Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Molecules ; 28(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37687259

ABSTRACT

Although loop epitopes at protein-protein binding interfaces often play key roles in mediating oligomer formation and interaction specificity, their binding sites are underexplored as drug targets owing to their high flexibility, relatively few hot spots, and solvent accessibility. Prior attempts to develop molecules that mimic loop epitopes to disrupt protein oligomers have had limited success. In this study, we used structure-based approaches to design and optimize cyclic-constrained peptides based on loop epitopes at the human phosphoglycerate dehydrogenase (PHGDH) dimer interface, which is an obligate homo-dimer with activity strongly dependent on the oligomeric state. The experimental validations showed that these cyclic peptides inhibit PHGDH activity by directly binding to the dimer interface and disrupting the obligate homo-oligomer formation. Our results demonstrate that loop epitope derived cyclic peptides with rationally designed affinity-enhancing substitutions can modulate obligate protein homo-oligomers, which can be used to design peptide inhibitors for other seemingly intractable oligomeric proteins.


Subject(s)
Dermatitis , Phosphoglycerate Dehydrogenase , Humans , Phosphoglycerate Dehydrogenase/genetics , Peptides, Cyclic/pharmacology , Binding Sites , Epitopes , Polymers
2.
Methods Mol Biol ; 2266: 279-298, 2021.
Article in English | MEDLINE | ID: mdl-33759133

ABSTRACT

Designing drugs that directly interact with multiple targets is a promising approach for treating complicated diseases. In order to successfully bind to multiple targets of different families and achieve the desired ligand efficiency, multi-target-directed ligands (MTDLs) require a higher level of diversity and complexity. De novo design strategies for creating more diverse chemical entities with desired properties may present an improved approach for developing MTDLs. In this chapter, we describe a computational protocol for developing MTDLs using the first reported multi-target de novo program, LigBuilder 3, which combines a binding site prediction module with de novo drug design and optimization modules. As an illustration of each detailed procedure, we design dual-functional compounds of two well-characterized virus enzymes, HIV protease and reverse transcriptase (PR and RT, respectively), using fragments extracted from known inhibitors. LigBuilder 3 is accessible at http://www.pkumdl.cn/ligbuilder3/ .


Subject(s)
Drug Design , HIV Protease Inhibitors/chemistry , HIV Protease/chemistry , HIV Reverse Transcriptase/chemistry , Software , Algorithms , Binding Sites , Databases, Protein , HIV Reverse Transcriptase/antagonists & inhibitors , Ligands , Models, Molecular , Protein Binding , Small Molecule Libraries/chemistry
3.
J Biol Chem ; 296: 100572, 2021.
Article in English | MEDLINE | ID: mdl-33753166

ABSTRACT

Human D-3-phosphoglycerate dehydrogenase (PHGDH), a key enzyme in de novo serine biosynthesis, is amplified in various cancers and serves as a potential target for anticancer drug development. To facilitate this process, more information is needed on the basic biochemistry of this enzyme. For example, PHGDH was found to form tetramers in solution and the structure of its catalytic unit (sPHGDH) was solved as a dimer. However, how the oligomeric states affect PHGDH enzyme activity remains elusive. We studied the dependence of PHGDH enzymatic activity on its oligomeric states. We found that sPHGDH forms a mixture of monomers and dimers in solution with a dimer dissociation constant of ∼0.58 µM, with the enzyme activity depending on the dimer content. We computationally identified hotspot residues at the sPHGDH dimer interface. Single-point mutants at these sites disrupt dimer formation and abolish enzyme activity. Molecular dynamics simulations showed that dimer formation facilitates substrate binding and maintains the correct conformation required for enzyme catalysis. We further showed that the full-length PHGDH exists as a dynamic mixture of monomers, dimers, and tetramers in solution with enzyme concentration-dependent activity. Mutations that can completely disrupt the sPHGDH dimer show different abilities to interrupt the full-length PHGDH tetramer. Among them, E108A and I121A can also disrupt the oligomeric structures of the full-length PHGDH and abolish its enzyme activity. Our study indicates that disrupting the oligomeric structure of PHGDH serves as a novel strategy for PHGDH drug design and the hotspot residues identified can guide the design process.


Subject(s)
Biocatalysis , Phosphoglycerate Dehydrogenase/chemistry , Phosphoglycerate Dehydrogenase/metabolism , Humans , Molecular Dynamics Simulation , Protein Multimerization , Protein Structure, Quaternary
4.
Biochem Biophys Res Commun ; 495(1): 375-381, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29127011

ABSTRACT

The response regulator PhoP, which is part of the PhoP/PhoQ two-component system, regulates the expression of multiple genes involved in controlling virulence in Salmonella enterica serovar Typhimurium and other species of Gram-negative bacteria. Modulating the phosphorylation-mediated dimerization in the receiver domain may interfere with the transcriptional function of PhoP. In this study, we analyzed the therapeutic potential of the PhoP receiver domain by exploring it as a potential target for drug design. The structural information was then applied to identify the first hit compounds from commercial chemical libraries by combining pharmacophore modelling and docking methods with a GFP (Green Fluorescent Protein)-based promoter-fusion bioassay. In total, one hundred and forty compounds were selected, purchased, and tested for biological activity. Several novel scaffolds showed acceptable potency to modulate the transcriptional function of PhoP, either by enhancing or inhibiting the expression of PhoP-dependent genes. These compounds may be used as the starting point for developing modulators that target the protein-protein interface of the PhoP protein as an alternative strategy against antibiotic resistance.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Drug Design , Molecular Docking Simulation , Peptides/chemistry , Repressor Proteins/chemistry , Transcriptional Activation , Binding Sites , Drug Evaluation, Preclinical , Protein Binding , Protein Interaction Mapping/methods , Repressor Proteins/ultrastructure
5.
Mol Inform ; 36(11)2017 11.
Article in English | MEDLINE | ID: mdl-28598557

ABSTRACT

The response regulator PhoP is part of the PhoP/PhoQ two-component system, which is responsible for regulating the expression of multiple genes involved in controlling virulence, biofilm formation, and resistance to antimicrobial peptides. Therefore, modulating the transcriptional function of the PhoP protein is a promising strategy for developing new antimicrobial agents. There is evidence suggesting that phosphorylation-mediated dimerization in the regulatory domain of PhoP is essential for its transcriptional function. Disruption or stabilization of protein-protein interactions at the dimerization interface may inhibit or enhance the expression of PhoP-dependent genes. In this study, we performed molecular dynamics simulations on the active and inactive dimers and monomers of the PhoP regulatory domains, followed by pocket-detecting screenings and a quantitative hot-spot analysis in order to assess the druggability of the protein. Consistent with prior hypothesis, the calculation of the binding free energy shows that phosphorylation enhances dimerization of PhoP. Furthermore, we have identified two different putative binding sites at the dimerization active site (the α4-ß5-α5 face) with energetic "hot-spot" areas, which could be used to search for modulators of protein-protein interactions. This study delivers insight into the dynamics and druggability of the dimerization interface of the PhoP regulatory domain, and may serve as a basis for the rational identification of new antimicrobial drugs.


Subject(s)
Bacterial Proteins/metabolism , Molecular Dynamics Simulation , Binding Sites , Gene Expression Regulation, Bacterial , Virulence
6.
J Biomol Struct Dyn ; 31(2): 215-23, 2013.
Article in English | MEDLINE | ID: mdl-22831459

ABSTRACT

C5aR antagonists have been thought as potential immune mediators in various inflammatory and autoimmune diseases, and discovery of C5aR antagonists has attracted much attention in recent years. The discovery of C5aR antagonists was usually achieved through high-throughput screening, which usually suffered a high cost and a low success rate. Currently, the fast developing computer-aided virtual screening (VS) methods provide economic and rapid approaches to the lead discovery. In this account, we proposed a hybrid ligand-based VS protocol that is based on support vector machine (SVM) classification and pharmacophore models for retrieving novel C5aR antagonists. Performance evaluation of this hybrid VS protocol in virtual screening against a large independent test set, T-CHEM, showed that the hybrid VS approach significantly increased the hit rate and enrichment factor compared with the individual SVM classification model-based VS and pharmacophore model-based VS, as well as molecular docking-based VS in that the receptor structure was created by homology modeling. The hybrid VS approach was then used to screen several large chemical libraries including PubChem, Specs, and Enamine. Finally, a total of 20 compounds were selected from the top ranking hits, and shifted to the subsequent in vitro and in vivo studies, which results will be reported in the near future.


Subject(s)
Complement Inactivating Agents/chemistry , Molecular Docking Simulation , Receptors, Complement/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Humans , Inhibitory Concentration 50 , Ligands , Models, Chemical , Receptor, Anaphylatoxin C5a , Receptors, Complement/chemistry , Small Molecule Libraries , Structural Homology, Protein , Support Vector Machine
7.
Molecules ; 17(6): 6317-30, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22634842

ABSTRACT

A novel series of N-methylpicolinamide-4-thiol derivatives were synthesized and evaluated on human cancer cell lines. Among them, compound 6p displayed potent and broad-spectrum anti-proliferative activities in vitro on some human cancer cell lines, even better than sorafenib. The advanced kinase inhibitory assays showed that compound 6p could selectively inhibit Aurora-B kinase. The biological results were rationalized by the molecular docking study, which indicated the stable interactions of 6p with the Aurora-B kinase.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Picolinic Acids/chemical synthesis , Picolinic Acids/pharmacology , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/pharmacology , Antineoplastic Agents/chemistry , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Inhibitory Concentration 50 , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry
8.
Acta Crystallogr Sect E Struct Rep Online ; 67(Pt 6): o1480, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21754849

ABSTRACT

There are two mol-ecules in the asymmetric unit mol-ecule of the title compound, C(8)H(10)ClN(3)O(2). Intra-molecular N-H⋯O hydrogen bonds stabilize the mol-ecular structure. There are no classical inter-molecular hydrogen bonds in the crystal structure.

9.
Acta Crystallogr Sect E Struct Rep Online ; 67(Pt 4): o856, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21754137

ABSTRACT

There are two independent mol-ecules in the asymmetric unit of the title compound, C(22)H(21)N(3)O(4)S. The central benzene ring makes dihedral angles of 74.28 (6) and 68.84 (6)° with the pyridine and 3,5-dimeth-oxy-phenyl rings, respectively, in one molecule [86.66 (6) and 81.14 (6)° respectively, in the other]. Each of the mol-ecules forms a centrosymmetric dimer with another mol-ecule via pairs of inter-molecular N-H⋯O hydrogen bonds. These hydrogen bonds connect the N-H groups and the O atoms of the carbonyl groups next to the 3,5-dimeth-oxy-phenyl rings. Additional inter-molecular N-H⋯O inter-actions link the dimers in the crystal structure.

SELECTION OF CITATIONS
SEARCH DETAIL
...