255 research outputs found

    Reduction method based on a new fuzzy rough set in fuzzy information system and its applications to scheduling problems

    Get PDF
    AbstractIn this paper, we present the concept of fuzzy information granule based on a relatively weaker fuzzy similarity relation called fuzzy TL-similarity relation for the first time. Then, according to the fuzzy information granule, we define the lower and upper approximations of fuzzy sets and a corresponding new fuzzy rough set. Furthermore, we construct a kind of new fuzzy information system based on the fuzzy TL-similarity relation and study its reduction using the fuzzy rough set. At last, we apply the reduction method based on the defined fuzzy rough set in the above fuzzy information system to the reduction of the redundant multiple fuzzy rule in the scheduling problems, and numerical computational results show that the reduction method based on the new fuzzy rough set is more suitable for the reduction of multiple fuzzy rules in the scheduling problems compared with the reduction methods based on the existing fuzzy rough set

    Examining the base stacking interaction in a dinucleotide context via reversible cyclobutane dimer analogue formation under UV irradiation

    Get PDF
    Substituted tolyl groups are considered as close isosteres of the thymine (T) residue. They can be recognized by DNA polymerases as if they were thymine. Although these toluene derivatives are relatively inert toward radical additions, our recent finding suggests that the dinucleotide analogue TpTo (To = 2'-deoxy-1-(3-tolyl)-Ī²-D-ribofuranose) supports an ortho photocycloaddition reaction upon UV irradiation, producing two cyclobutane pyrimidine dimer (CPD) analogues 2 and 3. Our report here further shows that formation of these CPD species is reversible under UVC irradiation, resembling the photochemical property of the CPD species formed between two Ts. Analyzing the stability of these CPD analogues suggests that one (2) is more stable than the other (3). The TpTo conformer responsible for 2 formation is also more stable than that responsible for 3 formation, as indicated by the Gibbs free energy change calculated from the constructed Bordwell thermodynamic cycle. These different stabilities are not due to the varying photochemical properties, as proved by quantum yields determined from the corresponding photoreactions. Instead, they are ascribed to the different stacking interaction between the T and the To rings both in the TpTo dinucleotide as well as in the formed CPD analogues. Factors contributing to the ring stacking interactions are also discussed. Our proof-of-concept approach suggests that a carefully designed Bordwell cycle coupled with reversible CPD formations under UV irradiation can be very useful in studying DNA base interactions

    A new class of orthosteric uPARĀ·uPA small-molecule antagonists are allosteric inhibitors of the uPARĀ·vitronectin interaction

    Get PDF
    The urokinase receptor (uPAR) is a GPI-anchored cell surface receptor that is at the center of an intricate network of protein-protein interactions. Its immediate binding partners are the serine proteinase urokinase (uPA), and vitronectin (VTN), a component of the extracellular matrix. uPA and VTN bind at distinct sites on uPAR to promote extracellular matrix degradation and integrin signaling, respectively. Here, we report the discovery of a new class of pyrrolone small-molecule inhibitors of the tight āˆ¼1 nM uPARĀ·uPA protein-protein interaction. These compounds were designed to bind to the uPA pocket on uPAR. The highest affinity compound, namely 7, displaced a fluorescently labeled Ī±-helical peptide (AE147-FAM) with an inhibition constant Ki of 0.7 Ī¼M and inhibited the tight uPARĀ·uPAATF interaction with an IC50 of 18 Ī¼M. Biophysical studies with surface plasmon resonance showed that VTN binding is highly dependent on uPA. This cooperative binding was confirmed as 7, which binds at the uPARĀ·uPA interface, also inhibited the distal VTNĀ·uPAR interaction. In cell culture, 7 blocked the uPARĀ·uPA interaction in uPAR-expressing human embryonic kidney (HEK-293) cells and impaired cell adhesion to VTN, a process that is mediated by integrins. As a result, 7 inhibited integrin signaling in MDA-MB-231 cancer cells as evidenced by a decrease in focal adhesion kinase (FAK) phosphorylation and Rac1 GTPase activation. Consistent with these results, 7 blocked breast MDA-MB-231 cancer cell invasion with IC50 values similar to those observed in ELISA and surface plasmon resonance competition studies. Explicit-solvent molecular dynamics simulations show that the cooperativity between uPA and VTN is attributed to stabilization of uPAR motion by uPA. In addition, free energy calculations revealed that uPA stabilizes the VTNSMBĀ·uPAR interaction through more favorable electrostatics and entropy. Disruption of the uPARĀ·VTNSMB interaction by 7 is consistent with the cooperative binding to uPAR by uPA and VTN. Interestingly, the VTNSMBĀ·uPAR interaction was less favorable in the VTNSMBĀ·uPARĀ·7 complex suggesting potential cooperativity between 7 and VTN. Compound 7 provides an excellent starting point for the development of more potent derivatives to explore uPAR biology

    Structure-Based Target-Specific Screening Leads to Small-Molecule CaMKII Inhibitors

    Get PDF
    Target-specific scoring methods are more commonly used to identify small-molecule inhibitors among compounds docked to a target of interest. Top candidates that emerge from these methods have rarely been tested for activity and specificity across a family of proteins. In this study we docked a chemical library into CaMKIIĪ“, a member of the Ca2+ /calmodulin (CaM)-dependent protein kinase (CaMK) family, and re-scored the resulting protein-compound structures using Support Vector Machine SPecific (SVMSP), a target-specific method that we developed previously. Among the 35 selected candidates, three hits were identified, such as quinazoline compound 1 (KIN-1; N4-[7-chloro-2-[(E)-styryl]quinazolin-4-yl]-N1,N1-diethylpentane-1,4-diamine), which was found to inhibit CaMKIIĪ“ kinase activity at single-digit micromolar IC50 . Activity across the kinome was assessed by profiling analogues of 1, namely 6 (KIN-236; N4-[7-chloro-2-[(E)-2-(2-chloro-4,5-dimethoxyphenyl)vinyl]quinazolin-4-yl]-N1,N1-diethylpentane-1,4-diamine), and an analogue of hit compound 2 (KIN-15; 2-[4-[(E)-[(5-bromobenzofuran-2-carbonyl)hydrazono]methyl]-2-chloro-6-methoxyphenoxy]acetic acid), namely 14 (KIN-332; N-[(E)-[4-(2-anilino-2-oxoethoxy)-3-chlorophenyl]methyleneamino]benzofuran-2-carboxamide), against 337 kinases. Interestingly, for compound 6, CaMKIIĪ“ and homologue CaMKIIĪ³ were among the top ten targets. Among the top 25 targets of 6, IC50 values ranged from 5 to 22ā€…Ī¼m. Compound 14 was found to be not specific toward CaMKII kinases, but it does inhibit two kinases with sub-micromolar IC50 values among the top 25. Derivatives of 1 were tested against several kinases including several members of the CaMK family. These data afforded a limited structure-activity relationship study. Molecular dynamics simulations with explicit solvent followed by end-point MM-GBSA free-energy calculations revealed strong engagement of specific residues within the ATP binding pocket, and also changes in the dynamics as a result of binding. This work suggests that target-specific scoring approaches such as SVMSP may hold promise for the identification of small-molecule kinase inhibitors that exhibit some level of specificity toward the target of interest across a large number of proteins

    RFAConv: Innovating Spatital Attention and Standard Convolutional Operation

    Full text link
    Spatial attention has been widely used to improve the performance of convolutional neural networks by allowing them to focus on important information. However, it has certain limitations. In this paper, we propose a new perspective on the effectiveness of spatial attention, which is that it can solve the problem of convolutional kernel parameter sharing. Despite this, the information contained in the attention map generated by spatial attention is not sufficient for large-size convolutional kernels. Therefore, we introduce a new attention mechanism called Receptive-Field Attention (RFA). While previous attention mechanisms such as the Convolutional Block Attention Module (CBAM) and Coordinate Attention (CA) only focus on spatial features, they cannot fully address the issue of convolutional kernel parameter sharing. In contrast, RFA not only focuses on the receptive-field spatial feature but also provides effective attention weights for large-size convolutional kernels. The Receptive-Field Attention convolutional operation (RFAConv), developed by RFA, represents a new approach to replace the standard convolution operation. It offers nearly negligible increment of computational cost and parameters, while significantly improving network performance. We conducted a series of experiments on ImageNet-1k, MS COCO, and VOC datasets, which demonstrated the superiority of our approach in various tasks including classification, object detection, and semantic segmentation. Of particular importance, we believe that it is time to shift focus from spatial features to receptive-field spatial features for current spatial attention mechanisms. By doing so, we can further improve network performance and achieve even better results. The code and pre-trained models for the relevant tasks can be found at https://github.com/Liuchen1997/RFAConv.Comment: 14 pages, 5 figure

    Small Molecules Engage Hot Spots through Cooperative Binding To Inhibit a Tight Proteinā€“Protein Interaction

    Get PDF
    Proteinā€“protein interactions drive every aspect of cell signaling, yet only a few small-molecule inhibitors of these interactions exist. Despite our ability to identify critical residues known as hot spots, little is known about how to effectively engage them to disrupt proteinā€“protein interactions. Here, we take advantage of the ease of preparation and stability of pyrrolinone 1, a small-molecule inhibitor of the tight interaction between the urokinase receptor (uPAR) and its binding partner, the urokinase-type plasminogen activator uPA, to synthesize more than 40 derivatives and explore their effect on the proteinā€“protein interaction. We report the crystal structure of uPAR bound to previously discovered pyrazole 3 and to pyrrolinone 12. While both 3 and 12 bind to uPAR and compete with a fluorescently labeled peptide probe, only 12 and its derivatives inhibit the full uPARĀ·uPA interaction. Compounds 3 and 12 mimic and engage different hot-spot residues on uPA and uPAR, respectively. Interestingly, 12 is involved in a Ļ€ā€“cation interaction with Arg-53, which is not considered a hot spot. Explicit-solvent molecular dynamics simulations reveal that 3 and 12 exhibit dramatically different correlations of motion with residues on uPAR. Free energy calculations for the wild-type and mutant uPAR bound to uPA or 12 show that Arg-53 interacts with uPA or with 12 in a highly cooperative manner, thereby altering the contributions of hot spots to uPAR binding. The direct engagement of peripheral residues not considered hot spots through Ļ€ā€“cation or salt-bridge interactions could provide new opportunities for enhanced small-molecule engagement of hot spots to disrupt challenging proteinā€“protein interactions

    A Computational Investigation of Small-Molecule Engagement of Hot Spots at Proteinā€“Protein Interaction Interfaces

    Get PDF
    The binding affinity of a proteinā€“protein interaction is concentrated at amino acids known as hot spots. It has been suggested that small molecules disrupt proteinā€“protein interactions by either (i) engaging receptor protein hot spots or (ii) mimicking hot spots of the protein ligand. Yet, no systematic studies have been done to explore how effectively existing small-molecule proteinā€“protein interaction inhibitors mimic or engage hot spots at protein interfaces. Here, we employ explicit-solvent molecular dynamics simulations and end-point MM-GBSA free energy calculations to explore this question. We select 36 compounds for which high-quality binding affinity and cocrystal structures are available. Five complexes that belong to three classes of proteinā€“protein interactions (primary, secondary, and tertiary) were considered, namely, BRD4ā€¢H4, XIAPā€¢Smac, MDM2ā€¢p53, Bcl-xLā€¢Bak, and IL-2ā€¢IL-2RĪ±. Computational alanine scanning using MM-GBSA identified hot-spot residues at the interface of these protein interactions. Decomposition energies compared the interaction of small molecules with individual receptor hot spots to those of the native protein ligand. Pharmacophore analysis was used to investigate how effectively small molecules mimic the position of hot spots of the protein ligand. Finally, we study whether small molecules mimic the effects of the native protein ligand on the receptor dynamics. Our results show that, in general, existing small-molecule inhibitors of proteinā€“protein interactions do not optimally mimic proteinā€“ligand hot spots, nor do they effectively engage protein receptor hot spots. The more effective use of hot spots in future drug design efforts may result in smaller compounds with higher ligand efficiencies that may lead to greater success in clinical trials

    Small-molecule CaVĪ±1ā‹…CaVĪ² antagonist suppresses neuronal voltage-gated calcium-channel trafficking

    Get PDF
    Extracellular calcium flow through neuronal voltage-gated CaV2.2 calcium channels converts action potential-encoded information to the release of pronociceptive neurotransmitters in the dorsal horn of the spinal cord, culminating in excitation of the postsynaptic central nociceptive neurons. The CaV2.2 channel is composed of a pore-forming Ī±1 subunit (CaVĪ±1) that is engaged in protein-protein interactions with auxiliary Ī±2/Ī“ and Ī² subunits. The high-affinity CaV2.2Ī±1ā‹…CaVĪ²3 protein-protein interaction is essential for proper trafficking of CaV2.2 channels to the plasma membrane. Here, structure-based computational screening led to small molecules that disrupt the CaV2.2Ī±1ā‹…CaVĪ²3 protein-protein interaction. The binding mode of these compounds reveals that three substituents closely mimic the side chains of hot-spot residues located on the Ī±-helix of CaV2.2Ī±1 Site-directed mutagenesis confirmed the critical nature of a salt-bridge interaction between the compounds and CaVĪ²3 Arg-307. In cells, compounds decreased trafficking of CaV2.2 channels to the plasma membrane and modulated the functions of the channel. In a rodent neuropathic pain model, the compounds suppressed pain responses. Small-molecule Ī±-helical mimetics targeting ion channel protein-protein interactions may represent a strategy for developing nonopioid analgesia and for treatment of other neurological disorders associated with calcium-channel trafficking
    • ā€¦
    corecore