results suggest that CDK-mediated phosphorylation of Cdt2 inactivates its ubiquitin ligase activity by reducing its affinity to PCNA, an important strategy for regulating the levels of key proteins in the cell cycle.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitina/metabolismo , Proteína Quinasa CDC2/genética , Células HEK293 , Células HeLa , Humanos , Fosforilación , Proteolisis , Fase S , Ubiquitinación
7.
Life Sci Alliance ; 1(6): e201800238, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30623174

RESUMEN

The CRL4Cdt2 ubiquitin ligase complex is an essential regulator of cell-cycle progression and genome stability, ubiquitinating substrates such as p21, Set8, and Cdt1, via a display of substrate degrons on proliferating cell nuclear antigens (PCNAs). Here, we examine the hierarchy of the ligase and substrate recruitment kinetics onto PCNA at sites of DNA replication. We demonstrate that the C-terminal end of Cdt2 bears a PCNA interaction protein motif (PIP box, Cdt2PIP), which is necessary and sufficient for the binding of Cdt2 to PCNA. Cdt2PIP binds PCNA directly with high affinity, two orders of magnitude tighter than the PIP box of Cdt1. X-ray crystallographic structures of PCNA bound to Cdt2PIP and Cdt1PIP show that the peptides occupy all three binding sites of the trimeric PCNA ring. Mutating Cdt2PIP weakens the interaction with PCNA, rendering CRL4Cdt2 less effective in Cdt1 ubiquitination and leading to defects in Cdt1 degradation. The molecular mechanism we present suggests a new paradigm for bringing substrates to the CRL4-type ligase, where the substrate receptor and substrates bind to a common multivalent docking platform to enable subsequent ubiquitination.

8.
Nat Commun ; 8: 16102, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28719581

RESUMEN

HBO1, a histone acetyl transferase, is a co-activator of DNA pre-replication complex formation. We recently reported that HBO1 is phosphorylated by ATM and/or ATR and binds to DDB2 after ultraviolet irradiation. Here, we show that phosphorylated HBO1 at cyclobutane pyrimidine dimer (CPD) sites mediates histone acetylation to facilitate recruitment of XPC at the damaged DNA sites. Furthermore, HBO1 facilitates accumulation of SNF2H and ACF1, an ATP-dependent chromatin remodelling complex, to CPD sites. Depletion of HBO1 inhibited repair of CPDs and sensitized cells to ultraviolet irradiation. However, depletion of HBO1 in cells derived from xeroderma pigmentosum patient complementation groups, XPE, XPC and XPA, did not lead to additional sensitivity towards ultraviolet irradiation. Our findings suggest that HBO1 acts in concert with SNF2H-ACF1 to make the chromosome structure more accessible to canonical nucleotide excision repair factors.


Asunto(s)
Reparación del ADN , Histona Acetiltransferasas/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Fosforilación , Dímeros de Pirimidina/metabolismo , Factores de Transcripción/metabolismo , Rayos Ultravioleta
9.
Cell Cycle ; 16(7): 673-684, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28278049

RESUMEN

Cdt1 is rapidly degraded by CRL4Cdt2 E3 ubiquitin ligase after UV (UV) irradiation. Previous reports revealed that the nucleotide excision repair (NER) pathway is responsible for the rapid Cdt1-proteolysis. Here, we show that mismatch repair (MMR) proteins are also involved in the degradation of Cdt1 after UV irradiation in the G1 phase. First, compared with the rapid (within ∼15 min) degradation of Cdt1 in normal fibroblasts, Cdt1 remained stable for ∼30 min in NER-deficient XP-A cells, but was degraded within ∼60 min. The delayed degradation was also dependent on PCNA and CRL4Cdt2. The MMR proteins Msh2 and Msh6 were recruited to the UV-damaged sites of XP-A cells in the G1 phase. Depletion of these factors with small interfering RNAs prevented Cdt1 degradation in XP-A cells. Similar to the findings in XP-A cells, depletion of XPA delayed Cdt1 degradation in normal fibroblasts and U2OS cells, and co-depletion of Msh6 further prevented Cdt1 degradation. Furthermore, depletion of Msh6 alone delayed Cdt1 degradation in both cell types. When Cdt1 degradation was attenuated by high Cdt1 expression, repair synthesis at the damaged sites was inhibited. Our findings demonstrate that UV irradiation induces multiple repair pathways that activate CRL4Cdt2 to degrade its target proteins in the G1 phase of the cell cycle, leading to efficient repair of DNA damage.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Reparación de la Incompatibilidad de ADN/efectos de la radiación , Fase G1/efectos de la radiación , Proteolisis/efectos de la radiación , Rayos Ultravioleta , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Células HeLa , Humanos , Cinética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Xerodermia Pigmentosa/metabolismo
10.
Genes Cells ; 22(4): 392-405, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28318075

RESUMEN

Thymine DNA glycosylase (TDG) is a base excision repair (BER) enzyme, which is implicated in correction of deamination-induced DNA mismatches, the DNA demethylation process and regulation of gene expression. Because of these pivotal roles associated, it is crucial to elucidate how the TDG functions are appropriately regulated in vivo. Here, we present evidence that the TDG protein undergoes degradation upon various types of DNA damage, including ultraviolet light (UV). The UV-induced degradation of TDG was dependent on proficiency in nucleotide excision repair and on CRL4CDT2 -mediated ubiquitination that requires a physical interaction between TDG and DNA polymerase clamp PCNA. Using the Tdg-deficient mouse embryonic fibroblasts, we found that ectopic expression of TDG compromised cellular survival after UV irradiation and repair of UV-induced DNA lesions. These negative effects on cellular UV responses were alleviated by introducing mutations in TDG that impaired its BER function. The expression of TDG induced a large-scale alteration in the gene expression profile independently of its DNA glycosylase activity, whereas a subset of genes was affected by the catalytic activity of TDG. Our results indicate the presence of BER-dependent and BER-independent functions of TDG, which are involved in regulation of cellular DNA damage responses and gene expression patterns.


Asunto(s)
Reparación del ADN , Timina ADN Glicosilasa/metabolismo , Secuencias de Aminoácidos , Línea Celular , Daño del ADN , Humanos , Mutación , Timina ADN Glicosilasa/química , Ubiquitina-Proteína Ligasas/metabolismo , Rayos Ultravioleta
11.
Genes (Basel) ; 8(2)2017 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-28134787

RESUMEN

During cell division, genome integrity is maintained by faithful DNA replication during S phase, followed by accurate segregation in mitosis. Many DNA metabolic events linked with DNA replication are also regulated throughout the cell cycle. In eukaryotes, the DNA sliding clamp, proliferating cell nuclear antigen (PCNA), acts on chromatin as a processivity factor for DNA polymerases. Since its discovery, many other PCNA binding partners have been identified that function during DNA replication, repair, recombination, chromatin remodeling, cohesion, and proteolysis in cell-cycle progression. PCNA not only recruits the proteins involved in such events, but it also actively controls their function as chromatin assembles. Therefore, control of PCNA-loading onto chromatin is fundamental for various replication-coupled reactions. PCNA is loaded onto chromatin by PCNA-loading replication factor C (RFC) complexes. Both RFC1-RFC and Ctf18-RFC fundamentally function as PCNA loaders. On the other hand, after DNA synthesis, PCNA must be removed from chromatin by Elg1-RFC. Functional defects in RFC complexes lead to chromosomal abnormalities. In this review, we summarize the structural and functional relationships among RFC complexes, and describe how the regulation of PCNA loading/unloading by RFC complexes contributes to maintaining genome integrity.

12.
PLoS One ; 10(3): e0120553, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25798850

RESUMEN

Cdt1 begins to accumulate in M phase and has a key role in establishing replication licensing at the end of mitosis or in early G1 phase. Treatments that damage the DNA of cells, such as UV irradiation, induce Cdt1 degradation through PCNA-dependent CRL4-Cdt2 ubiquitin ligase. How Cdt1 degradation is linked to cell cycle progression, however, remains unclear. In G1 phase, when licensing is established, UV irradiation leads to Cdt1 degradation, but has little effect on the licensing state. In M phase, however, UV irradiation does not induce Cdt1 degradation. When mitotic UV-irradiated cells were released into G1 phase, Cdt1 was degraded before licensing was established. Thus, these cells exhibited both defective licensing and G1 cell cycle arrest. The frequency of G1 arrest increased in cells expressing extra copies of Cdt2, and thus in cells in which Cdt1 degradation was enhanced, whereas the frequency of G1 arrest was reduced in cell expressing an extra copy of Cdt1. The G1 arrest response of cells irradiated in mitosis was important for cell survival by preventing the induction of apoptosis. Based on these observations, we propose that mammalian cells have a DNA replication-licensing checkpoint response to DNA damage induced during mitosis.


Asunto(s)
Replicación del ADN/efectos de la radiación , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de la radiación , Mitosis/genética , Mitosis/efectos de la radiación , Rayos Ultravioleta/efectos adversos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Cromatina/efectos de la radiación , Daño del ADN/genética , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Proteínas Nucleares/metabolismo , Estabilidad Proteica/efectos de la radiación , Proteolisis/efectos de la radiación , Ubiquitina-Proteína Ligasas/metabolismo
13.
J Alzheimers Dis ; 42(4): 1415-33, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25024339

RESUMEN

Proteolytic processing of the amyloid-ß protein precursor (AßPP) occurs via alternative pathways, culminating with the production of the AßPP intracellular domain (AICD). AICD can translocate to the nucleus and regulate transcription, but its activity is modulated by interactions with other proteins. In the nucleus, AICD, FE65, and Tip60 associate into AFT complexes, which are targeted to nuclear spots which correspond to transcription factories. Here we report that RanBP9 interacts with the cytoplasmic domain of AßPP, through the NPXY internalization motif. Moreover, RanBP9 interaction with Tip60 is also described. The RanBP9-Tip60 interaction dramatically relocated RanBP9 from a widespread cellular distribution to nuclear speckles. AßPP processing is a central aspect in determining the protein's function and that of its resulting proteolytic fragments, among them AICD. The latter results from the amyloidogenic pathway and is the peptidic species predominantly involved in nuclear signaling. Of note RanBP9 transfection was previously demonstrated to increase amyloid-ß generation. Here we show that RanBP9 relocates AICD to the Tip60-enriched nuclear speckles, and prevented the formation of nuclear spots formation, having therefore a negative effect on AICD mediated nuclear signaling and consequently AFT complex formation. Furthermore, by transfecting cells with increasing amounts of RanBP9, the expression of AICD-regulated genes, including AßPP itself, was reduced. Given the data presented, one can deduce that RanBP9 has an inhibitory regulatory effect on AICD-mediated transcription and the effect is mediated by relocating AICD away from transcription factories.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas del Citoesqueleto/metabolismo , Histona Acetiltransferasas/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Precursor de Proteína beta-Amiloide/genética , Núcleo Celular/metabolismo , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunohistoquímica , Lisina Acetiltransferasa 5 , Microscopía Confocal , Proteínas del Tejido Nervioso/metabolismo , Saccharomyces cerevisiae , Transcripción Genética/fisiología , Técnicas del Sistema de Dos Híbridos
14.
Methods Mol Biol ; 1170: 367-82, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24906324

RESUMEN

PCNA is a DNA clamp, acting on chromatin as a platform for various proteins involved in many aspects of DNA replication-linked processes. Most of these proteins have the PCNA-interaction protein motif (PIP box) that associates with PCNA. Recent works show that PCNA plays an important role as a matchmaker, connecting PCNA-interacting proteins to the ubiquitin ligase CRL4(Cdt2) for their degradation. Proteins degraded by CRL4(Cdt2) include Cdt1, p21, and Set8 in mammalian cells. These CRL4(Cdt2) substrates have a PIP degron that consists of the canonical PIP-box sequence and additional conserved amino acids required for ubiquitination. The degradation of these proteins is triggered when PCNA is loaded onto chromatin at the onset of S phase, and this process is important to prevent re-replication of DNA. These CRL4(Cdt2) substrates are also degraded through the same mechanism in response to DNA damage. In this chapter, we describe several approaches to investigate how PIP degron-containing proteins are degraded in a PCNA-dependent manner.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación , Animales , Línea Celular , Replicación del ADN , Células HeLa , Humanos , Insectos , Antígeno Nuclear de Célula en Proliferación/genética , Proteolisis , Interferencia de ARN , ARN Interferente Pequeño/genética , Ubiquitina-Proteína Ligasas/metabolismo
15.
Methods Mol Biol ; 1170: 357-65, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24906323

RESUMEN

Numerous cell cycle-regulating proteins are controlled by protein degradation. Recent work shows that ubiquitination-dependent proteolysis plays an important role in once-per-cell cycle control of DNA replication. Cdt1 is a licensing factor essential for assembling the pre-replicative complex on replication origins. Cdt1 is present in G1 phase, but after S phase ubiquitin-mediated proteolysis maintains Cdt1 at low levels. This is important to prevent the re-replication of chromosomal DNA. The cell cycle-dependent degradation of Cdt1 can be monitored by dual staining of the cell nuclei with antibodies against Cdt1- and S/G2-phase marker proteins, such as cyclin A or geminin.


Asunto(s)
Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Microscopía Fluorescente/métodos , Proteolisis , Anticuerpos/análisis , Ciclina A/análisis , Ciclina A/metabolismo , Geminina/análisis , Geminina/metabolismo , Células HeLa , Humanos , Imagen Óptica/métodos , Ubiquitina/metabolismo
16.
Methods Mol Biol ; 1170: 517-27, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24906333

RESUMEN

ORC, Cdc6, Cdt1, and MCM2-7 are replication-licensing factors, which play a central role in the once-per-cell cycle control of DNA replication. ORC, Cdc6, and Cdt1 collaborate to load MCM2-7 onto replication origins in order to license them for replication. MCM2-7 is a DNA helicase directly involved in DNA replication and dissociates from DNA as S phase progresses and each replicon is replicated. In the cell cycle, the loading of MCM2-7 is restricted during the end of mitosis and the G1 phase. Thus, the levels of chromatin-bound MCM2-7 and its loaders oscillate during the cell cycle. Chromatin association of these factors can be analyzed by separating a cell lysate into soluble and chromatin-enriched insoluble fractions in mammalian cells.


Asunto(s)
Proteínas de Ciclo Celular/aislamiento & purificación , Fraccionamiento Químico/métodos , Cromatina/aislamiento & purificación , Técnicas de Cultivo de Célula/métodos , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Células HeLa , Humanos , Proteínas de Mantenimiento de Minicromosoma/aislamiento & purificación , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Unión Proteica
17.
Methods Mol Biol ; 1170: 529-37, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24906334

RESUMEN

S-CDK and DDK protein kinases initiate DNA replication at replication origins. Prior to the activation of these kinases, origins must become competent for replication by loading MCM2-7 DNA helicase on chromatin. This process is known as replication licensing or pre-replicative complex (pre-RC) formation. After the onset of S phase, however, licensing is inhibited to prevent re-replication of DNA. In this chapter, we describe a method to analyze origin licensing by imaging the chromatin-bound licensing factor MCM2-7. In a normal cell cycle, MCM2-7 is loaded at the end of mitosis or early G1 phase. As S phase progresses, MCM2-7 is dissociated from the replicated regions. When DNA replication is completed, cells in G2 phase have no chromatin-bound MCM2-7. The analysis of chromatin-bound MCM2-7 in each cell provides an insight into cell cycle stage and condition for cell cycle.


Asunto(s)
Cromatina/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Microscopía Fluorescente/métodos , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Ciclo Celular , Células HeLa , Humanos , Proteínas de Mantenimiento de Minicromosoma/análisis , Imagen Óptica/métodos , Unión Proteica , Fijación del Tejido/métodos
18.
Genes Cells ; 18(11): 946-59, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23937667

RESUMEN

Proliferating cell nuclear antigen (PCNA) is loaded on chromatin upon initiation of the S phase and acts as a platform for a large number of proteins involved in chromosome duplication at the replication fork. As duplication is completed, PCNA dissociates from chromatin, and thus, chromatin-bound PCNA levels are regulated during the cell cycle. Although the mechanism of PCNA loading has been extensively investigated, the unloading mechanism has remained unclear. Here, we show that Elg1, an alternative replication factor C protein, is required for the regulation of chromatin-bound PCNA levels. When Elg1 was depleted by small interfering RNA, chromatin-bound PCNA levels were extremely increased during the S phase. The number of PCNA foci, regions in the nucleus normally representing DNA replication sites, was increased and PCNA remained on chromatin after DNA replication. Various chromatin-associated protein levels on chromatin were affected, and chromatin loop size was increased. During mitosis, cells with aberrant chromosomes and lagging chromosomes were frequently detected. Our findings suggest that Elg1 has an important role in maintaining chromosome integrity by regulating PCNA levels on chromatin, thereby acting as a PCNA unloading factor.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cromatina/metabolismo , Inestabilidad Cromosómica , Proteínas de Unión al ADN/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína de Replicación C/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Ciclo Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Fase G2 , Humanos , Fase S
19.
J Biol Chem ; 288(33): 23953-63, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23814078

RESUMEN

Once per cell cycle replication is crucial for maintaining genome integrity. Geminin interacts with the licensing factor Cdt1 to prevent untimely replication and is controlled by APC/C-dependent cell cycle specific proteolysis during mitosis and in G1. We show here that human geminin, when expressed in human cells in culture under a constitutive promoter, is excluded from the nucleus during part of the G1 phase and at the transition from G0 to G1. The N-terminal 30 amino acids of geminin, which contain its destruction box, are essential for nuclear exclusion. In addition, 30 amino acids within the central domain of geminin are required for both nuclear exclusion and nuclear accumulation. Cdt1 overexpression targets geminin to the nucleus, while reducing Cdt1 levels by RNAi leads to the appearance of endogenous geminin in the cytoplasm. Our data propose a novel means of regulating the balance of Cdt1/geminin in human cells, at the level of the subcellular localization of geminin.


Asunto(s)
Ciclo Celular , Replicación del ADN , Geminina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Fase G1 , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células MCF-7 , Modelos Biológicos , Proteínas Mutantes/metabolismo , Mutación/genética , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Fracciones Subcelulares/metabolismo
20.
PLoS One ; 7(9): e46480, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029527

RESUMEN

The DNA replication-licensing factor Cdt1 is present during the G1 phase of the cell cycle. When cells initiate S phase or are UV-irradiated, Cdt1 is recruited to chromatin-bound PCNA and ubiquitinated by CRL4(Cdt2) for degradation. In both situations, the substrate-recognizing subunit Cdt2 is detected as a highly phosphorylated form. Here, we show that both caffeine-sensitive kinase and MAP kinases are responsible for Cdt2 phosphorylation following UV irradiation. We found that Cdt1 degradation was attenuated in the presence of caffeine. This attenuation was also observed in cells depleted of ATR, but not ATM. Following UV irradiation, Cdt2 was phosphorylated at the S/TQ sites. ATR phosphorylated Cdt2 in vitro, mostly in the C-terminal region. Cdt1 degradation was also induced by DNA damaging chemicals such as methyl methanesulfonate (MMS) or zeocin, depending on PCNA and CRL4-Cdt2, though it was less caffeine-sensitive. These findings suggest that ATR, activated after DNA damage, phosphorylates Cdt2 and promotes the rapid degradation of Cdt1 after UV irradiation in the G1 phase of the cell cycle.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis/efectos de la radiación , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Cafeína/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas Cullin/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...