Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
J Am Chem Soc ; 145(6): 3682-3695, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36727591

RESUMEN

With easily accessible and operator-friendly reagents, shelf-stable ortho-methoxycarbonylethynylphenyl thioglycosides were efficiently prepared. Based on these MCEPT glycoside donors, a novel glycosylation protocol featuring mild and catalytic promotion conditions with Au(I) or Cu(II) complexes, expanded substrate scope encompassing challenging donors and acceptors and clinically used pharmaceuticals, and versatility in various strategies for highly efficient synthesis of glycosides has been established. The practicality of the MCEPT glycosylation protocol was fully exhibited by highly efficient and scalable synthesis of surface polysaccharide subunits of Acinetobacter baumannii via latent-active, reagent-controlled divergent orthogonal one-pot and orthogonal one-pot strategies. The underlying reaction mechanism was investigated systematically through control reactions, leading to the isolation and characterization of the vital catalyst species in MCEPT glycosylation, the benzothiophen-3-yl-gold(I) complex. Based on the results obtained both from control reactions and from studies leading to the glycosylation protocol establishment, an operative mechanism was proposed and the effect of the vital catalyst species reactivity on the results of metal-catalyzed alkyne-containing donor-involved glycosylation was disclosed. Moreover, the mechanism for C-glycosylation side product formation from ortho-(substituted)ethynylphenyl thioglycoside donors with electron-donating substituents was also illuminated.

3.
PLoS One ; 10(1): e0116852, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25617833

RESUMEN

Homologous recombination is involved in the repair of DNA damage and collapsed replication fork, and is critical for the maintenance of genomic stability. Its process involves a network of proteins with different enzymatic activities. Human DNA helicase B (HDHB) is a robust 5'-3' DNA helicase which accumulates on chromatin in cells exposed to DNA damage. HDHB facilitates cellular recovery from replication stress, but its role in DNA damage response remains unclear. Here we report that HDHB silencing results in reduced sister chromatid exchange, impaired homologous recombination repair, and delayed RPA late-stage foci formation induced by ionizing radiation. Ectopically expressed HDHB colocalizes with Rad51, Rad52, RPA, and ssDNA. In vitro, HDHB stimulates Rad51-mediated heteroduplex extension in 5'-3' direction. A helicase-defective mutant HDHB failed to promote this reaction. Our studies implicate HDHB promotes homologous recombination in vivo and stimulates 5'-3' heteroduplex extension during Rad51-mediated strand exchange in vitro.


Asunto(s)
ADN Helicasas/metabolismo , ADN/genética , Recombinación Homóloga , Ácidos Nucleicos Heterodúplex/genética , Recombinasa Rad51/metabolismo , Línea Celular Tumoral , Cromátides/genética , Cromátides/efectos de la radiación , Daño del ADN , Reparación del ADN/efectos de la radiación , ADN de Cadena Simple/genética , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Recombinación Homóloga/efectos de la radiación , Humanos , Transporte de Proteínas/efectos de la radiación , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Proteína de Replicación A/metabolismo
4.
J Biol Chem ; 287(9): 6469-81, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22194613

RESUMEN

Maintenance of genomic stability in proliferating cells depends on a network of proteins that coordinate chromosomal replication with DNA damage responses. Human DNA helicase B (HELB or HDHB) has been implicated in chromosomal replication, but its role in this coordinated network remains undefined. Here we report that cellular exposure to UV irradiation, camptothecin, or hydroxyurea induces accumulation of HDHB on chromatin in a dose- and time-dependent manner, preferentially in S phase cells. Replication stress-induced recruitment of HDHB to chromatin is independent of checkpoint signaling but correlates with the level of replication protein A (RPA) recruited to chromatin. We show using purified proteins that HDHB physically interacts with the N-terminal domain of the RPA 70-kDa subunit (RPA70N). NMR spectroscopy and site-directed mutagenesis reveal that HDHB docks on the same RPA70N surface that recruits S phase checkpoint signaling proteins to chromatin. Consistent with this pattern of recruitment, cells depleted of HDHB display reduced recovery from replication stress.


Asunto(s)
Daño del ADN/fisiología , ADN Helicasas/metabolismo , Replicación del ADN/fisiología , Proteína de Replicación A/metabolismo , Estrés Fisiológico/fisiología , Secuencia de Aminoácidos , Cromosomas/fisiología , ADN Helicasas/química , ADN Helicasas/genética , Células HCT116 , Células HeLa , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Osteosarcoma , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas/fisiología , Proteína de Replicación A/química , Proteína de Replicación A/genética , Puntos de Control de la Fase S del Ciclo Celular/fisiología
5.
Mol Biol Cell ; 15(7): 3320-32, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15146062

RESUMEN

Mutational studies of human DNA helicase B (HDHB) have suggested that its activity is critical for the G1/S transition of the cell cycle, but the nature of its role remains unknown. In this study, we show that during G1, ectopically expressed HDHB localizes in nuclear foci induced by DNA damaging agents and that this focal pattern requires active HDHB. During S and G2/M, HDHB localizes primarily in the cytoplasm. A carboxy-terminal domain from HDHB confers cell cycle-dependent localization, but not the focal pattern, to a reporter protein. A cluster of potential cyclin-dependent kinase phosphorylation sites in this domain was modified at the G1/S transition and maintained through G2/M of the cell cycle in vivo, coincident with nuclear export of HDHB. Serine 967 of HDHB was the major site phosphorylated in vivo and in vitro by cyclin-dependent kinases. Mutational analysis demonstrated that phosphorylation of serine 967 is crucial in regulating the subcellular localization of ectopically expressed HDHB. We propose that the helicase of HDHB operates primarily during G1 to process endogenous DNA damage before the G1/S transition, and it is largely sequestered in the cytoplasm during S/G2.


Asunto(s)
Ciclo Celular , Daño del ADN , ADN Helicasas/análisis , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Quinasas CDC2-CDC28/análisis , Quinasas CDC2-CDC28/metabolismo , Camptotecina/farmacología , Quinasa 2 Dependiente de la Ciclina , ADN/efectos de los fármacos , ADN Helicasas/genética , ADN Helicasas/metabolismo , Análisis Mutacional de ADN , Fase G1/fisiología , Humanos , Espacio Intracelular/inmunología , Espacio Intracelular/ultraestructura , Mitomicina/farmacología , Datos de Secuencia Molecular , Fosforilación , Serina/genética , Serina/metabolismo
6.
Protein Pept Lett ; 9(3): 265-8, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12144524

RESUMEN

A new member of class II chitinase from Phaseolus vulgaris was purified and crystallized. Diffraction data to 2.7A resolution have been collected and the preliminary crystallographic studies have been completed. The space group is P1 with unit cell parameters of a=36.32A, b=46.24A, c=70.36A, a =97.9 degrees, b=103.8 degrees and g =110.5 degrees. Molecular replacement and initial refinement statistics indicate there are two chitinase molecules in the crystallographic asymmetric unit.


Asunto(s)
Quitinasas/química , Fabaceae/enzimología , Secuencia de Aminoácidos , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Datos de Secuencia Molecular , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA