Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.760
Filter
Add more filters

Publication year range
1.
Cell ; 187(16): 4305-4317.e18, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38936360

ABSTRACT

Interleukin (IL)-23 and IL-17 are well-validated therapeutic targets in autoinflammatory diseases. Antibodies targeting IL-23 and IL-17 have shown clinical efficacy but are limited by high costs, safety risks, lack of sustained efficacy, and poor patient convenience as they require parenteral administration. Here, we present designed miniproteins inhibiting IL-23R and IL-17 with antibody-like, low picomolar affinities at a fraction of the molecular size. The minibinders potently block cell signaling in vitro and are extremely stable, enabling oral administration and low-cost manufacturing. The orally administered IL-23R minibinder shows efficacy better than a clinical anti-IL-23 antibody in mouse colitis and has a favorable pharmacokinetics (PK) and biodistribution profile in rats. This work demonstrates that orally administered de novo-designed minibinders can reach a therapeutic target past the gut epithelial barrier. With high potency, gut stability, and straightforward manufacturability, de novo-designed minibinders are a promising modality for oral biologics.


Subject(s)
Colitis , Interleukin-17 , Th17 Cells , Animals , Administration, Oral , Mice , Humans , Rats , Colitis/drug therapy , Interleukin-17/metabolism , Interleukin-17/antagonists & inhibitors , Th17 Cells/immunology , Receptors, Interleukin/metabolism , Receptors, Interleukin/antagonists & inhibitors , Mice, Inbred C57BL , Male , Interleukin-23/metabolism , Interleukin-23/antagonists & inhibitors , Tissue Distribution , Female , Rats, Sprague-Dawley
2.
Cell ; 187(14): 3726-3740.e43, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38861993

ABSTRACT

Many growth factors and cytokines signal by binding to the extracellular domains of their receptors and driving association and transphosphorylation of the receptor intracellular tyrosine kinase domains, initiating downstream signaling cascades. To enable systematic exploration of how receptor valency and geometry affect signaling outcomes, we designed cyclic homo-oligomers with up to 8 subunits using repeat protein building blocks that can be modularly extended. By incorporating a de novo-designed fibroblast growth factor receptor (FGFR)-binding module into these scaffolds, we generated a series of synthetic signaling ligands that exhibit potent valency- and geometry-dependent Ca2+ release and mitogen-activated protein kinase (MAPK) pathway activation. The high specificity of the designed agonists reveals distinct roles for two FGFR splice variants in driving arterial endothelium and perivascular cell fates during early vascular development. Our designed modular assemblies should be broadly useful for unraveling the complexities of signaling in key developmental transitions and for developing future therapeutic applications.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors , Receptors, Fibroblast Growth Factor , Signal Transduction , Animals , Humans , Receptors, Fibroblast Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Mice , Ligands , Calcium/metabolism , MAP Kinase Signaling System
3.
Cell ; 186(21): 4710-4727.e35, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37774705

ABSTRACT

Polarized cells rely on a polarized cytoskeleton to function. Yet, how cortical polarity cues induce cytoskeleton polarization remains elusive. Here, we capitalized on recently established designed 2D protein arrays to ectopically engineer cortical polarity of virtually any protein of interest during mitosis in various cell types. This enables direct manipulation of polarity signaling and the identification of the cortical cues sufficient for cytoskeleton polarization. Using this assay, we dissected the logic of the Par complex pathway, a key regulator of cytoskeleton polarity during asymmetric cell division. We show that cortical clustering of any Par complex subunit is sufficient to trigger complex assembly and that the primary kinetic barrier to complex assembly is the relief of Par6 autoinhibition. Further, we found that inducing cortical Par complex polarity induces two hallmarks of asymmetric cell division in unpolarized mammalian cells: spindle orientation, occurring via Par3, and central spindle asymmetry, depending on aPKC activity.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Polarity , Cytological Techniques , Mitosis , Animals , Cytoskeleton/metabolism , Mammals/metabolism , Microtubules/metabolism , Protein Kinase C/metabolism , Adaptor Proteins, Signal Transducing/metabolism
4.
Cell ; 186(22): 4803-4817.e13, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37683634

ABSTRACT

Patescibacteria, also known as the candidate phyla radiation (CPR), are a diverse group of bacteria that constitute a disproportionately large fraction of microbial dark matter. Its few cultivated members, belonging mostly to Saccharibacteria, grow as epibionts on host Actinobacteria. Due to a lack of suitable tools, the genetic basis of this lifestyle and other unique features of Patescibacteira remain unexplored. Here, we show that Saccharibacteria exhibit natural competence, and we exploit this property for their genetic manipulation. Imaging of fluorescent protein-labeled Saccharibacteria provides high spatiotemporal resolution of phenomena accompanying epibiotic growth, and a transposon-insertion sequencing (Tn-seq) genome-wide screen reveals the contribution of enigmatic Saccharibacterial genes to growth on their hosts. Finally, we leverage metagenomic data to provide cutting-edge protein structure-based bioinformatic resources that support the strain Southlakia epibionticum and its corresponding host, Actinomyces israelii, as a model system for unlocking the molecular underpinnings of the epibiotic lifestyle.


Subject(s)
Bacteria , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Metagenome , Metagenomics , Phylogeny , Actinobacteria/physiology
5.
Cell ; 185(19): 3520-3532.e26, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36041435

ABSTRACT

We use computational design coupled with experimental characterization to systematically investigate the design principles for macrocycle membrane permeability and oral bioavailability. We designed 184 6-12 residue macrocycles with a wide range of predicted structures containing noncanonical backbone modifications and experimentally determined structures of 35; 29 are very close to the computational models. With such control, we show that membrane permeability can be systematically achieved by ensuring all amide (NH) groups are engaged in internal hydrogen bonding interactions. 84 designs over the 6-12 residue size range cross membranes with an apparent permeability greater than 1 × 10-6 cm/s. Designs with exposed NH groups can be made membrane permeable through the design of an alternative isoenergetic fully hydrogen-bonded state favored in the lipid membrane. The ability to robustly design membrane-permeable and orally bioavailable peptides with high structural accuracy should contribute to the next generation of designed macrocycle therapeutics.


Subject(s)
Amides , Peptides , Amides/chemistry , Hydrogen , Hydrogen Bonding , Lipids , Peptides/chemistry
6.
Cell ; 176(6): 1420-1431.e17, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30849373

ABSTRACT

Respiratory syncytial virus (RSV) is a worldwide public health concern for which no vaccine is available. Elucidation of the prefusion structure of the RSV F glycoprotein and its identification as the main target of neutralizing antibodies have provided new opportunities for development of an effective vaccine. Here, we describe the structure-based design of a self-assembling protein nanoparticle presenting a prefusion-stabilized variant of the F glycoprotein trimer (DS-Cav1) in a repetitive array on the nanoparticle exterior. The two-component nature of the nanoparticle scaffold enabled the production of highly ordered, monodisperse immunogens that display DS-Cav1 at controllable density. In mice and nonhuman primates, the full-valency nanoparticle immunogen displaying 20 DS-Cav1 trimers induced neutralizing antibody responses ∼10-fold higher than trimeric DS-Cav1. These results motivate continued development of this promising nanoparticle RSV vaccine candidate and establish computationally designed two-component nanoparticles as a robust and customizable platform for structure-based vaccine design.


Subject(s)
Antibodies, Neutralizing/immunology , Respiratory Syncytial Viruses/immunology , Vaccination/methods , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/immunology , Caveolin 1 , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Nanoparticles/therapeutic use , Primary Cell Culture , Respiratory Syncytial Viruses/pathogenicity , Vaccines/immunology , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/physiology
7.
Cell ; 160(5): 952-962, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25723169

ABSTRACT

Bacteria use rapid contraction of a long sheath of the type VI secretion system (T6SS) to deliver effectors into a target cell. Here, we present an atomic-resolution structure of a native contracted Vibrio cholerae sheath determined by cryo-electron microscopy. The sheath subunits, composed of tightly interacting proteins VipA and VipB, assemble into a six-start helix. The helix is stabilized by a core domain assembled from four ß strands donated by one VipA and two VipB molecules. The fold of inner and middle layers is conserved between T6SS and phage sheaths. However, the structure of the outer layer is distinct and suggests a mechanism of interaction of the bacterial sheath with an accessory ATPase, ClpV, that facilitates multiple rounds of effector delivery. Our results provide a mechanistic insight into assembly of contractile nanomachines that bacteria and phages use to translocate macromolecules across membranes.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Secretion Systems , Vibrio cholerae/metabolism , Amino Acid Sequence , Cryoelectron Microscopy , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Vibrio cholerae/chemistry , Vibrio cholerae/cytology , Vibrio cholerae/ultrastructure
8.
Nature ; 632(8026): 911-920, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39143214

ABSTRACT

Allosteric modulation of protein function, wherein the binding of an effector to a protein triggers conformational changes at distant functional sites, plays a central part in the control of metabolism and cell signalling1-3. There has been considerable interest in designing allosteric systems, both to gain insight into the mechanisms underlying such 'action at a distance' modulation and to create synthetic proteins whose functions can be regulated by effectors4-7. However, emulating the subtle conformational changes distributed across many residues, characteristic of natural allosteric proteins, is a significant challenge8,9. Here, inspired by the classic Monod-Wyman-Changeux model of cooperativity10, we investigate the de novo design of allostery through rigid-body coupling of peptide-switchable hinge modules11 to protein interfaces12 that direct the formation of alternative oligomeric states. We find that this approach can be used to generate a wide variety of allosterically switchable systems, including cyclic rings that incorporate or eject subunits in response to peptide binding and dihedral cages that undergo effector-induced disassembly. Size-exclusion chromatography, mass photometry13 and electron microscopy reveal that these designed allosteric protein assemblies closely resemble the design models in both the presence and absence of peptide effectors and can have ligand-binding cooperativity comparable to classic natural systems such as haemoglobin14. Our results indicate that allostery can arise from global coupling of the energetics of protein substructures without optimized side-chain-side-chain allosteric communication pathways and provide a roadmap for generating allosterically triggerable delivery systems, protein nanomachines and cellular feedback control circuitry.


Subject(s)
Allosteric Regulation , Peptides , Proteins , Allosteric Regulation/drug effects , Chromatography , Feedback, Physiological , Ligands , Microscopy, Electron , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Protein Multimerization/drug effects , Proteins/chemistry , Proteins/drug effects , Proteins/metabolism , Proteins/ultrastructure
9.
Nature ; 631(8020): 449-458, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38898281

ABSTRACT

De novo design of complex protein folds using solely computational means remains a substantial challenge1. Here we use a robust deep learning pipeline to design complex folds and soluble analogues of integral membrane proteins. Unique membrane topologies, such as those from G-protein-coupled receptors2, are not found in the soluble proteome, and we demonstrate that their structural features can be recapitulated in solution. Biophysical analyses demonstrate the high thermal stability of the designs, and experimental structures show remarkable design accuracy. The soluble analogues were functionalized with native structural motifs, as a proof of concept for bringing membrane protein functions to the soluble proteome, potentially enabling new approaches in drug discovery. In summary, we have designed complex protein topologies and enriched them with functionalities from membrane proteins, with high experimental success rates, leading to a de facto expansion of the functional soluble fold space.


Subject(s)
Computer-Aided Design , Deep Learning , Membrane Proteins , Protein Folding , Solubility , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Protein Stability , Proteome/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Amino Acid Motifs , Proof of Concept Study
10.
Nature ; 627(8005): 898-904, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480887

ABSTRACT

A wooden house frame consists of many different lumber pieces, but because of the regularity of these building blocks, the structure can be designed using straightforward geometrical principles. The design of multicomponent protein assemblies, in comparison, has been much more complex, largely owing to the irregular shapes of protein structures1. Here we describe extendable linear, curved and angled protein building blocks, as well as inter-block interactions, that conform to specified geometric standards; assemblies designed using these blocks inherit their extendability and regular interaction surfaces, enabling them to be expanded or contracted by varying the number of modules, and reinforced with secondary struts. Using X-ray crystallography and electron microscopy, we validate nanomaterial designs ranging from simple polygonal and circular oligomers that can be concentrically nested, up to large polyhedral nanocages and unbounded straight 'train track' assemblies with reconfigurable sizes and geometries that can be readily blueprinted. Because of the complexity of protein structures and sequence-structure relationships, it has not previously been possible to build up large protein assemblies by deliberate placement of protein backbones onto a blank three-dimensional canvas; the simplicity and geometric regularity of our design platform now enables construction of protein nanomaterials according to 'back of an envelope' architectural blueprints.


Subject(s)
Nanostructures , Proteins , Crystallography, X-Ray , Nanostructures/chemistry , Proteins/chemistry , Proteins/metabolism , Microscopy, Electron , Reproducibility of Results
11.
Nature ; 626(7998): 435-442, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38109936

ABSTRACT

Many peptide hormones form an α-helix on binding their receptors1-4, and sensitive methods for their detection could contribute to better clinical management of disease5. De novo protein design can now generate binders with high affinity and specificity to structured proteins6,7. However, the design of interactions between proteins and short peptides with helical propensity is an unmet challenge. Here we describe parametric generation and deep learning-based methods for designing proteins to address this challenge. We show that by extending RFdiffusion8 to enable binder design to flexible targets, and to refining input structure models by successive noising and denoising (partial diffusion), picomolar-affinity binders can be generated to helical peptide targets by either refining designs generated with other methods, or completely de novo starting from random noise distributions without any subsequent experimental optimization. The RFdiffusion designs enable the enrichment and subsequent detection of parathyroid hormone and glucagon by mass spectrometry, and the construction of bioluminescence-based protein biosensors. The ability to design binders to conformationally variable targets, and to optimize by partial diffusion both natural and designed proteins, should be broadly useful.


Subject(s)
Computer-Aided Design , Deep Learning , Peptides , Proteins , Biosensing Techniques , Diffusion , Glucagon/chemistry , Glucagon/metabolism , Luminescent Measurements , Mass Spectrometry , Parathyroid Hormone/chemistry , Parathyroid Hormone/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Structure, Secondary , Proteins/chemistry , Proteins/metabolism , Substrate Specificity , Models, Molecular
12.
Nature ; 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39322662

ABSTRACT

Endocytosis and lysosomal trafficking of cell surface receptors can be triggered by endogenous ligands. Therapeutic approaches such as lysosome-targeting chimaeras1,2 (LYTACs) and cytokine receptor-targeting chimeras3 (KineTACs) have used this to target specific proteins for degradation by fusing modified native ligands to target binding proteins. Although powerful, these approaches can be limited by competition with native ligands and requirements for chemical modification that limit genetic encodability and can complicate manufacturing, and, more generally, there may be no native ligands that stimulate endocytosis through a given receptor. Here we describe computational design approaches for endocytosis-triggering binding proteins (EndoTags) that overcome these challenges. We present EndoTags for insulin-like growth factor 2 receptor (IGF2R) and asialoglycoprotein receptor (ASGPR), sortilin and transferrin receptors, and show that fusing these tags to soluble or transmembrane target protein binders leads to lysosomal trafficking and target degradation. As these receptors have different tissue distributions, the different EndoTags could enable targeting of degradation to different tissues. EndoTag fusion to a PD-L1 antibody considerably increases efficacy in a mouse tumour model compared to antibody alone. The modularity and genetic encodability of EndoTags enables AND gate control for higher-specificity targeted degradation, and the localized secretion of degraders from engineered cells. By promoting endocytosis, EndoTag fusion increases signalling through an engineered ligand-receptor system by nearly 100-fold. EndoTags have considerable therapeutic potential as targeted degradation inducers, signalling activators for endocytosis-dependent pathways, and cellular uptake inducers for targeted antibody-drug and antibody-RNA conjugates.

13.
Immunity ; 53(4): 733-744.e8, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32946741

ABSTRACT

Discovering potent human monoclonal antibodies (mAbs) targeting the Plasmodium falciparum circumsporozoite protein (PfCSP) on sporozoites (SPZ) and elucidating their mechanisms of neutralization will facilitate translation for passive prophylaxis and aid next-generation vaccine development. Here, we isolated a neutralizing human mAb, L9 that preferentially bound NVDP minor repeats of PfCSP with high affinity while cross-reacting with NANP major repeats. L9 was more potent than six published neutralizing human PfCSP mAbs at mediating protection against mosquito bite challenge in mice. Isothermal titration calorimetry and multiphoton microscopy showed that L9 and the other most protective mAbs bound PfCSP with two binding events and mediated protection by killing SPZ in the liver and by preventing their egress from sinusoids and traversal of hepatocytes. This study defines the subdominant PfCSP minor repeats as neutralizing epitopes, identifies an in vitro biophysical correlate of SPZ neutralization, and demonstrates that the liver is an important site for antibodies to prevent malaria.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Antimalarials/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Adolescent , Adult , Animals , Cell Line , Cell Line, Tumor , Epitopes/immunology , Female , HEK293 Cells , Hepatocytes/immunology , Hepatocytes/parasitology , Humans , Liver/immunology , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Young Adult
14.
Cell ; 157(5): 1117-29, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855948

ABSTRACT

Dynamic instability, the stochastic switching between growth and shrinkage, is essential for microtubule function. This behavior is driven by GTP hydrolysis in the microtubule lattice and is inhibited by anticancer agents like Taxol. We provide insight into the mechanism of dynamic instability, based on high-resolution cryo-EM structures (4.7-5.6 Å) of dynamic microtubules and microtubules stabilized by GMPCPP or Taxol. We infer that hydrolysis leads to a compaction around the E-site nucleotide at longitudinal interfaces, as well as movement of the α-tubulin intermediate domain and H7 helix. Displacement of the C-terminal helices in both α- and ß-tubulin subunits suggests an effect on interactions with binding partners that contact this region. Taxol inhibits most of these conformational changes, allosterically inducing a GMPCPP-like state. Lateral interactions are similar in all conditions we examined, suggesting that microtubule lattice stability is primarily modulated at longitudinal interfaces.


Subject(s)
Guanosine Triphosphate/metabolism , Microtubules/chemistry , Tubulin/chemistry , Animals , Cryoelectron Microscopy , Crystallography, X-Ray , Guanosine Triphosphate/analogs & derivatives , Humans , Hydrolysis , Microtubules/metabolism , Microtubules/ultrastructure , Models, Molecular , Paclitaxel/metabolism , Protein Conformation , Tubulin/metabolism
15.
Cell ; 157(7): 1644-1656, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949974

ABSTRACT

Because apoptosis of infected cells can limit virus production and spread, some viruses have co-opted prosurvival genes from the host. This includes the Epstein-Barr virus (EBV) gene BHRF1, a homolog of human Bcl-2 proteins that block apoptosis and are associated with cancer. Computational design and experimental optimization were used to generate a novel protein called BINDI that binds BHRF1 with picomolar affinity. BINDI recognizes the hydrophobic cleft of BHRF1 in a manner similar to other Bcl-2 protein interactions but makes many additional contacts to achieve exceptional affinity and specificity. BINDI induces apoptosis in EBV-infected cancer lines, and when delivered with an antibody-targeted intracellular delivery carrier, BINDI suppressed tumor growth and extended survival in a xenograft disease model of EBV-positive human lymphoma. High-specificity-designed proteins that selectively kill target cells may provide an advantage over the toxic compounds used in current generation antibody-drug conjugates.


Subject(s)
Herpesvirus 4, Human/chemistry , Protein Engineering , Proteins/pharmacology , Viral Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Apoptosis/drug effects , Computational Biology , Crystallography, X-Ray , Epstein-Barr Virus Infections/drug therapy , Herpesvirus 4, Human/physiology , Heterografts , Humans , Lymphoma, B-Cell/drug therapy , Mice , Models, Molecular , Molecular Sequence Data , Neoplasm Transplantation , Proteins/chemistry , Proteins/metabolism , Sequence Alignment , Viral Proteins/chemistry
16.
Nature ; 614(7949): 774-780, 2023 02.
Article in English | MEDLINE | ID: mdl-36813896

ABSTRACT

De novo enzyme design has sought to introduce active sites and substrate-binding pockets that are predicted to catalyse a reaction of interest into geometrically compatible native scaffolds1,2, but has been limited by a lack of suitable protein structures and the complexity of native protein sequence-structure relationships. Here we describe a deep-learning-based 'family-wide hallucination' approach that generates large numbers of idealized protein structures containing diverse pocket shapes and designed sequences that encode them. We use these scaffolds to design artificial luciferases that selectively catalyse the oxidative chemiluminescence of the synthetic luciferin substrates diphenylterazine3 and 2-deoxycoelenterazine. The designed active sites position an arginine guanidinium group adjacent to an anion that develops during the reaction in a binding pocket with high shape complementarity. For both luciferin substrates, we obtain designed luciferases with high selectivity; the most active of these is a small (13.9 kDa) and thermostable (with a melting temperature higher than 95 °C) enzyme that has a catalytic efficiency on diphenylterazine (kcat/Km = 106 M-1 s-1) comparable to that of native luciferases, but a much higher substrate specificity. The creation of highly active and specific biocatalysts from scratch with broad applications in biomedicine is a key milestone for computational enzyme design, and our approach should enable generation of a wide range of luciferases and other enzymes.


Subject(s)
Deep Learning , Luciferases , Biocatalysis , Catalytic Domain , Enzyme Stability , Hot Temperature , Luciferases/chemistry , Luciferases/metabolism , Luciferins/metabolism , Luminescence , Oxidation-Reduction , Substrate Specificity
17.
Nature ; 622(7983): 594-602, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37821698

ABSTRACT

Metagenomes encode an enormous diversity of proteins, reflecting a multiplicity of functions and activities1,2. Exploration of this vast sequence space has been limited to a comparative analysis against reference microbial genomes and protein families derived from those genomes. Here, to examine the scale of yet untapped functional diversity beyond what is currently possible through the lens of reference genomes, we develop a computational approach to generate reference-free protein families from the sequence space in metagenomes. We analyse 26,931 metagenomes and identify 1.17 billion protein sequences longer than 35 amino acids with no similarity to any sequences from 102,491 reference genomes or the Pfam database3. Using massively parallel graph-based clustering, we group these proteins into 106,198 novel sequence clusters with more than 100 members, doubling the number of protein families obtained from the reference genomes clustered using the same approach. We annotate these families on the basis of their taxonomic, habitat, geographical and gene neighbourhood distributions and, where sufficient sequence diversity is available, predict protein three-dimensional models, revealing novel structures. Overall, our results uncover an enormously diverse functional space, highlighting the importance of further exploring the microbial functional dark matter.


Subject(s)
Metagenome , Metagenomics , Microbiology , Proteins , Cluster Analysis , Metagenome/genetics , Metagenomics/methods , Proteins/chemistry , Proteins/classification , Proteins/genetics , Databases, Protein , Protein Conformation
18.
Nature ; 616(7957): 581-589, 2023 04.
Article in English | MEDLINE | ID: mdl-37020023

ABSTRACT

General approaches for designing sequence-specific peptide-binding proteins would have wide utility in proteomics and synthetic biology. However, designing peptide-binding proteins is challenging, as most peptides do not have defined structures in isolation, and hydrogen bonds must be made to the buried polar groups in the peptide backbone1-3. Here, inspired by natural and re-engineered protein-peptide systems4-11, we set out to design proteins made out of repeating units that bind peptides with repeating sequences, with a one-to-one correspondence between the repeat units of the protein and those of the peptide. We use geometric hashing to identify protein backbones and peptide-docking arrangements that are compatible with bidentate hydrogen bonds between the side chains of the protein and the peptide backbone12. The remainder of the protein sequence is then optimized for folding and peptide binding. We design repeat proteins to bind to six different tripeptide-repeat sequences in polyproline II conformations. The proteins are hyperstable and bind to four to six tandem repeats of their tripeptide targets with nanomolar to picomolar affinities in vitro and in living cells. Crystal structures reveal repeating interactions between protein and peptide interactions as designed, including ladders of hydrogen bonds from protein side chains to peptide backbones. By redesigning the binding interfaces of individual repeat units, specificity can be achieved for non-repeating peptide sequences and for disordered regions of native proteins.


Subject(s)
Peptides , Protein Engineering , Proteins , Amino Acid Sequence , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Proteins/chemistry , Proteins/metabolism , Protein Engineering/methods , Hydrogen Bonding , Protein Binding , Protein Folding , Protein Conformation
19.
Nature ; 620(7976): 1089-1100, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37433327

ABSTRACT

There has been considerable recent progress in designing new proteins using deep-learning methods1-9. Despite this progress, a general deep-learning framework for protein design that enables solution of a wide range of design challenges, including de novo binder design and design of higher-order symmetric architectures, has yet to be described. Diffusion models10,11 have had considerable success in image and language generative modelling but limited success when applied to protein modelling, probably due to the complexity of protein backbone geometry and sequence-structure relationships. Here we show that by fine-tuning the RoseTTAFold structure prediction network on protein structure denoising tasks, we obtain a generative model of protein backbones that achieves outstanding performance on unconditional and topology-constrained protein monomer design, protein binder design, symmetric oligomer design, enzyme active site scaffolding and symmetric motif scaffolding for therapeutic and metal-binding protein design. We demonstrate the power and generality of the method, called RoseTTAFold diffusion (RFdiffusion), by experimentally characterizing the structures and functions of hundreds of designed symmetric assemblies, metal-binding proteins and protein binders. The accuracy of RFdiffusion is confirmed by the cryogenic electron microscopy structure of a designed binder in complex with influenza haemagglutinin that is nearly identical to the design model. In a manner analogous to networks that produce images from user-specified inputs, RFdiffusion enables the design of diverse functional proteins from simple molecular specifications.


Subject(s)
Deep Learning , Proteins , Catalytic Domain , Cryoelectron Microscopy , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/ultrastructure , Protein Binding , Proteins/chemistry , Proteins/metabolism , Proteins/ultrastructure
20.
Nature ; 606(7912): 49-58, 2022 06.
Article in English | MEDLINE | ID: mdl-35650353

ABSTRACT

The ability to design efficient enzymes from scratch would have a profound effect on chemistry, biotechnology and medicine. Rapid progress in protein engineering over the past decade makes us optimistic that this ambition is within reach. The development of artificial enzymes containing metal cofactors and noncanonical organocatalytic groups shows how protein structure can be optimized to harness the reactivity of nonproteinogenic elements. In parallel, computational methods have been used to design protein catalysts for diverse reactions on the basis of fundamental principles of transition state stabilization. Although the activities of designed catalysts have been quite low, extensive laboratory evolution has been used to generate efficient enzymes. Structural analysis of these systems has revealed the high degree of precision that will be needed to design catalysts with greater activity. To this end, emerging protein design methods, including deep learning, hold particular promise for improving model accuracy. Here we take stock of key developments in the field and highlight new opportunities for innovation that should allow us to transition beyond the current state of the art and enable the robust design of biocatalysts to address societal needs.


Subject(s)
Biocatalysis , Biotechnology , Protein Engineering , Proteins , Biotechnology/methods , Biotechnology/trends , Protein Engineering/methods , Protein Engineering/trends , Proteins/chemistry , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL