Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 436(16): 168640, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38844044

ABSTRACT

Computational free energy-based methods have the potential to significantly improve throughput and decrease costs of protein design efforts. Such methods must reach a high level of reliability, accuracy, and automation to be effectively deployed in practical industrial settings in a way that impacts protein design projects. Here, we present a benchmark study for the calculation of relative changes in protein-protein binding affinity for single point mutations across a variety of systems from the literature, using free energy perturbation (FEP+) calculations. We describe a method for robust treatment of alternate protonation states for titratable amino acids, which yields improved correlation with and reduced error compared to experimental binding free energies. Following careful analysis of the largest outlier cases in our dataset, we assess limitations of the default FEP+ protocols and introduce an automated script which identifies probable outlier cases that may require additional scrutiny and calculates an empirical correction for a subset of charge-related outliers. Through a series of three additional case study systems, we discuss how Protein FEP+ can be applied to real-world protein design projects, and suggest areas of further study.

2.
bioRxiv ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38712280

ABSTRACT

Computational free energy-based methods have the potential to significantly improve throughput and decrease costs of protein design efforts. Such methods must reach a high level of reliability, accuracy, and automation to be effectively deployed in practical industrial settings in a way that impacts protein design projects. Here, we present a benchmark study for the calculation of relative changes in protein-protein binding affinity for single point mutations across a variety of systems from the literature, using free energy perturbation (FEP+) calculations. We describe a method for robust treatment of alternate protonation states for titratable amino acids, which yields improved correlation with and reduced error compared to experimental binding free energies. Following careful analysis of the largest outlier cases in our dataset, we assess limitations of the default FEP+ protocols and introduce an automated script which identifies probable outlier cases that may require additional scrutiny and calculates an empirical correction for a subset of charge-related outliers. Through a series of three additional case study systems, we discuss how protein FEP+ can be applied to real-world protein design projects, and suggest areas of further study.

3.
Nat Commun ; 15(1): 1509, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38374079

ABSTRACT

In contemporary drug discovery, enhancing the sp3-hybridized character of molecular structures is paramount, necessitating innovative synthetic methods. Herein, we introduce a deoxygenative cross-electrophile coupling technique that pairs easily accessible carboxylic acid-derived redox-active esters with aldehyde sulfonyl hydrazones, employing Eosin Y as an organophotocatalyst under visible light irradiation. This approach serves as a versatile, metal-free C(sp3)-C(sp3) cross-coupling platform. We demonstrate its synthetic value as a safer, broadly applicable C1 homologation of carboxylic acids, offering an alternative to the traditional Arndt-Eistert reaction. Additionally, our method provides direct access to cyclic and acyclic ß-arylethylamines using diverse aldehyde-derived sulfonyl hydrazones. Notably, the methodology proves to be compatible with the late-stage functionalization of peptides on solid-phase, streamlining the modification of intricate peptides without the need for exhaustive de-novo synthesis.

4.
Methods Mol Biol ; 2355: 35-48, 2021.
Article in English | MEDLINE | ID: mdl-34386948

ABSTRACT

Synthetic molecular probes have recently been in focus for their potential use in target deconvolution, target engagement studies, and imaging. With the field expanding, new strategies to develop such tools are in high demand. While traditional conjugation techniques relying on inherently nucleophilic amino acids such as cysteine (Cys) and lysine (Lys) or pre-incorporated non-natural amino acids are still heavily used, novel methodologies for the direct and site-selective modification of peptides are attracting increasing attention. Of particular interest are Late-Stage Functionalization (LSF) approaches based on radical chemistry as they afford mild and biocompatible alternatives to transition-metal catalysis. A recent synthetic method, which leverages the unique reactivity of histidine (His), has proven to be a promising new strategy for LSF and site-selective conjugation of unprotected peptides. In this chapter, detailed step-by-step protocols depicting the C2-alkylation of His-containing peptides, the unveiling of a ketone as handle for hydrazone conjugation, and its use to site-selectively introduce a fluorophore at this residue are discussed. In addition to its application toward the synthesis of molecular probes, this methodology can be employed in peptide-based drug discovery programs, offering the possibility to rapidly explore the chemical space surrounding peptide hits. Finally, this strategy is also amenable to the preparation of novel peptide-ASO/small molecule drug conjugates.


Subject(s)
Peptides/chemistry , Amino Acids , Histidine , Hydrazones , Molecular Probes , Oximes
5.
Nat Commun ; 11(1): 5425, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110077

ABSTRACT

Transcription factors are key protein effectors in the regulation of gene transcription, and in many cases their activity is regulated via a complex network of protein-protein interactions (PPI). The chemical modulation of transcription factor activity is a long-standing goal in drug discovery but hampered by the difficulties associated with the targeting of PPIs, in particular when extended and flat protein interfaces are involved. Peptidomimetics have been applied to inhibit PPIs, however with variable success, as for certain interfaces the mimicry of a single secondary structure element is insufficient to obtain high binding affinities. Here, we describe the design and characterization of a stabilized protein tertiary structure that acts as an inhibitor of the interaction between the transcription factor TEAD and its co-repressor VGL4, both playing a central role in the Hippo signalling pathway. Modification of the inhibitor with a cell-penetrating entity yielded a cell-permeable proteomimetic that activates cell proliferation via regulation of the Hippo pathway, highlighting the potential of protein tertiary structure mimetics as an emerging class of PPI modulators.


Subject(s)
Peptidomimetics , Transcription Factors/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Hippo Signaling Pathway , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Angew Chem Int Ed Engl ; 58(52): 19096-19102, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31609503

ABSTRACT

The late-stage functionalization (LSF) of peptides represents a valuable strategy for the design of potent peptide pharmaceuticals by enabling rapid exploration of chemical diversity and offering novel opportunities for peptide conjugation. While the C(sp2 )-H activation of tryptophan (Trp) is well documented, the resurgence of radical chemistry is opening new avenues for the C-H functionalization of other aromatic side-chains. Herein, we report the first example of LSF at C2 of histidine (His) utilizing a broad scope of aliphatic sulfinate salts as radical precursors. In this work, the exquisite selectivity for histidine functionalization was demonstrated through the alkylation of complex unprotected peptides in aqueous media. Finally, this methodology was extended for the installation of a ketone handle, providing an unprecedented anchor for selective oxime/hydrazone conjugation at histidine.


Subject(s)
Histidine/chemistry , Peptides/chemistry , Humans
7.
Angew Chem Int Ed Engl ; 56(35): 10294-10323, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28186380

ABSTRACT

Our ever-increasing understanding of biological systems is providing a range of exciting novel biological targets, whose modulation may enable novel therapeutic options for many diseases. These targets include protein-protein and protein-nucleic acid interactions, which are, however, often refractory to classical small-molecule approaches. Other types of molecules, or modalities, are therefore required to address these targets, which has led several academic research groups and pharmaceutical companies to increasingly use the concept of so-called "new modalities". This Review defines for the first time the scope of this term, which includes novel peptidic scaffolds, oligonucleotides, hybrids, molecular conjugates, as well as new uses of classical small molecules. We provide the most representative examples of these modalities to target large binding surface areas such as those found in protein-protein interactions and for biological processes at the center of cell regulation.


Subject(s)
Drug Discovery , Nucleic Acids/antagonists & inhibitors , Oligonucleotides/pharmacology , Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Humans , Nucleic Acids/chemistry , Oligonucleotides/chemistry , Proteins/chemistry , Small Molecule Libraries/chemistry , Surface Properties
8.
J Med Chem ; 59(21): 9599-9621, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27362955

ABSTRACT

Over the past decade, foldamers have progressively emerged as useful architectures to mimic secondary structures of proteins. Peptidic foldamers, consisting of various amino acid based backbones, have been the most studied from a therapeutic perspective, while polyaromatic foldamers have barely evolved from their nascency and remain perplexing for medicinal chemists due to their poor drug-like nature. Despite these limitations, this compound class may still offer opportunities to study challenging targets or provide chemical biology tools. The potential of foldamer drug candidates reaching the clinic is still a stretch. Nevertheless, advances in the field have demonstrated their potential for the discovery of next generation therapeutics. In this perspective, the current knowledge of foldamers is reviewed in a drug discovery context. Recent advances in the early phases of drug discovery including hit finding, target validation, and optimization and molecular modeling are discussed. In addition, challenges and focus areas are debated and gaps highlighted.


Subject(s)
Drug Discovery , Peptides/pharmacology , Polycyclic Aromatic Hydrocarbons/pharmacology , Protein Folding/drug effects , Humans , Models, Molecular , Molecular Structure , Peptides/chemistry , Peptides/therapeutic use , Polycyclic Aromatic Hydrocarbons/chemistry , Polycyclic Aromatic Hydrocarbons/therapeutic use
9.
PLoS One ; 11(2): e0148608, 2016.
Article in English | MEDLINE | ID: mdl-26863431

ABSTRACT

The aim of this study was to design, synthesize and validate a multifunctional antidepressant probe that is modified at two distinct positions. The purpose of these modifications was to allow covalent linkage of the probe to interaction partners, and decoration of probe-target complexes with fluorescent reporter molecules. The strategy for the design of such a probe (i.e., azidobupramine) was guided by the need for the introduction of additional functional groups, conveying the required properties while keeping the additional moieties as small as possible. This should minimize the risk of changing antidepressant-like properties of the new probe azidobupramine. To control for this, we evaluated the binding parameters of azidobupramine to known target sites such as the transporters for serotonin (SERT), norepinephrine (NET), and dopamine (DAT). The binding affinities of azidobupramine to SERT, NET, and DAT were in the range of structurally related and clinically active antidepressants. Furthermore, we successfully visualized azidobupramine-SERT complexes not only in SERT-enriched protein material but also in living cells stably overexpressing SERT. To our knowledge, azidobupramine is the first structural analogue of a tricyclic antidepressant that can be covalently linked to target structures and further attached to reporter molecules while preserving antidepressant-like properties and avoiding radioactive isotopes.


Subject(s)
Antidepressive Agents, Tricyclic/chemistry , Azepines/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Fluorescent Dyes/chemistry , Molecular Probes/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Amines/chemistry , Antidepressive Agents, Tricyclic/chemical synthesis , Azepines/chemical synthesis , Binding Sites , Cell Line , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/chemistry , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorescent Dyes/chemical synthesis , Gene Expression , Humans , Kinetics , Ligands , Molecular Probes/chemical synthesis , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Protein Binding , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry
10.
Nat Chem ; 6(11): 1009-16, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25343607

ABSTRACT

The disulfide bonds that form between two cysteine residues are important in defining and rigidifying the structures of proteins and peptides. In polypeptides containing multiple cysteine residues, disulfide isomerization can lead to multiple products with different biological activities. Here, we describe the development of a dithiol amino acid (Dtaa) that can form two disulfide bridges at a single amino acid site. Application of Dtaas to a serine protease inhibitor and a nicotinic acetylcholine receptor inhibitor that contain disulfide constraints enhanced their inhibitory activities 40- and 7.6-fold, respectively. X-ray crystallographic and NMR structure analysis show that the peptide ligands containing Dtaas have retained their native tertiary structures. We furthermore show that replacement of two cysteines by Dtaas can avoid the formation of disulfide bond isomers. With these properties, Dtaas are likely to have broad application in the rational design or directed evolution of peptides and proteins with high activity and stability.


Subject(s)
Amino Acids/chemistry , Peptides/chemistry , Amino Acids/metabolism , Binding Sites , Catalytic Domain , Conotoxins/chemistry , Conotoxins/metabolism , Crystallography, X-Ray , Disulfides/chemistry , Isomerism , Ligands , Molecular Dynamics Simulation , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/metabolism , Peptides/metabolism , Protein Binding , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/metabolism
11.
J Med Chem ; 56(10): 3922-35, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23647266

ABSTRACT

The design of efficient ligands remains a key challenge in drug discovery. In the quest for lead-like ligands for the FK506-binding protein 51 (FKBP51), we designed two new classes of bicyclic sulfonamides to probe the contribution of conformational energy in these ligands. The [4.3.1] scaffold had consistently higher affinity compared to the [3.3.1] or monocyclic scaffolds, which could be attributed to better preorganization of two key recognition motifs. Surprisingly, the binding of the rigid [4.3.1] scaffold was enthalpy-driven and entropically disfavored compared to the flexible analogues. Cocrystal structures at atomic resolution revealed that the sulfonamide nitrogen in the bicyclic scaffolds can accept an unusual hydrogen bond from Tyr(113) that mimics the putative FKBP transition state. This resulted in the first lead-like, functionally active ligand for FKBP51. Our work exemplifies how atom-efficient ligands can be achieved by careful conformational control even in very open and thus difficult binding sites such as FKBP51.


Subject(s)
Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Tacrolimus Binding Proteins/drug effects , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Binding Sites , Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/pharmacology , Calorimetry , Chromatography, High Pressure Liquid , Crystallography , Drug Design , Humans , Indicators and Reagents , Ligands , Protein Conformation , Receptors, Gonadotropin/drug effects , Receptors, Gonadotropin/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacology , Tacrolimus/chemistry
12.
J Med Chem ; 55(9): 4123-31, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22455398

ABSTRACT

FK506-binding proteins (FKBP) 51 and 52 are cochaperones that modulate the signal transduction of steroid hormone receptors. Single nucleotide polymorphisms in the gene encoding FKBP51 have been associated with a variety of psychiatric disorders. Rapamycin and FK506 are two macrocyclic natural products, which tightly bind to most FKBP family members, including FKBP51 and FKBP52. A bioisosteric replacement of the α-ketoamide moiety of rapamycin and FK506 with a sulfonamide was envisaged with the retention of the conserved hydrogen bonds. A focused solid support-based synthesis protocol was developed, which led to ligands with submicromolar affinity for FKBP51 and FKBP52. The molecular binding mode for one sulfonamide analogue was confirmed by X-ray crystallography.


Subject(s)
Pipecolic Acids/chemical synthesis , Sulfonamides/chemical synthesis , Tacrolimus Binding Proteins/metabolism , Tacrolimus/analogs & derivatives , Crystallography, X-Ray , Fluorescence Polarization , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Pipecolic Acids/chemistry , Pipecolic Acids/pharmacology , Protein Binding , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Tacrolimus/chemical synthesis , Tacrolimus/chemistry , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/antagonists & inhibitors
13.
J Med Chem ; 55(9): 4114-22, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22455444

ABSTRACT

The FK506-binding proteins (FKBP) 51 and 52 are cochaperones that modulate the signal transduction of steroid hormone receptors. Both proteins have been implicated in prostate cancer. Furthermore, single nucleotide polymorphisms in the gene encoding FKBP51 have been associated with a variety of psychiatric disorders. Rapamycin and FK506 are two macrocyclic natural products that bind to these proteins indiscriminately but with nanomolar affinity. We here report the cocrystal structure of FKBP51 with a simplified α-ketoamide analogue derived from FK506 and the first structure-activity relationship analysis for FKBP51 and FKBP52 based on this compound. In particular, the tert-pentyl group of this ligand was systematically replaced by a cyclohexyl ring system, which more closely resembles the pyranose ring in the high-affinity ligands rapamycin and FK506. The interaction with FKBPs was found to be surprisingly tolerant to the stereochemistry of the attached cyclohexyl substituents. The molecular basis for this tolerance was elucidated by X-ray cocrystallography.


Subject(s)
Tacrolimus Binding Proteins/metabolism , Tacrolimus/analogs & derivatives , Crystallography, X-Ray , Humans , Ligands , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Structure , Structure-Activity Relationship , Tacrolimus/chemical synthesis , Tacrolimus/chemistry , Tacrolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...