Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Psychol ; 14: 1140959, 2023.
Article in English | MEDLINE | ID: mdl-37384187

ABSTRACT

Based on previous literature, the present study examines the effects of background music on English reading comprehension using eye tracking techniques. All the participants, whose first language was Chinese, were selected from a foreign language college and all of them were sophomores who majored in English. The experiment in this study was a 2 (music tempo: fast and slow) × 2 (text difficulty: difficult and easy) × 2 (background music preference: high and low) mixed design. Both musical tempo and English reading passage were within-subjects factors, and the level of music listening preference was a between-subjects factor. The results showed that the main effect of the music tempo was statistically significant, which indicated that participants read texts more quickly in the fast-tempo music condition than in the slow-tempo music condition. Furthermore, the main effect of the text difficulty was statistically significant. Additionally, the interaction between the text difficulty and music tempo was statistically significant. The music tempo had a greater effect on easy texts than on difficult texts. The results of this study reveal that it is beneficial for people who have a stronger preference for music listening to conduct English reading tasks with fast-tempo music. It is detrimental for people who have little preference for background music listening to complete difficult English reading tasks with slow-tempo music.

2.
Biochem J ; 473(8): 1027-35, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26846349

ABSTRACT

Historically, drugs used in the treatment of cancers also tend to cause damage to healthy cells while affecting cancer cells. Therefore, the identification of novel agents that act specifically against cancer cells remains a high priority in the search for new therapies. In contrast with normal cells, most cancer cells contain multiple centrosomes which are associated with genome instability and tumorigenesis. Cancer cells can avoid multipolar mitosis, which can cause cell death, by clustering the extra centrosomes into two spindle poles, thereby enabling bipolar division. Kinesin-like protein KIFC1 plays a critical role in centrosome clustering in cancer cells, but is not essential for normal cells. Therefore, targeting KIFC1 may provide novel insight into selective killing of cancer cells. In the present study, we identified a small-molecule KIFC1 inhibitor, SR31527, which inhibited microtubule (MT)-stimulated KIFC1 ATPase activity with an IC50 value of 6.6 µM. By using bio layer interferometry technology, we further demonstrated that SR31527 bound directly to KIFC1 with high affinity (Kd=25.4 nM). Our results from computational modelling and saturation-transfer difference (STD)-NMR experiments suggest that SR31527 bound to a novel allosteric site of KIFC1 that appears suitable for developing selective inhibitors of KIFC1. Importantly, SR31527 prevented bipolar clustering of extra centrosomes in triple negative breast cancer (TNBC) cells and significantly reduced TNBC cell colony formation and viability, but was less toxic to normal fibroblasts. Therefore, SR31527 provides a valuable tool for studying the biological function of KIFC1 and serves as a potential lead for the development of novel therapeutic agents for breast cancer treatment.


Subject(s)
Drug Discovery , Kinesins/antagonists & inhibitors , Kinesins/metabolism , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Discovery/methods , Humans , Kinesins/chemistry , Protein Binding/physiology , Protein Structure, Secondary , Thiadiazoles/pharmacology
3.
J Psycholinguist Res ; 45(4): 945-59, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26187379

ABSTRACT

This study examined the mediating role of working memory (WM) in the relation between rapid automatized naming (RAN) and Chinese reading comprehension. Three tasks assessing differentially visual and verbal components of WM were programmed by E-prime 2.0. Data collected from 55 Chinese college students were analyzed using correlations and hierarchical regression methods to determine the connection among RAN, reading comprehension, and WM components. Results showed that WM played a significant mediating role in the RAN-reading relation and that auditory WM made stronger contributions than visual WM. Taking into account of the multi-component nature of WM and the specificity of Chinese reading processing, this study discussed the mediating powers of the WM components, particularly auditory WM, further clarifying the possible components involved in the RAN-reading relation and thus providing some insight into the complicated Chinese reading process.


Subject(s)
Comprehension/physiology , Memory, Short-Term/physiology , Psycholinguistics , Reading , Adult , China , Humans , Young Adult
4.
Mol Cell Biol ; 33(23): 4685-700, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24081328

ABSTRACT

Our previous study showed that Akt phosphorylates TopBP1 at the Ser-1159 residue and induces its oligomerization. Oligomerization is required for TopBP1 to bind and repress E2F1 activity. However, the mechanism through which phosphorylation of TopBP1 by Akt leads to its oligomerization remains to be determined. Here, we demonstrate that binding between the phosphorylated Ser-1159 (pS1159) residue and the 7th and 8th BRCT domains of TopBP1 mediates TopBP1 oligomerization. Mutations within the 7th and 8th BRCT domains of TopBP1 that block binding to a pS1159-containing peptide block TopBP1 oligomerization and its ability to bind and repress E2F1 activities. The Akt-induced TopBP1 oligomerization is also directly demonstrated in vitro by size exclusion chromatography. Importantly, oligomerization perturbs the checkpoint-activating function of TopBP1 by preventing its recruitment to chromatin and ATR binding upon replicative stress. Hyperactivation of Akt inhibits Chk1 phosphorylation after hydroxyurea treatment, and this effect is dependent on TopBP1 phosphorylation at Ser-1159. Thus, Akt can switch the TopBP1 function from checkpoint activation to transcriptional regulation by regulating its quaternary structure. This pathway of regulation is clinically significant, since treatment of a specific Akt inhibitor in PTEN-mutated cancer cells inhibits TopBP1 oligomerization and causes its function to revert from promoting survival to checkpoint activation.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Checkpoints , DNA-Binding Proteins/physiology , Gene Expression Regulation , Nuclear Proteins/physiology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Substitution , Carrier Proteins/chemistry , Cell Line, Tumor , Chromatin/metabolism , DNA-Binding Proteins/chemistry , E2F1 Transcription Factor/metabolism , HEK293 Cells , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Hydroxyurea/pharmacology , Molecular Dynamics Simulation , Nuclear Proteins/chemistry , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Serine/metabolism , Stress, Physiological , Transcription, Genetic
5.
ChemMedChem ; 8(10): 1629-33, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23894090

ABSTRACT

Singling out the truth: A combined application of STD-NMR, molecular docking, and CORCEMA-ST calculations is described as an attractive, easily applicable tool for the identification and validation of the binding site for allosteric ligands, with potential application as an aid in drug discovery research.


Subject(s)
Ligands , Molecular Docking Simulation , Nuclear Magnetic Resonance, Biomolecular , Allosteric Regulation , Binding Sites , Humans , Kinesins/chemistry , Kinesins/metabolism , Kinetics , Protein Binding , Protein Structure, Tertiary
6.
Cancer Res ; 68(15): 6232-40, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676847

ABSTRACT

Clinical studies have shown that decreased tamoxifen effectiveness correlates with elevated levels of vascular endothelial growth factor (VEGF)-A(165) in biopsy samples of breast cancers. To investigate the mechanisms underlying tamoxifen resistance and metastasis, we engineered the estrogen receptor (ER)-positive MCF-7 human breast cancer cell line to express VEGF to clinically relevant levels in a doxycycline-regulated manner. Induction of VEGF expression in orthotopically implanted xenografts that were initially tamoxifen responsive and noninvasive resulted in tamoxifen-resistant tumor growth and metastasis to the lungs. Lung metastases were also observed in a VEGF-dependent manner following tail vein injection of tumor cells. At both primary and metastatic sites, VEGF-overexpressing tumors exhibited extensive fibroblastic stromal content, a clinical feature called desmoplasia. VEGF-induced metastatic colonies were surrounded by densely packed stromal cells before detectable angiogenesis, suggesting that VEGF is involved in the initiation of desmoplasia. Because expression of VEGF receptors R1 and R2 was undetectable in these tumor cells, the observed VEGF effects on reduction of tamoxifen efficacy and metastatic colonization are most likely mediated by paracrine signaling that enhances tumor/stromal cell interactions and increases the level of desmoplasia. This study reveals new roles for VEGF in breast cancer progression and suggests that combination of antiestrogens and VEGF inhibitors may prolong tamoxifen sensitivity and prevent metastasis in patients with ER-positive tumors.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Neoplasm Metastasis , Tamoxifen/pharmacology , Vascular Endothelial Growth Factor A/physiology , Humans , Signal Transduction/drug effects
7.
Proteins ; 67(3): 709-19, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17335005

ABSTRACT

Atypical protein kinase C zeta (PKCzeta) plays an important role in cell proliferation and survival. PKCzeta and its truncated form containing only the kinase domain, CATzeta, have been reported to be activated by the phosphorylation of threonine 410 in the activation loop. We expressed both the full length PKCzeta and CATzeta in a baculovirus/insect cell over-expression system and purified the proteins for biochemical characterization. Ion exchange chromatography of CATzeta revealed three species with different levels of phosphorylation at Thr-410 and allowed the isolation of the CATzeta protein devoid of phosphorylation at Thr-410. All three species of CATzeta were active and their activity was not correlated with phosphorylation at Thr-410, indicating that the kinase activity of CATzeta did not depend solely on activation loop phosphorylation. Tyrosine phosphorylation was detected in all three species of CATzeta and the full length PKCzeta. Homology structural modeling of PKCzeta revealed a conserved, predicted-to-be phosphorylated tyrosine residue, Tyr-428, in the close proximity of the RD motif of the catalytic loop and of Thr-410 in the activation loop. The structural analysis indicated that phospho-Tyr-428 would interact with two key, positively-charged residues to form a triad conformation similar to that formed by phospho-Thr-410. Based on these observations, it is possible that the Thr-410 phosphorylation-independent kinase activity of CATzeta is regulated by the phosphorylation of Tyr-428. This alternative mode of PKCzeta activation is supported by the observed stimulation of PKCzeta kinase activity upon phosphorylation at the equivalent site by Abl, and may be involved in resistance to drug-induced apoptosis.


Subject(s)
Protein Kinase C/chemistry , Protein Kinase C/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , Catalytic Domain , Cell Line , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spodoptera , Structure-Activity Relationship , Threonine/chemistry , Threonine/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
8.
Mol Cancer Ther ; 5(9): 2218-24, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985055

ABSTRACT

4'-Thio-beta-D-arabinofuranosylcytosine (T-araC), a new-generation deoxycytidine nucleoside analogue, showed significant efficacy against numerous solid tumors in preclinical studies and entered clinical development for cancer therapy. It is a structural analogue of cytarabine (araC), a clinically used drug in the treatment of acute myelogenous leukemia, which has no or very limited efficacy against solid tumors. In comparison with araC, the excellent in vivo activity of T-araC against solid tumors suggests that, in addition to inhibition of DNA synthesis, T-araC may target cellular signaling pathways, such as angiogenesis, in solid tumors. We studied T-araC and araC for their antiangiogenic activities in vitro and in vivo. Both compounds inhibited human endothelial cell proliferation with similar IC50s. However, only T-araC inhibited endothelial cell migration and differentiation into capillary tubules. T-araC also abrogated endothelial cell extracellular signal-regulated kinase (ERK) 1/2 phosphorylation, a key signaling molecule involved in cellular processes of angiogenesis. Results from chick chorioallantoic membrane angiogenesis assays revealed that T-araC significantly inhibited the development of new blood vessels in vivo, whereas araC showed much less effect. The findings of this study show a role of T-araC in antiangiogenesis and suggest that T-araC combines antiproliferative and antiangiogenic activity in one molecule for a dual mechanism of drug action to achieve the excellent in vivo efficacy against several solid tumors. This study also provides important information for optimizing dosage and sequence of T-araC administration in clinical investigations by considering T-araC as both an antiproliferative and an antiangiogenic agent.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Arabinonucleosides/pharmacology , Animals , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/blood supply , Endothelial Cells/drug effects , Humans , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/drug therapy
9.
Article in English | MEDLINE | ID: mdl-16511094

ABSTRACT

Adenosine kinase from Mycobacterium tuberculosis is the only prokaryotic adenosine kinase that has been isolated and characterized. The enzyme catalyzes the phosphorylation of adenosine to adenosine monophosphate and is involved in the activation of 2-methyladenosine, a compound that has demonstrated selective activity against M. tuberculosis. The mechanism of action of 2-methyladenosine is likely to be different from those of current tuberculosis treatments and this compound (or other adenosine analogs) may prove to be a novel therapeutic intervention for this disease. The M. tuberculosis adenosine kinase was overexpressed in Escherichia coli and the enzyme was purified with activity comparable to that reported previously. The protein was crystallized in the presence of adenosine using the vapour-diffusion method. The crystals diffracted X-rays to high resolution and a complete data set was collected to 2.2 A using synchrotron radiation. The crystal belonged to space group P3(1)21, with unit-cell parameters a = 70.2, c = 111.6 A, and contained a single protein molecule in the asymmetric unit. An initial structural model of the protein was obtained by the molecular-replacement method, which revealed a dimeric structure. The monomers of the dimer were related by twofold crystallographic symmetry. An understanding of how the M. tuberculosis adenosine kinase differs from the human homolog should aid in the design of more potent and selective antimycobacterial agents that are selectively activated by this enzyme.


Subject(s)
Adenosine Kinase/chemistry , Mycobacterium tuberculosis/enzymology , Adenosine Kinase/genetics , Adenosine Kinase/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Cloning, Molecular , Crystallization/methods , Dimerization , Escherichia coli/genetics , Volatilization , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...