469 research outputs found

    Hydrophobicity Directed Chiral Self-Assembly and Aggregation-Induced Emission:Diacetylene-Cored Pseudopeptide Chiral Dopants

    Get PDF
    Here we delineate simple and tunable hydrophobically driven chiral functional assemblies of diacetylene cored pseudopeptides. These amino acid appended, rigid core dialkynes constitute promising chiral supramolecular building blocks for materials properties engineering. The chiral appended amino acid elements allow for simple tuning of solubility and interaction properties as well as governing chirality, while the central dialkyne core can impart hydrophobically driven assembly and Aggregation Induced Emission (AIE) properties. The self-assembly of these rod-like dialkynes can be regulated by tuning the solvent environment, with for example self-assembly into vesicles in acetonitrile and into helical organization with AIE in a H 2 O/DMSO mixture. Of additional high interest, these supramolecular materials, themselves devoid of liquid crystal (LC) properties, can induce chirality into non-chiral LC matrices with high helical twisting power. </p

    Carboraneā€“Ī²-cyclodextrin complexes as a supramolecular connector for bioactive surfaces

    Get PDF
    Supramolecular chemistry provides an attractive entry to generate dynamic and well-controlled bioactive surfaces. Novel hostā€“guest systems are urgently needed to provide a broader affinity and applicability portfolio. A synthetic strategy to carboraneā€“peptide bioconjugates was therefore developed to provide an entry to monovalent supramolecular functionalization of Ī²-cyclodextrin coated surfaces. The Ī²-cyclodextrinĀ·carboraneā€“cRGD surfaces are formed efficiently and with high affinity as demonstrated by IR-RAS, WCA, and QCM-D, compare favourable to existing bio-active hostā€“guest surface assemblies, and display an efficient bioactivity, as illustrated by a strong functional effect of the supramolecular system on the cell adhesion and spreading properties. Cells seeded on the supramolecular surface displaying bioactive peptide epitopes exhibited a more elongated morphology, focal adhesions, and stronger cell adhesion compared to control surfaces. This highlights the macroscopic functionality of the novel supramolecular immobilization strategy

    Discovery of 14-3-3 PPI Stabilizers by Extension of an Amidine-Substituted Thiophene Fragment

    Get PDF
    Protein-protein interaction (PPI) modulation is a promising approach in drug discovery with the potential to expand the 'druggable' proteome and develop new therapeutic strategies. While there have been significant advancements in methodologies for developing PPI inhibitors, there is a relative scarcity of literature describing the 'bottom-up' development of PPI stabilizers (Molecular Glues). The hub protein 14-3-3 and its interactome provide an excellent platform for exploring conceptual approaches to PPI modulation, including evolution of chemical matter for Molecular Glues. In this study, we employed a fragment extension strategy to discover stabilizers for the complex of 14-3-3 protein and an Estrogen Receptor alpha-derived peptide (ERĪ±). A focused library of analogues derived from an amidine-substituted thiophene fragment enhanced the affinity of the 14-3-3/ERĪ± complex up to 6.2-fold. Structure-activity relationship (SAR) analysis underscored the importance of the newly added, aromatic side chain with a certain degree of rigidity. X-ray structural analysis revealed a unique intermolecular Ļ€-Ļ€ stacking binding mode of the most active analogues, resulting in the simultaneous binding of two molecules to the PPI binding pocket. Notably, analogue 11 displayed selective stabilization of the 14-3-3/ERĪ± complex.</p

    Reversible Dual-Covalent Molecular Locking of the 14-3-3/ERRĪ³ Protein-Protein Interaction as a Molecular Glue Drug Discovery Approach

    Get PDF
    Molecules that stabilize protein-protein interactions (PPIs) are invaluable as tool compounds for biophysics and (structural) biology, and as starting points for molecular glue drug discovery. However, identifying initial starting points for PPI stabilizing matter is highly challenging, and chemical optimization is labor-intensive. Inspired by chemical crosslinking and reversible covalent fragment-based drug discovery, we developed an approach that we term ā€œmolecular locksā€ to rapidly access molecular glue-like tool compounds. These dual-covalent small molecules reversibly react with a nucleophilic amino acid on each of the partner proteins to dynamically crosslink the protein complex. The PPI between the hub protein 14-3-3 and estrogen-related receptor Ī³ (ERRĪ³) was used as a pharmacologically relevant case study. Based on a focused library of dual-reactive small molecules, a molecular glue tool compound was rapidly developed. Biochemical assays and X-ray crystallographic studies validated the ternary covalent complex formation and overall PPI stabilization via dynamic covalent crosslinking. The molecular lock approach is highly selective for the specific 14-3-3/ERRĪ³ complex, over other 14-3-3 complexes. This selectivity is driven by the interplay of molecular reactivity and molecular recognition of the composite PPI binding interface. The long lifetime of the dual-covalent locks enabled the selective stabilization of the 14-3-3/ERRĪ³ complex even in the presence of several other competing 14-3-3 clients with higher intrinsic binding affinities. The molecular lock approach enables systematic, selective, and potent stabilization of protein complexes to support molecular glue drug discovery.</p

    Molecular basis and dual ligand regulation of tetrameric Estrogen Receptor Ī±/14-3-3Ī¶ protein complex

    Get PDF
    Therapeutic strategies targeting Nuclear Receptors (NRs) beyond their endogenous ligand binding pocket have gained significant scientific interest, driven by a need to circumvent problems associated with drug resistance and pharmacological profile. The hub protein 14-3-3 is an endogenous regulator of various NRs, providing a novel entry point for small molecule modulation of NR activity. Exemplified, 14-3-3 binding to the C-terminal F-domain of the Estrogen Receptor alpha (ERĪ±), and small molecule stabilization of the ERĪ±/14-3-3Ī¶ protein complex by the natural product Fusicoccin A (FC-A), was demonstrated to downregulate ERĪ±-mediated breast cancer proliferation. This presents a novel drug discovery approach to target ERĪ±, however, structural and mechanistic insights into ERĪ±/14-3-3 complex formation are lacking. Here, we provide an in-depth molecular understanding of the ERĪ±/14-3-3Ī¶ complex by isolating 14-3-3Ī¶ in complex with an ERĪ± protein construct comprising its Ligand Binding Domain (LBD) and phosphorylated F-domain. Bacterial co-expression and co-purification of the ERĪ±/14-3-3Ī¶ complex, followed by extensive biophysical and structural characterization, revealed a tetrameric complex between the ERĪ± homodimer and the 14-3-3Ī¶ homodimer. 14-3-3Ī¶ binding to ERĪ±, and ERĪ±/14-3-3Ī¶ complex stabilization by FC-A, appeared to be orthogonal to ERĪ± endogenous agonist (E2) binding, E2-induced conformational changes, and cofactor recruitment. Similarly, the ERĪ± antagonist 4-hydroxytamoxifen inhibited cofactor recruitment to the ERĪ± LBD while ERĪ± was bound to 14-3-3Ī¶. Furthermore, stabilization of the ERĪ±/14-3-3Ī¶ protein complex by FC-A was not influenced by the disease-associated and 4-hydroxytamoxifen resistant ERĪ±-Y537S mutant. Together, these molecular and mechanistic insights provide direction for targeting ERĪ± via the ERĪ±/14-3-3 complex as an alternative drug discovery approach.</p

    Enzymatic Regulation of Protein-Protein Interactions in Artificial Cells

    Get PDF
    Membraneless organelles are important for spatial organization of proteins and regulation of intracellular processes. Proteins can be recruited to these condensates by specific proteinā€“protein or proteinā€“nucleic acid interactions, which are often regulated by post-translational modifications. However, the mechanisms behind these dynamic, affinity-based protein recruitment events are not well understood. Here, a coacervate system that incorporates the 14-3-3 scaffold protein to study enzymatically regulated recruitment of 14-3-3-binding proteins is presented, which mostly bind in a phosphorylation-dependent manner. Synthetic coacervates are efficiently loaded with 14-3-3, and phosphorylated binding partners, such as the c-Raf pS233/pS259 peptide (c-Raf), show 14-3-3-dependent sequestration with up to 161-fold increase in local concentration. The c-Raf domain is fused to green fluorescent protein (GFP-c-Raf) to demonstrate recruitment of proteins. In situ phosphorylation of GFP-c-Raf by a kinase leads to enzymatically regulated uptake. The introduction of a phosphatase into coacervates preloaded with the phosphorylated 14-3-3-GFP-c-Raf complex results in a significant cargo efflux mediated by dephosphorylation. Finally, the general applicability of this platform to study proteinā€“protein interactions is demonstrated by the phosphorylation-dependent and 14-3-3-mediated active reconstitution of a split-luciferase inside artificial cells. This work presents an approach to study dynamically regulated protein recruitment in condensates, using native interaction domains.</p

    Glucocorticoid receptor Thr524 phosphorylation by MINK1 induces interactions with 14-3-3 protein regulators

    Get PDF
    The glucocorticoid receptor (GR) is a ligand-dependent transcription factor that plays a central role in inflammation. The GR activity is also modulated via protein-protein interactions, including binding of 14-3-3 proteins induced by GR phosphorylation. However, the specific phosphorylation sites on the GR that trigger these interactions and their functional consequences are less clear. Hence, we sought to examine this system in more detail. We used phosphorylated GR peptides, biophysical studies, and X-ray crystallography to identify key residues within the ligand-binding domain of the GR, T524 and S617, whose phosphorylation results in binding of the representative 14-3-3 protein 14-3-3Ī¶. A kinase screen identified misshapen-like kinase 1 (MINK1) as responsible for phosphorylating T524 and Rho-associated protein kinase 1 for phosphorylating S617; cell-based approaches confirmed the importance of both GR phosphosites and MINK1 but not Rhoassociated protein kinase 1 alone in inducing GR-14-3-3 binding. Together our results provide molecular-level insight into 14-3-3-mediated regulation of the GR and highlight both MINK1 and the GR-14-3-3 axis as potential targets for future therapeutic intervention

    Structure-Based Optimization of Covalent, Small-Molecule Stabilizers of the 14-3-3Ļƒ/ERĪ± Protein-Protein Interaction from Nonselective Fragments

    Get PDF
    The stabilization of protein-protein interactions (PPIs) has emerged as a promising strategy in chemical biology and drug discovery. The identification of suitable starting points for stabilizing native PPIs and their subsequent elaboration into selective and potent molecular glues lacks structure-guided optimization strategies. We have previously identified a disulfide fragment that stabilized the hub protein 14-3-3Ļƒ bound to several of its clients, including ERĪ± and C-RAF. Here, we show the structure-based optimization of the nonselective fragment toward selective and highly potent small-molecule stabilizers of the 14-3-3Ļƒ/ERĪ± complex. The more elaborated molecular glues, for example, show no stabilization of 14-3-3Ļƒ/C-RAF up to 150 Ī¼M compound. Orthogonal biophysical assays, including mass spectrometry and fluorescence anisotropy, were used to establish structure-activity relationships. The binding modes of 37 compounds were elucidated with X-ray crystallography, which further assisted the concomitant structure-guided optimization. By targeting specific amino acids in the 14-3-3Ļƒ/ERĪ± interface and locking the conformation with a spirocycle, the optimized covalent stabilizer 181 achieved potency, cooperativity, and selectivity similar to the natural product Fusicoccin-A. This case study showcases the value of addressing the structure, kinetics, and cooperativity for molecular glue development. </p

    From Tethered to Freestanding Stabilizers of 14-3-3 Protein-Protein Interactions through Fragment Linking

    Get PDF
    Small-molecule stabilization of protein-protein interactions (PPIs) is a promising strategy in chemical biology and drug discovery. However, the systematic discovery of PPI stabilizers remains a largely unmet challenge. Herein we report a fragment-linking approach targeting the interface of 14-3-3 and a peptide derived from the estrogen receptor alpha (ERĪ±) protein. Two classes of fragmentsā€”a covalent and a noncovalent fragmentā€”were co-crystallized and subsequently linked, resulting in a noncovalent hybrid molecule in which the original fragment interactions were largely conserved. Supported by 20 crystal structures, this initial hybrid molecule was further optimized, resulting in selective, 25-fold stabilization of the 14-3-3/ERĪ± interaction. The high-resolution structures of both the single fragments, their co-crystal structures and those of the linked fragments document a feasible strategy to develop orthosteric PPI stabilizers by linking to an initial tethered fragment.</p
    • ā€¦
    corecore