Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Publication year range
1.
Cells ; 12(20)2023 10 17.
Article in English | MEDLINE | ID: mdl-37887313

ABSTRACT

De novo mutations in GNAO1, the gene encoding the major neuronal G protein Gαo, cause a spectrum of pediatric encephalopathies with seizures, motor dysfunction, and developmental delay. Of the >80 distinct missense pathogenic variants, many appear to uniformly destabilize the guanine nucleotide handling of the mutant protein, speeding up GTP uptake and deactivating GTP hydrolysis. Zinc supplementation emerges as a promising treatment option for this disease, as Zn2+ ions reactivate the GTP hydrolysis on the mutant Gαo and restore cellular interactions for some of the mutants studied earlier. The molecular etiology of GNAO1 encephalopathies needs further elucidation as a prerequisite for the development of efficient therapeutic approaches. In this work, we combine clinical and medical genetics analysis of a novel GNAO1 mutation with an in-depth molecular dissection of the resultant protein variant. We identify two unrelated patients from Norway and France with a previously unknown mutation in GNAO1, c.509C>G that results in the production of the Pro170Arg mutant Gαo, leading to severe developmental and epileptic encephalopathy. Molecular investigations of Pro170Arg identify this mutant as a unique representative of the pathogenic variants. Its 100-fold-accelerated GTP uptake is not accompanied by a loss in GTP hydrolysis; Zn2+ ions induce a previously unseen effect on the mutant, forcing it to lose the bound GTP. Our work combining clinical and molecular analyses discovers a novel, biochemically distinct pathogenic missense variant of GNAO1 laying the ground for personalized treatment development.


Subject(s)
Brain Diseases , Humans , Child , Mutation/genetics , GTP-Binding Proteins/metabolism , Ions/metabolism , Guanosine Triphosphate , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
2.
Biosci Rep ; 43(10)2023 10 31.
Article in English | MEDLINE | ID: mdl-37815922

ABSTRACT

YY1 is a ubiquitously expressed, intrinsically disordered transcription factor involved in neural development. The oligomeric state of YY1 varies depending on the environment. These structural changes may alter its DNA binding ability and hence its transcriptional activity. Just as YY1's oligomeric state can impact its role in transcription, so does its interaction with other proteins such as FOXP2. The aim of this work is to study the structure and dynamics of YY1 so as to determine the influence of oligomerisation and associations with FOXP2 on its DNA binding mechanism. The results confirm that YY1 is primarily a disordered protein, but it does consist of certain specific structured regions. We observed that YY1 quaternary structure is a heterogenous mixture of oligomers, the overall size of which is dependent on ionic strength. Both YY1 oligomerisation and its dynamic behaviour are further subject to changes upon DNA binding, whereby increases in DNA concentration result in a decrease in the size of YY1 oligomers. YY1 and the FOXP2 forkhead domain were found to interact with each other both in isolation and in the presence of YY1-specific DNA. The heterogeneous, dynamic multimerisation of YY1 identified in this work is, therefore likely to be important for its ability to make heterologous associations with other proteins such as FOXP2. The interactions that YY1 makes with itself, FOXP2 and DNA form part of an intricate mechanism of transcriptional regulation by YY1, which is vital for appropriate neural development.


Subject(s)
Intrinsically Disordered Proteins , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism , DNA/metabolism , Gene Expression Regulation
3.
Protein Sci ; 32(10): e4781, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37703013

ABSTRACT

The 11 lytic transglycosylases of Pseudomonas aeruginosa have overlapping activities in the turnover of the cell-wall peptidoglycan. Rare lipoprotein A (RlpA) is distinct among the 11 by its use of only peptidoglycan lacking peptide stems. The spatial localization of RlpA and its interactome within P. aeruginosa are unknown. We employed suppression of introduced amber codons at sites in the rlpA gene for the introduction of the unnatural-amino-acids Νζ -[(2-azidoethoxy)carbonyl]-l-lysine (compound 1) and Nζ -[[[3-(3-methyl-3H-diazirin-3-yl)propyl]amino]carbonyl]-l-lysine (compound 2). In live P. aeruginosa, full-length RlpA incorporating compound 1 into its sequence was fluorescently tagged using strained-promoted alkyne-azide cycloaddition and examined by fluorescence microscopy. RlpA is present at low levels along the sidewall length of the bacterium, and at higher levels at the nascent septa of replicating bacteria. In intact P. aeruginosa, UV photolysis of full-length RlpA having compound 2 within its sequence generated a transient reactive carbene, which engaged in photoaffinity capture of neighboring proteins. Thirteen proteins were identified. Three of these proteins-PBP1a, PBP5, and MreB-are members of the bacterial divisome. The use of the complementary methodologies of non-canonical amino-acid incorporation, photoaffinity proximity analysis, and fluorescent microscopy confirm a dominant septal location for the RlpA enzyme of P. aeruginosa, as a divisome-associated activity. This accomplishment adds to the emerging recognition of the value of these methodologies for identification of the intracellular localization of bacterial proteins.


Subject(s)
Lipoprotein(a) , Pseudomonas aeruginosa , Lipoprotein(a)/metabolism , Codon, Terminator/metabolism , Peptidoglycan/metabolism , Lysine/metabolism
4.
Proc Natl Acad Sci U S A ; 120(30): e2218826120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37463207

ABSTRACT

Development of a simple, label-free screening technique capable of precisely and directly sensing interaction-in-solution over a size range from small molecules to large proteins such as antibodies could offer an important tool for researchers and pharmaceutical companies in the field of drug development. In this work, we present a thermostable Raman interaction profiling (TRIP) technique that facilitates low-concentration and low-dose screening of binding between protein and ligand in physiologically relevant conditions. TRIP was applied to eight protein-ligand systems, and produced reproducible high-resolution Raman measurements, which were analyzed by principal component analysis. TRIP was able to resolve time-depending binding between 2,4-dinitrophenol and transthyretin, and analyze biologically relevant SARS-CoV-2 spike-antibody interactions. Mixtures of the spike receptor-binding domain with neutralizing, nonbinding, or binding but nonneutralizing antibodies revealed distinct and reproducible Raman signals. TRIP holds promise for the future developments of high-throughput drug screening and real-time binding measurements between protein and drug.


Subject(s)
COVID-19 , Microscopy , Humans , SARS-CoV-2 , Drug Evaluation, Preclinical , Ligands , Antibodies, Viral , Drug Interactions , Spike Glycoprotein, Coronavirus/metabolism , Antibodies, Neutralizing
5.
Carbohydr Polym ; 298: 120038, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36241312

ABSTRACT

Pectin has been used as a gel strengthening agent, but its role in pea protein gels remains unclear. The present study investigated the effects of low-methyl pectin on the physicochemical and rheological properties of pea protein gels at neutral pH and elucidated underlying gelling mechanisms. Pectin increased the stability and viscosity of pea protein dispersions and induced the formation of large protein aggregates. After heating, the storage modulus of pea protein gel containing 0.5 % pectin increased by nearly six times compared to those without pectin, and the gel microstructures became more heterogeneous and compact. Molecular docking revealed that pectin interacted with proteins via mainly electrostatic and hydrogen-bonding interactions. Solvent extraction found that pectin increased the hydrogen bonding, hydrophobic interactions, and disulfide bonds of the gel systems, thus boosting their strength. The current work provided a practical simple approach to increasing the strength of pea protein gels at a neutral pH.


Subject(s)
Pea Proteins , Pectins , Disulfides , Gels/chemistry , Hydrogen , Hydrogen-Ion Concentration , Molecular Docking Simulation , Pectins/chemistry , Protein Aggregates , Rheology , Solvents
6.
Biomolecules ; 12(9)2022 09 06.
Article in English | MEDLINE | ID: mdl-36139085

ABSTRACT

Machine learning (ML) has been an important arsenal in computational biology used to elucidate protein function for decades. With the recent burgeoning of novel ML methods and applications, new ML approaches have been incorporated into many areas of computational biology dealing with protein function. We examine how ML has been integrated into a wide range of computational models to improve prediction accuracy and gain a better understanding of protein function. The applications discussed are protein structure prediction, protein engineering using sequence modifications to achieve stability and druggability characteristics, molecular docking in terms of protein-ligand binding, including allosteric effects, protein-protein interactions and protein-centric drug discovery. To quantify the mechanisms underlying protein function, a holistic approach that takes structure, flexibility, stability, and dynamics into account is required, as these aspects become inseparable through their interdependence. Another key component of protein function is conformational dynamics, which often manifest as protein kinetics. Computational methods that use ML to generate representative conformational ensembles and quantify differences in conformational ensembles important for function are included in this review. Future opportunities are highlighted for each of these topics.


Subject(s)
Computational Biology , Proteins , Computational Biology/methods , Ligands , Machine Learning , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Proteins/chemistry
7.
J Virol ; 96(19): e0129722, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36102648

ABSTRACT

Human respiratory syncytial virus (RSV) is the leading cause of severe acute lower respiratory tract infections in infants worldwide. Nonstructural protein NS1 of RSV modulates the host innate immune response by acting as an antagonist of type I and type III interferon (IFN) production and signaling in multiple ways. Likely, NS1 performs this function by interacting with different host proteins. In order to obtain a comprehensive overview of the NS1 interaction partners, we performed three complementary protein-protein interaction screens, i.e., BioID, MAPPIT, and KISS. To closely mimic a natural infection, the BioID proximity screen was performed using a recombinant RSV in which the NS1 protein is fused to a biotin ligase. Remarkably, MED25, a subunit of the Mediator complex, was identified in all three performed screening methods as a potential NS1-interacting protein. We confirmed the interaction between MED25 and RSV NS1 by coimmunoprecipitation, not only upon overexpression of NS1 but also with endogenous NS1 during RSV infection. We also demonstrate that the replication of RSV can be enhanced in MED25 knockout A549 cells, suggesting a potential antiviral role of MED25 during RSV infection. Mediator subunits function as transcriptional coactivators and are involved in transcriptional regulation of their target genes. Therefore, the interaction between RSV NS1 and cellular MED25 might be beneficial for RSV during infection by affecting host transcription and the host immune response to infection. IMPORTANCE Innate immune responses, including the production of type I and III interferons, play a crucial role in the first line of defense against RSV infection. However, only a poor induction of type I IFNs is observed during RSV infection, suggesting that RSV has evolved mechanisms to prevent type I IFN expression by the infected host cell. A unique RSV protein, NS1, is largely responsible for this effect, probably through interaction with multiple host proteins. A better understanding of the interactions that occur between RSV NS1 and host proteins may help to identify targets for an effective antiviral therapy. We addressed this question by performing three complementary protein-protein interaction screens and identified MED25 as an RSV NS1-interacting protein. We propose a role in innate anti-RSV defense for this Mediator complex subunit.


Subject(s)
Mediator Complex , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Viral Nonstructural Proteins , A549 Cells , Humans , Interferons/metabolism , Mediator Complex/genetics , Mediator Complex/metabolism , Respiratory Syncytial Virus Infections/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
8.
Biomolecules ; 12(6)2022 06 11.
Article in English | MEDLINE | ID: mdl-35740944

ABSTRACT

It is important for elucidating the regulation mechanism of life activities, as well as for the prevention, diagnosis, and drug design of diseases, to study protein-protein interactions (PPIs). Here, we investigated the interactions of human serum albumin (HSA) in the presence of tyrosine kinase inhibitors (TKIs: imatinib, nilotinib, dasatinib, bosutinib, and ponatinib) using atomic force microscopy (AFM). The distribution of rupture events including the specific interaction force Fi and the non-specific interaction force F0 between HSA pairs was analyzed. Based on the force measurements, Fi and F0 between HSA pairs in the control experiment were calculated to be 47 ± 1.5 and 116.1 ± 1.3 pN. However, Fi was significantly decreased in TKIs, while F0 was slightly decreased. By measuring the rupture forces at various loading rates and according to the Bell equation, the kinetic parameters of the complexes were investigated in greater detail. Molecular docking was used as a complementary means by which to explore the force of this effect. The whole measurements indicated that TKIs influenced PPIs in a variety of ways, among which hydrogen bonding and hydrophobic interactions were the most important. In conclusion, these outcomes give us a better insight into the mechanisms of PPIs when there are exogenous compounds present as well as in different liquid environments.


Subject(s)
Protein Kinase Inhibitors , Serum Albumin, Human , Dasatinib/pharmacology , Humans , Microscopy, Atomic Force , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
9.
Chembiochem ; 23(17): e202200178, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35767695

ABSTRACT

The development of protein-protein interaction (PPI) inhibitors has been a successful strategy in drug development. However, the identification of PPI stabilizers has proven much more challenging. Here we report a fragment-based drug screening approach using the regulatory hub-protein 14-3-3 as a platform for identifying PPI stabilizers. A homogenous time-resolved FRET assay was used to monitor stabilization of 14-3-3/peptide binding using the known interaction partner estrogen receptor alpha. Screening of an in-house fragment library identified fragment 2 (VUF15640) as a putative PPI stabilizer capable of cooperatively stabilizing 14-3-3 PPIs in a cooperative fashion with Fusicoccin-A. Mechanistically, fragment 2 appears to enhance 14-3-3 dimerization leading to increased client-protein binding. Functionally, fragment 2 enhanced potency of 14-3-3 in a cell-free system inhibiting the enzyme activity of the nitrate reductase. In conclusion, we identified a general PPI stabilizer targeting 14-3-3, which could be used as a tool compound for investigating 14-3-3 client protein interactions.


Subject(s)
14-3-3 Proteins , 14-3-3 Proteins/chemistry , Drug Evaluation, Preclinical , Humans , Protein Binding
10.
Mol Cell Proteomics ; 21(7): 100243, 2022 07.
Article in English | MEDLINE | ID: mdl-35577067

ABSTRACT

Protein arginine (R) methylation is a post-translational modification involved in various biological processes, such as RNA splicing, DNA repair, immune response, signal transduction, and tumor development. Although several advancements were made in the study of this modification by mass spectrometry, researchers still face the problem of a high false discovery rate. We present a dataset of high-quality methylations obtained from several different heavy methyl stable isotope labeling with amino acids in cell culture experiments analyzed with a machine learning-based tool and show that this model allows for improved high-confidence identification of real methyl-peptides. Overall, our results are consistent with the notion that protein R methylation modulates protein-RNA interactions and suggest a role in rewiring protein-protein interactions, for which we provide experimental evidence for a representative case (i.e., NONO [non-POU domain-containing octamer-binding protein]-paraspeckle component 1 [PSPC1]). Upon intersecting our R-methyl-sites dataset with the PhosphoSitePlus phosphorylation dataset, we observed that R methylation correlates differently with S/T-Y phosphorylation in response to various stimuli. Finally, we explored the application of heavy methyl stable isotope labeling with amino acids in cell culture to identify unconventional methylated residues and successfully identified novel histone methylation marks on serine 28 and threonine 32 of H3. The database generated, named ProMetheusDB, is freely accessible at https://bioserver.ieo.it/shiny/app/prometheusdb.


Subject(s)
Protein Processing, Post-Translational , Proteome , Amino Acids/metabolism , Humans , Isotope Labeling/methods , Mass Spectrometry , Methylation , Proteome/metabolism , RNA-Binding Proteins/metabolism
11.
Curr Comput Aided Drug Des ; 18(3): 228-239, 2022.
Article in English | MEDLINE | ID: mdl-35388762

ABSTRACT

BACKGROUND: The compound Sophora flavescenes (Kushen) decoction was found to reduce the inflammatory symptom of Ulcerative Colitis (UC). However, there exists a very limited understanding of the molecular pharmacological mechanisms. OBJECTIVE: This study aimed to explore the mechanism of compound Sophora flavescens (Kushen) decoction in treating ulcerative colitis from the perspective of network pharmacology. METHODS: Active components and potential targets of compound Sophora flavescens (Kushen) decoction were obtained through the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) database. GeneCards and other databases were used to predict and screen ulcerative colitis-related genes. Cytoscape software was applied to construct the "drugactive component-disease-target" network. GO function and KEGG pathway enrichment analyses revealed the potential pathway of the compound Sophora flavescenes (Kushen) decoction for UC. RESULTS: After the screening, a total of 124 active ingredients and 163 potential therapeutic targets for UC were obtained from the compound Sophora flavescens (Kushen) decoction. Protein interaction network analysis showed that 15 key targets could be identified for the possible treatment of UC. GO and KEGG analyses showed that the active ingredients in the compound Sophora flavescens (Kushen) decoction were mainly enriched in 2556 biological processes and 172 signaling pathways. CONCLUSION: The study showed that the compound Sophora flavescens (Kushen) decoction has therapeutic effects on UC through multi-component, multi-target, and multi-pathway.


Subject(s)
Colitis, Ulcerative , Sophora , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Drugs, Chinese Herbal , Molecular Docking Simulation , Network Pharmacology , Technology
12.
J Cell Mol Med ; 26(7): 2049-2062, 2022 04.
Article in English | MEDLINE | ID: mdl-35229974

ABSTRACT

Through a comprehensive review and in silico analysis of reported data on STAT-linked diseases, we analysed the communication pathways and interactome of the seven STATs in major cancer categories and proposed rational targeting approaches for therapeutic intervention to disrupt critical pathways and addictions to hyperactive JAK/STAT in neoplastic states. Although all STATs follow a similar molecular activation pathway, STAT1, STAT2, STAT4 and STAT6 exert specific biological profiles associated with a more restricted pattern of activation by cytokines. STAT3 and STAT5A as well as STAT5B have pleiotropic roles in the body and can act as critical oncogenes that promote many processes involved in cancer development. STAT1, STAT3 and STAT5 also possess tumour suppressive action in certain mutational and cancer type context. Here, we demonstrated member-specific STAT activity in major cancer types. Through systems biology approaches, we found surprising roles for EGFR family members, sex steroid hormone receptor ESR1 interplay with oncogenic STAT function and proposed new drug targeting approaches of oncogenic STAT pathway addiction.


Subject(s)
Neoplasms , STAT Transcription Factors , Cytokines/metabolism , ErbB Receptors/metabolism , Humans , Neoplasms/genetics , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism
13.
Curr Opin Plant Biol ; 66: 102191, 2022 04.
Article in English | MEDLINE | ID: mdl-35220088

ABSTRACT

Plant-derived oils are a major agricultural product that exist in both ubiquitous forms such as common vegetable oils and in specialized forms such as castor oil and coconut oil. These specialized oils are the result of lineage-specific metabolic pathways that create oils rich in unusual fatty acids. Considerable progress has been made toward understanding the enzymes that mediate fatty acid biosynthesis, triacylglycerol assembly, and oil storage. However, efforts to translate this knowledge into renewable bioproducts via engineered oil-producing plants and algae have had limited success. Here, we review recent evidence that protein-protein interactions in each of the three major phases of oil formation appear to have profound effects on specialized oil accumulation. We suggest that furthering our knowledge of the noncatalytic attributes of enzymes and other proteins involved in oil formation will be a critical step toward creating renewable bioproducts derived from high performing, engineered oilseeds.


Subject(s)
Plant Oils , Seeds , Fatty Acids/metabolism , Plant Oils/metabolism , Plants/metabolism , Seeds/metabolism , Triglycerides/metabolism
14.
Biomolecules ; 12(1)2022 01 15.
Article in English | MEDLINE | ID: mdl-35053288

ABSTRACT

After more than fifteen years from the first high-throughput experiments for human protein-protein interaction (PPI) detection, we are still wondering how close the completion of the genome-scale human PPI network reconstruction is, what needs to be further explored and whether the biological insights gained from the holistic investigation of the current network are valid and useful. The unique structure of PICKLE, a meta-database of the human experimentally determined direct PPI network developed by our group, presently covering ~80% of the UniProtKB/Swiss-Prot reviewed human complete proteome, enables the evaluation of the interactome expansion by comparing the successive PICKLE releases since 2013. We observe a gradual overall increase of 39%, 182%, and 67% in protein nodes, PPIs, and supporting references, respectively. Our results indicate that, in recent years, (a) the PPI addition rate has decreased, (b) the new PPIs are largely determined by high-throughput experiments and mainly concern existing protein nodes and (c), as we had predicted earlier, most of the newly added protein nodes have a low degree. These observations, combined with a largely overlapping k-core between PICKLE releases and a network density increase, imply that an almost complete picture of a structurally defined network has been reached. The comparative unsupervised application of two clustering algorithms indicated that exploring the full interactome topology can reveal the protein neighborhoods involved in closely related biological processes as transcriptional regulation, cell signaling and multiprotein complexes such as the connexon complex associated with cancers. A well-reconstructed human protein interactome is a powerful tool in network biology and medicine research forming the basis for multi-omic and dynamic analyses.


Subject(s)
Protein Interaction Mapping , Protein Interaction Maps , Algorithms , Cluster Analysis , Databases, Protein , Humans , Protein Interaction Mapping/methods , Proteome/metabolism
15.
Methods Mol Biol ; 2354: 95-110, 2021.
Article in English | MEDLINE | ID: mdl-34448156

ABSTRACT

Yeast two-hybrid (Y2H) is a technique used to identify protein-protein interactions. It relies on interacting proteins bringing the two domains of a split transcription factor into close proximity, thereby reconstituting its ability to activate reporter genes. There are many variations on this technique. Here we provide an adapted protocol based on the Invitrogen ProQuest™ Two-Hybrid system, which has been used successfully to screen over 60 Phytophthora infestans pathogen effector proteins to identify candidate-interacting proteins in Solanum tuberosum, and has been used to identify many potato-potato protein-protein interactions.


Subject(s)
Solanum tuberosum , Phytophthora infestans , Plant Diseases , Saccharomyces cerevisiae , Solanum tuberosum/genetics , Transcription Factors , Two-Hybrid System Techniques
16.
Methods Mol Biol ; 2281: 117-133, 2021.
Article in English | MEDLINE | ID: mdl-33847955

ABSTRACT

The bacterial single-stranded DNA-binding protein (SSB) uses an acidic C-terminal tail to interact with over a dozen proteins, acting as a genome maintenance hub. These SSB-protein interactions are essential, as mutations to the C-terminal tail that disrupt these interactions are lethal in Escherichia coli. While the roles of individual SSB-protein interactions have been dissected with mutational studies, small-molecule inhibitors of these interactions could serve as valuable research tools and have potential as novel antimicrobial agents. This chapter describes a high-throughput screening campaign used to identify inhibitors of SSB-protein interactions. A screen targeting the PriA-SSB interface from Klebsiella pneumoniae is presented as an example, but the methods may be adapted to target nearly any SSB interaction.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Klebsiella pneumoniae/metabolism , Small Molecule Libraries/pharmacology , Binding Sites , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Models, Molecular , Protein Binding/drug effects , Protein Conformation
17.
SLAS Discov ; 26(8): 1020-1028, 2021 09.
Article in English | MEDLINE | ID: mdl-33899548

ABSTRACT

Biophysical methods are widely employed in academia and the pharmaceutical industry to detect and quantify weak molecular interactions. Such methods find broad application in fragment-based drug discovery (FBDD). In an FBDD campaign, a suitable affinity determination method is key to advancing a project beyond the initial screening phase. Protein-observed (PO) nuclear magnetic resonance (NMR) finds widespread use due to its ability to sensitively detect very weak interactions at residue-level resolution. When there are issues precluding the use of PO-NMR, ligand-observed (LO) NMR reporter assays can be a useful alternative. Such assays can measure affinities in a similar range to PO-NMR while offering some distinct advantages, especially with regard to protein consumption and compound throughput. In this paper, we take a closer look at setting up such assays for routine use, with the aim of getting high-quality, accurate data and good throughput. We assess some of the key characteristics of these assays in the mathematical framework established for fluorescence polarization assays with which the readers may be more familiar. We also provide guidance on setting up such assays and compare their performance with other affinity determination methods that are commonly used in drug discovery.


Subject(s)
Drug Discovery/methods , Genes, Reporter , Ligands , Magnetic Resonance Spectroscopy/methods , Proteins/chemistry , Biological Assay , Drug Evaluation, Preclinical , Fluorescence Polarization/methods , Humans , Protein Binding , Proteins/metabolism
18.
SLAS Discov ; 26(5): 698-711, 2021 06.
Article in English | MEDLINE | ID: mdl-33345679

ABSTRACT

ETV6 is an ETS family transcriptional repressor for which head-to-tail polymerization of its PNT domain facilitates cooperative binding to DNA by its ETS domain. Chromosomal translocations frequently fuse the ETV6 PNT domain to one of several protein tyrosine kinases. The resulting chimeric oncoproteins undergo ligand-independent self-association, autophosphorylation, and aberrant stimulation of downstream signaling pathways, leading to a variety of cancers. Currently, no small-molecule inhibitors of ETV6 PNT domain polymerization are known and no assays targeting PNT domain polymerization have been described. In this study, we developed complementary experimental and computational approaches for identifying such inhibitory compounds. One mammalian cellular approach utilized a mutant PNT domain heterodimer system covalently attached to split Gaussia luciferase fragments. In this protein-fragment complementation assay, inhibition of PNT domain heterodimerization reduces luminescence. A yeast assay took advantage of activation of the reporter HIS3 gene upon heterodimerization of mutant PNT domains fused to DNA-binding and transactivation domains. In this two-hybrid screen, inhibition of PNT domain heterodimerization prevents cell growth in medium lacking histidine. The Bristol University Docking Engine (BUDE) was used to identify virtual ligands from the ZINC8 library predicted to bind the PNT domain polymerization interfaces. More than 75 hits from these three assays were tested by nuclear magnetic resonance spectroscopy for binding to the purified ETV6 PNT domain. Although none were found to bind, the lessons learned from this study may facilitate future approaches for developing therapeutics that act against ETV6 oncoproteins by disrupting PNT domain polymerization.


Subject(s)
Drug Discovery/methods , Drug Screening Assays, Antitumor/methods , Protein Interaction Domains and Motifs/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/chemistry , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Biological Assay/methods , Genes, Reporter , Humans , Protein Binding , Structure-Activity Relationship , ETS Translocation Variant 6 Protein
19.
ChemMedChem ; 16(7): 1163-1171, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33332774

ABSTRACT

Phosphorylation-dependent protein-protein interactions play a significant role in biological signaling pathways; therefore, small molecules that are capable of influencing these interactions can be valuable research tools and have potential as pharmaceutical agents. MEMO1 (mediator of ErbB2-cell driven motility) is a phosphotyrosine-binding protein that interacts with a variety of protein partners and has been found to be upregulated in breast cancer patients. Herein, we report the first small-molecule inhibitors of MEMO1 interactions identified through a virtual screening platform and validated in a competitive fluorescence polarization assay. Initial structure-activity relationships have been investigated for these phenazine-core inhibitors and the binding sites have been postulated using molecular dynamics simulations. The most potent biochemical inhibitor is capable of disrupting the large protein interface with a KI of 2.7 µm. In addition, the most promising phenazine core compounds slow the migration of breast cancer cell lines in a scratch assay.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Phenazines/pharmacology , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Female , Fluorescence Polarization , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Dynamics Simulation , Molecular Structure , Phenazines/chemical synthesis , Phenazines/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
20.
ChemMedChem ; 16(6): 949-954, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33305877

ABSTRACT

Inhibition of PSD-95 has emerged as a promising strategy for the treatment of ischemic stroke, as shown with peptide-based compounds that target the PDZ domains of PSD-95. In contrast, developing potent and drug-like small molecules against the PSD-95 PDZ domains has so far been unsuccessful. Here, we explore the druggability of the PSD-95 PDZ1-2 domain and use fragment screening to investigate if this protein is prone to binding small molecules. We screened 2500 fragments by fluorescence polarization (FP) and validated the hits by surface plasmon resonance (SPR), including an inhibition counter-test, and found four promising fragments. Three ligand efficient fragments were shown by 1 H,15 N HSQC NMR to bind in the small hydrophobic P0 pockets of PDZ1-2, and one of them underwent structure-activity relationship (SAR) studies. Overall, we demonstrate that fragment screening can successfully be applied to PDZ1-2 of PSD-95 and disclose novel fragments that can serve as starting points for optimization towards small-molecule PDZ domain inhibitors.


Subject(s)
Disks Large Homolog 4 Protein/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Drug Evaluation, Preclinical , Fluorescence Polarization , Humans , Ligands , Models, Molecular , Molecular Structure , PDZ Domains/drug effects , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL