Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Protein Sci ; 32(11): e4780, 2023 11.
Article in English | MEDLINE | ID: mdl-37695922

ABSTRACT

Predicting the effects of mutations on protein function and stability is an outstanding challenge. Here, we assess the performance of a variant of RoseTTAFold jointly trained for sequence and structure recovery, RFjoint , for mutation effect prediction. Without any further training, we achieve comparable accuracy in predicting mutation effects for a diverse set of protein families using RFjoint to both another zero-shot model (MSA Transformer) and a model that requires specific training on a particular protein family for mutation effect prediction (DeepSequence). Thus, although the architecture of RFjoint was developed to address the protein design problem of scaffolding functional motifs, RFjoint acquired an understanding of the mutational landscapes of proteins during model training that is equivalent to that of recently developed large protein language models. The ability to simultaneously reason over protein structure and sequence could enable even more precise mutation effect predictions following supervised training on the task. These results suggest that RFjoint has a quite broad understanding of protein sequence-structure landscapes, and can be viewed as a joint model for protein sequence and structure which could be broadly useful for protein modeling.


Subject(s)
Proteins , Proteins/genetics , Proteins/chemistry , Mutation , Amino Acid Sequence , Protein Stability
3.
Science ; 381(6659): 754-760, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37590357

ABSTRACT

In nature, proteins that switch between two conformations in response to environmental stimuli structurally transduce biochemical information in a manner analogous to how transistors control information flow in computing devices. Designing proteins with two distinct but fully structured conformations is a challenge for protein design as it requires sculpting an energy landscape with two distinct minima. Here we describe the design of "hinge" proteins that populate one designed state in the absence of ligand and a second designed state in the presence of ligand. X-ray crystallography, electron microscopy, double electron-electron resonance spectroscopy, and binding measurements demonstrate that despite the significant structural differences the two states are designed with atomic level accuracy and that the conformational and binding equilibria are closely coupled.


Subject(s)
Protein Engineering , Crystallography, X-Ray , Ligands , Protein Engineering/methods , Protein Conformation
4.
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
5.
bioRxiv ; 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38187589

ABSTRACT

A general method for designing proteins to bind and sense any small molecule of interest would be widely useful. Due to the small number of atoms to interact with, binding to small molecules with high affinity requires highly shape complementary pockets, and transducing binding events into signals is challenging. Here we describe an integrated deep learning and energy based approach for designing high shape complementarity binders to small molecules that are poised for downstream sensing applications. We employ deep learning generated psuedocycles with repeating structural units surrounding central pockets; depending on the geometry of the structural unit and repeat number, these pockets span wide ranges of sizes and shapes. For a small molecule target of interest, we extensively sample high shape complementarity pseudocycles to generate large numbers of customized potential binding pockets; the ligand binding poses and the interacting interfaces are then optimized for high affinity binding. We computationally design binders to four diverse molecules, including for the first time polar flexible molecules such as methotrexate and thyroxine, which are expressed at high levels and have nanomolar affinities straight out of the computer. Co-crystal structures are nearly identical to the design models. Taking advantage of the modular repeating structure of pseudocycles and central location of the binding pockets, we constructed low noise nanopore sensors and chemically induced dimerization systems by splitting the binders into domains which assemble into the original pseudocycle pocket upon target molecule addition.

6.
Science ; 377(6604): 387-394, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35862514

ABSTRACT

The binding and catalytic functions of proteins are generally mediated by a small number of functional residues held in place by the overall protein structure. Here, we describe deep learning approaches for scaffolding such functional sites without needing to prespecify the fold or secondary structure of the scaffold. The first approach, "constrained hallucination," optimizes sequences such that their predicted structures contain the desired functional site. The second approach, "inpainting," starts from the functional site and fills in additional sequence and structure to create a viable protein scaffold in a single forward pass through a specifically trained RoseTTAFold network. We use these two methods to design candidate immunogens, receptor traps, metalloproteins, enzymes, and protein-binding proteins and validate the designs using a combination of in silico and experimental tests.


Subject(s)
Deep Learning , Protein Engineering , Proteins , Binding Sites , Catalysis , Protein Binding , Protein Engineering/methods , Protein Folding , Protein Structure, Secondary , Proteins/chemistry
7.
Analyst ; 146(9): 2851-2861, 2021 May 04.
Article in English | MEDLINE | ID: mdl-33949378

ABSTRACT

The number of people living with HIV continues to increase with the current total near 38 million, of which about 26 million are receiving antiretroviral therapy (ART). These treatment regimens are highly effective when properly managed, requiring routine viral load monitoring to assess successful viral suppression. Efforts to expand access by decentralizing HIV nucleic acid testing in low- and middle-income countries (LMICs) has been hampered by the cost and complexity of current tests. Sample preparation of blood samples has traditionally relied on cumbersome RNA extraction methods, and it continues to be a key bottleneck for developing low-cost POC nucleic acid tests. We present a microfluidic paper-based analytical device (µPAD) for extracting RNA and detecting HIV in serum, leveraging low-cost materials, simple buffers, and an electric field. We detect HIV virions and MS2 bacteriophage internal control in human serum using a novel lysis and RNase inactivation method, paper-based isotachophoresis (ITP) for RNA extraction, and duplexed reverse transcription recombinase polymerase amplification (RT-RPA) for nucleic acid amplification. We design a specialized ITP system to extract and concentrate RNA, while excluding harsh reagents used for lysis and RNase inactivation. We found the ITP µPAD can extract and purify 5000 HIV RNA copies per mL of serum. We then demonstrate detection of HIV virions and MS2 bacteriophage in human serum within 45-minutes.


Subject(s)
HIV Infections , Isotachophoresis , HIV Infections/diagnosis , Humans , Nucleic Acid Amplification Techniques , RNA/genetics , RNA, Viral/genetics , Recombinases/genetics , Recombinases/metabolism , Reverse Transcription , Sensitivity and Specificity
8.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33712545

ABSTRACT

The protein design problem is to identify an amino acid sequence that folds to a desired structure. Given Anfinsen's thermodynamic hypothesis of folding, this can be recast as finding an amino acid sequence for which the desired structure is the lowest energy state. As this calculation involves not only all possible amino acid sequences but also, all possible structures, most current approaches focus instead on the more tractable problem of finding the lowest-energy amino acid sequence for the desired structure, often checking by protein structure prediction in a second step that the desired structure is indeed the lowest-energy conformation for the designed sequence, and typically discarding a large fraction of designed sequences for which this is not the case. Here, we show that by backpropagating gradients through the transform-restrained Rosetta (trRosetta) structure prediction network from the desired structure to the input amino acid sequence, we can directly optimize over all possible amino acid sequences and all possible structures in a single calculation. We find that trRosetta calculations, which consider the full conformational landscape, can be more effective than Rosetta single-point energy estimations in predicting folding and stability of de novo designed proteins. We compare sequence design by conformational landscape optimization with the standard energy-based sequence design methodology in Rosetta and show that the former can result in energy landscapes with fewer alternative energy minima. We show further that more funneled energy landscapes can be designed by combining the strengths of the two approaches: the low-resolution trRosetta model serves to disfavor alternative states, and the high-resolution Rosetta model serves to create a deep energy minimum at the design target structure.


Subject(s)
Neural Networks, Computer , Proteins/chemistry , Models, Molecular , Protein Conformation , Protein Folding , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...