Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biochim Biophys Acta Gen Subj ; 1868(3): 130549, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38158023

ABSTRACT

BACKGROUND: Chitosanases (EC 3.2.1.132) hydrolyze chitosan which is a polymer of glucosamine (GlcN) linked by ß - 1,4 bonds, and show cleavage specificity against partially acetylated chitosan containing N-acetylglucosamine (GlcNAc) residues. Chitosanases' structural underpinnings for cleavage specificity and the conformational switch from open to closed structures are still a mystery. METHODS: The GH-46 subclass III chitosanase from Bacillus circulans MH-K1 (MH-K1 chitosanase), which also catalyzes the hydrolysis of GlcN-GlcNAc bonds in addition to GlcN-GlcN, has had its chitotetraose [(GlcN)4]-complexed crystal structure solved at 1.35 Å resolution. RESULTS: The MH-K1 chitosanase's (GlcN)4-bound structure has numerous structural similarities to other GH-46 chitosanases in terms of substrate binding and catalytic processes. However, subsite -1, which is absolutely specific for GlcN, seems to characterize the structure of a subclass III chitosanase due to its distinctive length and angle of a flexible loop. According to a comparison of the (GlcN)4-bound and apo-form structures, the particular binding of a GlcN residue at subsite -2 through Asp77 causes the backbone helix to kink, which causes the upper- and lower-domains to approach closely when binding a substrate. CONCLUSIONS: Although GH-46 chitosanases vary in the finer details of the subsites defining cleavage specificity, they share similar structural characteristics in substrate-binding, catalytic processes, and potentially in conformational change. GENERAL SIGNIFICANCE: The precise binding of a GlcN residue to the -2 subsite is essential for the conformational shift that occurs in all GH-46 chitosanases, as shown by the crystal structures of the apo- and substrate-bound forms of MH-K1 chitosanase.


Subject(s)
Bacillus , Chitosan , Oligosaccharides , Glycoside Hydrolases/metabolism , Glucosamine/metabolism
2.
Acta Crystallogr D Struct Biol ; 79(Pt 5): 435-441, 2023 May 01.
Article in English | MEDLINE | ID: mdl-37098908

ABSTRACT

Structure determination of G-protein-coupled receptors (GPCRs) is key for the successful development of efficient drugs targeting GPCRs. BRIL is a thermostabilized apocytochrome b562 (with M7W/H102I/R106L mutations) from Escherichia coli and is often used as a GPCR fusion protein for expression and crystallization. SRP2070Fab, an anti-BRIL antibody Fab fragment, has been reported to facilitate and enhance the crystallization of BRIL-fused GPCRs as a crystallization chaperone. This study was conducted to characterize the high-resolution crystal structure of the BRIL-SRP2070Fab complex. The structure of the BRIL-SRP2070Fab complex was determined at 2.1 Šresolution. This high-resolution structure elucidates the binding interaction between BRIL and SRP2070Fab. When binding to BRIL, SRP2070Fab recognizes conformational epitopes, not linear epitopes, on the surface of BRIL helices III and IV, thereby binding perpendicularly to the helices, which indicates stable binding. Additionally, the packing contacts of the BRIL-SRP2070Fab co-crystal are largely due to SRP2070Fab rather than BRIL. The accumulation of SRP2070Fab molecules by stacking is remarkable and is consistent with the finding that stacking of SRP2070Fab is predominant in known crystal structures of BRIL-fused GPCRs complexed with SRP2070Fab. These findings clarified the mechanism of SRP2070Fab as a crystallization chaperone. Moreover, these data will be useful in the structure-based drug design of membrane-protein drug targets.


Subject(s)
Immunoglobulin Fab Fragments , Receptors, G-Protein-Coupled , Crystallization , Receptors, G-Protein-Coupled/chemistry , Protein Structure, Secondary , Immunoglobulin Fab Fragments/chemistry , Escherichia coli , Epitopes
3.
Mol Pharmacol ; 103(6): 311-324, 2023 06.
Article in English | MEDLINE | ID: mdl-36894319

ABSTRACT

KW-6356 is a novel adenosine A2A (A2A) receptor antagonist/inverse agonist, and its efficacy as monotherapy in Parkinson's disease (PD) patients has been reported. Istradefylline is a first-generation A2A receptor antagonist approved for use as adjunct treatment to levodopa/decarboxylase inhibitor in adult PD patients experiencing "OFF" episodes. In this study, we investigated the in vitro pharmacological profile of KW-6356 as an A2A receptor antagonist/inverse agonist and the mode of antagonism and compared them with istradefylline. In addition, we determined cocrystal structures of A2A receptor in complex with KW-6356 and istradefylline to explore the structural basis of the antagonistic properties of KW-6356. Pharmacological studies have shown that KW-6356 is a potent and selective ligand for the A2A receptor (the -log of inhibition constant = 9.93 ± 0.01 for human receptor) with a very low dissociation rate from the receptor (the dissociation kinetic rate constant = 0.016 ± 0.006 minute-1 for human receptor). In particular, in vitro functional studies indicated that KW-6356 exhibits insurmountable antagonism and inverse agonism, whereas istradefylline exhibits surmountable antagonism. Crystallography of KW-6356- and istradefylline-bound A2A receptor have indicated that interactions with His2506.52 and Trp2466.48 are essential for the inverse agonism, whereas the interactions at both deep inside the orthosteric pocket and the pocket lid stabilizing the extracellular loop conformation may contribute to the insurmountable antagonism of KW-6356. These profiles may reflect important differences in vivo and help predict better clinical performance. SIGNIFICANCE STATEMENT: KW-6356 is a potent and selective adenosine A2A receptor antagonist/inverse agonist and exhibits insurmountable antagonism, whereas istradefylline, a first-generation adenosine A2A receptor antagonist, exhibits surmountable antagonism. Structural studies of adenosine A2A receptor in complex with KW-6356 and istradefylline explain the characteristic differences in the pharmacological properties of KW-6356 and istradefylline.


Subject(s)
Adenosine A2 Receptor Antagonists , Drug Inverse Agonism , Parkinson Disease , Receptor, Adenosine A2A , Humans , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Levodopa/pharmacology , Levodopa/therapeutic use , Receptor, Adenosine A2A/physiology
4.
Am J Cancer Res ; 11(1): 264-276, 2021.
Article in English | MEDLINE | ID: mdl-33520373

ABSTRACT

The Wnt/ß-catenin pathway, which is associated with disease progression, is activated in many cancers. Tankyrase (TNKS) has received attention as a target molecule for Wnt/ß-catenin pathway inhibition. We identified K-476, a novel TNKS inhibitor, a dual pocket binder that binds to both the nicotinamide and ADP-ribose pockets. In a human colon cancer cell line, K-476 specifically and potently inhibited TNKS and led to stabilization of the Axin protein, resulting in Wnt/ß-catenin pathway suppression. Aberrant Wnt/ß-catenin pathway activation was recently reported as a possible mechanism of ineffectiveness in immune checkpoint inhibitor (ICI) treatment. Because the Wnt/ß-catenin pathway activation causes dendritic cell inactivation and suppresses chemokine production, resulting in a paucity of CD8+ T cells in tumor tissue, which is an important effector of ICIs. Thus, TNKS inhibitors may enhance the efficacy of ICIs. To examine whether K-476 enhances the antitumor effect of anti-PD-L1 antibodies, K-476 was administered orally with an anti-PD-L1 antibody to melanoma-bearing C57BL/6J mice. Although K-476 was ineffective as a monotherapy, it significantly enhanced the antitumor effect in combination with anti-PD-L1 antibody. In mice, intra-tumor infiltration of CD8+ T cells was increased by combination treatment. K-476 upregulated the chemokine expression (e.g., Ccl3 and Ccl4), which attracted CD8+ T cells. This was considered to contribute to the increased CD8+ T cells in the tumor microenvironment. Furthermore, while the potential gastrointestinal toxicity of TNKS inhibitors has been reported, it was not observed at effective doses. Thus, K-476 could be an attractive therapeutic option to enhance the efficacy of ICIs.

5.
RNA ; 27(2): 151-162, 2021 02.
Article in English | MEDLINE | ID: mdl-33177187

ABSTRACT

A key approach for improving siRNA efficacy is chemical modifications. Through an in silico screening of modifications at the 5'-end nucleobase of the guide strand, an adenine-derived compound called 6-(3-(2-carboxyethyl)phenyl)-purine (6-mCEPh-purine) was identified to improve the RNAi activity in cultured human cells and in vivo mouse models. Nevertheless, it remains unclear how this chemical modification enhances the siRNA potency. Here, we used a series of biochemical approaches to quantitatively evaluate the effect of the 6-mCEPh-purine modification at each step in the assembly of the RNAi effector complex called RISC. We found that the modification improves the formation of mature RISC at least in two different ways, by fixing the loading orientation of siRNA duplexes and increasing the stability of mature RISC after passenger strand ejection. Our data will provide a molecular platform for further development of chemically modified siRNA drugs.


Subject(s)
Adenine/pharmacology , Argonaute Proteins/genetics , RNA Interference/drug effects , RNA, Double-Stranded/genetics , RNA, Small Interfering/agonists , RNA-Induced Silencing Complex/agonists , Adenine/analogs & derivatives , Adenine/chemical synthesis , Argonaute Proteins/metabolism , Base Pairing , Base Sequence , HEK293 Cells , Humans , Methylation , Protein Binding , RNA, Double-Stranded/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism
6.
RNA ; 27(2): 163-173, 2021 02.
Article in English | MEDLINE | ID: mdl-33177188

ABSTRACT

Small interfering RNAs (siRNAs) can be utilized not only as functional biological research tools but also as therapeutic agents. For the clinical use of siRNA as drugs, various chemical modifications have been used to improve the activity of siRNA drugs, and further chemical modifications are expected to improve the utility of siRNA therapeutics. As the 5' nucleobase of the guide strand affects the interaction between an siRNA and AGO2 and target cleavage activity, structural optimization of this specific position may be a useful strategy for improving siRNA activity. Here, using the in silico model of the complex between human AGO2 MID domain and nucleoside monophosphates, we screened and synthesized an original adenine-derived analog, 6-(3-(2-carboxyethyl)phenyl)purine (6-mCEPh-purine), that fits better than the natural nucleotide bases into the MID domain of AGO2. Introduction of the 6-mCEPh-purine analog at the 5'-end of the siRNA guide strand significantly enhanced target knockdown activity in both cultured cell lines and in vivo animal models. Our findings can help expand strategies for rationally optimizing siRNA activity via chemical modifications of nucleotide bases.


Subject(s)
Adenine/pharmacology , Argonaute Proteins/genetics , RNA Interference/drug effects , RNA, Double-Stranded/genetics , RNA, Small Interfering/agonists , RNA-Induced Silencing Complex/agonists , Adenine/analogs & derivatives , Adenine/chemical synthesis , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Animals , Apolipoprotein B-100/antagonists & inhibitors , Apolipoprotein B-100/blood , Apolipoprotein B-100/chemistry , Apolipoprotein B-100/genetics , Argonaute Proteins/metabolism , Base Pairing , Base Sequence , Binding Sites , Cholesterol/blood , HeLa Cells , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Male , Methylation , Mice , Mice, Knockout , Models, Molecular , Protein Binding , RNA, Double-Stranded/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Uridine Monophosphate/chemistry , Uridine Monophosphate/metabolism
7.
PLoS One ; 15(7): e0236710, 2020.
Article in English | MEDLINE | ID: mdl-32735606

ABSTRACT

Argonaute (AGO) proteins are the key component of the RNA interference machinery that suppresses gene expression by forming an RNA-induced silencing complex (RISC) with microRNAs (miRNAs). Each miRNA is involved in various cellular processes, such as development, differentiation, tumorigenesis, and viral infection. Thus, molecules that regulate miRNA function are expected to have therapeutic potential. In addition, the biogenesis of miRNA is a multistep process involving various proteins, although the complete pathway remains to be elucidated. Therefore, identification of molecules that can specifically modulate each step will help understand the mechanism of gene suppression. To date, several AGO2 inhibitors have been identified. However, these molecules were identified through a single screening method, and no studies have specifically evaluated a combinatorial strategy. Here, we demonstrated a combinatorial screening (SCR) approach comprising an in silico molecular docking study, surface plasmon resonance (SPR) analysis, and nuclear magnetic resonance (NMR) analysis, focusing on the strong binding between the 5'-terminal phosphate of RNA and the AGO2 middle (MID) domain. By combining SPR and NMR, we identified binding modes of amino acid residues binding to AGO2. First, using a large chemical library (over 6,000,000 compounds), 171 compounds with acidic functional groups were screened using in silico SCR. Next, we constructed an SPR inhibition system that could analyze only the 5'-terminal binding site of RNA, and nine molecules that strongly bound to the AGO2 MID domain were selected. Finally, using NMR, three molecules that bound to the desired site were identified. The RISC inhibitory ability of the "hit" compounds was analyzed in human cell lysate, and all three hit compounds strongly inhibited the binding between double-stranded RNA and AGO2.


Subject(s)
Argonaute Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Magnetic Resonance Spectroscopy , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Surface Plasmon Resonance , Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Computer Simulation , HeLa Cells , Humans , Molecular Docking Simulation , Protein Conformation , Small Molecule Libraries/metabolism
8.
Sci Rep ; 10(1): 11669, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32669569

ABSTRACT

G-protein-coupled receptors (GPCRs)-the largest family of cell-surface membrane proteins-mediate the intracellular signal transduction of many external ligands. Thus, GPCRs have become important drug targets. X-ray crystal structures of GPCRs are very useful for structure-based drug design (SBDD). Herein, we produced a new antibody (SRP2070) targeting the thermostabilised apocytochrome b562 from Escherichia coli M7W/H102I/R106L (BRIL). We found that a fragment of this antibody (SRP2070Fab) facilitated the crystallisation of the BRIL-tagged, ligand bound GPCRs, 5HT1B and AT2R. Furthermore, the electron densities of the ligands were resolved, suggesting that SPR2070Fab is versatile and adaptable for GPCR SBDD. We anticipate that this new tool will significantly accelerate structure determination of other GPCRs and the design of small molecular drugs targeting them.


Subject(s)
Antibodies, Monoclonal/chemistry , Cytochrome b Group/chemistry , Escherichia coli Proteins/chemistry , Immunoglobulin Fab Fragments/chemistry , Receptor, Angiotensin, Type 2/chemistry , Receptor, Serotonin, 5-HT1B/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Angiotensin II/chemistry , Angiotensin II/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Ergotamine/chemistry , Ergotamine/metabolism , Escherichia coli/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/metabolism , Mice , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/metabolism , Receptor, Serotonin, 5-HT1B/genetics , Receptor, Serotonin, 5-HT1B/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sf9 Cells , Spodoptera
9.
Am J Cancer Res ; 10(12): 4399-4415, 2020.
Article in English | MEDLINE | ID: mdl-33415007

ABSTRACT

The Hippo signaling pathway regulates cell fate and organ development. In the Hippo pathway, transcriptional enhanced associate domain (TEAD) which is a transcription factor is activated by forming a complex with yes-associated protein 1 (YAP1) or transcriptional coactivator with PDZ-binding motif (TAZ, also called WWTR1). Hyper-activation of YAP1/TAZ, leading to the activation of TEAD, has been reported in many cancers, including malignant pleural mesothelioma (MPM). Therefore, the YAP1/TAZ-TEAD complex is considered a novel therapeutic target for cancer treatment. However, few reports have described YAP1/TAZ-TEAD inhibitors, and their efficacy and selectivity are poor. In this study, we performed a high-throughput screening of a neurofibromin 2 (NF2)-deficient MPM cell line and a large tumor suppressor kinase 1/2 (LATS1/2)-deficient non-small-cell lung cancer cell line using a transcriptional reporter assay. After screening and optimization, K-975 was successfully identified as a potent inhibitor of YAP1/TAZ-TEAD signaling. X-ray crystallography revealed that K-975 was covalently bound to an internal cysteine residue located in the palmitate-binding pocket of TEAD. K-975 had a strong inhibitory effect against protein-protein interactions between YAP1/TAZ and TEAD in cell-free and cell-based assays. Furthermore, K-975 potently inhibited the proliferation of NF2-non-expressing MPM cell lines compared with NF2-expressing MPM cell lines. K-975 also suppressed tumor growth and provided significant survival benefit in MPM xenograft models. These findings indicate that K-975 is a strong and selective TEAD inhibitor with the potential to become an effective drug candidate for MPM therapy.

10.
Bioorg Med Chem ; 27(22): 115122, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31623970

ABSTRACT

We previously identified dibenzooxepine derivative 1 as a potent PPARγ ligand with a unique binding mode owing to its non-thiazolidinedione scaffold. However, while 1 showed remarkably potent MKN-45 gastric cancer cell aggregation activity, an indicator of cancer differentiation-inducing activity induced by PPARγ activation, we recognized that 1 was metabolically unstable. In the present study, we identified a metabolically soft spot, and successfully discovered 3-fluoro dibenzooxepine derivative 9 with better metabolic stability. Further optimization provided imidazo[1,2-a]pyridine derivative 17, which showed potent MKN-45 gastric cancer cell aggregation activity and excellent PK profiles compared with 9. Compound 17 exerted a growth inhibitory effect on AsPC-1/AG1 pancreatic tumor in mice. Furthermore, the decrease in the hematocrit (an indicator of localized edema, a serious adverse effect of PPARγ ligands) was tolerable even with oral administration at 200 mg/kg in healthy mice.


Subject(s)
Antineoplastic Agents/therapeutic use , PPAR gamma/therapeutic use , Antineoplastic Agents/pharmacology , Humans , Ligands , PPAR gamma/pharmacology
11.
J Med Chem ; 61(22): 10067-10083, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30351933

ABSTRACT

A novel class of PPARγ ligand 1 (EC50 = 197 nM) with a dibenzoazepin scaffold was identified through high-throughput screening campaign. To avoid the synthetically troublesome chiral center of 1, its conformational analysis using the MacroModel was conducted, focusing on conformational flip of the tricyclic ring and the conformational restriction by the methyl group at the chiral center. On the basis of this analysis, scaffold hopping of dibenzoazepine into dibenzo[ b, e]oxepine by replacing the chiral structures with the corresponding olefinic E/ Z isomers was performed. Consequently, dibenzo[ b, e]oxepine scaffold 9 was developed showing extremely potent PPARγ reporter activity (EC50 = 2.4 nM, efficacy = 9.5%) as well as differentiation-inducing activity against a gastric cancer cell line MKN-45 that was more potent than any other well-known PPARγ agonists in vitro (94% at 30 nM). The X-ray crystal structure analysis of 9 complexed with PPARγ showed that it had a unique binding mode to PPARγ ligand-binding domain that differed from that of any other PPARγ agonists identified thus far.


Subject(s)
Alkenes/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Drug Design , Oxepins/metabolism , Oxepins/pharmacology , PPAR gamma/metabolism , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Line, Tumor , Humans , Ligands , Models, Molecular , Oxepins/chemistry , PPAR gamma/chemistry , Protein Binding , Protein Domains , Stereoisomerism , Structure-Activity Relationship
12.
Mol Cancer Ther ; 15(7): 1525-34, 2016 07.
Article in English | MEDLINE | ID: mdl-27196752

ABSTRACT

The Wnt/ß-catenin pathway is a well-known oncogenic pathway. Its suppression has long been considered as an important challenge in treating cancer patients. Among colon cancer patients in particular, most patients carry an adenomatous polyposis coli (APC) mutation that leads to an aberration of Wnt/ß-catenin pathway. To discover the small molecule inhibitors of the Wnt/ß-catenin pathway, we conducted high-throughput screening in APC-mutant colon cancer DLD-1 cells using a transcriptional reporter assay, which identified a selective Wnt/ß-catenin pathway inhibitor, K-756. K-756 stabilizes Axin and reduces active ß-catenin, and inhibits the genes downstream of endogenous Wnt/ß-catenin. We subsequently identified that K-756 is a tankyrase (TNKS) inhibitor. TNKS, which belongs to the PARP family, poly-ADP ribosylates Axin and promotes Axin degradation via the proteasome pathway. K-756 binds to the induced pocket of TNKS and inhibits its enzyme activity. Moreover, PARP family enzyme assays showed that K-756 is a selective TNKS inhibitor. K-756 inhibited the cell growth of APC-mutant colorectal cancer COLO 320DM and SW403 cells by inhibiting the Wnt/ß-catenin pathway. An in vivo study showed that the oral administration of K-756 inhibited the Wnt/ß-catenin pathway in colon cancer xenografts in mice. To further explore the therapeutic potential of K-756, we also evaluated the effects of K-756 in non-small cell lung cancer cells. Although a single treatment of K-756 did not induce antiproliferative activity, when K-756 was combined with an EGFR inhibitor (gefitinib), it showed a strong synergistic effect. Therefore, K-756, a novel selective TNKS inhibitor, could be a leading compound in the development of anticancer agents. Mol Cancer Ther; 15(7); 1525-34. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Tankyrases/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Animals , Antineoplastic Agents/chemistry , Axin Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Gene Expression Profiling , Genes, APC , Humans , Mice , Molecular Conformation , Mutation , Protein Stability , RNA, Small Interfering/genetics , Tankyrases/chemistry , Transcriptome , beta Catenin/genetics
13.
J Phys Chem B ; 120(14): 3527-39, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27008416

ABSTRACT

Suspension state of a titanium nanoparticle in the liquid sodium was quantum chemically characterized by comparing physical characteristics, viz., electronic state, viscosity, and surface tension, with those of liquid sodium. The exterior titanium atoms on the topmost facet of the nanoparticle were found to constitute a stable Na-Ti layer, and the Brownian motion of a titanium nanoparticle could be seen in tandem with the surrounding sodium atoms. An electrochemical gradient due to the differences in electronegativity of both titanium and sodium causes electron flow from liquid sodium atoms to a titanium nanoparticle, Ti + Na → Ti(δ-) + Na(δ+), making the exothermic reaction possible. In other words, the titanium nanoparticle takes a role as electron-reservoir by withdrawing free electrons from sodium atoms and makes liquid sodium electropositive. The remaining electrons in the liquid sodium still make Na-Na bonds and become more stabilized. With increasing size of the titanium nanoparticle, the deeper electrostatic potential, the steeper electric field, and the larger Debye atmosphere are created in the electric double layer shell. Owing to electropositive sodium-to-sodium electrostatic repulsion between the external shells, naked titanium nanoparticles cannot approach each other, thus preventing the agglomeration.

14.
J Radiat Res ; 56(4): 663-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25887043

ABSTRACT

The mammalian target of rapamycin (mTOR) correlates with cell survival under hypoxia and regulates hypoxia-inducible factor-1α (HIF-1α), a key protein in hypoxia-related events. However, the role of mTOR in radio-resistance has not been fully investigated. Therefore, the effect of mTOR on the radio-resistance of cancer cells under hypoxia was evaluated using the mTOR inhibitor temsirolimus. Clonogenic survival was examined in the A549 human lung adenocarcinoma cell line under normoxia or hypoxia, with or without temsirolimus. An oxygen enhancement ratio (OER) was calculated using the D(10) values, the doses giving 10% survival. Western blotting was performed to investigate the effect of temsirolimus on mTOR and the HIF-1α pathway under normoxia and hypoxia. A549 cells showed a radio-resistance of 5.1 and 14.2 Gy, as indicated by D(10) values under normoxia and hypoxia, respectively; the OER was 2.8. The cell survival rates under hypoxia and with temsirolimus remarkably decreased compared with those under normoxia. The D(10) values of the cells under normoxia and hypoxia were 4.8 and 5.4 Gy, respectively (OER = 1.1). mTOR expression was suppressed by temsirolimus under both normoxia and hypoxia. HIF-1α expression decreased under hypoxia in the presence of temsirolimus. These results suggest that temsirolimus can overcome the radio-resistance induced by hypoxia. When the fact that mTOR acts upstream of HIF-1α is considered, our data suggest that the restoration of radiation sensitivity by temsirolimus under hypoxia may be associated with the suppression of the HIF-1α pathway. Temsirolimus could therefore be used as a hypoxic cell radio-sensitizer.


Subject(s)
Cell Hypoxia/radiation effects , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Radiation-Sensitizing Agents/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Hypoxia/drug effects , Cell Line, Tumor , Dose-Response Relationship, Radiation , Humans , Radiation Tolerance , Radiotherapy Dosage , Sirolimus/administration & dosage , Treatment Outcome
15.
Anticancer Res ; 34(9): 5099-104, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25202098

ABSTRACT

AIM: To evaluate dosimetric differences between carbon ion radiotherapy (C-ion RT) and stereotactic body radiotherapy (SBRT) for stage I non-small cell lung cancer (NSCLC). PATIENTS AND METHODS: Thirteen stage I NSCLC cases were planned with C-ion RT and SBRT. Prescription of the dose and fractionation (fr) for stage IA and IB in C-ion RT were 52.8 Gy (RBE)/4fr and 60.0 Gy (RBE)/4fr, respectively and those in SBRT were 52.8 Gy/4fr and 60.0 Gy/4fr, respectively. RESULTS: The conformity index (CI) for planning target volume of C-ion RT was significantly lower than that of SBRT. The normal lung doses in C-ion RT were significantly lower those that in SBRT. In particularly, for a larger tumor, C-ion RT was lower CI and normal lung dose than SBRT. CONCLUSION: C-ion RT has an advantage in both target conformity and sparing of normal lung in stage I NSCLC.


Subject(s)
Heavy Ion Radiotherapy , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Radiometry , Radiosurgery , Carbon Radioisotopes , Combined Modality Therapy , Humans , Neoplasm Staging , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted
16.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 11): 1535-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23090402

ABSTRACT

L-Amino-acid ligases (LALs) are enzymes which catalyze the formation of dipeptides by linking two L-amino acids. Although many dipeptides are known and expected to have medical and nutritional benefits, their practical use has been limited owing to their low availability and high expense. LALs are potentially desirable tools for the efficient production of dipeptides; however, the molecular basis of substrate recognition by LAL has not yet been sufficiently elucidated for the design of ideal LALs for the desired dipeptides. This report presents the crystal structure of the LAL BL00235 derived from Bacillus licheniformis NBRC 12200 determined at 1.9 Å resolution using the multi-wavelength anomalous dispersion method. The overall structure of BL00235 is fairly similar to that of YwfE, the only LAL with a known structure, but the structure around the catalytic site contains some significant differences. Detailed structural comparison of BL00235 with YwfE sheds some light on the molecular basis of the substrate specificities.


Subject(s)
Bacillus/enzymology , Peptide Synthases/chemistry , Adenosine Triphosphate/metabolism , Bacillus/chemistry , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Peptide Synthases/metabolism , Protein Conformation , Substrate Specificity
17.
Protein Sci ; 21(5): 707-16, 2012 May.
Article in English | MEDLINE | ID: mdl-22407814

ABSTRACT

BacD is an ATP-dependent dipeptide ligase responsible for the biosynthesis of L-alanyl-L-anticapsin, a precursor of an antibiotic produced by Bacillus spp. In contrast to the well-studied and phylogenetically related D-alanine: D-alanine ligase (Ddl), BacD synthesizes dipeptides using L-amino acids as substrates and has a low substrate specificity in vitro. The enzyme is of great interest because of its potential application in industrial protein engineering for the environmentally friendly biological production of useful peptide compounds, such as physiologically active peptides, artificial sweeteners and antibiotics, but the determinants of its substrate specificity and its catalytic mechanism have not yet been established due to a lack of structural information. In this study, we report the crystal structure of BacD in complex with ADP and an intermediate analog, phosphorylated phosphinate L-alanyl-L-phenylalanine, refined to 2.5-Å resolution. The complex structure reveals that ADP and two magnesium ions bind in a manner similar to that of Ddl. However, the dipeptide orientation is reversed, and, concomitantly, the entrance to the amino acid binding cavity differs in position. Enzymatic characterization of two mutants, Y265F and S185A, demonstrates that these conserved residues are not catalytic residues at least in the reaction where L-phenylalanine is used as a substrate. On the basis of the biochemical and the structural data, we propose a reaction scheme and a catalytic mechanism for BacD.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/chemistry , Dipeptides/chemistry , Ligases/chemistry , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Catalytic Domain , Dipeptides/metabolism , Ligases/antagonists & inhibitors , Ligases/metabolism , Models, Molecular , Protein Conformation
18.
Anticancer Res ; 31(11): 3963-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22110227

ABSTRACT

BACKGROUND: The safety of regional hyperthermia has been tested in locally advanced rectal cancer. The aim of this study was to assess the effects of shorter distal margins on local control and survival in rectal cancer patients who were treated with preoperative hyperthermochemoradiation therapy (HCRT) and underwent rectal resection by using the total mesorectal excision (TME) method. PATIENTS AND METHODS: Ninety-three patients with rectal adenocarcinoma who received neoadjuvant HCRT (total radiation: 50 Gy) were included in this study. Surgery was performed 8 weeks after HCRT, and each resected specimen was evaluated histologically. Length of distal surgical margins, status of circumferential margins, pathological response, and tumor node metastasis stage were examined for their effects on recurrence and survival. RESULTS: Fifty-eight (62.4%) patients had tumor regression, and 20 (21.5%) had a pathological complete response. Distal margin length ranged from 1 to 55 mm (median, 21 mm) and did not correlate with local recurrence (p=0.57) or survival (p=0.75) by univariate analysis. Kaplan-Meier estimates of recurrence-free survival and local recurrence for the <10 mm versus ≥10 mm groups were not significantly different. Positive circumferential margins and failure of tumors to respond were unfavorable factors in survival. CONCLUSION: Distal resection margins that are shorter than 10 mm but are not positive appear to be equivalent to longer margins in patients who undergo HCRT followed by rectal resection with TME. To improve the down-staging rate, additional studies are needed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Digestive System Surgical Procedures , Gamma Rays , Hyperthermia, Induced , Neoplasm Recurrence, Local/diagnosis , Rectal Neoplasms/mortality , Rectal Neoplasms/therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Leucovorin/administration & dosage , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Rectal Neoplasms/pathology , Survival Rate , Treatment Outcome
19.
J Chem Phys ; 134(5): 054513, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21303144

ABSTRACT

Orientational correlations in Langmuir monolayers of nematic and smectic-C liquid crystal (LC) phases are investigated by molecular dynamics simulation. In both phases, the orientational correlation functions decay algebraically yet with the different exponents of 1.9 and 0.2 for the nematic and the smectic-C monolayers, respectively. The power law decay, i.e., the absence of long-range orientational order, means the both monolayers should be the ideal 2D system with a continuous symmetry, whereas the large difference in the exponents of power law gives rise to the crucial difference in their optical properties; the nematic monolayer is optically isotropic while the smectic-C monolayer exhibits an anisotropy on the length scale of visible light. Since the exponent is inversely proportional to the molecular exchange energy, the averaged molecular interaction in the nematic monolayer should be an order of magnitude smaller than that in the smectic-C monolayer, which is ascribed to the low molecular density and the weak molecular dipole due to the water molecule. The relation between the molecular interaction and the orientational correlation calculated for the 2D LC system offers much information not only about the 2D LCs but also on the bulk system.

20.
Anticancer Res ; 30(10): 4223-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21036745

ABSTRACT

AIM: To evaluate whether expression of L-type amino acid transporter 1 (LAT1) in pretreatment rectal cancer biopsies is predictive of tumour response to neoadjuvant hyperthermo-chemoradiotherapy (HCRT). PATIENTS AND METHODS: Forty-four patients with rectal adenocarcinoma who received neoadjuvant HCRT were investigated. LAT1 expression was immunohistochemically evaluated using pretreatment biopsies. The operation was performed after 2-3 months following HCRT and each resected specimen was graded by the histological criteria of the Japanese Classification of Colorectal Carcinoma. RESULTS: A positive LAT1 expression was recognized in 50.0% (22/44) of patients. Resected specimens were divided into 2 groups according to the histological grading criteria: good response (n=29) and poor response (n=15). LAT1-negative tumours had an 81.8% probability of good response and 18.2% probability of poor response. LAT1 expression showed marginally significant association with response to HCRT (p=0.05). CONCLUSION: LAT1 may be a useful predictive marker of response to HCRT in rectal cancer.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Large Neutral Amino Acid-Transporter 1/biosynthesis , Rectal Neoplasms/metabolism , Rectal Neoplasms/therapy , Adenocarcinoma/pathology , Adult , Aged , Biopsy , Cell Membrane/metabolism , Fluorouracil/therapeutic use , Humans , Hyperthermia, Induced , Leucovorin/therapeutic use , Male , Middle Aged , Neoadjuvant Therapy , Predictive Value of Tests , Radiotherapy , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...