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










Publication year range
1.
Nanoscale ; 13(16): 7667-7677, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33928964

ABSTRACT

Surface-Enhanced Infrared Absorption (SEIRA) has been proposed as a valuable tool for protein binding studies, but its performances have been often proven on model proteins undergoing severe secondary structure rearrangements, while ligand binding only marginally involves the protein backbone in the vast majority of the biologically relevant cases. In this study we demonstrate the potential of SEIRA microscopy for highlighting the very subtle secondary structure modifications associated with the binding of Lapatinib, a tyrosine kinase inhibitor (TKI), to epidermal growth factor receptor (EGFR), a well-known driver of tumorigenesis in pathological settings such as lung, breast and brain cancers. By boosting the performances of Mid-IR plasmonic devices based on nanoantennas cross-geometry, accustoming the protein purification protocols, carefully tuning the protein anchoring methodology and optimizing the data analysis, we were able to detect EGFR secondary structure modification associated with few amino acids. A nano-patterned platform with this kind of sensitivity bridges biophysical and structural characterization methods, thus opening new possibilities in studying of proteins of biomedical interest, particularly for drug-screening purposes.


Subject(s)
Lung Neoplasms , Microscopy , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Mutation , Protein Kinase Inhibitors/pharmacology
2.
Int J Biol Macromol ; 182: 502-511, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33848543

ABSTRACT

High-resolution structural data of complexes between antibodies and membrane receptors still represent a demanding task. In this study, we used complementary sets of experimental data to obtain a structural model of the complex formed by the human epidermal growth factor receptor 2 (HER2) and its specific nanobody A10. First we identified by NMR the residues that bind or rearrange as a consequence of the complex formation. In parallel, the complex was cross-linked, digested and the resulting peptides were characterized by mass-spectrometry to define maximal distance restraints between HER2 and A10 amino acids in their complex. These independent datasets guided a docking process, refined by molecular dynamics simulations, to develop a model of the complex and estimate per-residue free-energy contributions. Such a model explains the experimental data and identifies a second, non-canonical paratope, located in the region opposite to the conventional nanobody paratope, formed by the hypervariable loop regions LH1 and LH3. Both paratopes contributed substantially to the overall affinity by binding to independent HER2 epitopes. Nanobody mutants with substitution of key interaction residues, as indicated by the model, possess significantly lower affinity for HER2. This is the first described case of a "natural" biparatopic nanobody, directly selected by in-vitro panning.


Subject(s)
Binding Sites, Antibody , Receptor, ErbB-2/chemistry , Single-Chain Antibodies/chemistry , Humans , Molecular Docking Simulation , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Protein Binding , Receptor, ErbB-2/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
3.
J Mol Biol ; 433(13): 166964, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33781758

ABSTRACT

Recent years have seen a dramatic improvement in protein-design methodology. Nevertheless, most methods demand expert intervention, limiting their widespread adoption. By contrast, the PROSS algorithm for improving protein stability and heterologous expression levels has been successfully applied to a range of challenging enzymes and binding proteins. Here, we benchmark the application of PROSS as a stand-alone tool for protein scientists with no or limited experience in modeling. Twelve laboratories from the Protein Production and Purification Partnership in Europe (P4EU) challenged the PROSS algorithm with 14 unrelated protein targets without support from the PROSS developers. For each target, up to six designs were evaluated for expression levels and in some cases, for thermal stability and activity. In nine targets, designs exhibited increased heterologous expression levels either in prokaryotic and/or eukaryotic expression systems under experimental conditions that were tailored for each target protein. Furthermore, we observed increased thermal stability in nine of ten tested targets. In two prime examples, the human Stem Cell Factor (hSCF) and human Cadherin-Like Domain (CLD12) from the RET receptor, the wild type proteins were not expressible as soluble proteins in E. coli, yet the PROSS designs exhibited high expression levels in E. coli and HEK293 cells, respectively, and improved thermal stability. We conclude that PROSS may improve stability and expressibility in diverse cases, and that improvement typically requires target-specific expression conditions. This study demonstrates the strengths of community-wide efforts to probe the generality of new methods and recommends areas for future research to advance practically useful algorithms for protein science.


Subject(s)
Algorithms , Protein Stability , Animals , Escherichia coli/metabolism , HEK293 Cells , High-Throughput Screening Assays , Humans , Models, Molecular , Proteins/chemistry , Proteins/metabolism , Solubility , Temperature , Zebrafish
4.
Molecules ; 25(4)2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32093112

ABSTRACT

Alzheimer's disease is the most common type of dementia, affecting millions of people worldwide. One of its main consequences is memory loss, which is related to downstream effectors of cyclic adenosine monophosphate (cAMP). A well-established strategy to avoid cAMP degradation is the inhibition of phosphodiesterase (PDE). In recent years, GEBR-32a has been shown to possess selective inhibitory properties against PDE type 4 family members, resulting in an improvement in spatial memory processes without the typical side effects that are usually correlated with this mechanism of action. In this work, we performed the HPLC chiral resolution and absolute configuration assignment of GEBR-32a. We developed an efficient analytical and semipreparative chromatographic method exploiting an amylose-based stationary phase, we studied the chiroptical properties of both enantiomers and we assigned their absolute configuration by 1H-NMR (nuclear magnetic resonance). Lastly, we measured the IC50 values of both enantiomers against both the PDE4D catalytic domain and the long PDE4D3 isoform. Results strongly support the notion that GEBR-32a inhibits the PDE4D enzyme by interacting with both the catalytic pocket and the regulatory domains.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Phosphodiesterase 4 Inhibitors/chemistry , Humans , Nuclear Magnetic Resonance, Biomolecular
5.
Protein Expr Purif ; 166: 105505, 2020 02.
Article in English | MEDLINE | ID: mdl-31563543

ABSTRACT

Recombinant antibodies can be expressed as fusion constructs in combination with tags which simplify their engineering into reliable and homogeneous immunoreagents by allowing site-specific, 1:1 functionalization. Several tags and corresponding reagents for recombinant protein derivatization have been proposed but benchmarking surveys for the evaluation of their effect on the characteristics of recombinant antibodies have not been reported. In this work we evaluated the impact on expression yields, shelf-stability, thermostability and binding affinity of a set of C-terminal tags fused to the same anti-Her2 nanobody. Furthermore, we assessed the efficiency of the derivatization process. The constructs always bore a 6xHis tag plus either the controls (EGFP and C-tag) or CLIP, HALO, AviTag, the LEPTG sequence recognized by Sortase A (Sortase tag), or a free cysteine. The advantages and drawbacks of the different systems were analyzed and discussed.


Subject(s)
Recombinant Fusion Proteins/genetics , Single-Domain Antibodies/genetics , Binding, Competitive , Cysteine/metabolism , Escherichia coli , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Oxidoreductases/chemistry , Oxidoreductases/genetics , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/genetics , Protein Stability , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/chemistry , Single-Domain Antibodies/chemistry
6.
Chem Commun (Camb) ; 55(93): 14043-14046, 2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31690899

ABSTRACT

We present an in silico mutagenetic protocol for improving the binding affinity of single domain antibodies (or nanobodies, VHHs). The method iteratively attempts random mutations in the interacting region of the protein and evaluates the resulting binding affinity towards the target by scoring, with a collection of scoring functions, short explicit solvent molecular dynamics trajectories of the binder-target complexes. The acceptance/rejection of each attempted mutation is carried out by a consensus decision-making algorithm, which considers all individual assessments derived from each scoring function. The method was benchmarked by evolving a single complementary determining region (CDR) of an anti-HER2 VHH hit obtained by direct panning of a phage display library. The optimised VHH mutant showed significantly enhanced experimental affinity with respect to the original VHH it matured from. The protocol can be employed as it is for the optimization of peptides, antibody fragments, and (given enough computational power) larger antibodies.


Subject(s)
Antigen-Antibody Reactions , Computer Simulation , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Algorithms , Humans , Molecular Dynamics Simulation , Mutation , Receptor, ErbB-2/immunology , Single-Domain Antibodies/genetics
7.
ChemMedChem ; 13(24): 2627-2634, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30457710

ABSTRACT

Following the discovery of a type III allosteric modulator of cyclin-dependent kinase 2 (CDK2) characterized by a hexahydrocyclopenta[c]quinolone scaffold, three different series of its derivatives were synthesized and biologically evaluated. Docking of the synthesized compounds into the allosteric pocket of CDK2 allowed the elucidation of structure-activity relationships (SARs). Moreover, the compounds were tested on the wild-type epidermal growth factor receptor (EGFR) kinase domain (KD) and its clinically relevant T790M/L858R mutant form. Herein we describe the first SAR investigation of allosteric ligands that bind to the type III inhibitor pocket of CDK2 and EGFR-KD. Although the activity of the synthesized inhibitors needs to be improved, the obtained results provide clear-cut indications about pharmacophore requirements and selectivity determinants. Remarkably, this study led to the identification of a selective T790M/L858R EGFR allosteric inhibitor that is inactive toward both wild-type EGFR and CDK2. Finally, docking into the T790M/L858R EGFR-KD led us to hypothesize that the compounds bind to the double-mutant EGFR-KD by adopting a binding mode different from that in CDK2, thus rationalizing the observed selectivity profile.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Quinolines/chemistry , Allosteric Regulation , Animals , ErbB Receptors/genetics , Escherichia coli , Ligands , Models, Molecular , Mutation , Protein Binding , Quinolines/pharmacology , Sf9 Cells , Structure-Activity Relationship
8.
Future Med Chem ; 10(13): 1545-1553, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29766737

ABSTRACT

AIM: The EGFR inhibitors represent the first-line treatment of non-small-cell lung cancer. However, the emergence of resistance urgently requires the development of new inhibitors targeting drug-resistant mutants. METHODOLOGY: A recently released structure of an EGFR kinase domain in complex with an allosteric inhibitor and a mutant protein model derived from it were used to set up a low-cost high-throughput docking protocol for the fast identification of EGFR allosteric inhibitors. CONCLUSION: The virtual screening of commercially available compounds led to the identification of interesting new hit compounds. The most promising hit was confirmed to be a new allosteric inhibitor of wild-type and T790M/L858R double mutant EGFR which was able to inhibit the growth of  non-small-cell lung cancer cell lines.


Subject(s)
Molecular Docking Simulation/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Drug Discovery , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , High-Throughput Screening Assays/economics , High-Throughput Screening Assays/methods , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Molecular Docking Simulation/economics , Mutation
9.
Biochemistry ; 57(19): 2876-2888, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29652483

ABSTRACT

Selected members of the large rolipram-related GEBR family of type 4 phosphodiesterase (PDE4) inhibitors have been shown to facilitate long-term potentiation and to improve memory functions without causing emetic-like behavior in rodents. Despite their micromolar-range binding affinities and their promising pharmacological and toxicological profiles, few if any structure-activity relationship studies have been performed to elucidate the molecular bases of their action. Here, we report the crystal structure of a number of GEBR library compounds in complex with the catalytic domain of PDE4D as well as their inhibitory profiles for both the long PDE4D3 isoform and the catalytic domain alone. Furthermore, we assessed the stability of the observed ligand conformations in the context of the intact enzyme using molecular dynamics simulations. The longer and more flexible ligands appear to be capable of forming contacts with the regulatory portion of the enzyme, thus possibly allowing some degree of selectivity between the different PDE4 isoforms.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Memory/drug effects , Phosphodiesterase 4 Inhibitors/chemistry , Structure-Activity Relationship , Animals , Catalytic Domain , Crystallography, X-Ray , Humans , Ligands , Memory/physiology , Molecular Dynamics Simulation , Phosphodiesterase 4 Inhibitors/therapeutic use , Rolipram/chemistry , Rolipram/therapeutic use
10.
Nat Struct Mol Biol ; 24(3): 270-278, 2017 03.
Article in English | MEDLINE | ID: mdl-28165509

ABSTRACT

Protein modification by ubiquitin and ubiquitin-like modifiers (Ubls) is counteracted by ubiquitin proteases and Ubl proteases, collectively termed DUBs. In contrast to other proteases of the ubiquitin-specific protease (USP) family, USP18 shows no reactivity toward ubiquitin but specifically deconjugates the interferon-induced Ubl ISG15. To identify the molecular determinants of this specificity, we solved the crystal structures of mouse USP18 alone and in complex with mouse ISG15. USP18 was crystallized in an open and a closed conformation, thus revealing high flexibility of the enzyme. Structural data, biochemical and mutational analysis showed that only the C-terminal ubiquitin-like domain of ISG15 is recognized and essential for USP18 activity. A critical hydrophobic patch in USP18 interacts with a hydrophobic region unique to ISG15, thus providing evidence that USP18's ISG15 specificity is mediated by a small interaction interface. Our results may provide a structural basis for the development of new drugs modulating ISG15 linkage.


Subject(s)
Cytokines/metabolism , Endopeptidases/chemistry , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Zebrafish Proteins/chemistry , Animals , Crystallization , Crystallography, X-Ray , Cytokines/chemistry , Endopeptidases/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Conformation , Protein Domains , Structure-Activity Relationship , Substrate Specificity , Ubiquitin/metabolism , Ubiquitins/chemistry , Ubiquitins/metabolism , Zebrafish , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL