Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 964
Filter
1.
Org Biomol Chem ; 22(23): 4625-4636, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38804977

ABSTRACT

Both natural and unnatural amino acids, peptides, and proteins are widely recognized as green and sustainable organic chemicals, not only in the field of biological sciences but also in materials science. It has been discovered that artificially designed unnatural peptides and proteins exhibit advanced properties in medical and materials science. In this context, the development of precise chemical modification methods for amino acids and peptides is acknowledged as an important research project in the field of organic synthesis. While a wide variety of modification methods for amino acid residues have been developed to artificially modify peptides and proteins, the representative methods for modifying amino acid residues have traditionally relied on the nucleophilic properties of the functionalities on the residues. In this context, the development of different modification methods using an umpolung-like approach by utilizing the electrophilic nature of amino acid derivatives appears to be very attractive. One of the promising electrophilic amino acid compounds for realizing important modification methods of amino acid derivatives is α,ß-dehydroamino acids, which possess an α,ß-unsaturated carbonyl structure. This review article summarizes methods for the preparation of α,ß-dehydroamino acids derived from natural and unnatural amino acid derivatives. The utilities of α,ß-dehydroamino acid derivatives, including peptides and proteins containing dehydroalanine units, in bioconjugations are also discussed.


Subject(s)
Amino Acids , Amino Acids/chemistry , Amino Acids/chemical synthesis , Proteins/chemistry , Proteins/chemical synthesis , Materials Science , Peptides/chemistry , Peptides/chemical synthesis , Green Chemistry Technology , Chemistry Techniques, Synthetic/methods , Alanine/chemistry , Alanine/analogs & derivatives , Alanine/chemical synthesis
2.
ChemMedChem ; 19(13): e202300692, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38572578

ABSTRACT

Glycosylation is one of the most ubiquitous post-translational modifications. It affects the structure and function of peptides/proteins and consequently has a significant impact on various biological events. However, the structural complexity and heterogeneity of glycopeptides/proteins caused by the diversity of glycan structures and glycosylation sites complicates the detailed elucidation of glycan function and hampers their clinical applications. To address these challenges, chemical and/or enzyme-assisted synthesis methods have been developed to realize glycopeptides/proteins with well-defined glycan morphologies. In particular, N-glycans are expected to be useful for improving the solubility, in vivo half-life and aggregation of bioactive peptides/proteins that have had limited clinical applications so far due to their short duration of action in the blood and unsuitable physicochemical properties. Chemical glycosylation performed in a post-synthetic procedure can be used to facilitate the development of glycopeptide/protein analogues or mimetics that are superior to the original molecules in terms of physicochemical and pharmacokinetic properties. N-glycans are used to modify targets because they are highly biodegradable and biocompatible and have structures that already exist in the human body. On the practical side, from a quality control perspective, close attention should be paid to their structural homogeneity when they are to be applied to pharmaceuticals.


Subject(s)
Polysaccharides , Polysaccharides/chemistry , Polysaccharides/chemical synthesis , Humans , Glycosylation , Peptides/chemistry , Peptides/chemical synthesis , Proteins/chemistry , Proteins/chemical synthesis , Proteins/metabolism , Glycopeptides/chemical synthesis , Glycopeptides/chemistry
3.
Angew Chem Int Ed Engl ; 63(19): e202403396, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38490953

ABSTRACT

Although solid-phase peptide synthesis combining with chemical ligation provides a way to build up customized polypeptides in general, many targets are still presenting challenges for the conventional synthetic process, such as hydrophobic proteins. New methods and strategies are still required to overcome these obstacles. In this study, kinetic studies of Cys/Pen ligation and its acidolysis were performed, from which the fast acidolysis of substituted N,S-benzylidene thioacetals (NBTs) was discovered. The study demonstrates the potential of NBTs as a promising Cys switchable protection, facilitating the chemical synthesis of peptides and proteins by efficiently disrupting peptide aggregation. The compatibility of NBTs with other commonly adopted Cys protecting groups and their applications in sequential disulfide bond formation were also investigated. The first chemical synthesis of the native human programmed death ligand 1 immunoglobulin V-like (PD-L1 IgV) domain was achieved using the NBT strategy, showcasing its potential in difficult protein synthesis.


Subject(s)
Cysteine , Peptides , Cysteine/chemistry , Peptides/chemistry , Peptides/chemical synthesis , Humans , Acetals/chemistry , Benzylidene Compounds/chemistry , Benzylidene Compounds/chemical synthesis , Proteins/chemistry , Proteins/chemical synthesis
4.
Nature ; 626(8001): 1019-1024, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38418914

ABSTRACT

The single chirality of biological molecules is a signature of life. Yet, rationalizing how single chirality emerged remains a challenging goal1. Research has commonly focused on initial symmetry breaking and subsequent enantioenrichment of monomer building blocks-sugars and amino acids-that compose the genetic polymers RNA and DNA as well as peptides. If these building blocks are only partially enantioenriched, however, stalling of chain growth may occur, whimsically termed in the case of nucleic acids as "the problem of original syn"2. Here, in studying a new prebiotically plausible route to proteinogenic peptides3-5, we discovered that the reaction favours heterochiral ligation (that is, the ligation of L monomers with D monomers). Although this finding seems problematic for the prebiotic emergence of homochiral L-peptides, we demonstrate, paradoxically, that this heterochiral preference provides a mechanism for enantioenrichment in homochiral chains. Symmetry breaking, chiral amplification and chirality transfer processes occur for all reactants and products in multicomponent competitive reactions even when only one of the molecules in the complex mixture exhibits an imbalance in enantiomer concentrations (non-racemic). Solubility considerations rationalize further chemical purification and enhanced chiral amplification. Experimental data and kinetic modelling support this prebiotically plausible mechanism for the emergence of homochiral biological polymers.


Subject(s)
Biopolymers , Evolution, Chemical , Peptides , Proteins , Stereoisomerism , Biopolymers/chemistry , Nucleic Acids/chemical synthesis , Nucleic Acids/chemistry , Origin of Life , Peptides/chemistry , Proteins/chemical synthesis , Proteins/chemistry , Solubility
5.
Int J Mol Sci ; 23(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35008947

ABSTRACT

The prevailing current view of protein folding is the thermodynamic hypothesis, under which the native folded conformation of a protein corresponds to the global minimum of Gibbs free energy G. We question this concept and show that the empirical evidence behind the thermodynamic hypothesis of folding is far from strong. Furthermore, physical theory-based approaches to the prediction of protein folds and their folding pathways so far have invariably failed except for some very small proteins, despite decades of intensive theory development and the enormous increase of computer power. The recent spectacular successes in protein structure prediction owe to evolutionary modeling of amino acid sequence substitutions enhanced by deep learning methods, but even these breakthroughs provide no information on the protein folding mechanisms and pathways. We discuss an alternative view of protein folding, under which the native state of most proteins does not occupy the global free energy minimum, but rather, a local minimum on a fluctuating free energy landscape. We further argue that ΔG of folding is likely to be positive for the majority of proteins, which therefore fold into their native conformations only through interactions with the energy-dependent molecular machinery of living cells, in particular, the translation system and chaperones. Accordingly, protein folding should be modeled as it occurs in vivo, that is, as a non-equilibrium, active, energy-dependent process.


Subject(s)
Protein Conformation , Protein Folding , Proteins/chemistry , Thermodynamics , Algorithms , Kinetics , Models, Molecular , Models, Theoretical , Protein Refolding , Protein Stability , Proteins/chemical synthesis , Proteome , Proteomics/methods , Recombinant Proteins/chemistry , Solubility , Species Specificity
6.
Int J Mol Sci ; 22(23)2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34884714

ABSTRACT

As an important component that constitutes all the cells and tissues of the human body, protein is involved in most of the biological processes. Inspired by natural protein systems, considerable efforts covering many discipline fields were made to design artificial protein assemblies and put them into application in recent decades. The rapid development of structural DNA nanotechnology offers significant means for protein assemblies and promotes their application. Owing to the programmability, addressability and accurate recognition ability of DNA, many protein assemblies with unprecedented structures and improved functions have been successfully fabricated, consequently creating many brand-new researching fields. In this review, we briefly introduced the DNA-based protein assemblies, and highlighted the limitations in application process and corresponding strategies in four aspects, including biological catalysis, protein detection, biomedicine treatment and other applications.


Subject(s)
DNA/chemistry , Nanotechnology , Proteins/chemical synthesis , Catalysis , Drug Delivery Systems , Proteins/analysis
7.
J Chem Phys ; 154(19): 194101, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34240891

ABSTRACT

Protein assembly is often studied in a three-dimensional solution, but a significant fraction of binding events involve proteins that can reversibly bind and diffuse along a two-dimensional surface. In a recent study, we quantified how proteins can exploit the reduced dimensionality of the membrane to trigger complex formation. Here, we derive a single expression for the characteristic timescale of this multi-step assembly process, where the change in dimensionality renders rates and concentrations effectively time-dependent. We find that proteins can accelerate dimer formation due to an increase in relative concentration, driving more frequent collisions, which often win out over slow-downs due to diffusion. Our model contains two protein populations that dimerize with one another and use a distinct site to bind membrane lipids, creating a complex reaction network. However, by identifying two major rate-limiting pathways to reach an equilibrium steady-state, we derive an excellent approximation for the mean first passage time when lipids are in abundant supply. Our theory highlights how the "sticking rate" or effective adsorption coefficient of the membrane is central in controlling timescales. We also derive a corrected localization rate to quantify how the geometry of the system and diffusion can reduce rates of membrane localization. We validate and test our results using kinetic and particle-based reaction-diffusion simulations. Our results establish how the speed of key assembly steps can shift by orders-of-magnitude when membrane localization is possible, which is critical to understanding mechanisms used in cells.


Subject(s)
Cell Membrane/chemistry , Proteins/chemical synthesis , Proteins/chemistry
8.
Chembiochem ; 22(20): 2966-2972, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34265138

ABSTRACT

The reversible nature of disulfide functionality has been exploited to design intelligent materials such as nanocapsules, micelles, vesicles, inorganic nanoparticles, peptide and nucleic acid nanodevices. Herein, we report a new chemical methodology for the construction redox-sensitive protein assemblies using monodisperse facially amphiphilic protein-dendron bioconjugates. The disulfide functionality is strategically placed between the dendron and protein domains. The custom designed bioconjugates self-assembled into nanoscopic objects of a defined size dictated by the nature of dendron domain. The stimuli-responsive behavior of the protein assemblies is demonstrated using a suitable redox trigger.


Subject(s)
Disulfides/chemistry , Nanostructures/chemistry , Protein Engineering , Proteins/chemical synthesis , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Structure , Oxidation-Reduction , Proteins/chemistry
9.
Bioconjug Chem ; 32(8): 1570-1575, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34232618

ABSTRACT

5-(Alkynyl)dibenzothiophenium triflates are introduced as new reagents to prepare different protein conjugates through site-selective cysteine alkynylation. The protocol developed allows a highly efficient label of free cysteine-containing proteins with relevant biological roles, such as ubiquitin, the C2A domain of Synaptotagmin-I, or HER2 targeting nanobodies. An electrophilic bis-alkynylating reagent was also designed. The second alkynylating handle thus introduced in the desired protein enables access to protein-thiol, protein-peptide, and protein-protein conjugates, and even diubiquitin dimers can be prepared through this approach. The low excess of reagent needed, mild reaction conditions used, short reaction times, and stability of the S-C(alkyne) bonds at physiological conditions make this approach an interesting addition to the toolbox of classical, site-selective cysteine-conjugation methods.


Subject(s)
Alkynes/chemistry , Proteins/chemistry , Thiophenes/chemistry , Alkynes/chemical synthesis , Animals , Chemistry Techniques, Synthetic , Cysteine/chemical synthesis , Cysteine/chemistry , Humans , Indicators and Reagents , Mesylates/chemical synthesis , Mesylates/chemistry , Models, Molecular , Proteins/chemical synthesis , Sulfhydryl Compounds/chemistry , Thiophenes/chemical synthesis
10.
J Am Chem Soc ; 143(31): 11919-11926, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34323481

ABSTRACT

Here we report a nonenzymatic glycosylation reaction that builds axial S-glycosidic bonds under biorelevant conditions. This strategy is enabled by the design and use of allyl glycosyl sulfones as precursors to glycosyl radicals and exploits the exceptional functional group tolerance of radical processes. Our method introduces a variety of unprotected glycosyl units to the cysteine residues of peptides in a highly selective fashion. Through developing the second-generation protocol, we applied our method in the direct glycosylation of complex polypeptides and proteins. Computational studies were performed to elucidate the reaction mechanism.


Subject(s)
Peptides/chemical synthesis , Proteins/chemical synthesis , Glycosylation , Molecular Structure , Peptides/chemistry , Proteins/chemistry , Stereoisomerism
11.
Chem Commun (Camb) ; 57(58): 7083-7095, 2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34180471

ABSTRACT

Proteins provide an excellent means to monitor and regulate biological processes. Hence, a precise chemical toolbox for their modification becomes indispensable. In this perspective, this feature article outlines our efforts to establish the core principles of chemoselectivity, site-selectivity, site-specificity, site-modularity, residue-modularity, and protein-specificity. With the knowledge to systematically regulate these parameters, the field has access to technological platforms that can address multiple challenges at the interface of chemistry, biology, and medicine.


Subject(s)
Proteins/chemistry , Animals , Chemistry Techniques, Synthetic , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Immunoconjugates/chemistry , Models, Molecular , Proteins/chemical synthesis
12.
Chem Rec ; 21(8): 1941-1956, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34184826

ABSTRACT

Are chemical methods capable of precisely engineering the native proteins? Is it possible to develop platforms that can empower the regulation of chemoselectivity, site-selectivity, modularity, protein-specificity, and site-specificity? This account delineates our research journey in the last ten years on the developments revolving around these questions. It will range from the realization of chemoselective and site-selective labeling of reactivity hotspots to modular linchpin directed modification (LDM®) platform and site-specific Gly-tag® technology. Also, we outline a few biotechnology tools, including Maspecter®, that accelerated the detailed analysis of the bioconjugates and rendered a powerful toolbox for homogeneous antibody-drug conjugates (ADCs).


Subject(s)
Proteins/chemistry , Staining and Labeling/methods , Copper/chemistry , Glycine/chemistry , Histidine/chemistry , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Ligands , Lysine/chemistry , Proteins/chemical synthesis , Proteins/metabolism
13.
Acc Chem Res ; 54(11): 2670-2682, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34014638

ABSTRACT

Bioinspired self-assembly has been explored with diverse synthetic scaffolds, among which amphiphiles are perhaps the most extensively studied systems. Classical surfactants or amphiphilic block copolymers, depending on the hydrophobic-hydrophilic balance, produce distinct nanostructures, which hold promise for applications ranging from biology to materials sciences. Nevertheless, their immiscibility-driven aggregation does not provide the opportunity to precisely regulate the internal order, morphology, or functional group display, which is highly desirable, especially in the context of biological applications.A new class of amphiphiles have emerged in the recent past in which the hydrophilic segment(s) is appended with a hydrophobic supramolecular-structure-directing-unit (SSDU), consisting of a π-conjugated chromophore and a H-bonding group. Self-recognition of the SSDU by attractive directional interactions governs the supramolecular assembly, which is fundamentally different than the repulsive solvent-immiscibility driven aggregation of traditional amphiphiles. Such SSDU-appended hydrophilic polymers exhibit entropy-driven highly stable self-assembly producing distinct nanostructures depending on the H-bonding functional group. For example, polymers with the hydrazide-functionalized SSDU attached form a polymersome, while in a sharp contrast, the same polymers when connected to an amide containing SSDU produce a cylindrical micelle via a spherical-micelle intermediate. This relationship holds true for a series of SSDU-attached hydrophilic polymers irrespective of the hydrophobic/hydrophilic balance or chemical structure, indicating that the supramolecular-assembly is primarily controlled by the specific molecular-recognition motif of the SSDU, instead of the packing parameter-based norms. Beyond synthetic polymers, SSDU-attached proteins also exhibit similar molecular-recognition driven self-assembly as well as coassembly with SSDU-attached polymers or hydrophilic wedges, producing multi-stimuli-responsive nanostructures in which the protein gains remarkable protection from thermal denaturation or enzymatic hydrolysis and exhibits redox-responsive enzymatic activity.Furthermore, SSDU-derived bola-shape π-amphiphiles have been recognized as a useful scaffold for the synthesis of unsymmetric polymersomes, rarely reported in the literature. The building block consists of a hydrophobic naphthalene-diimide (NDI) π-system attached to a hydrophilic functional group (ionic or nonionic) and a nonionic wedge on its two opposite arms. Extended H-bonding among the hydrazide groups, placed only on one side of the central chromophore by design, ensures stacking of the NDIs with parallel orientation and induces a preferred direction of curvature so that the H-bonded chain and consequently the functional groups attached to the same side remain at the inner-wall of the supramolecular polymersome. Automatically, the functional groups, located on the other side, are displayed at the outer surface. This design works for different amphiphiles, which by virtue of efficient and predictable functional group display, strongly influences the multivalent binding with different biological targets resulting in efficient enzyme inhibition, glycocluster effect, or antibacterial activity, depending on the nature of the functional group. By taking advantage of the electron accepting nature of the NDI, electron rich pyrene-containing amphiphiles can be costacked in alternating sequence, producing temperature and redox-responsive supramolecular polymers with NDI/pyrene stoichiometry-dependent morphology, lower critical solution temperature (LCST), functional group display, and antibacterial activity.


Subject(s)
Proteins/chemical synthesis , Surface-Active Agents/chemical synthesis , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Molecular Structure , Proteins/chemistry , Surface-Active Agents/chemistry , Temperature
14.
Chem Commun (Camb) ; 57(49): 6015-6018, 2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34032224

ABSTRACT

A new miniprotein built from three helices, including one structure based on the ααßαααß sequence pattern was developed. Its crystal structure revealed a compact conformation with a well-packed hydrophobic core of unprecedented structure. The miniprotein formed dimers that were stabilized by the interaction of their hydrophobic surfaces.


Subject(s)
Amino Acids/chemistry , Proteins/chemical synthesis , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Proteins/chemistry
15.
Bioorg Med Chem ; 41: 116210, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34022527

ABSTRACT

The elaboration of peptides and proteins containing non-proteinogenic amino acids has been realized using several complementary strategies, including chemical synthesis, ribosome- or non-ribosome-mediated elaboration, intein-mediated polypeptide rearrangements, or some combination of these strategies. All of these have strengths and limitations, and significant efforts have been focused on minimizing the effects of limitations, to improve the overall utility of individual strategies. Our laboratory has studied ribosomally mediated peptide and protein synthesis involving a wide variety of non-proteinogenic amino acids, and in recent years we have described a novel strategy for the selection of modified bacterial ribosomes. These modified ribosomes have enabled the incorporation into peptides and proteins of numerous modified amino acids not accessible using wild-type ribosomes. This has included d-amino acids, ß-amino acids, dipeptides and dipeptidomimetic species, as well as phosphorylated amino acids. Presently, we have considered novel strategies for incorporating non-proteinogenic amino acids in improved yields. This has included the incorporation of non-proteinogenic amino acids into contiguous positions, a transformation known to be challenging. We demonstrate the preparation of this type of protein modification by utilizing a suppressor tRNACUA activated with a dipeptide consisting of two identical non-proteinogenic amino acids, in the presence of modified ribosomes selected to recognize such dipeptides. Also, we demonstrate that the use of bis-aminoacylated suppressor tRNAs, shown previously to increase protein yields significantly in vitro, can be extended to the use of non-proteinogenic amino acids.


Subject(s)
Dipeptides/chemistry , Proteins/chemical synthesis , Amino Acids/chemistry , Escherichia coli , Protein Conformation , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/chemistry , Ribosomes
16.
Chempluschem ; 86(2): 284-290, 2021 02.
Article in English | MEDLINE | ID: mdl-33605561

ABSTRACT

While the protein assemblies have been found widely existing and playing significant roles in biological systems, their imitation and re-construction is further boosting more applications in biomedical research, such as enzymatic reaction regulation, sensing, and biomedicine. DNA nanotechnology provides a programmable strategy for the fabrication of nanostructures with unprecedented accuracy on the nanoscale. By linking the DNA nanotechnology with proteins of different functions, the precise construction of DNA-guided protein assemblies can be achieved for various biomedical applications. This minireview summarizes the recent advances in the programmable protein assemblies on DNA nanoplatforms and discusses the outlook of DNA-guided protein assemblies in the biomedical research.


Subject(s)
Biomedical Research , DNA/chemistry , Proteins/chemical synthesis , Animals , Humans , Nanostructures , Proteins/chemistry
17.
Nat Commun ; 12(1): 870, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558523

ABSTRACT

Despite six decades of efforts to synthesize peptides and proteins bearing multiple disulfide bonds, this synthetic challenge remains an unsolved problem in most targets (e.g., knotted mini proteins). Here we show a de novo general synthetic strategy for the ultrafast, high-yielding formation of two and three disulfide bonds in peptides and proteins. We develop an approach based on the combination of a small molecule, ultraviolet-light, and palladium for chemo- and regio-selective activation of cysteine, which enables the one-pot formation of multiple disulfide bonds in various peptides and proteins. We prepare bioactive targets of high therapeutic potential, including conotoxin, RANTES, EETI-II, and plectasin peptides and the linaclotide drug. We anticipate that this strategy will be a game-changer in preparing millions of inaccessible targets for drug discovery.


Subject(s)
Disulfides/chemistry , Disulfides/metabolism , Peptides/metabolism , Proteins/metabolism , Chromatography, High Pressure Liquid , Peptides/chemical synthesis , Peptides/chemistry , Proteins/chemical synthesis , Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism
18.
Biosci Biotechnol Biochem ; 85(1): 53-60, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33577657

ABSTRACT

N-Acyl imidazoles are unique electrophiles that exhibit moderate reactivity, relatively long-half life, and high solubility in water. Thanks to their tunable reactivity and chemical selectivity, the application of N-acyl imidazole derivatives has launched to a number of chemical biology researches, which include chemical synthesis of peptide/protein, chemical labeling of native proteins of interest (POIs), and structural analysis and functional manipulation of RNAs. Since proteins and RNAs play pivotal roles in numerous biological events in all living organisms, the methods that enable the chemical modification of endogenously existing POIs and RNAs in live cells may offer a variety of opportunities not only for fundamental scientific study but also for biotechnology and drug development. In this review, we discuss the recent progress of N-acyl imidazole chemistry that contributes to the chemical labeling and functional control of endogenous proteins and RNAs under multimolecularly crowded biological conditions of live cells.


Subject(s)
Biology/methods , Imidazoles/chemistry , Acylation , Animals , Humans , Peptides/chemical synthesis , Proteins/chemical synthesis
19.
mSphere ; 6(1)2021 01 06.
Article in English | MEDLINE | ID: mdl-33408232

ABSTRACT

Structure-guided vaccine design provides a route to elicit a focused immune response against the most functionally important regions of a pathogen surface. This can be achieved by identifying epitopes for neutralizing antibodies through structural methods and recapitulating these epitopes by grafting their core structural features onto smaller scaffolds. In this study, we conducted a modified version of this protocol. We focused on the PfEMP1 protein family found on the surfaces of erythrocytes infected with Plasmodium falciparum A subset of PfEMP1 proteins bind to endothelial protein C receptor (EPCR), and their expression correlates with development of the symptoms of severe malaria. Structural studies revealed that PfEMP1 molecules present a helix-kinked-helix motif that forms the core of the EPCR-binding site. Using Rosetta-based design, we successfully grafted this motif onto a three-helical bundle scaffold. We show that this synthetic binder interacts with EPCR with nanomolar affinity and adopts the expected structure. We also assessed its ability to bind to antibodies found in immunized animals and in humans from malaria-endemic regions. Finally, we tested the capacity of the synthetic binder to effectively elicit antibodies that prevent EPCR binding and analyzed the degree of cross-reactivity of these antibodies across a diverse repertoire of EPCR-binding PfEMP1 proteins. Despite our synthetic binder adopting the correct structure, we find that it is not as effective as the CIDRα domain on which it is based for inducing adhesion-inhibitory antibodies. This cautions against the rational design of focused immunogens that contain the core features of a ligand-binding site of a protein family, rather than those of a neutralizing antibody epitope.IMPORTANCE Vaccines train our immune systems to generate antibodies which recognize pathogens. Some of these antibodies are highly protective, preventing infection, while others are ineffective. Structure-guided rational approaches allow design of synthetic molecules which contain only the regions of a pathogen required to induce production of protective antibodies. On the surfaces of red blood cells infected by the malaria parasite Plasmodium falciparum are parasite molecules called PfEMP1 proteins. PfEMP1 proteins, which bind to human receptor EPCR, are linked to development of severe malaria. We have designed a synthetic protein on which we grafted the EPCR-binding surface of a PfEMP1 protein. We use this molecule to show which fraction of protective antibodies recognize the EPCR-binding surface and test its effectiveness as a vaccine immunogen.


Subject(s)
Antibodies, Protozoan/metabolism , Endothelial Protein C Receptor/metabolism , Proteins/chemical synthesis , Proteins/metabolism , Protozoan Proteins/agonists , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Motifs , Animals , Antibodies, Protozoan/immunology , Binding Sites , Cell Adhesion , Endothelial Protein C Receptor/immunology , Erythrocytes/parasitology , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Protein Binding , Proteins/chemistry , Proteins/immunology , Rats
20.
Nature ; 589(7842): 468-473, 2021 01.
Article in English | MEDLINE | ID: mdl-33408408

ABSTRACT

Ordered two-dimensional arrays such as S-layers1,2 and designed analogues3-5 have intrigued bioengineers6,7, but with the exception of a single lattice formed with flexible linkers8, they are constituted from just one protein component. Materials composed of two components have considerable potential advantages for modulating assembly dynamics and incorporating more complex functionality9-12. Here we describe a computational method to generate co-assembling binary layers by designing rigid interfaces between pairs of dihedral protein building blocks, and use it to design a p6m lattice. The designed array components are soluble at millimolar concentrations, but when combined at nanomolar concentrations, they rapidly assemble into nearly crystalline micrometre-scale arrays nearly identical to the computational design model in vitro and in cells without the need for a two-dimensional support. Because the material is designed from the ground up, the components can be readily functionalized and their symmetry reconfigured, enabling formation of ligand arrays with distinguishable surfaces, which we demonstrate can drive extensive receptor clustering, downstream protein recruitment and signalling. Using atomic force microscopy on supported bilayers and quantitative microscopy on living cells, we show that arrays assembled on membranes have component stoichiometry and structure similar to arrays formed in vitro, and that our material can therefore impose order onto fundamentally disordered substrates such as cell membranes. In contrast to previously characterized cell surface receptor binding assemblies such as antibodies and nanocages, which are rapidly endocytosed, we find that large arrays assembled at the cell surface suppress endocytosis in a tunable manner, with potential therapeutic relevance for extending receptor engagement and immune evasion. Our work provides a foundation for a synthetic cell biology in which multi-protein macroscale materials are designed to modulate cell responses and reshape synthetic and living systems.


Subject(s)
Drug Design , Protein Engineering , Proteins/chemical synthesis , Proteins/metabolism , 3T3 Cells , Animals , Cell Biology , Cell Survival , Computational Biology , Endocytosis , Escherichia coli/genetics , Escherichia coli/metabolism , In Vitro Techniques , Kinetics , Ligands , Mice , Microscopy, Atomic Force , Models, Molecular , Synthetic Biology
SELECTION OF CITATIONS
SEARCH DETAIL
...