Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
J Am Chem Soc ; 145(6): 3682-3695, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36727591

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-25617833

ABSTRACT

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.


Subject(s)
DNA Helicases/metabolism , DNA/genetics , Homologous Recombination , Nucleic Acid Heteroduplexes/genetics , Rad51 Recombinase/metabolism , Cell Line, Tumor , Chromatids/genetics , Chromatids/radiation effects , DNA Damage , DNA Repair/radiation effects , DNA, Single-Stranded/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Homologous Recombination/radiation effects , Humans , Protein Transport/radiation effects , Rad52 DNA Repair and Recombination Protein/metabolism , Replication Protein A/metabolism
4.
J Biol Chem ; 287(9): 6469-81, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22194613

ABSTRACT

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.


Subject(s)
DNA Damage/physiology , DNA Helicases/metabolism , DNA Replication/physiology , Replication Protein A/metabolism , Stress, Physiological/physiology , Amino Acid Sequence , Chromosomes/physiology , DNA Helicases/chemistry , DNA Helicases/genetics , HCT116 Cells , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Osteosarcoma , Protein Folding , Protein Interaction Domains and Motifs/physiology , Replication Protein A/chemistry , Replication Protein A/genetics , S Phase Cell Cycle Checkpoints/physiology
5.
Mol Biol Cell ; 15(7): 3320-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15146062

ABSTRACT

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.


Subject(s)
Cell Cycle , DNA Damage , DNA Helicases/analysis , Amino Acid Motifs/genetics , Amino Acid Sequence , CDC2-CDC28 Kinases/analysis , CDC2-CDC28 Kinases/metabolism , Camptothecin/pharmacology , Cyclin-Dependent Kinase 2 , DNA/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Mutational Analysis , G1 Phase/physiology , Humans , Intracellular Space/immunology , Intracellular Space/ultrastructure , Mitomycin/pharmacology , Molecular Sequence Data , Phosphorylation , Serine/genetics , Serine/metabolism
6.
Protein Pept Lett ; 9(3): 265-8, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12144524

ABSTRACT

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.


Subject(s)
Chitinases/chemistry , Fabaceae/enzymology , Amino Acid Sequence , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL