1
|
Maillie CA, Golden K, Wilson IA, Ward AB, Mravic M. Ab initio prediction of specific phospholipid complexes and membrane association of HIV-1 MPER antibodies by multi-scale simulations. eLife 2025; 12:RP90139. [PMID: 40192122 PMCID: PMC11975376 DOI: 10.7554/elife.90139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
A potent class of HIV-1 broadly neutralizing antibodies (bnAbs) targets the envelope glycoprotein's membrane proximal exposed region (MPER) through a proposed mechanism where hypervariable loops embed into lipid bilayers and engage headgroup moieties alongside the epitope. We address the feasibility and determinant molecular features of this mechanism using multi-scale modeling. All-atom simulations of 4E10, PGZL1, 10E8, and LN01 docked onto HIV-like membranes consistently form phospholipid complexes at key complementarity-determining region loop sites, solidifying that stable and specific lipid interactions anchor bnAbs to membrane surfaces. Ancillary protein-lipid contacts reveal surprising contributions from antibody framework regions. Coarse-grained simulations effectively capture antibodies embedding into membranes. Simulations estimating protein-membrane interaction strength for PGZL1 variants along an inferred maturation pathway show bilayer affinity is evolved and correlates with neutralization potency. The modeling demonstrated here uncovers insights into lipid participation in antibodies' recognition of membrane proteins and highlights antibody features to prioritize in vaccine design.
Collapse
Affiliation(s)
- Colleen A Maillie
- Department of Integrative Structural and Computational Biology, The Scripps Research InstituteLa JollaUnited States
| | - Kiana Golden
- Department of Integrative Structural and Computational Biology, The Scripps Research InstituteLa JollaUnited States
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research InstituteLa JollaUnited States
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research InstituteLa JollaUnited States
| | - Marco Mravic
- Department of Integrative Structural and Computational Biology, The Scripps Research InstituteLa JollaUnited States
| |
Collapse
|
2
|
Zhu J, Liang M, Sun K, Wei Y, Guo R, Zhang L, Shi J, Ma D, Hu Q, Huang G, Lu P. De novo design of transmembrane fluorescence-activating proteins. Nature 2025; 640:249-257. [PMID: 39972138 DOI: 10.1038/s41586-025-08598-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/09/2024] [Indexed: 02/21/2025]
Abstract
The recognition of ligands by transmembrane proteins is essential for the exchange of materials, energy and information across biological membranes. Progress has been made in the de novo design of transmembrane proteins1-6, as well as in designing water-soluble proteins to bind small molecules7-12, but de novo design of transmembrane proteins that tightly and specifically bind to small molecules remains an outstanding challenge13. Here we present the accurate design of ligand-binding transmembrane proteins by integrating deep learning and energy-based methods. We designed pre-organized ligand-binding pockets in high-quality four-helix backbones for a fluorogenic ligand, and generated a transmembrane span using gradient-guided hallucination. The designer transmembrane proteins specifically activated fluorescence of the target fluorophore with mid-nanomolar affinity, exhibiting higher brightness and quantum yield compared to those of enhanced green fluorescent protein. These proteins were highly active in the membrane fraction of live bacterial and eukaryotic cells following expression. The crystal and cryogenic electron microscopy structures of the designer protein-ligand complexes were very close to the structures of the design models. We showed that the interactions between ligands and transmembrane proteins within the membrane can be accurately designed. Our work paves the way for the creation of new functional transmembrane proteins, with a wide range of applications including imaging, ligand sensing and membrane transport.
Collapse
Affiliation(s)
- Jingyi Zhu
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Mingfu Liang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Ke Sun
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yu Wei
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Ruiying Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Lijing Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Junhui Shi
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Dan Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Qi Hu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Gaoxingyu Huang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Peilong Lu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China.
| |
Collapse
|
3
|
Chen Y, Bhattacharya S, Bergmann L, Correy GJ, Tan S, Hou K, Biel J, Lu L, Bakanas I, Polizzi NF, Fraser JS, DeGrado WF. Emergence of specific binding and catalysis from a designed generalist binding protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635804. [PMID: 39975260 PMCID: PMC11838529 DOI: 10.1101/2025.01.30.635804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The evolution of binding and catalysis played a central role in the emergence of life. While natural proteins have finely tuned affinities for their primary ligands, they also bind weakly and promiscuously to other molecules, which serve as starting points for stepwise, incremental evolution of entirely new specificities. Thus, modern proteins emerged from the joint exploration of sequence and structural space. The ability of natural proteins to bind promiscuously to small molecule fragments has been widely evaluated using methods including crystallographic fragment screening. However, this approach had not been applied to de novo proteins. Here, we apply this method to explore the promiscuity of a de novo small molecule-binding protein ABLE. As in Nature, we found ABLE was capable of forming weak complexes, which were found to be excellent starting points for evolving entirely new functions, including a binder of a turn-on fluorophore and a highly efficient and specific Kemp eliminase enzyme. This work shows how Nature and protein designers can take advantage of promiscuous binding interactions to evolve new proteins with specialized functions.
Collapse
Affiliation(s)
- Yuda Chen
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Sagar Bhattacharya
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Lena Bergmann
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Galen J. Correy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Sophia Tan
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Kaipeng Hou
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Justin Biel
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Lei Lu
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Ian Bakanas
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Nicholas F. Polizzi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
4
|
Rosen PC, Horwitz SM, Brooks DJ, Kim E, Ambarian JA, Waidmann L, Davis KM, Yellen G. State-dependent motion of a genetically encoded fluorescent biosensor. Proc Natl Acad Sci U S A 2025; 122:e2426324122. [PMID: 40048274 PMCID: PMC11912384 DOI: 10.1073/pnas.2426324122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
Genetically encoded biosensors can measure biochemical properties such as small-molecule concentrations with single-cell resolution, even in vivo. Despite their utility, these sensors are "black boxes": Very little is known about the structures of their low- and high-fluorescence states or what features are required to transition between them. We used LiLac, a lactate biosensor with a quantitative fluorescence-lifetime readout, as a model system to address these questions. X-ray crystal structures and engineered high-affinity metal bridges demonstrate that LiLac exhibits a large interdomain twist motion that pulls the fluorescent protein away from a "sealed," high-lifetime state in the absence of lactate to a "cracked," low-lifetime state in its presence. Understanding the structures and dynamics of LiLac will help to think about and engineer other fluorescent biosensors.
Collapse
Affiliation(s)
- Paul C Rosen
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Daniel J Brooks
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Erica Kim
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | | | - Lidia Waidmann
- Department of Chemistry, Emory University, Atlanta, GA 30322
| | | | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
5
|
Han J, Dang B. Optimizing Solid-Phase Protein Synthesis Using CPG-2000 and a Nickel-Cleavable SNAC-tag Linker. Org Lett 2025; 27:2104-2109. [PMID: 39988888 DOI: 10.1021/acs.orglett.5c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Solid-phase chemical ligation (SPCL) is a powerful method for simplifying protein synthesis but faces challenges with orthogonal protection strategies, suboptimal solid supports, and limited cleavable linkers. In this study, we optimized SPCL by combining low-loading controlled-pore glass (CPG-2000) with a nickel-cleavable SNAC-tag linker. This system enabled the successful assembly of five peptide fragments and the efficient synthesis of a 131-amino-acid de novo protein, achieving a 25% yield.
Collapse
Affiliation(s)
- Jianyi Han
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Research Center for Industries of the Future and Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study; Hangzhou, Zhejiang 310024, China
| | - Bobo Dang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Research Center for Industries of the Future and Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study; Hangzhou, Zhejiang 310024, China
| |
Collapse
|
6
|
Mann SI, Lin Z, Tan SK, Zhu J, Widel ZXW, Bakanas I, Mansergh JP, Liu R, Kelly MJS, Wu Y, Wells JA, Therien MJ, DeGrado WF. De Novo Design of Proteins That Bind Naphthalenediimides, Powerful Photooxidants with Tunable Photophysical Properties. J Am Chem Soc 2025; 147:7849-7858. [PMID: 39982408 PMCID: PMC11972567 DOI: 10.1021/jacs.4c18151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
De novo protein design provides a framework to test our understanding of protein function and build proteins with cofactors and functions not found in nature. Here, we report the design of proteins designed to bind powerful photooxidants and the evaluation of the use of these proteins to generate diffusible small-molecule reactive species. Because excited-state dynamics are influenced by the dynamics and hydration of a photooxidant's environment, it was important to not only design a binding site but also to evaluate its dynamic properties. Thus, we used computational design in conjunction with molecular dynamics (MD) simulations to design a protein, designated NBP (NDI Binding Protein), that held a naphthalenediimide (NDI), a powerful photooxidant, in a programmable molecular environment. Solution NMR confirmed the structure of the complex. We evaluated two NDI cofactors in this de novo protein using ultrafast pump-probe spectroscopy to evaluate light-triggered intra- and intermolecular electron transfer function. Moreover, we demonstrated the utility of this platform to activate multiple molecular probes for protein labeling.
Collapse
Affiliation(s)
- Samuel I Mann
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
- The Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158-9001, United States
| | - Zhi Lin
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
| | - Sophia K Tan
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
- The Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158-9001, United States
| | - Jiaqi Zhu
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Zachary X W Widel
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Ian Bakanas
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
- The Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158-9001, United States
| | - Jarrett P Mansergh
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Rui Liu
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Mark J S Kelly
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
| | - Yibing Wu
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California 94158, United States
| | - Michael J Therien
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94158-9001, United States
- The Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158-9001, United States
| |
Collapse
|
7
|
Grau B, Kormos R, Bañó-Polo M, Chen K, García-Murria MJ, Hajredini F, Sánchez del Pino MM, Jo H, Martínez-Gil L, von Heijne G, DeGrado WF, Mingarro I. Sequence-dependent scale for translocon-mediated insertion of interfacial helices in membranes. SCIENCE ADVANCES 2025; 11:eads6804. [PMID: 39970206 PMCID: PMC11837994 DOI: 10.1126/sciadv.ads6804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Biological membranes consist of a lipid bilayer studded with integral and peripheral membrane proteins. Most α-helical membrane proteins require protein-conducting insertases known as translocons to assist in their membrane insertion and folding. While the sequence-dependent propensities for a helix to either translocate through the translocon or insert into the membrane have been codified into numerical hydrophobicity scales, the corresponding propensity to partition into the membrane interface remains unrevealed. By engineering diagnostic glycosylation sites around test peptide sequences inserted into a host protein, we devised a system that can differentiate between water-soluble, surface-bound, and transmembrane (TM) states of the sequence based on its glycosylation pattern. Using this system, we determined the sequence-dependent propensities for transfer from the translocon to a TM, interfacial, or extramembrane space and compared these propensities with the corresponding probability distributions determined from the sequences and structures of experimentally determined proteins.
Collapse
Affiliation(s)
- Brayan Grau
- Institute for Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, E-46100 Burjassot, Spain
| | - Rian Kormos
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Manuel Bañó-Polo
- Institute for Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, E-46100 Burjassot, Spain
| | - Kehan Chen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - María J. García-Murria
- Institute for Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, E-46100 Burjassot, Spain
| | - Fatlum Hajredini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Manuel M. Sánchez del Pino
- Institute for Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, E-46100 Burjassot, Spain
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Luis Martínez-Gil
- Institute for Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, E-46100 Burjassot, Spain
| | - Gunnar von Heijne
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, SE-10691 Stockholm, Sweden
| | - William F. DeGrado
- Institute for Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, E-46100 Burjassot, Spain
| | - Ismael Mingarro
- Institute for Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, E-46100 Burjassot, Spain
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
8
|
Petrenas R, Hawkins OA, Jones JF, Scott DA, Fletcher JM, Obst U, Lombardi L, Pirro F, Leggett GJ, Oliver TA, Woolfson DN. Confinement and Catalysis within De Novo Designed Peptide Barrels. J Am Chem Soc 2025; 147:3796-3803. [PMID: 39813445 PMCID: PMC11783595 DOI: 10.1021/jacs.4c16633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
De novo protein design has advanced such that many peptide assemblies and protein structures can be generated predictably and quickly. The drive now is to bring functions to these structures, for example, small-molecule binding and catalysis. The formidable challenge of binding and orienting multiple small molecules to direct chemistry is particularly important for paving the way to new functionalities. To address this, here we describe the design, characterization, and application of small-molecule:peptide ternary complexes in aqueous solution. This uses α-helical barrel (αHB) peptide assemblies, which comprise 5 or more α helices arranged around central channels. These channels are solvent accessible, and their internal dimensions and chemistries can be altered predictably. Thus, αHBs are analogous to "molecular flasks" made in supramolecular, polymer, and materials chemistry. Using Förster resonance energy transfer as a readout, we demonstrate that specific αHBs can accept two different organic dyes, 1,6-diphenyl-1,3,5-hexatriene and Nile red, in close proximity. In addition, two anthracene molecules can be accommodated within an αHB to promote anthracene photodimerization. However, not all ternary complexes are productive, either in energy transfer or photodimerization, illustrating the control that can be exerted by judicious choice and design of the αHB.
Collapse
Affiliation(s)
- Rokas Petrenas
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Olivia A. Hawkins
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Jacob F. Jones
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - D. Arne Scott
- Rosa
Biotech, Science Creates St Philips, Albert Road, Bristol BS2 0XJ, U.K.
| | - Jordan M. Fletcher
- Rosa
Biotech, Science Creates St Philips, Albert Road, Bristol BS2 0XJ, U.K.
| | - Ulrike Obst
- Rosa
Biotech, Science Creates St Philips, Albert Road, Bristol BS2 0XJ, U.K.
| | - Lucia Lombardi
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- Department
of Chemical Engineering, Imperial College
London, London SW7 2AZ, U.K.
| | - Fabio Pirro
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Graham J. Leggett
- School
of Mathematical and Physical Sciences, University
of Sheffield, Brook Hill, Sheffield S3 7HF, U.K.
| | - Thomas A.A. Oliver
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
| | - Derek N. Woolfson
- School
of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- Max
Planck-Bristol Centre for Minimal Biology, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- Bristol BioDesign
Institute, University of Bristol, Cantock’s Close, Bristol BS8 1TS, U.K.
- School
of Biochemistry, University of Bristol, Medical Sciences Building, Bristol BS8 1TD, U.K.
| |
Collapse
|
9
|
Litschko C, Di Domenico V, Schulze J, Li S, Ovchinnikova OG, Voskuilen T, Bethe A, Cifuente JO, Marina A, Budde I, Mast TA, Sulewska M, Berger M, Buettner FFR, Lowary TL, Whitfield C, Codée JDC, Schubert M, Guerin ME, Fiebig T. Transition transferases prime bacterial capsule polymerization. Nat Chem Biol 2025; 21:120-130. [PMID: 38951648 PMCID: PMC11666461 DOI: 10.1038/s41589-024-01664-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
Capsules are long-chain carbohydrate polymers that envelop the surfaces of many bacteria, protecting them from host immune responses. Capsule biosynthesis enzymes are potential drug targets and valuable biotechnological tools for generating vaccine antigens. Despite their importance, it remains unknown how structurally variable capsule polymers of Gram-negative pathogens are linked to the conserved glycolipid anchoring these virulence factors to the bacterial membrane. Using Actinobacillus pleuropneumoniae as an example, we demonstrate that CpsA and CpsC generate a poly(glycerol-3-phosphate) linker to connect the glycolipid with capsules containing poly(galactosylglycerol-phosphate) backbones. We reconstruct the entire capsule biosynthesis pathway in A. pleuropneumoniae serotypes 3 and 7, solve the X-ray crystal structure of the capsule polymerase CpsD, identify its tetratricopeptide repeat domain as essential for elongating poly(glycerol-3-phosphate) and show that CpsA and CpsC stimulate CpsD to produce longer polymers. We identify the CpsA and CpsC product as a wall teichoic acid homolog, demonstrating similarity between the biosynthesis of Gram-positive wall teichoic acid and Gram-negative capsules.
Collapse
Affiliation(s)
- Christa Litschko
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Valerio Di Domenico
- Structural Glycobiology Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, Spain
- Structural Glycobiology Laboratory, Department of Structural and Molecular Biology; Molecular Biology Institute of Barcelona, Spanish National Research Council, Barcelona Science Park, Tower R, Barcelona, Spain
| | - Julia Schulze
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Sizhe Li
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Olga G Ovchinnikova
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Thijs Voskuilen
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Andrea Bethe
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Javier O Cifuente
- Structural Glycobiology Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, Spain
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences, Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| | - Alberto Marina
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences, Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| | - Insa Budde
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Tim A Mast
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Małgorzata Sulewska
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Monika Berger
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Falk F R Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Proteomics, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Chris Whitfield
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Mario Schubert
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, Spain.
- Structural Glycobiology Laboratory, Department of Structural and Molecular Biology; Molecular Biology Institute of Barcelona, Spanish National Research Council, Barcelona Science Park, Tower R, Barcelona, Spain.
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences, Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| | - Timm Fiebig
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.
- German Center for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany.
| |
Collapse
|
10
|
Glögl M, Krishnakumar A, Ragotte RJ, Goreshnik I, Coventry B, Bera AK, Kang A, Joyce E, Ahn G, Huang B, Yang W, Chen W, Sanchez MG, Koepnick B, Baker D. Target-conditioned diffusion generates potent TNFR superfamily antagonists and agonists. Science 2024; 386:1154-1161. [PMID: 39636970 DOI: 10.1126/science.adp1779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024]
Abstract
Despite progress in designing protein-binding proteins, the shape matching of designs to targets is lower than in many native protein complexes, and design efforts have failed for the tumor necrosis factor receptor 1 (TNFR1) and other protein targets with relatively flat and polar surfaces. We hypothesized that free diffusion from random noise could generate shape-matched binders for challenging targets and tested this approach on TNFR1. We obtain designs with low picomolar affinity whose specificity can be completely switched to other family members using partial diffusion. Designs function as antagonists or as superagonists when presented at higher valency for OX40 and 4-1BB. The ability to design high-affinity and high-specificity antagonists and agonists for pharmacologically important targets in silico presages a coming era in protein design in which binders are made by computation rather than immunization or random screening approaches.
Collapse
MESH Headings
- Humans
- Drug Design
- Protein Binding
- Receptors, Tumor Necrosis Factor, Type I/agonists
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/chemistry
- Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists
- Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
- Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry
- Receptors, OX40/agonists
- Receptors, OX40/antagonists & inhibitors
- Receptors, OX40/chemistry
Collapse
Affiliation(s)
- Matthias Glögl
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Aditya Krishnakumar
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Robert J Ragotte
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Asim K Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Emily Joyce
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Green Ahn
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Buwei Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Wei Yang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Wei Chen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Mariana Garcia Sanchez
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Koepnick
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
11
|
Nguyen VTD, Nguyen ND, Hy TS. ProteinReDiff: Complex-based ligand-binding proteins redesign by equivariant diffusion-based generative models. STRUCTURAL DYNAMICS (MELVILLE, N.Y.) 2024; 11:064102. [PMID: 39629167 PMCID: PMC11614476 DOI: 10.1063/4.0000271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
Proteins, serving as the fundamental architects of biological processes, interact with ligands to perform a myriad of functions essential for life. Designing functional ligand-binding proteins is pivotal for advancing drug development and enhancing therapeutic efficacy. In this study, we introduce ProteinReDiff, an diffusion framework targeting the redesign of ligand-binding proteins. Using equivariant diffusion-based generative models, ProteinReDiff enables the creation of high-affinity ligand-binding proteins without the need for detailed structural information, leveraging instead the potential of initial protein sequences and ligand SMILES strings. Our evaluations across sequence diversity, structural preservation, and ligand binding affinity underscore ProteinReDiff's potential to advance computational drug discovery and protein engineering.
Collapse
Affiliation(s)
| | - Nhan D Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois 60637, USA
| | - Truong Son Hy
- Department of Computer Science, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| |
Collapse
|
12
|
Liu Y, Wang S, Dong J, Chen L, Wang X, Wang L, Li F, Wang C, Zhang J, Wang Y, Wei S, Chen Q, Liu H. De novo protein design with a denoising diffusion network independent of pretrained structure prediction models. Nat Methods 2024; 21:2107-2116. [PMID: 39384986 DOI: 10.1038/s41592-024-02437-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 08/30/2024] [Indexed: 10/11/2024]
Abstract
The recent success of RFdiffusion, a method for protein structure design with a denoising diffusion probabilistic model, has relied on fine-tuning the RoseTTAFold structure prediction network for protein backbone denoising. Here, we introduce SCUBA-diffusion (SCUBA-D), a protein backbone denoising diffusion probabilistic model freshly trained by considering co-diffusion of sequence representation to enhance model regularization and adversarial losses to minimize data-out-of-distribution errors. While matching the performance of the pretrained RoseTTAFold-based RFdiffusion in generating experimentally realizable protein structures, SCUBA-D readily generates protein structures with not-yet-observed overall folds that are different from those predictable with RoseTTAFold. The accuracy of SCUBA-D was confirmed by the X-ray structures of 16 designed proteins and a protein complex, and by experiments validating designed heme-binding proteins and Ras-binding proteins. Our work shows that deep generative models of images or texts can be fruitfully extended to complex physical objects like protein structures by addressing outstanding issues such as the data-out-of-distribution errors.
Collapse
Affiliation(s)
- Yufeng Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sheng Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jixin Dong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | | | - Xinyu Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lei Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fudong Li
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Anhui Basic Discipline Research Center of Artificial Intelligence Biotechnology and Synthetic Biology, University of Science and Technology of China, Hefei, China
| | - Chenchen Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiahai Zhang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Anhui Basic Discipline Research Center of Artificial Intelligence Biotechnology and Synthetic Biology, University of Science and Technology of China, Hefei, China
| | - Yuzhu Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Si Wei
- iFLYTEK Research, Hefei, China
| | - Quan Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China.
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Oristruct Biotech Co. Ltd, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, Anhui Basic Discipline Research Center of Artificial Intelligence Biotechnology and Synthetic Biology, University of Science and Technology of China, Hefei, China.
| | - Haiyan Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Oristruct Biotech Co. Ltd, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, Anhui Basic Discipline Research Center of Artificial Intelligence Biotechnology and Synthetic Biology, University of Science and Technology of China, Hefei, China.
- School of Biomedical Engineering, Suzhou Institute for Advanced Research, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
13
|
Hatstat AK, Kormos R, Xu V, DeGrado WF. A designed Zn 2+ sensor domain transmits binding information to transmembrane histidine kinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621206. [PMID: 39553995 PMCID: PMC11565981 DOI: 10.1101/2024.10.30.621206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Generating stimulus-responsive, allosteric signaling de novo is a significant challenge in protein design. In natural systems like bacterial histidine kinases (HKs), signal transduction occurs when ligand binding initiates a signal that is amplified across biological membranes over long distances to induce large-scale rearrangements and phosphorylation relays. Here, we ask whether our understanding of protein design and multi-domain, intramolecular signaling has progressed sufficiently to enable engineering of a HK with tunable de novo components. We generated de novo metal-binding sensor domains and substituted them for the native sensor domain of a transmembrane HK, affording chimeras that transduce signals initiated from a de novo sensor. Signaling depended on the designed sensor's stability and the interdomain linker's phase and length. These results show the usefulness of de novo design to elucidate biochemical mechanisms and principles for design of new signaling systems.
Collapse
|
14
|
Durant G, Boyles F, Birchall K, Deane CM. The future of machine learning for small-molecule drug discovery will be driven by data. NATURE COMPUTATIONAL SCIENCE 2024; 4:735-743. [PMID: 39407003 DOI: 10.1038/s43588-024-00699-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/03/2024] [Indexed: 10/25/2024]
Abstract
Many studies have prophesied that the integration of machine learning techniques into small-molecule therapeutics development will help to deliver a true leap forward in drug discovery. However, increasingly advanced algorithms and novel architectures have not always yielded substantial improvements in results. In this Perspective, we propose that a greater focus on the data for training and benchmarking these models is more likely to drive future improvement, and explore avenues for future research and strategies to address these data challenges.
Collapse
Affiliation(s)
- Guy Durant
- Department of Statistics, University of Oxford, Oxford, UK
| | - Fergus Boyles
- Department of Statistics, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
15
|
Zhang Z, Shen WX, Liu Q, Zitnik M. Efficient Generation of Protein Pockets with PocketGen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.25.581968. [PMID: 38464121 PMCID: PMC10925136 DOI: 10.1101/2024.02.25.581968] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Designing protein-binding proteins is critical for drug discovery. However, the AI-based design of such proteins is challenging due to the complexity of ligand-protein interactions, the flexibility of ligand molecules and amino acid side chains, and sequence-structure dependencies. We introduce PocketGen, a deep generative model that simultaneously produces both the residue sequence and atomic structure of the protein regions where ligand interactions occur. PocketGen ensures consistency between sequence and structure by using a graph transformer for structural encoding and a sequence refinement module based on a protein language model. The bilevel graph transformer captures interactions at multiple scales, including atom, residue, and ligand levels. To enhance sequence refinement, PocketGen integrates a structural adapter into the protein language model, ensuring that structure-based predictions align with sequence-based predictions. PocketGen can generate high-fidelity protein pockets with superior binding affinity and structural validity. It operates ten times faster than physics-based methods and achieves a 95% success rate, defined as the percentage of generated pockets with higher binding affinity than reference pockets. Additionally, it attains an amino acid recovery rate exceeding 64%.
Collapse
Affiliation(s)
- Zaixi Zhang
- State Key Laboratory of Cognitive Intelligence, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Wan Xiang Shen
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Qi Liu
- State Key Laboratory of Cognitive Intelligence, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| |
Collapse
|
16
|
Leonard AC, Friedman AJ, Chayer R, Petersen BM, Woojuh J, Xing Z, Cutler SR, Kaar JL, Shirts MR, Whitehead TA. Rationalizing Diverse Binding Mechanisms to the Same Protein Fold: Insights for Ligand Recognition and Biosensor Design. ACS Chem Biol 2024; 19:1757-1772. [PMID: 39017707 DOI: 10.1021/acschembio.4c00243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The engineering of novel protein-ligand binding interactions, particularly for complex drug-like molecules, is an unsolved problem, which could enable many practical applications of protein biosensors. In this work, we analyzed two engineered biosensors, derived from the plant hormone sensor PYR1, to recognize either the agrochemical mandipropamid or the synthetic cannabinoid WIN55,212-2. Using a combination of quantitative deep mutational scanning experiments and molecular dynamics simulations, we demonstrated that mutations at common positions can promote protein-ligand shape complementarity and revealed prominent differences in the electrostatic networks needed to complement diverse ligands. MD simulations indicate that both PYR1 protein-ligand complexes bind a single conformer of their target ligand that is close to the lowest free-energy conformer. Computational design using a fixed conformer and rigid body orientation led to new WIN55,212-2 sensors with nanomolar limits of detection. This work reveals mechanisms by which the versatile PYR1 biosensor scaffold can bind diverse ligands. This work also provides computational methods to sample realistic ligand conformers and rigid body alignments that simplify the computational design of biosensors for novel ligands of interest.
Collapse
Affiliation(s)
- Alison C Leonard
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| | - Anika J Friedman
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| | - Rachel Chayer
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| | - Brian M Petersen
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| | - Janty Woojuh
- Department of Botany and Plant Sciences, University of California, Riverside, California 92521-9800, United States
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, California 92521, United States
- Center for Plant Cell Biology, University of California, Riverside, Riverside, California 92521, United States
| | - Zenan Xing
- Department of Botany and Plant Sciences, University of California, Riverside, California 92521-9800, United States
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, California 92521, United States
- Center for Plant Cell Biology, University of California, Riverside, Riverside, California 92521, United States
| | - Sean R Cutler
- Department of Botany and Plant Sciences, University of California, Riverside, California 92521-9800, United States
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, California 92521, United States
- Center for Plant Cell Biology, University of California, Riverside, Riverside, California 92521, United States
| | - Joel L Kaar
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| | - Michael R Shirts
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| | - Timothy A Whitehead
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80305, United States
| |
Collapse
|
17
|
Listov D, Goverde CA, Correia BE, Fleishman SJ. Opportunities and challenges in design and optimization of protein function. Nat Rev Mol Cell Biol 2024; 25:639-653. [PMID: 38565617 PMCID: PMC7616297 DOI: 10.1038/s41580-024-00718-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
The field of protein design has made remarkable progress over the past decade. Historically, the low reliability of purely structure-based design methods limited their application, but recent strategies that combine structure-based and sequence-based calculations, as well as machine learning tools, have dramatically improved protein engineering and design. In this Review, we discuss how these methods have enabled the design of increasingly complex structures and therapeutically relevant activities. Additionally, protein optimization methods have improved the stability and activity of complex eukaryotic proteins. Thanks to their increased reliability, computational design methods have been applied to improve therapeutics and enzymes for green chemistry and have generated vaccine antigens, antivirals and drug-delivery nano-vehicles. Moreover, the high success of design methods reflects an increased understanding of basic rules that govern the relationships among protein sequence, structure and function. However, de novo design is still limited mostly to α-helix bundles, restricting its potential to generate sophisticated enzymes and diverse protein and small-molecule binders. Designing complex protein structures is a challenging but necessary next step if we are to realize our objective of generating new-to-nature activities.
Collapse
Affiliation(s)
- Dina Listov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Casper A Goverde
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bruno E Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Sarel Jacob Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
18
|
An L, Said M, Tran L, Majumder S, Goreshnik I, Lee GR, Juergens D, Dauparas J, Anishchenko I, Coventry B, Bera AK, Kang A, Levine PM, Alvarez V, Pillai A, Norn C, Feldman D, Zorine D, Hicks DR, Li X, Sanchez MG, Vafeados DK, Salveson PJ, Vorobieva AA, Baker D. Binding and sensing diverse small molecules using shape-complementary pseudocycles. Science 2024; 385:276-282. [PMID: 39024436 PMCID: PMC11542606 DOI: 10.1126/science.adn3780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/03/2024] [Indexed: 07/20/2024]
Abstract
We describe an approach for designing high-affinity small molecule-binding proteins poised for downstream sensing. We use deep learning-generated pseudocycles with repeating structural units surrounding central binding pockets with widely varying shapes that depend on the geometry and number of the repeat units. We dock small molecules of interest into the most shape complementary of these pseudocycles, design the interaction surfaces for high binding affinity, and experimentally screen to identify designs with the highest affinity. We obtain binders to four diverse molecules, including the polar and flexible methotrexate and thyroxine. Taking advantage of the modular repeat structure and central binding pockets, we construct chemically induced dimerization systems and low-noise nanopore sensors by splitting designs into domains that reassemble upon ligand addition.
Collapse
Affiliation(s)
- Linna An
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Meerit Said
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Long Tran
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Sagardip Majumder
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Gyu Rie Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Juergens
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Graduate Program in Molecular Engineering, University of Washington, Seattle, WA, USA
| | - Justas Dauparas
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Ivan Anishchenko
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Asim K. Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Paul M. Levine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Valentina Alvarez
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Arvind Pillai
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | | | - Dmitri Zorine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Derrick R. Hicks
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinting Li
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Dionne K. Vafeados
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Patrick J. Salveson
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Anastassia A. Vorobieva
- VIB-VUB Center for Structural Biology, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
19
|
Krishna R, Wang J, Ahern W, Sturmfels P, Venkatesh P, Kalvet I, Lee GR, Morey-Burrows FS, Anishchenko I, Humphreys IR, McHugh R, Vafeados D, Li X, Sutherland GA, Hitchcock A, Hunter CN, Kang A, Brackenbrough E, Bera AK, Baek M, DiMaio F, Baker D. Generalized biomolecular modeling and design with RoseTTAFold All-Atom. Science 2024; 384:eadl2528. [PMID: 38452047 DOI: 10.1126/science.adl2528] [Citation(s) in RCA: 182] [Impact Index Per Article: 182.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
Deep-learning methods have revolutionized protein structure prediction and design but are presently limited to protein-only systems. We describe RoseTTAFold All-Atom (RFAA), which combines a residue-based representation of amino acids and DNA bases with an atomic representation of all other groups to model assemblies that contain proteins, nucleic acids, small molecules, metals, and covalent modifications, given their sequences and chemical structures. By fine-tuning on denoising tasks, we developed RFdiffusion All-Atom (RFdiffusionAA), which builds protein structures around small molecules. Starting from random distributions of amino acid residues surrounding target small molecules, we designed and experimentally validated, through crystallography and binding measurements, proteins that bind the cardiac disease therapeutic digoxigenin, the enzymatic cofactor heme, and the light-harvesting molecule bilin.
Collapse
Affiliation(s)
- Rohith Krishna
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Jue Wang
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Woody Ahern
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA 98105, USA
| | - Pascal Sturmfels
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA 98105, USA
| | - Preetham Venkatesh
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
- Graduate Program in Biological Physics, Structure and Design, University of Washington, Seattle, WA 98105, USA
| | - Indrek Kalvet
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98105, USA
| | - Gyu Rie Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98105, USA
| | | | - Ivan Anishchenko
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Ian R Humphreys
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Ryan McHugh
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
- Graduate Program in Biological Physics, Structure and Design, University of Washington, Seattle, WA 98105, USA
| | - Dionne Vafeados
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Xinting Li
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | | | - Andrew Hitchcock
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - C Neil Hunter
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Alex Kang
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Evans Brackenbrough
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Asim K Bera
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - Minkyung Baek
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98105, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
20
|
Lu L, Gou X, Tan SK, Mann SI, Yang H, Zhong X, Gazgalis D, Valdiviezo J, Jo H, Wu Y, Diolaiti ME, Ashworth A, Polizzi NF, DeGrado WF. De novo design of drug-binding proteins with predictable binding energy and specificity. Science 2024; 384:106-112. [PMID: 38574125 PMCID: PMC11290694 DOI: 10.1126/science.adl5364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/28/2024] [Indexed: 04/06/2024]
Abstract
The de novo design of small molecule-binding proteins has seen exciting recent progress; however, high-affinity binding and tunable specificity typically require laborious screening and optimization after computational design. We developed a computational procedure to design a protein that recognizes a common pharmacophore in a series of poly(ADP-ribose) polymerase-1 inhibitors. One of three designed proteins bound different inhibitors with affinities ranging from <5 nM to low micromolar. X-ray crystal structures confirmed the accuracy of the designed protein-drug interactions. Molecular dynamics simulations informed the role of water in binding. Binding free energy calculations performed directly on the designed models were in excellent agreement with the experimentally measured affinities. We conclude that de novo design of high-affinity small molecule-binding proteins with tuned interaction energies is feasible entirely from computation.
Collapse
Affiliation(s)
- Lei Lu
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Xuxu Gou
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Sophia K Tan
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Samuel I Mann
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
- Department of Chemistry, University of California, Riverside, CA 92521, USA
| | - Hyunjun Yang
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Xiaofang Zhong
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA
| | - Dimitrios Gazgalis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Jesús Valdiviezo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Yibing Wu
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Morgan E Diolaiti
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Nicholas F Polizzi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - William F DeGrado
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
21
|
Xu Y, Hu X, Wang C, Liu Y, Chen Q, Liu H. De novo design of cavity-containing proteins with a backbone-centered neural network energy function. Structure 2024; 32:424-432.e4. [PMID: 38325370 DOI: 10.1016/j.str.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 10/04/2023] [Accepted: 01/11/2024] [Indexed: 02/09/2024]
Abstract
The design of small-molecule-binding proteins requires protein backbones that contain cavities. Previous design efforts were based on naturally occurring cavity-containing backbone architectures. Here, we designed diverse cavity-containing backbones without predefined architectures by introducing tailored restraints into the backbone sampling driven by SCUBA (Side Chain-Unknown Backbone Arrangement), a neural network statistical energy function. For 521 out of 5816 designs, the root-mean-square deviations (RMSDs) of the Cα atoms for the AlphaFold2-predicted structures and our designed structures are within 2.0 Å. We experimentally tested 10 designed proteins and determined the crystal structures of two of them. One closely agrees with the designed model, while the other forms a domain-swapped dimer, where the partial structures are in agreement with the designed structures. Our results indicate that data-driven methods such as SCUBA hold great potential for designing de novo proteins with tailored small-molecule-binding function.
Collapse
Affiliation(s)
- Yang Xu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Centre for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiuhong Hu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Centre for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Chenchen Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yongrui Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Quan Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Centre for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Haiyan Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui 230027, China; School of Data Science, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
22
|
Leonard AC, Friedman AJ, Chayer R, Petersen BM, Kaar J, Shirts MR, Whitehead TA. Rationalizing diverse binding mechanisms to the same protein fold: insights for ligand recognition and biosensor design. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.586677. [PMID: 38586024 PMCID: PMC10996623 DOI: 10.1101/2024.03.25.586677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The engineering of novel protein-ligand binding interactions, particularly for complex drug-like molecules, is an unsolved problem which could enable many practical applications of protein biosensors. In this work, we analyzed two engineer ed biosensors, derived from the plant hormone sensor PYR1, to recognize either the agrochemical mandipropamid or the synthetic cannabinoid WIN55,212-2. Using a combination of quantitative deep mutational scanning experiments and molecular dynamics simulations, we demonstrated that mutations at common positions can promote protein-ligand shape complementarity and revealed prominent differences in the electrostatic networks needed to complement diverse ligands. MD simulations indicate that both PYR1 protein-ligand complexes bind a single conformer of their target ligand that is close to the lowest free energy conformer. Computational design using a fixed conformer and rigid body orientation led to new WIN55,212-2 sensors with nanomolar limits of detection. This work reveals mechanisms by which the versatile PYR1 biosensor scaffold can bind diverse ligands. This work also provides computational methods to sample realistic ligand conformers and rigid body alignments that simplify the computational design of biosensors for novel ligands of interest.
Collapse
|
23
|
Zhang K, Tang Y, Yu H, Yang J, Tao L, Xiang P. Discovery of lupus nephritis targeted inhibitors based on De novo molecular design: comprehensive application of vinardo scoring, ADMET analysis, and molecular dynamics simulation. J Biomol Struct Dyn 2024:1-14. [PMID: 38501728 DOI: 10.1080/07391102.2024.2329293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
Lupus Nephritis (LN) is an autoimmune disease affecting the kidneys, and conventional drug studies have limitations due to its imprecise and complex pathogenesis. Therefore, the aim of this study was to design a novel Lupus Nephritis-targeted drug with good clinical due potential, high potency and selectivity by computer-assisted approach.NIK belongs to the serine/threonine protein kinase, which is gaining attention as a drug target for Lupus Nephritis. we used bioinformatics, homology modelling and sequence comparison analysis, small molecule ab initio design, ADMET analysis, molecular docking, molecular dynamics simulation, and MM/PBSA analysis to design and explore the selectivity and efficiency of a novel Lupus Nephritis-targeting drug, ClImYnib, and a classical NIK inhibitor, NIK SMI1. We used bioinformatics techniques to determine the correlation between lupus nephritis and the NF-κB signaling pathway. De novo drugs design was used to create a NIK-targeted inhibitor, ClImYnib, with lower toxicity, after which we used molecular dynamics to simulate NIK SMI1 against ClImYnib, and the simulation results showed that ClImYnib had better selectivity and efficiency. Our research delves into the molecular mechanism of protein ligands, and we have designed and validated an excellent NIK inhibitor using multiple computational simulation methods. More importantly, it provides an idea of target designing small molecules.
Collapse
Affiliation(s)
- Kaiyuan Zhang
- School of Clinical Medicine, Bengbu Medical College, China
| | - Yingkai Tang
- Department of Anatomy, School of basic Medicine, Bengbu Medical College, China
| | - Haiyue Yu
- School of Clinical Medicine, Bengbu Medical College, China
| | - Jingtao Yang
- School of Clinical Medicine, Bengbu Medical College, China
| | - Lu Tao
- Central Laboratory, The Frist Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Ping Xiang
- Central Laboratory, The Frist Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
24
|
Corso G, Deng A, Fry B, Polizzi N, Barzilay R, Jaakkola T. Deep Confident Steps to New Pockets: Strategies for Docking Generalization. ARXIV 2024:arXiv:2402.18396v1. [PMID: 38463508 PMCID: PMC10925391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Accurate blind docking has the potential to lead to new biological breakthroughs, but for this promise to be realized, docking methods must generalize well across the proteome. Existing benchmarks, however, fail to rigorously assess generalizability. Therefore, we develop DockGen, a new benchmark based on the ligand-binding domains of proteins, and we show that existing machine learning-based docking models have very weak generalization abilities. We carefully analyze the scaling laws of ML-based docking and show that, by scaling data and model size, as well as integrating synthetic data strategies, we are able to significantly increase the generalization capacity and set new state-of-the-art performance across benchmarks. Further, we propose Confidence Bootstrapping, a new training paradigm that solely relies on the interaction between diffusion and confidence models and exploits the multi-resolution generation process of diffusion models. We demonstrate that Confidence Bootstrapping significantly improves the ability of ML-based docking methods to dock to unseen protein classes, edging closer to accurate and generalizable blind docking methods.
Collapse
Affiliation(s)
| | | | - Benjamin Fry
- Dana-Farber Cancer Institute and Harvard Medical School
| | | | | | | |
Collapse
|
25
|
Zhao C, Li X, Han X, Li Z, Bian S, Zeng W, Ding M, Liang J, Jiang Q, Zhou Z, Fan Y, Zhang X, Sun Y. Molecular co-assembled strategy tuning protein conformation for cartilage regeneration. Nat Commun 2024; 15:1488. [PMID: 38374253 PMCID: PMC10876949 DOI: 10.1038/s41467-024-45703-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
The assembly of oligopeptide and polypeptide molecules can reconstruct various ordered advanced structures through intermolecular interactions to achieve protein-like biofunction. Here, we develop a "molecular velcro"-inspired peptide and gelatin co-assembly strategy, in which amphiphilic supramolecular tripeptides are attached to the molecular chain of gelatin methacryloyl via intra-/intermolecular interactions. We perform molecular docking and dynamics simulations to demonstrate the feasibility of this strategy and reveal the advanced structural transition of the co-assembled hydrogel, which brings more ordered β-sheet content and 10-fold or more compressive strength improvement. We conduct transcriptome analysis to reveal the role of co-assembled hydrogel in promoting cell proliferation and chondrogenic differentiation. Subcutaneous implantation evaluation confirms considerably reduced inflammatory responses and immunogenicity in comparison with type I collagen. We demonstrate that bone mesenchymal stem cells-laden co-assembled hydrogel can be stably fixed in rabbit knee joint defects by photocuring, which significantly facilitates hyaline cartilage regeneration after three months. This co-assembly strategy provides an approach for developing cartilage regenerative biomaterials.
Collapse
Affiliation(s)
- Chengkun Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Xing Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Xiaowen Han
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, Sichuan, 621099, P. R. China
| | - Zhulian Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Shaoquan Bian
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Weinan Zeng
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu, 610064, P. R. China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Zongke Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, P. R. China.
| |
Collapse
|
26
|
Chu AE, Lu T, Huang PS. Sparks of function by de novo protein design. Nat Biotechnol 2024; 42:203-215. [PMID: 38361073 PMCID: PMC11366440 DOI: 10.1038/s41587-024-02133-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
Information in proteins flows from sequence to structure to function, with each step causally driven by the preceding one. Protein design is founded on inverting this process: specify a desired function, design a structure executing this function, and find a sequence that folds into this structure. This 'central dogma' underlies nearly all de novo protein-design efforts. Our ability to accomplish these tasks depends on our understanding of protein folding and function and our ability to capture this understanding in computational methods. In recent years, deep learning-derived approaches for efficient and accurate structure modeling and enrichment of successful designs have enabled progression beyond the design of protein structures and towards the design of functional proteins. We examine these advances in the broader context of classical de novo protein design and consider implications for future challenges to come, including fundamental capabilities such as sequence and structure co-design and conformational control considering flexibility, and functional objectives such as antibody and enzyme design.
Collapse
Affiliation(s)
- Alexander E Chu
- Biophysics Program, Stanford University, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
- Google DeepMind, London, UK
| | - Tianyu Lu
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | - Po-Ssu Huang
- Biophysics Program, Stanford University, Palo Alto, CA, USA.
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
27
|
Kortemme T. De novo protein design-From new structures to programmable functions. Cell 2024; 187:526-544. [PMID: 38306980 PMCID: PMC10990048 DOI: 10.1016/j.cell.2023.12.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/03/2023] [Accepted: 12/19/2023] [Indexed: 02/04/2024]
Abstract
Methods from artificial intelligence (AI) trained on large datasets of sequences and structures can now "write" proteins with new shapes and molecular functions de novo, without starting from proteins found in nature. In this Perspective, I will discuss the state of the field of de novo protein design at the juncture of physics-based modeling approaches and AI. New protein folds and higher-order assemblies can be designed with considerable experimental success rates, and difficult problems requiring tunable control over protein conformations and precise shape complementarity for molecular recognition are coming into reach. Emerging approaches incorporate engineering principles-tunability, controllability, and modularity-into the design process from the beginning. Exciting frontiers lie in deconstructing cellular functions with de novo proteins and, conversely, constructing synthetic cellular signaling from the ground up. As methods improve, many more challenges are unsolved.
Collapse
Affiliation(s)
- Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
28
|
Liu Y, Liu H. Protein sequence design on given backbones with deep learning. Protein Eng Des Sel 2024; 37:gzad024. [PMID: 38157313 DOI: 10.1093/protein/gzad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/08/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
Deep learning methods for protein sequence design focus on modeling and sampling the many- dimensional distribution of amino acid sequences conditioned on the backbone structure. To produce physically foldable sequences, inter-residue couplings need to be considered properly. These couplings are treated explicitly in iterative methods or autoregressive methods. Non-autoregressive models treating these couplings implicitly are computationally more efficient, but still await tests by wet experiment. Currently, sequence design methods are evaluated mainly using native sequence recovery rate and native sequence perplexity. These metrics can be complemented by sequence-structure compatibility metrics obtained from energy calculation or structure prediction. However, existing computational metrics have important limitations that may render the generalization of computational test results to performance in real applications unwarranted. Validation of design methods by wet experiments should be encouraged.
Collapse
Affiliation(s)
- Yufeng Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haiyan Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui 230027, China
- School of Biomedical Engineering, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu 215004, China
| |
Collapse
|
29
|
Lu L, Gou X, Tan SK, Mann SI, Yang H, Zhong X, Gazgalis D, Valdiviezo J, Jo H, Wu Y, Diolaiti ME, Ashworth A, Polizzi NF, DeGrado WF. De novo design of drug-binding proteins with predictable binding energy and specificity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.23.573178. [PMID: 38187746 PMCID: PMC10769398 DOI: 10.1101/2023.12.23.573178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The de novo design of small-molecule-binding proteins has seen exciting recent progress; however, the ability to achieve exquisite affinity for binding small molecules while tuning specificity has not yet been demonstrated directly from computation. Here, we develop a computational procedure that results in the highest affinity binders to date with predetermined relative affinities, targeting a series of PARP1 inhibitors. Two of four designed proteins bound with affinities ranging from < 5 nM to low μM, in a predictable manner. X-ray crystal structures confirmed the accuracy of the designed protein-drug interactions. Molecular dynamics simulations informed the role of water in binding. Binding free-energy calculations performed directly on the designed models are in excellent agreement with the experimentally measured affinities, suggesting that the de novo design of small-molecule-binding proteins with tuned interaction energies is now feasible entirely from computation. We expect these methods to open many opportunities in biomedicine, including rapid sensor development, antidote design, and drug delivery vehicles.
Collapse
Affiliation(s)
- Lei Lu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Xuxu Gou
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Sophia K Tan
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Samuel I. Mann
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hyunjun Yang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Dimitrios Gazgalis
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jesús Valdiviezo
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yibing Wu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Morgan E. Diolaiti
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | | | - William F. DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
30
|
An L, Said M, Tran L, Majumder S, Goreshnik I, Lee GR, Juergens D, Dauparas J, Anishchenko I, Coventry B, Bera AK, Kang A, Levine PM, Alvarez V, Pillai A, Norn C, Feldman D, Zorine D, Hicks DR, Li X, Sanchez MG, Vafeados DK, Salveson PJ, Vorobieva AA, Baker D. De novo design of diverse small molecule binders and sensors using Shape Complementary Pseudocycles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572602. [PMID: 38187589 PMCID: PMC10769206 DOI: 10.1101/2023.12.20.572602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
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.
Collapse
Affiliation(s)
- Linna An
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Meerit Said
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Long Tran
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Sagardip Majumder
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Gyu Rie Lee
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Juergens
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justas Dauparas
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Ivan Anishchenko
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Asim K. Bera
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Paul M. Levine
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Valentina Alvarez
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Arvind Pillai
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - David Feldman
- BioInnovation Institute, DK2200 Copenhagen N, Denmark
| | - Dmitri Zorine
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Derrick R. Hicks
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinting Li
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Dionne K. Vafeados
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Patrick J. Salveson
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - David Baker
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
31
|
Lee GR, Pellock SJ, Norn C, Tischer D, Dauparas J, Anischenko I, Mercer JAM, Kang A, Bera A, Nguyen H, Goreshnik I, Vafeados D, Roullier N, Han HL, Coventry B, Haddox HK, Liu DR, Yeh AHW, Baker D. Small-molecule binding and sensing with a designed protein family. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.565201. [PMID: 37961294 PMCID: PMC10635051 DOI: 10.1101/2023.11.01.565201] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Despite transformative advances in protein design with deep learning, the design of small-molecule-binding proteins and sensors for arbitrary ligands remains a grand challenge. Here we combine deep learning and physics-based methods to generate a family of proteins with diverse and designable pocket geometries, which we employ to computationally design binders for six chemically and structurally distinct small-molecule targets. Biophysical characterization of the designed binders revealed nanomolar to low micromolar binding affinities and atomic-level design accuracy. The bound ligands are exposed at one edge of the binding pocket, enabling the de novo design of chemically induced dimerization (CID) systems; we take advantage of this to create a biosensor with nanomolar sensitivity for cortisol. Our approach provides a general method to design proteins that bind and sense small molecules for a wide range of analytical, environmental, and biomedical applications.
Collapse
|
32
|
An L, Hicks DR, Zorine D, Dauparas J, Wicky BIM, Milles LF, Courbet A, Bera AK, Nguyen H, Kang A, Carter L, Baker D. Hallucination of closed repeat proteins containing central pockets. Nat Struct Mol Biol 2023; 30:1755-1760. [PMID: 37770718 PMCID: PMC10643118 DOI: 10.1038/s41594-023-01112-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/28/2023] [Indexed: 09/30/2023]
Abstract
In pseudocyclic proteins, such as TIM barrels, β barrels, and some helical transmembrane channels, a single subunit is repeated in a cyclic pattern, giving rise to a central cavity that can serve as a pocket for ligand binding or enzymatic activity. Inspired by these proteins, we devised a deep-learning-based approach to broadly exploring the space of closed repeat proteins starting from only a specification of the repeat number and length. Biophysical data for 38 structurally diverse pseudocyclic designs produced in Escherichia coli are consistent with the design models, and the three crystal structures we were able to obtain are very close to the designed structures. Docking studies suggest the diversity of folds and central pockets provide effective starting points for designing small-molecule binders and enzymes.
Collapse
Affiliation(s)
- Linna An
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| | - Derrick R Hicks
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Dmitri Zorine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justas Dauparas
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Basile I M Wicky
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lukas F Milles
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alexis Courbet
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Asim K Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Hannah Nguyen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
33
|
Hutchins GH, Noble CEM, Bunzel HA, Williams C, Dubiel P, Yadav SKN, Molinaro PM, Barringer R, Blackburn H, Hardy BJ, Parnell AE, Landau C, Race PR, Oliver TAA, Koder RL, Crump MP, Schaffitzel C, Oliveira ASF, Mulholland AJ, Anderson JLR. An expandable, modular de novo protein platform for precision redox engineering. Proc Natl Acad Sci U S A 2023; 120:e2306046120. [PMID: 37487099 PMCID: PMC10400981 DOI: 10.1073/pnas.2306046120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
The electron-conducting circuitry of life represents an as-yet untapped resource of exquisite, nanoscale biomolecular engineering. Here, we report the characterization and structure of a de novo diheme "maquette" protein, 4D2, which we subsequently use to create an expanded, modular platform for heme protein design. A well-folded monoheme variant was created by computational redesign, which was then utilized for the experimental validation of continuum electrostatic redox potential calculations. This demonstrates how fundamental biophysical properties can be predicted and fine-tuned. 4D2 was then extended into a tetraheme helical bundle, representing a 7 nm molecular wire. Despite a molecular weight of only 24 kDa, electron cryomicroscopy illustrated a remarkable level of detail, indicating the positioning of the secondary structure and the heme cofactors. This robust, expressible, highly thermostable and readily designable modular platform presents a valuable resource for redox protein design and the future construction of artificial electron-conducting circuitry.
Collapse
Affiliation(s)
- George H. Hutchins
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - Claire E. M. Noble
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
- BrisSynBio Synthetic Biology Research Centre, Life Sciences Building, University of Bristol, BristolBS8 1TQ, United Kingdom
| | - H. Adrian Bunzel
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | | | - Paulina Dubiel
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - Sathish K. N. Yadav
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - Paul M. Molinaro
- Department of Physics, The City College of New York, New York, NY10031
- Graduate Programs of Physics, Biology, Chemistry and Biochemistry, The Graduate Center of The City University of New York, New York, NY10016
| | - Rob Barringer
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - Hector Blackburn
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - Benjamin J. Hardy
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - Alice E. Parnell
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
- BrisSynBio Synthetic Biology Research Centre, Life Sciences Building, University of Bristol, BristolBS8 1TQ, United Kingdom
| | - Charles Landau
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - Paul R. Race
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
- BrisSynBio Synthetic Biology Research Centre, Life Sciences Building, University of Bristol, BristolBS8 1TQ, United Kingdom
| | | | - Ronald L. Koder
- Department of Physics, The City College of New York, New York, NY10031
- Graduate Programs of Physics, Biology, Chemistry and Biochemistry, The Graduate Center of The City University of New York, New York, NY10016
| | - Matthew P. Crump
- School of Chemistry, University of Bristol, BristolBS8 1TS, United Kingdom
| | - Christiane Schaffitzel
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
| | - A. Sofia F. Oliveira
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
- School of Chemistry, University of Bristol, BristolBS8 1TS, United Kingdom
| | - Adrian J. Mulholland
- BrisSynBio Synthetic Biology Research Centre, Life Sciences Building, University of Bristol, BristolBS8 1TQ, United Kingdom
- School of Chemistry, University of Bristol, BristolBS8 1TS, United Kingdom
| | - J. L. Ross Anderson
- School of Biochemistry, University of Bristol, University Walk, BristolBS8 1TD, United Kingdom
- BrisSynBio Synthetic Biology Research Centre, Life Sciences Building, University of Bristol, BristolBS8 1TQ, United Kingdom
| |
Collapse
|
34
|
Kratochvil HT, Watkins LC, Mravic M, Thomaston JL, Nicoludis JM, Somberg NH, Liu L, Hong M, Voth GA, DeGrado WF. Transient water wires mediate selective proton transport in designed channel proteins. Nat Chem 2023; 15:1012-1021. [PMID: 37308712 PMCID: PMC10475958 DOI: 10.1038/s41557-023-01210-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 04/19/2023] [Indexed: 06/14/2023]
Abstract
Selective proton transport through proteins is essential for forming and using proton gradients in cells. Protons are conducted along hydrogen-bonded 'wires' of water molecules and polar side chains, which, somewhat surprisingly, are often interrupted by dry apolar stretches in the conduction pathways, inferred from static protein structures. Here we hypothesize that protons are conducted through such dry spots by forming transient water wires, often highly correlated with the presence of the excess protons in the water wire. To test this hypothesis, we performed molecular dynamics simulations to design transmembrane channels with stable water pockets interspersed by apolar segments capable of forming flickering water wires. The minimalist designed channels conduct protons at rates similar to viral proton channels, and they are at least 106-fold more selective for H+ over Na+. These studies inform the mechanisms of biological proton conduction and the principles for engineering proton-conductive materials.
Collapse
Affiliation(s)
- Huong T Kratochvil
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA.
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Laura C Watkins
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics and James Franck Institute, The University of Chicago, Chicago, IL, USA
- Kemper Insurance, Chicago, IL, USA
| | - Marco Mravic
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
- Department of Integrative Structural and Computational Biology Scripps Research Institute, La Jolla, CA, USA
| | - Jessica L Thomaston
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
| | - John M Nicoludis
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA
- Genentech, San Francisco, CA, USA
| | - Noah H Somberg
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gregory A Voth
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics and James Franck Institute, The University of Chicago, Chicago, IL, USA.
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
35
|
Smith A, Naudin EA, Edgell CL, Baker EG, Mylemans B, FitzPatrick L, Herman A, Rice HM, Andrews DM, Tigue N, Woolfson DN, Savery NJ. Design and Selection of Heterodimerizing Helical Hairpins for Synthetic Biology. ACS Synth Biol 2023; 12:1845-1858. [PMID: 37224449 PMCID: PMC10278171 DOI: 10.1021/acssynbio.3c00231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Indexed: 05/26/2023]
Abstract
Synthetic biology applications would benefit from protein modules of reduced complexity that function orthogonally to cellular components. As many subcellular processes depend on peptide-protein or protein-protein interactions, de novo designed polypeptides that can bring together other proteins controllably are particularly useful. Thanks to established sequence-to-structure relationships, helical bundles provide good starting points for such designs. Typically, however, such designs are tested in vitro and function in cells is not guaranteed. Here, we describe the design, characterization, and application of de novo helical hairpins that heterodimerize to form 4-helix bundles in cells. Starting from a rationally designed homodimer, we construct a library of helical hairpins and identify complementary pairs using bimolecular fluorescence complementation in E. coli. We characterize some of the pairs using biophysics and X-ray crystallography to confirm heterodimeric 4-helix bundles. Finally, we demonstrate the function of an exemplar pair in regulating transcription in both E. coli and mammalian cells.
Collapse
Affiliation(s)
- Abigail
J. Smith
- School
of Biochemistry, University of Bristol, Bristol BS8 1TD, U.K.
| | - Elise A. Naudin
- School
of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.
| | - Caitlin L. Edgell
- School
of Biochemistry, University of Bristol, Bristol BS8 1TD, U.K.
- School
of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.
| | - Emily G. Baker
- School
of Biochemistry, University of Bristol, Bristol BS8 1TD, U.K.
- School
of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.
| | - Bram Mylemans
- School
of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.
| | | | - Andrew Herman
- Flow
Cytometry Facility, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, U.K.
| | - Helen M. Rice
- Flow
Cytometry Facility, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, U.K.
| | | | - Natalie Tigue
- BioPharmaceuticals
R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Derek N. Woolfson
- School
of Biochemistry, University of Bristol, Bristol BS8 1TD, U.K.
- School
of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.
| | - Nigel J. Savery
- School
of Biochemistry, University of Bristol, Bristol BS8 1TD, U.K.
- BrisEngBio,
School of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.
| |
Collapse
|
36
|
Di Rienzo L, Miotto M, Milanetti E, Ruocco G. Computational structural-based GPCR optimization for user-defined ligand: Implications for the development of biosensors. Comput Struct Biotechnol J 2023; 21:3002-3009. [PMID: 37249971 PMCID: PMC10220229 DOI: 10.1016/j.csbj.2023.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Organisms have developed effective mechanisms to sense the external environment. Human-designed biosensors exploit this natural optimization, where different biological machinery have been adapted to detect the presence of user-defined molecules. Specifically, the pheromone pathway in the model organism Saccharomyces cerevisiae represents a suitable candidate as a synthetic signaling system. Indeed, it expresses just one G-Protein Coupled Receptor (GPCR), Ste2, able to recognize pheromone and initiate the expression of pheromone-dependent genes. To date, the standard procedure to engineer this system relies on the substitution of the yeast GPCR with another one and on the modification of the yeast G-protein to bind the inserted receptor. Here, we propose an innovative computational procedure, based on geometrical and chemical optimization of protein binding pockets, to select the amino acid substitutions required to make the native yeast GPCR able to recognize a user-defined ligand. This procedure would allow the yeast to recognize a wide range of ligands, without a-priori knowledge about a GPCR recognizing them or the corresponding G protein. We used Monte Carlo simulations to design on Ste2 a binding pocket able to recognize epinephrine, selected as a test ligand. We validated Ste2 mutants via molecular docking and molecular dynamics. We verified that the amino acid substitutions we identified make Ste2 able to accommodate and remain firmly bound to epinephrine. Our results indicate that we sampled efficiently the huge space of possible mutants, proposing such a strategy as a promising starting point for the development of a new kind of S.cerevisiae-based biosensors.
Collapse
Affiliation(s)
- Lorenzo Di Rienzo
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Mattia Miotto
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Edoardo Milanetti
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
37
|
Kynast JP, Höcker B. Atligator Web: A Graphical User Interface for Analysis and Design of Protein-Peptide Interactions. BIODESIGN RESEARCH 2023; 5:0011. [PMID: 37849459 PMCID: PMC10521702 DOI: 10.34133/bdr.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/14/2023] [Indexed: 10/19/2023] Open
Abstract
A key functionality of proteins is based on their ability to form interactions with other proteins or peptides. These interactions are neither easily described nor fully understood, which is why the design of specific protein-protein interaction interfaces remains a challenge for protein engineering. We recently developed the software ATLIGATOR to extract common interaction patterns between different types of amino acids and store them in a database. The tool enables the user to better understand frequent interaction patterns and find groups of interactions. Furthermore, frequent motifs can be directly transferred from the database to a user-defined scaffold as a starting point for the engineering of new binding capabilities. Since three-dimensional visualization is a crucial part of ATLIGATOR, we created ATLIGATOR web-a web server offering an intuitive graphical user interface (GUI) available at https://atligator.uni-bayreuth.de. This new interface empowers users to apply ATLIGATOR by providing easy access with having all parts directly connected. Moreover, we extended the web by a design functionality so that, overall, ATLIGATOR web facilitates the use of ATLIGATOR with a more intuitive UI and advanced design options.
Collapse
Affiliation(s)
- Josef Paul Kynast
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Birte Höcker
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| |
Collapse
|
38
|
Tellechea-Luzardo J, Stiebritz MT, Carbonell P. Transcription factor-based biosensors for screening and dynamic regulation. Front Bioeng Biotechnol 2023; 11:1118702. [PMID: 36814719 PMCID: PMC9939652 DOI: 10.3389/fbioe.2023.1118702] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
Advances in synthetic biology and genetic engineering are bringing into the spotlight a wide range of bio-based applications that demand better sensing and control of biological behaviours. Transcription factor (TF)-based biosensors are promising tools that can be used to detect several types of chemical compounds and elicit a response according to the desired application. However, the wider use of this type of device is still hindered by several challenges, which can be addressed by increasing the current metabolite-activated transcription factor knowledge base, developing better methods to identify new transcription factors, and improving the overall workflow for the design of novel biosensor circuits. These improvements are particularly important in the bioproduction field, where researchers need better biosensor-based approaches for screening production-strains and precise dynamic regulation strategies. In this work, we summarize what is currently known about transcription factor-based biosensors, discuss recent experimental and computational approaches targeted at their modification and improvement, and suggest possible future research directions based on two applications: bioproduction screening and dynamic regulation of genetic circuits.
Collapse
Affiliation(s)
- Jonathan Tellechea-Luzardo
- Institute of Industrial Control Systems and Computing (AI2), Universitat Politècnica de València (UPV), Valencia, Spain
| | - Martin T. Stiebritz
- Institute of Industrial Control Systems and Computing (AI2), Universitat Politècnica de València (UPV), Valencia, Spain
| | - Pablo Carbonell
- Institute of Industrial Control Systems and Computing (AI2), Universitat Politècnica de València (UPV), Valencia, Spain
- Institute for Integrative Systems Biology I2SysBio, Universitat de València-CSIC, Paterna, Spain
| |
Collapse
|
39
|
Beltrán J, Steiner PJ, Bedewitz M, Wei S, Peterson FC, Li Z, Hughes BE, Hartley Z, Robertson NR, Medina-Cucurella AV, Baumer ZT, Leonard AC, Park SY, Volkman BF, Nusinow DA, Zhong W, Wheeldon I, Cutler SR, Whitehead TA. Rapid biosensor development using plant hormone receptors as reprogrammable scaffolds. Nat Biotechnol 2022; 40:1855-1861. [PMID: 35726092 PMCID: PMC9750858 DOI: 10.1038/s41587-022-01364-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 05/17/2022] [Indexed: 01/14/2023]
Abstract
A general method to generate biosensors for user-defined molecules could provide detection tools for a wide range of biological applications. Here, we describe an approach for the rapid engineering of biosensors using PYR1 (Pyrabactin Resistance 1), a plant abscisic acid (ABA) receptor with a malleable ligand-binding pocket and a requirement for ligand-induced heterodimerization, which facilitates the construction of sense-response functions. We applied this platform to evolve 21 sensors with nanomolar to micromolar sensitivities for a range of small molecules, including structurally diverse natural and synthetic cannabinoids and several organophosphates. X-ray crystallography analysis revealed the mechanistic basis for new ligand recognition by an evolved cannabinoid receptor. We demonstrate that PYR1-derived receptors are readily ported to various ligand-responsive outputs, including enzyme-linked immunosorbent assay (ELISA)-like assays, luminescence by protein-fragment complementation and transcriptional circuits, all with picomolar to nanomolar sensitivity. PYR1 provides a scaffold for rapidly evolving new biosensors for diverse sense-response applications.
Collapse
Affiliation(s)
- Jesús Beltrán
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA
| | - Paul J Steiner
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Matthew Bedewitz
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Shuang Wei
- Department of Biochemistry, University of California, Riverside, Riverside, CA, USA
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zongbo Li
- Department of Chemistry, University of California, Riverside, Riverside, CA, USA
| | - Brigid E Hughes
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zachary Hartley
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA
| | - Nicholas R Robertson
- Department of Bioengineering, University of California, Riverside, Riverside, USA
| | | | - Zachary T Baumer
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Alison C Leonard
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Sang-Youl Park
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Wenwan Zhong
- Department of Chemistry, University of California, Riverside, Riverside, CA, USA
| | - Ian Wheeldon
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA.
- Department of Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA, USA.
| | - Sean R Cutler
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA.
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA.
- Center for Plant Cell Biology, University of California, Riverside, Riverside, CA, USA.
| | - Timothy A Whitehead
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
40
|
Heide F, Stetefeld J. A Structural Analysis of Proteinaceous Nanotube Cavities and Their Applications in Nanotechnology. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4080. [PMID: 36432365 PMCID: PMC9698212 DOI: 10.3390/nano12224080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 06/16/2023]
Abstract
Protein nanotubes offer unique properties to the materials science field that allow them to fulfill various functions in drug delivery, biosensors and energy storage. Protein nanotubes are chemically diverse, modular, biodegradable and nontoxic. Furthermore, although the initial design or repurposing of such nanotubes is highly complex, the field has matured to understand underlying chemical and physical properties to a point where applications are successfully being developed. An important feature of a nanotube is its ability to bind ligands via its internal cavities. As ligands of interest vary in size, shape and chemical properties, cavities have to be able to accommodate very specific features. As such, understanding cavities on a structural level is essential for their effective application. The objective of this review is to present the chemical and physical diversity of protein nanotube cavities and highlight their potential applications in materials science, specifically in biotechnology.
Collapse
Affiliation(s)
- Fabian Heide
- Correspondence: (F.H.); (J.S.); Tel.: +1-(204)-332-0853 (F.H.); +1-(204)-474-9731 (J.S.)
| | - Jörg Stetefeld
- Correspondence: (F.H.); (J.S.); Tel.: +1-(204)-332-0853 (F.H.); +1-(204)-474-9731 (J.S.)
| |
Collapse
|
41
|
Kynast JP, Schwägerl F, Höcker B. ATLIGATOR: editing protein interactions with an atlas-based approach. Bioinformatics 2022; 38:5199-5205. [PMID: 36259946 PMCID: PMC9710554 DOI: 10.1093/bioinformatics/btac685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/24/2022] [Accepted: 10/17/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Recognition of specific molecules by proteins is a fundamental cellular mechanism and relevant for many applications. Being able to modify binding is a key interest and can be achieved by repurposing established interaction motifs. We were specifically interested in a methodology for the design of peptide binding modules. By leveraging interaction data from known protein structures, we plan to accelerate the design of novel protein or peptide binders. RESULTS We developed ATLIGATOR-a computational method to support the analysis and design of a protein's interaction with a single side chain. Our program enables the building of interaction atlases based on structures from the PDB. From these atlases pocket definitions are extracted that can be searched for frequent interactions. These searches can reveal similarities in unrelated proteins as we show here for one example. Such frequent interactions can then be grafted onto a new protein scaffold as a starting point of the design process. The ATLIGATOR tool is made accessible through a python API as well as a CLI with python scripts. AVAILABILITY AND IMPLEMENTATION Source code can be downloaded at github (https://www.github.com/Hoecker-Lab/atligator), installed from PyPI ('atligator') and is implemented in Python 3.
Collapse
Affiliation(s)
- Josef Paul Kynast
- Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany
| | - Felix Schwägerl
- Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany
| | | |
Collapse
|
42
|
Naudin EA, Albanese KI, Smith AJ, Mylemans B, Baker EG, Weiner OD, Andrews DM, Tigue N, Savery NJ, Woolfson DN. From peptides to proteins: coiled-coil tetramers to single-chain 4-helix bundles. Chem Sci 2022; 13:11330-11340. [PMID: 36320580 PMCID: PMC9533478 DOI: 10.1039/d2sc04479j] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
The design of completely synthetic proteins from first principles-de novo protein design-is challenging. This is because, despite recent advances in computational protein-structure prediction and design, we do not understand fully the sequence-to-structure relationships for protein folding, assembly, and stabilization. Antiparallel 4-helix bundles are amongst the most studied scaffolds for de novo protein design. We set out to re-examine this target, and to determine clear sequence-to-structure relationships, or design rules, for the structure. Our aim was to determine a common and robust sequence background for designing multiple de novo 4-helix bundles. In turn, this could be used in chemical and synthetic biology to direct protein-protein interactions and as scaffolds for functional protein design. Our approach starts by analyzing known antiparallel 4-helix coiled-coil structures to deduce design rules. In terms of the heptad repeat, abcdefg -i.e., the sequence signature of many helical bundles-the key features that we identify are: a = Leu, d = Ile, e = Ala, g = Gln, and the use of complementary charged residues at b and c. Next, we implement these rules in the rational design of synthetic peptides to form antiparallel homo- and heterotetramers. Finally, we use the sequence of the homotetramer to derive in one step a single-chain 4-helix-bundle protein for recombinant production in E. coli. All of the assembled designs are confirmed in aqueous solution using biophysical methods, and ultimately by determining high-resolution X-ray crystal structures. Our route from peptides to proteins provides an understanding of the role of each residue in each design.
Collapse
Affiliation(s)
- Elise A Naudin
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Katherine I Albanese
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Abigail J Smith
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
| | - Bram Mylemans
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Emily G Baker
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
| | - Orion D Weiner
- Cardiovascular Research Institute, Department of Biochemistry and Biophysics, University of California 555 Mission Bay Blvd. South San Francisco CA 94158 USA
| | - David M Andrews
- Oncology R&D, AstraZeneca Cambridge Science Park, Darwin Building Cambridge CB4 0WG UK
| | - Natalie Tigue
- BioPharmaceuticals R&D, AstraZeneca Granta Park Cambridge CB21 6GH UK
| | - Nigel J Savery
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
- BrisEngBio, School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Derek N Woolfson
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol Cantock's Close Bristol BS8 1TS UK
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
- BrisEngBio, School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| |
Collapse
|
43
|
Liu X, Liu P, Li H, Xu Z, Jia L, Xia Y, Yu M, Tang W, Zhu X, Chen C, Zhang Y, Nango E, Tanaka R, Luo F, Kato K, Nakajima Y, Kishi S, Yu H, Matsubara N, Owada S, Tono K, Iwata S, Yu LJ, Shen JR, Wang J. Excited-state intermediates in a designer protein encoding a phototrigger caught by an X-ray free-electron laser. Nat Chem 2022; 14:1054-1060. [DOI: 10.1038/s41557-022-00992-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/01/2022] [Indexed: 11/09/2022]
|
44
|
McCann JJ, Pike DH, Brown MC, Crouse DT, Nanda V, Koder RL. Computational design of a sensitive, selective phase-changing sensor protein for the VX nerve agent. SCIENCE ADVANCES 2022; 8:eabh3421. [PMID: 35857443 PMCID: PMC9258810 DOI: 10.1126/sciadv.abh3421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
The VX nerve agent is one of the deadliest chemical warfare agents. Specific, sensitive, real-time detection methods for this neurotoxin have not been reported. The creation of proteins that use biological recognition to fulfill these requirements using directed evolution or library screening methods has been hampered because its toxicity makes laboratory experimentation extraordinarily expensive. A pair of VX-binding proteins were designed using a supercharged scaffold that couples a large-scale phase change from unstructured to folded upon ligand binding, enabling fully internal binding sites that present the maximum surface area possible for high affinity and specificity in target recognition. Binding site residues were chosen using a new distributed evolutionary algorithm implementation in protCAD. Both designs detect VX at parts per billion concentrations with high specificity. Computational design of fully buried molecular recognition sites, in combination with supercharged phase-changing chassis proteins, enables the ready development of a new generation of small-molecule biosensors.
Collapse
Affiliation(s)
- James J. McCann
- Department of Physics, The City College of New York, New York, NY 10031, USA
| | - Douglas H. Pike
- Center for Advanced Biotechnology and Medicine and the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Mia C. Brown
- Department of Physics, The City College of New York, New York, NY 10031, USA
| | - David T. Crouse
- Department of Electrical and Computer Engineering, Clarkson University, Potsdam, NY 13699, USA
| | - Vikas Nanda
- Center for Advanced Biotechnology and Medicine and the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ronald L. Koder
- Department of Physics, The City College of New York, New York, NY 10031, USA
- Graduate Programs of Physics, Biology, Chemistry, and Biochemistry, The Graduate Center of CUNY, New York, NY 10016, USA
| |
Collapse
|
45
|
Ding W, Nakai K, Gong H. Protein design via deep learning. Brief Bioinform 2022; 23:bbac102. [PMID: 35348602 PMCID: PMC9116377 DOI: 10.1093/bib/bbac102] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 12/11/2022] Open
Abstract
Proteins with desired functions and properties are important in fields like nanotechnology and biomedicine. De novo protein design enables the production of previously unseen proteins from the ground up and is believed as a key point for handling real social challenges. Recent introduction of deep learning into design methods exhibits a transformative influence and is expected to represent a promising and exciting future direction. In this review, we retrospect the major aspects of current advances in deep-learning-based design procedures and illustrate their novelty in comparison with conventional knowledge-based approaches through noticeable cases. We not only describe deep learning developments in structure-based protein design and direct sequence design, but also highlight recent applications of deep reinforcement learning in protein design. The future perspectives on design goals, challenges and opportunities are also comprehensively discussed.
Collapse
Affiliation(s)
- Wenze Ding
- School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing 210044, China
- School of Future Technology, Nanjing University of Information Science and Technology, Nanjing 210044, China
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Kenta Nakai
- Institute of Medical Science, the University of Tokyo, Tokyo 1088639, Japan
| | - Haipeng Gong
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
46
|
Kretschmer S, Kortemme T. Advances in the Computational Design of Small-Molecule-Controlled Protein-Based Circuits for Synthetic Biology. PROCEEDINGS OF THE IEEE. INSTITUTE OF ELECTRICAL AND ELECTRONICS ENGINEERS 2022; 110:659-674. [PMID: 36531560 PMCID: PMC9754107 DOI: 10.1109/jproc.2022.3157898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Synthetic biology approaches living systems with an engineering perspective and promises to deliver solutions to global challenges in healthcare and sustainability. A critical component is the design of biomolecular circuits with programmable input-output behaviors. Such circuits typically rely on a sensor module that recognizes molecular inputs, which is coupled to a functional output via protein-level circuits or regulating the expression of a target gene. While gene expression outputs can be customized relatively easily by exchanging the target genes, sensing new inputs is a major limitation. There is a limited repertoire of sensors found in nature, and there are often difficulties with interfacing them with engineered circuits. Computational protein design could be a key enabling technology to address these challenges, as it allows for the engineering of modular and tunable sensors that can be tailored to the circuit's application. In this article, we review recent computational approaches to design protein-based sensors for small-molecule inputs with particular focus on those based on the widely used Rosetta software suite. Furthermore, we review mechanisms that have been harnessed to couple ligand inputs to functional outputs. Based on recent literature, we illustrate how the combination of protein design and synthetic biology enables new sensors for diverse applications ranging from biomedicine to metabolic engineering. We conclude with a perspective on how strategies to address frontiers in protein design and cellular circuit design may enable the next generation of sense-response networks, which may increasingly be assembled from de novo components to display diverse and engineerable input-output behaviors.
Collapse
Affiliation(s)
- Simon Kretschmer
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, CA 94158 USA, and affiliated with the California Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, CA 94158 USA
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, CA 94158 USA, and affiliated with the California Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, CA 94158 USA
| |
Collapse
|
47
|
ElGamacy M. Accelerating therapeutic protein design. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:85-118. [PMID: 35534117 DOI: 10.1016/bs.apcsb.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Protein structures provide for defined microenvironments that can support complex pharmacological functions, otherwise unachievable by small molecules. The advent of therapeutic proteins has thus greatly broadened the range of manageable disorders. Leveraging the knowledge and recent advances in de novo protein design methods has the prospect of revolutionizing how protein drugs are discovered and developed. This review lays out the main challenges facing therapeutic proteins discovery and development, and how present and future advancements of protein design can accelerate the protein drug pipelines.
Collapse
Affiliation(s)
- Mohammad ElGamacy
- University Hospital Tübingen, Division of Translational Oncology, Tübingen, Germany; Max Planck Institute for Biology, Tübingen, Germany.
| |
Collapse
|
48
|
Gonzalez NA, Li BA, McCully ME. The stability and dynamics of computationally designed proteins. Protein Eng Des Sel 2022; 35:gzac001. [PMID: 35174855 PMCID: PMC9214642 DOI: 10.1093/protein/gzac001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/11/2022] Open
Abstract
Protein stability, dynamics and function are intricately linked. Accordingly, protein designers leverage dynamics in their designs and gain insight to their successes and failures by analyzing their proteins' dynamics. Molecular dynamics (MD) simulations are a powerful computational tool for quantifying both local and global protein dynamics. This review highlights studies where MD simulations were applied to characterize the stability and dynamics of designed proteins and where dynamics were incorporated into computational protein design. First, we discuss the structural basis underlying the extreme stability and thermostability frequently observed in computationally designed proteins. Next, we discuss examples of designed proteins, where dynamics were not explicitly accounted for in the design process, whose coordinated motions or active site dynamics, as observed by MD simulation, enhanced or detracted from their function. Many protein functions depend on sizeable or subtle conformational changes, so we finally discuss the computational design of proteins to perform a specific function that requires consideration of motion by multi-state design.
Collapse
Affiliation(s)
- Natali A Gonzalez
- Department of Biology, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA
| | - Brigitte A Li
- Department of Biology, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA
| | - Michelle E McCully
- Department of Biology, Santa Clara University, 500 El Camino Real, Santa Clara, CA 95053, USA
| |
Collapse
|
49
|
Chen Y, Chen Q, Liu H. DEPACT and PACMatch: A Workflow of Designing De Novo Protein Pockets to Bind Small Molecules. J Chem Inf Model 2022; 62:971-985. [PMID: 35171604 DOI: 10.1021/acs.jcim.1c01398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Engineering of new functional proteins such as enzymes and biosensors involves the design of new protein pockets for the specific binding of small molecules. Here, we report a workflow composed of two new computational methods to execute this task. The DEPACT (Design Pocket as a Cluster based on Templates) method is a data-driven approach to design and evaluate small-molecule-binding pockets as isolated clusters, while the PACMatch method is a computational approach to match pocket residues in a cluster model to positions on given protein scaffolds. Using DEPACT and its scoring function, pocket clusters of natural-pocket-like chemical compositions and protein-ligand interaction strength can be designed. DEPACT can design pocket clusters containing water- or metal-ion-mediated protein-ligand interactions. While being able to efficiently treat relatively large pocket cluster models (e.g., of around 10 pocket residues), PACMatch outperforms previous methods in test cases of recovering the native positions of pocket residues in natural enzyme-substrate complexes.
Collapse
Affiliation(s)
- Yaoxi Chen
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Quan Chen
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.,Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, Anhui 230027, China
| | - Haiyan Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.,Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, Anhui 230027, China.,School of Data Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
50
|
Koehl A, Jagota M, Erdmann-Pham DD, Fung A, Song YS. Transferability of Geometric Patterns from Protein Self-Interactions to Protein-Ligand Interactions. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2022; 27:22-33. [PMID: 34890133 PMCID: PMC8669734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is significant interest in developing machine learning methods to model protein-ligand interactions but a scarcity of experimentally resolved protein-ligand structures to learn from. Protein self-contacts are a much larger source of structural data that could be leveraged, but currently it is not well understood how this data source differs from the target domain. Here, we characterize the 3D geometric patterns of protein self-contacts as probability distributions. We then present a flexible statistical framework to assess the transferability of these patterns to protein-ligand contacts. We observe that the level of transferability from protein self-contacts to protein-ligand contacts depends on contact type, with many contact types exhibiting high transferability. We then demonstrate the potential of leveraging information from these geometric patterns to aid in ligand pose-selection problems in protein-ligand docking. We publicly release our extracted data on geometric interaction patterns to enable further exploration of this problem.
Collapse
Affiliation(s)
- Antoine Koehl
- Department of Statistics, University of California, Berkeley, CA 94720, USA
| | - Milind Jagota
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, USA
| | | | - Alexander Fung
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, USA
| | - Yun S. Song
- Department of Statistics, University of California, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158
| |
Collapse
|