1
|
Islam S, Chauhan VM, Pantazes RJ. Analysis of how antigen mutations disrupt antibody binding interactions toward enabling rapid and reliable antibody repurposing. MAbs 2025; 17:2440586. [PMID: 39690439 DOI: 10.1080/19420862.2024.2440586] [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: 06/26/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Antibody repurposing is the process of changing a known antibody so that it binds to a mutated antigen. One of the findings to emerge from the Coronavirus Disease 2019 (COVID-19) pandemic was that it was possible to repurpose neutralizing antibodies for Severe Acute Respiratory Syndrome, a related disease, to work for COVID-19. Thus, antibody repurposing is a possible pathway to prepare for and respond to future pandemics, as well as personalizing cancer therapies. For antibodies to be successfully repurposed, it is necessary to know both how antigen mutations disrupt their binding and how they should be mutated to recover binding, with this work describing an analysis to address the first of these topics. Every possible antigen point mutation in the interface of 246 antibody-protein complexes were analyzed using the Rosetta molecular mechanics force field. The results highlight a number of features of how antigen mutations affect antibody binding, including the effects of mutating critical hotspot residues versus other positions, how many mutations are necessary to be likely to disrupt binding, the prevalence of indirect effects of mutations on binding, and the relative importance of changing attractive versus repulsive energies. These data are expected to be useful in guiding future antibody repurposing experiments.
Collapse
Affiliation(s)
- Sumaiya Islam
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| | - Varun M Chauhan
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| | - Robert J Pantazes
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| |
Collapse
|
2
|
Henshey B, Carneiro A, Lei K, Schaffer D, Boulis NM. Adeno-associated viral vector targeted evolution for neurofibromatosis gene delivery. Trends Mol Med 2025; 31:388-398. [PMID: 39890493 PMCID: PMC11985305 DOI: 10.1016/j.molmed.2025.01.004] [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: 09/08/2024] [Revised: 12/29/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
Neurofibromatosis type 1 (NF1) is an inherited genetic disease resulting from pathogenic mutations in NF1 that drive tumor formation along peripheral nerves, leading to many functional consequences. Tumor removal or treatment often results in regrowth and/or nerve damage. Addressing NF1 pathogenic variations at the cellular level through gene therapy holds great potential for long-term treatment of patients with NF1. Adeno-associated viruses (AAVs) are broadly used gene delivery vehicles for gene therapies because of their low pathogenicity, ability to transduce nondividing cells, and potential for long-term gene expression. This article explores the landscape of AAV-mediated gene delivery strategies for NF1, discusses the challenges of efficient delivery to relevant cell types, and highlights the progress in vector design strategies.
Collapse
Affiliation(s)
- Brett Henshey
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Ana Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kecheng Lei
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
| | - David Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
3
|
Frei L, Gao B, Han J, Taft JM, Irvine EB, Weber CR, Kumar RK, Eisinger BN, Ignatov A, Yang Z, Reddy ST. Deep mutational learning for the selection of therapeutic antibodies resistant to the evolution of Omicron variants of SARS-CoV-2. Nat Biomed Eng 2025; 9:552-565. [PMID: 40044817 PMCID: PMC12003156 DOI: 10.1038/s41551-025-01353-4] [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: 10/11/2023] [Accepted: 01/16/2025] [Indexed: 04/18/2025]
Abstract
Most antibodies for treating COVID-19 rely on binding the receptor-binding domain (RBD) of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2). However, Omicron and its sub-lineages, as well as other heavily mutated variants, have rendered many neutralizing antibodies ineffective. Here we show that antibodies with enhanced resistance to the evolution of SARS-CoV-2 can be identified via deep mutational learning. We constructed a library of full-length RBDs of Omicron BA.1 with high mutational distance and screened it for binding to the angiotensin-converting-enzyme-2 receptor and to neutralizing antibodies. After deep-sequencing the library, we used the data to train ensemble deep-learning models for the prediction of the binding and escape of a panel of eight therapeutic antibody candidates targeting a diverse range of RBD epitopes. By using in silico evolution to assess antibody breadth via the prediction of the binding and escape of the antibodies to millions of Omicron sequences, we found combinations of two antibodies with enhanced and complementary resistance to viral evolution. Deep learning may enable the development of therapeutic antibodies that remain effective against future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Lester Frei
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Beichen Gao
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Jiami Han
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Joseph M Taft
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Edward B Irvine
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Cédric R Weber
- Alloy Therapeutics (Switzerland) AG, Allschwil, Switzerland
| | - Rachita K Kumar
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Benedikt N Eisinger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Andrey Ignatov
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Zhouya Yang
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- Basel Research Centre for Child Health, Basel, Switzerland.
- Botnar Institute of Immune Engineering, Basel, Switzerland.
| |
Collapse
|
4
|
Papamichail D, Febinger M, Almeda S, Aberbach T, Papamichail G. Synthesis cost-optimal targeted mutant protein libraries. Comput Biol Chem 2024; 110:108068. [PMID: 38669847 DOI: 10.1016/j.compbiolchem.2024.108068] [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: 09/15/2023] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
Protein variant libraries produced by site-directed mutagenesis are a useful tool utilized by protein engineers to explore variants with potentially improved properties, such as activity and stability. These libraries are commonly built by selecting residue positions and alternative beneficial mutations for each position. All possible combinations are then constructed and screened, by incorporating degenerate codons at mutation sites. These degenerate codons often encode additional unwanted amino acids or even STOP codons. Our study aims to take advantage of annealing based recombination of oligonucleotides during synthesis and utilize multiple degenerate codons per mutation site to produce targeted protein libraries devoid of unwanted variants. Toward this goal we created an algorithm to calculate the minimum number of degenerate codons necessary to specify any given amino acid set, and a dynamic programming method that uses this algorithm to optimally partition a DNA target sequence with degeneracies into overlapping oligonucleotides, such that the total cost of synthesis of the target mutant protein library is minimized. Computational experiments show that, for a modest increase in DNA synthesis costs, beneficial variant yields in produced mutant libraries are increased by orders of magnitude, an effect particularly pronounced in large combinatorial libraries.
Collapse
Affiliation(s)
- Dimitris Papamichail
- Department of Computer Science, The College of New Jersey, 2000 Pennington Road, Ewing, 08628, NJ, USA.
| | - Madeline Febinger
- Department of Computer Science, The College of New Jersey, 2000 Pennington Road, Ewing, 08628, NJ, USA
| | - Shm Almeda
- Department of Computer Science, The College of New Jersey, 2000 Pennington Road, Ewing, 08628, NJ, USA
| | - Tomer Aberbach
- Department of Computer Science, The College of New Jersey, 2000 Pennington Road, Ewing, 08628, NJ, USA
| | | |
Collapse
|
5
|
Guo J, Lin LF, Oraskovich SV, Rivera de Jesús JA, Listgarten J, Schaffer DV. Computationally guided AAV engineering for enhanced gene delivery. Trends Biochem Sci 2024; 49:457-469. [PMID: 38531696 PMCID: PMC11456259 DOI: 10.1016/j.tibs.2024.03.002] [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: 11/23/2023] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/28/2024]
Abstract
Gene delivery vehicles based on adeno-associated viruses (AAVs) are enabling increasing success in human clinical trials, and they offer the promise of treating a broad spectrum of both genetic and non-genetic disorders. However, delivery efficiency and targeting must be improved to enable safe and effective therapies. In recent years, considerable effort has been invested in creating AAV variants with improved delivery, and computational approaches have been increasingly harnessed for AAV engineering. In this review, we discuss how computationally designed AAV libraries are enabling directed evolution. Specifically, we highlight approaches that harness sequences outputted by next-generation sequencing (NGS) coupled with machine learning (ML) to generate new functional AAV capsids and related regulatory elements, pushing the frontier of what vector engineering and gene therapy may achieve.
Collapse
Affiliation(s)
- Jingxuan Guo
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720, USA
| | - Li F Lin
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Sydney V Oraskovich
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, CA 94720, USA
| | - Julio A Rivera de Jesús
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, CA 94720, USA; Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Jennifer Listgarten
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA 94720, USA
| | - David V Schaffer
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
6
|
Biggs BW, de Paz AM, Bhan NJ, Cybulski TR, Church GM, Tyo KEJ. Engineering Ca 2+-Dependent DNA Polymerase Activity. ACS Synth Biol 2023; 12:3301-3311. [PMID: 37856140 DOI: 10.1021/acssynbio.3c00302] [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: 10/20/2023]
Abstract
Advancements in synthetic biology have provided new opportunities in biosensing, with applications ranging from genetic programming to diagnostics. Next generation biosensors aim to expand the number of accessible environments for measurements, increase the number of measurable phenomena, and improve the quality of the measurement. To this end, an emerging area in the field has been the integration of DNA as an information storage medium within biosensor outputs, leveraging nucleic acids to record the biosensor state over time. However, slow signal transduction steps, due to the time scales of transcription and translation, bottleneck many sensing-DNA recording approaches. DNA polymerases (DNAPs) have been proposed as a solution to the signal transduction problem by operating as both the sensor and responder, but there is presently a lack of DNAPs with functional sensitivity to many desirable target ligands. Here, we engineer components of the Pol δ replicative polymerase complex of Saccharomyces cerevisiae to sense and respond to Ca2+, a metal cofactor relevant to numerous biological phenomena. Through domain insertion and binding site grafting to Pol δ subunits, we demonstrate functional allosteric sensitivity to Ca2+. Together, this work provides an important foundation for future efforts in the development of DNAP-based biosensors.
Collapse
Affiliation(s)
- Bradley W Biggs
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - Alexandra M de Paz
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - Namita J Bhan
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - Thaddeus R Cybulski
- Interdepartmental Neuroscience Program, Northwestern University, Chicago, Illinois 60611, United States
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Keith E J Tyo
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
7
|
Cox J, Jennings M, Lenahan C, Manion M, Courville S, Blazeck J. Rational engineering of an improved adenosine deaminase 2 enzyme for weaponizing T-cell therapies. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 19:100394. [PMID: 37519414 PMCID: PMC10374970 DOI: 10.1016/j.iotech.2023.100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Adenosine is a potent immunosuppressive metabolite that accumulates in the extracellular space within solid tumors and inhibits the antitumor function of native immune cell responses as well as chimeric antigen receptor (CAR) T-cell therapies. Here, we show that engineered human cells can degrade extracellular adenosine through secretion of adenosine deaminase (ADA) enzymes-a possible therapeutic enhancement for CAR T cells. We first determine that the high-activity ADA1 isoform is naturally intracellularly restricted and show that the addition of canonical or computationally predicted secretory peptides did not allow for improved secretion. We did, however, determine that the lower-activity ADA2 isoform is naturally secreted. Thus, we utilized phylogenetic-based structural comparisons to guide a mutational survey of ADA2 active site residues, which when coupled with a high-throughput screen for enhanced ADA2-mediated extracellular adenosine rate allowed isolation of the most catalytically efficient ADA2 variant reported to date. When expressed by human cells, this variant exhibits 30× higher extracellular adenosine degradation activity than the wild-type enzyme. Finally, we demonstrate that Jurkat and CAR T cells engineered to express this secreted, high-activity ADA2 variant can degrade significant amounts of extracellular adenosine in vitro.
Collapse
Affiliation(s)
- J.R. Cox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, USA
| | - M. Jennings
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, USA
| | - C. Lenahan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, USA
| | - M. Manion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, USA
| | - S. Courville
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, USA
| | - J. Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, USA
| |
Collapse
|
8
|
Spirov AV, Myasnikova EM. Problem of Domain/Building Block Preservation in the Evolution of Biological Macromolecules and Evolutionary Computation. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:1345-1362. [PMID: 35594219 DOI: 10.1109/tcbb.2022.3175908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Structurally and functionally isolated domains in biological macromolecular evolution, both natural and artificial, are largely similar to "schemata", building blocks (BBs), in evolutionary computation (EC). The problem of preserving in subsequent evolutionary searches the already found domains / BBs is well known and quite relevant in biology as well as in EC. Both biology and EC are seeing parallel and independent development of several approaches to identifying and preserving previously identified domains / BBs. First, we notice the similarity of DNA shuffling methods in synthetic biology and multi-parent recombination algorithms in EC. Furthermore, approaches to computer identification of domains in proteins that are being developed in biology can be aligned with BB identification methods in EC. Finally, approaches to chimeric protein libraries optimization in biology can be compared to evolutionary search methods based on probabilistic models in EC. We propose to validate the prospects of mutual exchange of ideas and transfer of algorithms and approaches between evolutionary systems biology and EC in these three principal directions. A crucial aim of this transfer is the design of new advanced experimental techniques capable of solving more complex problems of in vitro evolution.
Collapse
|
9
|
Iqbal Z, Sadaf S. A patent-based consideration of latest platforms in the art of directed evolution: a decade long untold story. Biotechnol Genet Eng Rev 2022; 38:133-246. [PMID: 35200115 DOI: 10.1080/02648725.2021.2017638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Directed (or in vitro) evolution of proteins and metabolic pathways requires tools for creating genetic diversity and identifying protein variants with new or improved functional properties. Besides simplicity, reliability, speed, versatility, universal applicability and economy of the technique, the new science of synthetic biology requires improved means for construction of smart and high-quality mutant libraries to better navigate the sequence diversity. In vitro CRISPR/Cas9-mediated mutagenic (ICM) system and machine-learning (ML)-assisted approaches to directed evolution are now in the field to achieve the goal. This review describes the gene diversification strategies, screening and selection methods, in silico (computer-aided), Cas9-mediated and ML-based approaches to mutagenesis, developed especially in the last decade, and their patent position. The objective behind is to emphasize researchers the need for noting which mutagenesis, screening or selection method is patented and then selecting a suitable restriction-free approach to sequence diversity. Techniques and evolved products subject to patent rights need commercial license if their use is for purposes other than private or experimental research.
Collapse
Affiliation(s)
- Zarina Iqbal
- IP Litigation Department, PakPat World Intellectual Property Protection Services, Lahore, Pakistan
| | - Saima Sadaf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
10
|
de Klerk A, Swanepoel P, Lourens R, Zondo M, Abodunran I, Lytras S, MacLean OA, Robertson D, Kosakovsky Pond SL, Zehr JD, Kumar V, Stanhope MJ, Harkins G, Murrell B, Martin DP. Conserved recombination patterns across coronavirus subgenera. Virus Evol 2022; 8:veac054. [PMID: 35814334 PMCID: PMC9261289 DOI: 10.1093/ve/veac054] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/03/2022] [Accepted: 06/10/2022] [Indexed: 11/12/2022] Open
Abstract
Recombination contributes to the genetic diversity found in coronaviruses and is known to be a prominent mechanism whereby they evolve. It is apparent, both from controlled experiments and in genome sequences sampled from nature, that patterns of recombination in coronaviruses are non-random and that this is likely attributable to a combination of sequence features that favour the occurrence of recombination break points at specific genomic sites, and selection disfavouring the survival of recombinants within which favourable intra-genome interactions have been disrupted. Here we leverage available whole-genome sequence data for six coronavirus subgenera to identify specific patterns of recombination that are conserved between multiple subgenera and then identify the likely factors that underlie these conserved patterns. Specifically, we confirm the non-randomness of recombination break points across all six tested coronavirus subgenera, locate conserved recombination hot- and cold-spots, and determine that the locations of transcriptional regulatory sequences are likely major determinants of conserved recombination break-point hotspot locations. We find that while the locations of recombination break points are not uniformly associated with degrees of nucleotide sequence conservation, they display significant tendencies in multiple coronavirus subgenera to occur in low guanine-cytosine content genome regions, in non-coding regions, at the edges of genes, and at sites within the Spike gene that are predicted to be minimally disruptive of Spike protein folding. While it is apparent that sequence features such as transcriptional regulatory sequences are likely major determinants of where the template-switching events that yield recombination break points most commonly occur, it is evident that selection against misfolded recombinant proteins also strongly impacts observable recombination break-point distributions in coronavirus genomes sampled from nature.
Collapse
Affiliation(s)
- Arné de Klerk
- Institute of Infectious Diseases and Molecular Medicine, Division Of Computational Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town 7701, South Africa
| | - Phillip Swanepoel
- Institute of Infectious Diseases and Molecular Medicine, Division Of Computational Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town 7701, South Africa
| | - Rentia Lourens
- Division of Neurosurgery, Neuroscience Institute, Department of Surgery, University of Cape Town, Cape Town, 7701, South Africa
| | - Mpumelelo Zondo
- Institute of Infectious Diseases and Molecular Medicine, Division Of Computational Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town 7701, South Africa
| | - Isaac Abodunran
- Institute of Infectious Diseases and Molecular Medicine, Division Of Computational Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town 7701, South Africa
| | - Spyros Lytras
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G61 1QH, UK
| | - Oscar A MacLean
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G61 1QH, UK
| | - David Robertson
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G61 1QH, UK
| | - Sergei L Kosakovsky Pond
- Department of Biology, Temple University, Institute for Genomics and Evolutionary Medicine, Philadelphia, PA 19122, USA
| | - Jordan D Zehr
- Department of Biology, Temple University, Institute for Genomics and Evolutionary Medicine, Philadelphia, PA 19122, USA
| | - Venkatesh Kumar
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 14186, Sweden
| | - Michael J Stanhope
- Department of Population and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gordon Harkins
- South African National Bioinformatics Institute, University of the Western Cape, Cape Town, 7535, South Africa
| | - Ben Murrell
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 14186, Sweden
| | - Darren P Martin
- Institute of Infectious Diseases and Molecular Medicine, Division Of Computational Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town 7701, South Africa
| |
Collapse
|
11
|
DeBenedictis EA, Chory EJ, Gretton DW, Wang B, Golas S, Esvelt KM. Systematic molecular evolution enables robust biomolecule discovery. Nat Methods 2022; 19:55-64. [PMID: 34969982 PMCID: PMC11655129 DOI: 10.1038/s41592-021-01348-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/09/2021] [Indexed: 11/09/2022]
Abstract
Evolution occurs when selective pressures from the environment shape inherited variation over time. Within the laboratory, evolution is commonly used to engineer proteins and RNA, but experimental constraints have limited the ability to reproducibly and reliably explore factors such as population diversity, the timing of environmental changes and chance on outcomes. We developed a robotic system termed phage- and robotics-assisted near-continuous evolution (PRANCE) to comprehensively explore biomolecular evolution by performing phage-assisted continuous evolution in high-throughput. PRANCE implements an automated feedback control system that adjusts the stringency of selection in response to real-time measurements of each molecular activity. In evolving three distinct types of biomolecule, we find that evolution is reproducibly altered by both random chance and the historical pattern of environmental changes. This work improves the reliability of protein engineering and enables the systematic analysis of the historical, environmental and random factors governing biomolecular evolution.
Collapse
Affiliation(s)
- Erika A DeBenedictis
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Emma J Chory
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dana W Gretton
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brian Wang
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stefan Golas
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kevin M Esvelt
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
12
|
Alejaldre L, Pelletier JN, Quaglia D. Methods for enzyme library creation: Which one will you choose?: A guide for novices and experts to introduce genetic diversity. Bioessays 2021; 43:e2100052. [PMID: 34263468 DOI: 10.1002/bies.202100052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022]
Abstract
Enzyme engineering allows to explore sequence diversity in search for new properties. The scientific literature is populated with methods to create enzyme libraries for engineering purposes, however, choosing a suitable method for the creation of mutant libraries can be daunting, in particular for the novices. Here, we address both novices and experts: how can one enter the arena of enzyme library design and what guidelines can advanced users apply to select strategies best suited to their purpose? Section I is dedicated to the novices and presents an overview of established and standard methods for library creation, as well as available commercial solutions. The expert will discover an up-to-date tool to freshen up their repertoire (Section I) and learn of the newest methods that are likely to become a mainstay (Section II). We focus primarily on in vitro methods, presenting the advantages of each method. Our ultimate aim is to offer a selection of methods/strategies that we believe to be most useful to the enzyme engineer, whether a first-timer or a seasoned user.
Collapse
Affiliation(s)
- Lorea Alejaldre
- Département de biochimie and Center for Green Chemistry and Catalysis (CGCC), Université de Montréal, Montréal, Quebec, Canada.,PROTEO, The Québec Network for Research on Protein Function, Engineering and Applications, Québec, Quebec, Canada
| | - Joelle N Pelletier
- Département de biochimie and Center for Green Chemistry and Catalysis (CGCC), Université de Montréal, Montréal, Quebec, Canada.,PROTEO, The Québec Network for Research on Protein Function, Engineering and Applications, Québec, Quebec, Canada.,Département de chimie, Université de Montréal, Montréal, Quebec, Canada
| | - Daniela Quaglia
- Département de chimie, Université de Montréal, Montréal, Quebec, Canada.,School of Chemistry, University of Nottingham, Nottingham, UK
| |
Collapse
|
13
|
Herrmann KR, Hofmann I, Jungherz D, Wittwer M, Infanzón B, Hamer SN, Davari MD, Ruff AJ, Schwaneberg U. Generation of phytase chimeras with low sequence identities and improved thermal stability. J Biotechnol 2021; 339:14-21. [PMID: 34271055 DOI: 10.1016/j.jbiotec.2021.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 11/18/2022]
Abstract
Being able to recombine more than two genes with four or more crossover points in a sequence independent manner is still a challenge in protein engineering and limits our capabilities in tailoring enzymes for industrial applications. By computational analysis employing multiple sequence alignments and homology modeling, five fragments of six phytase genes (sequence identities 31-64 %) were identified and efficiently recombined through phosphorothioate-based cloning using the PTRec method. By combinatorial recombination, functional phytase chimeras containing fragments of up to four phytases were obtained. Two variants (PTRec 74 and PTRec 77) with up to 32 % improved residual activity (90 °C, 60 min) and retained specific activities of > 1100 U/mg were identified. Both variants are composed of fragments from the phytases of Citrobacter braakii, Hafnia alvei and Yersinia mollaretii. They exhibit sequence identities of ≤ 80 % to their parental enzymes, highlighting the great potential of DNA recombination strategies to generate new enzymes with low sequences identities that offer opportunities for property right claims.
Collapse
Affiliation(s)
- Kevin R Herrmann
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Isabell Hofmann
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Dennis Jungherz
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Malte Wittwer
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Belén Infanzón
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Stefanie Nicole Hamer
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Mehdi D Davari
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany
| | - Anna Joëlle Ruff
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany.
| | - Ulrich Schwaneberg
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074, Aachen, Germany; DWI-Leibniz Institut für Interaktive Materialien, Forckenbeckstraße 50, 52056, Aachen, Germany.
| |
Collapse
|
14
|
Ferruz N, Noske J, Höcker B. Protlego: A Python package for the analysis and design of chimeric proteins. Bioinformatics 2021; 37:3182-3189. [PMID: 33901273 PMCID: PMC8504633 DOI: 10.1093/bioinformatics/btab253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/05/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
Motivation Duplication and recombination of protein fragments have led to the highly diverse protein space that we observe today. By mimicking this natural process, the design of protein chimeras via fragment recombination has proven experimentally successful and has opened a new era for the design of customizable proteins. The in silico building of structural models for these chimeric proteins, however, remains a manual task that requires a considerable degree of expertise and is not amenable for high-throughput studies. Energetic and structural analysis of the designed proteins often require the use of several tools, each with their unique technical difficulties and available in different programming languages or web servers. Results We implemented a Python package that enables automated, high-throughput design of chimeras and their structural analysis. First, it fetches evolutionarily conserved fragments from a built-in database (also available at fuzzle.uni-bayreuth.de). These relationships can then be represented via networks or further selected for chimera construction via recombination. Designed chimeras or natural proteins are then scored and minimized with the Charmm and Amber forcefields and their diverse structural features can be analyzed at ease. Here, we showcase Protlego’s pipeline by exploring the relationships between the P-loop and Rossmann superfolds, building and characterizing their offspring chimeras. We believe that Protlego provides a powerful new tool for the protein design community. Availability and implementation Protlego runs on the Linux platform and is freely available at (https://hoecker-lab.github.io/protlego/) with tutorials and documentation. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Noelia Ferruz
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Jakob Noske
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Birte Höcker
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| |
Collapse
|
15
|
Campbell IJ, Kahanda D, Atkinson JT, Sparks ON, Kim J, Tseng CP, Verduzco R, Bennett GN, Silberg JJ. Recombination of 2Fe-2S Ferredoxins Reveals Differences in the Inheritance of Thermostability and Midpoint Potential. ACS Synth Biol 2020; 9:3245-3253. [PMID: 33226772 DOI: 10.1021/acssynbio.0c00303] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Recombination can be used in the laboratory to overcome component limitations in synthetic biology by creating enzymes that exhibit distinct activities and stabilities from native proteins. To investigate how recombination affects the properties of an oxidoreductase that transfers electrons in cells, we created ferredoxin (Fd) chimeras by recombining distantly related cyanobacterial and cyanomyophage Fds (53% identity) that present similar midpoint potentials but distinct thermostabilities. Fd chimeras having a wide range of amino acid substitutions retained the ability to coordinate an iron-sulfur cluster, although their thermostabilities varied with the fraction of residues inherited from each parent. The midpoint potentials of chimeric Fds also varied. However, all of the synthetic Fds exhibited midpoint potentials outside of the parental protein range. Each of the chimeric Fds could also support electron transfer between Fd-NADP reductase and sulfite reductase in Escherichia coli, although the chimeric Fds varied in the expression required for similar levels of cellular electron transfer. These results show how Fds can be diversified through recombination and reveal differences in the inheritance of thermostability and electrochemical properties. Furthermore, they illustrate how electron transfer efficiencies of chimeric Fds can be rapidly evaluated using a synthetic metabolic pathway.
Collapse
Affiliation(s)
- Ian J. Campbell
- Department of BioSciences, Rice University, 6100 Main Street, MS-140, Houston, Texas 77005, United States
| | - Dimithree Kahanda
- Department of BioSciences, Rice University, 6100 Main Street, MS-140, Houston, Texas 77005, United States
| | - Joshua T. Atkinson
- Department of BioSciences, Rice University, 6100 Main Street, MS-140, Houston, Texas 77005, United States
| | - Othneil Noble Sparks
- Department of BioSciences, Rice University, 6100 Main Street, MS-140, Houston, Texas 77005, United States
| | - Jinyoung Kim
- Department of BioSciences, Rice University, 6100 Main Street, MS-140, Houston, Texas 77005, United States
| | - Chia-Ping Tseng
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main Street, MS-362, Houston, Texas 77005, United States
| | - Rafael Verduzco
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main Street, MS-362, Houston, Texas 77005, United States
| | - George N. Bennett
- Department of BioSciences, Rice University, 6100 Main Street, MS-140, Houston, Texas 77005, United States
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main Street, MS-362, Houston, Texas 77005, United States
| | - Jonathan J. Silberg
- Department of BioSciences, Rice University, 6100 Main Street, MS-140, Houston, Texas 77005, United States
- Department of Chemical and Biomolecular Engineering, Rice University, 6100 Main Street, MS-362, Houston, Texas 77005, United States
- Department of Bioengineering, Rice University, 6100 Main Street, MS-142, Houston, Texas 77005, United States
| |
Collapse
|
16
|
Collins CH, Cirino PC. Commemorating Frances Arnold. AIChE J 2020. [DOI: 10.1002/aic.16924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Cynthia H. Collins
- Department of Chemical and Biological EngineeringRensselaer Polytechnic Institute Troy New York
| | - Patrick C. Cirino
- Department of Chemical & Biomolecular EngineeringUniversity of Houston Houston Texas
| |
Collapse
|
17
|
Tsai CH, Wei SC, Jan JT, Liao LL, Chang CJ, Chao YC. Generation of Stable Influenza Virus Hemagglutinin through Structure-Guided Recombination. ACS Synth Biol 2019; 8:2472-2482. [PMID: 31565926 DOI: 10.1021/acssynbio.9b00094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hemagglutinin (HA) is the major surface antigen of influenza virus and the most promising influenza vaccine immunogen. In 2013, the devastating H7N9 influenza virus was identified in China, which induced high mortality. The HA of this virus (H7) is relatively unstable, making it challenging to produce an effective vaccine. To improve the stability of HA protein from H7N9 influenza virus for better vaccine antigens without impairing immunogenicity, we recombined the HA from H7N9 (H7) with a more stable HA from H3N2 (H3) by structure-guided recombination, resulting in six chimeric HAs, FrA-FrF. Two of these chimeric HAs, FrB and FrC, exhibited proper hemagglutination activity and presented improved thermal stability compared to the original H7. Mice immunized with FrB and FrC elicited H7-specific antibodies comparable to those induced by parental H7, and the antisera collected from these immunized mice successfully inhibited H7N9 infection in a microneutralization assay. These results suggest that our structural-recombination approach can create stabilizing chimeric antigens while maintaining proper immunogenicity, which may not only benefit the construction of more stable HA vaccines to fight against H7N9 infection, but also facilitate effective vaccine improvements for other influenza viruses or infectious pathogens. In addition, this study also demonstrates the potential for better engineering of multimeric protein complexes like HA to achieve improved function, which are often immunologically or pharmaceutically important but difficult to modify.
Collapse
Affiliation(s)
- Chih-Hsuan Tsai
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei 115, Taiwan, ROC
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan, ROC
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan, ROC
| | - Sung-Chan Wei
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan, ROC
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan, ROC
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, ROC
| | - Lin-Li Liao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan, ROC
| | - Chia-Jung Chang
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan, ROC
| | - Yu-Chan Chao
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei 115, Taiwan, ROC
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan, ROC
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan, ROC
- Department of Plant Pathology and Microbiology, College of Bioresources and Agriculture, National Taiwan University, Taipei 106, Taiwan, ROC
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan, ROC
| |
Collapse
|
18
|
J B, M M B, Chanda K. Evolutionary approaches in protein engineering towards biomaterial construction. RSC Adv 2019; 9:34720-34734. [PMID: 35530663 PMCID: PMC9074691 DOI: 10.1039/c9ra06807d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/01/2019] [Indexed: 11/29/2022] Open
Abstract
The tailoring of proteins for specific applications by evolutionary methods is a highly active area of research. Rational design and directed evolution are the two main strategies to reengineer proteins or create chimeric structures. Rational engineering is often limited by insufficient knowledge about proteins' structure-function relationships; directed evolution overcomes this restriction but poses challenges in the screening of candidates. A combination of these protein engineering approaches will allow us to create protein variants with a wide range of desired properties. Herein, we focus on the application of these approaches towards the generation of protein biomaterials that are known for biodegradability, biocompatibility and biofunctionality, from combinations of natural, synthetic, or engineered proteins and protein domains. Potential applications depend on the enhancement of biofunctional, mechanical, or other desired properties. Examples include scaffolds for tissue engineering, thermostable enzymes for industrial biocatalysis, and other therapeutic applications.
Collapse
Affiliation(s)
- Brindha J
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Chennai Campus Vandalur-Kelambakkam Road Chennai-600 127 Tamil Nadu India
| | - Balamurali M M
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Chennai Campus Vandalur-Kelambakkam Road Chennai-600 127 Tamil Nadu India
| | - Kaushik Chanda
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology Vellore-632014 Tamil Nadu India
| |
Collapse
|
19
|
Engqvist MKM, Rabe KS. Applications of Protein Engineering and Directed Evolution in Plant Research. PLANT PHYSIOLOGY 2019; 179:907-917. [PMID: 30626612 PMCID: PMC6393796 DOI: 10.1104/pp.18.01534] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/25/2018] [Indexed: 05/06/2023]
Abstract
Engineered proteins can be used to optimize desired traits in plants; even though recent advances have resulted in new application areas, certain methodological challenges remain.
Collapse
Affiliation(s)
- Martin K M Engqvist
- Department of Biology and Biological Engineering, Chalmers University of Technology, Division of Systems and Synthetic Biology, Gothenburg, Sweden
| | - Kersten S Rabe
- Institute for Biological Interfaces (IBG 1), Karlsruhe Institute of Technology (KIT), Group for Molecular Evolution, Karlsruhe, Germany
| |
Collapse
|
20
|
Atkinson JT, Wu B, Segatori L, Silberg JJ. Overcoming component limitations in synthetic biology through transposon-mediated protein engineering. Methods Enzymol 2019; 621:191-212. [DOI: 10.1016/bs.mie.2019.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
21
|
Seo JH, Min WK, Lee SG, Yun H, Kim BG. To the Final Goal: Can We Predict and Suggest Mutations for Protein to Develop Desired Phenotype? BIOTECHNOL BIOPROC E 2018. [DOI: 10.1007/s12257-018-0064-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
22
|
Ordered chimerogenesis applied to CYP2B P450 enzymes. Biochim Biophys Acta Gen Subj 2016; 1860:1395-403. [DOI: 10.1016/j.bbagen.2016.03.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/12/2016] [Accepted: 03/20/2016] [Indexed: 12/11/2022]
|
23
|
Gobeil SMC, Gagné D, Doucet N, Pelletier JN. 15N, 13C and 1H backbone resonance assignments of an artificially engineered TEM-1/PSE-4 class A β-lactamase chimera and its deconvoluted mutant. BIOMOLECULAR NMR ASSIGNMENTS 2016; 10:93-99. [PMID: 26386961 PMCID: PMC5419827 DOI: 10.1007/s12104-015-9645-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/09/2015] [Indexed: 06/05/2023]
Abstract
The widespread use of β-lactam antibiotics has given rise to a dramatic increase in clinically-relevant β-lactamases. Understanding the structure/function relation in these variants is essential to better address the ever-growing incidence of antibiotic resistance. We previously reported the backbone resonance assignments of a chimeric protein constituted of segments of the class A β-lactamases TEM-1 and PSE-4 (Morin et al. in Biomol NMR Assign 4:127-130, 2010. doi: 10.1007/s12104-010-9227-8 ). That chimera, cTEM17m, held 17 amino acid substitutions relative to TEM-1 β-lactamase, resulting in a well-folded and fully functional protein with increased dynamics. Here we report the (1)H, (13)C and (15)N backbone resonance assignments of chimera cTEM-19m, which includes 19 substitutions and exhibits increased active-site perturbation, as well as one of its deconvoluted variants, as the first step in the analysis of their dynamic behaviours.
Collapse
Affiliation(s)
- Sophie M C Gobeil
- Department of Biochemistry, Université de Montréal, Montréal, QC, Canada
- PROTEO, The Québec Network for Research on Protein Function, Engineering and Applications, Québec, QC, Canada
| | - Donald Gagné
- PROTEO, The Québec Network for Research on Protein Function, Engineering and Applications, Québec, QC, Canada
- INRS-Institut Armand-Frappier, Université du Québec, Québec, QC, Canada
- GRASP, Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montréal, QC, Canada
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, USA
| | - Nicolas Doucet
- PROTEO, The Québec Network for Research on Protein Function, Engineering and Applications, Québec, QC, Canada
- INRS-Institut Armand-Frappier, Université du Québec, Québec, QC, Canada
- GRASP, Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montréal, QC, Canada
| | - Joelle N Pelletier
- Department of Biochemistry, Université de Montréal, Montréal, QC, Canada.
- PROTEO, The Québec Network for Research on Protein Function, Engineering and Applications, Québec, QC, Canada.
- Department of Chemistry, Université de Montréal, Montréal, Canada.
| |
Collapse
|
24
|
Pottel J, Moitessier N. Single-Point Mutation with a Rotamer Library Toolkit: Toward Protein Engineering. J Chem Inf Model 2015; 55:2657-71. [PMID: 26623941 DOI: 10.1021/acs.jcim.5b00525] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Protein engineers have long been hard at work to harness biocatalysts as a natural source of regio-, stereo-, and chemoselectivity in order to carry out chemistry (reactions and/or substrates) not previously achieved with these enzymes. The extreme labor demands and exponential number of mutation combinations have induced computational advances in this domain. The first step in our virtual approach is to predict the correct conformations upon mutation of residues (i.e., rebuilding side chains). For this purpose, we opted for a combination of molecular mechanics and statistical data. In this work, we have developed automated computational tools to extract protein structural information and created conformational libraries for each amino acid dependent on a variable number of parameters (e.g., resolution, flexibility, secondary structure). We have also developed the necessary tool to apply the mutation and optimize the conformation accordingly. For side-chain conformation prediction, we obtained overall average root-mean-square deviations (RMSDs) of 0.91 and 1.01 Å for the 18 flexible natural amino acids within two distinct sets of over 3000 and 1500 side-chain residues, respectively. The commonly used dihedral angle differences were also evaluated and performed worse than the state of the art. These two metrics are also compared. Furthermore, we generated a family-specific library for kinases that produced an average 2% lower RMSD upon side-chain reconstruction and a residue-specific library that yielded a 17% improvement. Ultimately, since our protein engineering outlook involves using our docking software, Fitted/Impacts, we applied our mutation protocol to a benchmarked data set for self- and cross-docking. Our side-chain reconstruction does not hinder our docking software, demonstrating differences in pose prediction accuracy of approximately 2% (RMSD cutoff metric) for a set of over 200 protein/ligand structures. Similarly, when docking to a set of over 100 kinases, side-chain reconstruction (using both general and biased conformation libraries) had minimal detriment to the docking accuracy.
Collapse
Affiliation(s)
- Joshua Pottel
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, QC, Canada H3A 0B8
| | - Nicolas Moitessier
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, QC, Canada H3A 0B8
| |
Collapse
|
25
|
Porter JL, Rusli RA, Ollis DL. Directed Evolution of Enzymes for Industrial Biocatalysis. Chembiochem 2015; 17:197-203. [DOI: 10.1002/cbic.201500280] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Joanne L. Porter
- Research School of Chemistry; Australian National University; Canberra ACT 2601 Australia
| | - Rukhairul A. Rusli
- Research School of Chemistry; Australian National University; Canberra ACT 2601 Australia
| | - David L. Ollis
- Research School of Chemistry; Australian National University; Canberra ACT 2601 Australia
| |
Collapse
|
26
|
Affiliation(s)
- Andreas S. Bommarius
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30318;
| |
Collapse
|
27
|
Johnson LB, Gintner LP, Park S, Snow CD. Discriminating between stabilizing and destabilizing protein design mutations via recombination and simulation. Protein Eng Des Sel 2015; 28:259-67. [PMID: 26080450 DOI: 10.1093/protein/gzv030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/15/2015] [Indexed: 11/13/2022] Open
Abstract
Accuracy of current computational protein design (CPD) methods is limited by inherent approximations in energy potentials and sampling. These limitations are often used to qualitatively explain design failures; however, relatively few studies provide specific examples or quantitative details that can be used to improve future CPD methods. Expanding the design method to include a library of sequences provides data that is well suited for discriminating between stabilizing and destabilizing design elements. Using thermophilic endoglucanase E1 from Acidothermus cellulolyticus as a model enzyme, we computationally designed a sequence with 60 mutations. The design sequence was rationally divided into structural blocks and recombined with the wild-type sequence. Resulting chimeras were assessed for activity and thermostability. Surprisingly, unlike previous chimera libraries, regression analysis based on one- and two-body effects was not sufficient for predicting chimera stability. Analysis of molecular dynamics simulations proved helpful in distinguishing stabilizing and destabilizing mutations. Reverting to the wild-type amino acid at destabilized sites partially regained design stability, and introducing predicted stabilizing mutations in wild-type E1 significantly enhanced thermostability. The ability to isolate stabilizing and destabilizing elements in computational design offers an opportunity to interpret previous design failures and improve future CPD methods.
Collapse
Affiliation(s)
- Lucas B Johnson
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Lucas P Gintner
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Sehoo Park
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Christopher D Snow
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
28
|
Abstract
Faced with a protein engineering challenge, a contemporary researcher can choose from myriad design strategies. Library-scale computational protein design (LCPD) is a hybrid method suitable for the engineering of improved protein variants with diverse sequences. This chapter discusses the background and merits of several practical LCPD techniques. First, LCPD methods suitable for delocalized protein design are presented in the context of example design calculations for cellobiohydrolase II. Second, localized design methods are discussed in the context of an example design calculation intended to shift the substrate specificity of a ketol-acid reductoisomerase Rossmann domain from NADPH to NADH.
Collapse
|
29
|
Directed evolution of cytochrome P450 enzymes for biocatalysis: exploiting the catalytic versatility of enzymes with relaxed substrate specificity. Biochem J 2015; 467:1-15. [DOI: 10.1042/bj20141493] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cytochrome P450 enzymes are renowned for their ability to insert oxygen into an enormous variety of compounds with a high degree of chemo- and regio-selectivity under mild conditions. This property has been exploited in Nature for an enormous variety of physiological functions, and representatives of this ancient enzyme family have been identified in all kingdoms of life. The catalytic versatility of P450s makes them well suited for repurposing for the synthesis of fine chemicals such as drugs. Although these enzymes have not evolved in Nature to perform the reactions required for modern chemical industries, many P450s show relaxed substrate specificity and exhibit some degree of activity towards non-natural substrates of relevance to applications such as drug development. Directed evolution and other protein engineering methods can be used to improve upon this low level of activity and convert these promiscuous generalist enzymes into specialists capable of mediating reactions of interest with exquisite regio- and stereo-selectivity. Although there are some notable successes in exploiting P450s from natural sources in metabolic engineering, and P450s have been proven repeatedly to be excellent material for engineering, there are few examples to date of practical application of engineered P450s. The purpose of the present review is to illustrate the progress that has been made in altering properties of P450s such as substrate range, cofactor preference and stability, and outline some of the remaining challenges that must be overcome for industrial application of these powerful biocatalysts.
Collapse
|
30
|
Currin A, Swainston N, Day PJ, Kell DB. Synthetic biology for the directed evolution of protein biocatalysts: navigating sequence space intelligently. Chem Soc Rev 2015; 44:1172-239. [PMID: 25503938 PMCID: PMC4349129 DOI: 10.1039/c4cs00351a] [Citation(s) in RCA: 258] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Indexed: 12/21/2022]
Abstract
The amino acid sequence of a protein affects both its structure and its function. Thus, the ability to modify the sequence, and hence the structure and activity, of individual proteins in a systematic way, opens up many opportunities, both scientifically and (as we focus on here) for exploitation in biocatalysis. Modern methods of synthetic biology, whereby increasingly large sequences of DNA can be synthesised de novo, allow an unprecedented ability to engineer proteins with novel functions. However, the number of possible proteins is far too large to test individually, so we need means for navigating the 'search space' of possible protein sequences efficiently and reliably in order to find desirable activities and other properties. Enzymologists distinguish binding (Kd) and catalytic (kcat) steps. In a similar way, judicious strategies have blended design (for binding, specificity and active site modelling) with the more empirical methods of classical directed evolution (DE) for improving kcat (where natural evolution rarely seeks the highest values), especially with regard to residues distant from the active site and where the functional linkages underpinning enzyme dynamics are both unknown and hard to predict. Epistasis (where the 'best' amino acid at one site depends on that or those at others) is a notable feature of directed evolution. The aim of this review is to highlight some of the approaches that are being developed to allow us to use directed evolution to improve enzyme properties, often dramatically. We note that directed evolution differs in a number of ways from natural evolution, including in particular the available mechanisms and the likely selection pressures. Thus, we stress the opportunities afforded by techniques that enable one to map sequence to (structure and) activity in silico, as an effective means of modelling and exploring protein landscapes. Because known landscapes may be assessed and reasoned about as a whole, simultaneously, this offers opportunities for protein improvement not readily available to natural evolution on rapid timescales. Intelligent landscape navigation, informed by sequence-activity relationships and coupled to the emerging methods of synthetic biology, offers scope for the development of novel biocatalysts that are both highly active and robust.
Collapse
Affiliation(s)
- Andrew Currin
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- School of Chemistry , The University of Manchester , Manchester M13 9PL , UK
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
| | - Neil Swainston
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
- School of Computer Science , The University of Manchester , Manchester M13 9PL , UK
| | - Philip J. Day
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
- Faculty of Medical and Human Sciences , The University of Manchester , Manchester M13 9PT , UK
| | - Douglas B. Kell
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- School of Chemistry , The University of Manchester , Manchester M13 9PL , UK
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
| |
Collapse
|
31
|
Guenther CM, Kuypers BE, Lam MT, Robinson TM, Zhao J, Suh J. Synthetic virology: engineering viruses for gene delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2014; 6:548-58. [PMID: 25195922 PMCID: PMC4227300 DOI: 10.1002/wnan.1287] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/16/2014] [Accepted: 07/20/2014] [Indexed: 12/13/2022]
Abstract
The success of gene therapy relies heavily on the performance of vectors that can effectively deliver transgenes to desired cell populations. As viruses have evolved to deliver genetic material into cells, a prolific area of research has emerged over the last several decades to leverage the innate properties of viruses as well as to engineer new features into them. Specifically, the field of synthetic virology aims to capitalize on knowledge accrued from fundamental virology research in order to design functionally enhanced gene delivery vectors. The enhanced viral vectors, or 'bionic' viruses, feature engineered components, or 'parts', that are natural (intrinsic to viruses or from other organisms) and synthetic (such as man-made polymers or inorganic nanoparticles). Various design strategies--rational, combinatorial, and pseudo-rational--have been pursued to create the hybrid viruses. The gene delivery vectors of the future will likely criss-cross the boundaries between natural and synthetic domains to harness the unique strengths afforded by the various functional parts that can be grafted onto virus capsids. Such research endeavors will further expand and enable enhanced control over the functional capacity of these nanoscale devices for biomedicine.
Collapse
Affiliation(s)
| | - Brianna E. Kuypers
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, 77005
| | - Michael T. Lam
- Department of Bioengineering, Rice University, Houston, TX, 77005
| | | | - Julia Zhao
- Department of Chemistry, Rice University, Houston, TX, 77005
| | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, TX, 77005
| |
Collapse
|
32
|
Gobeil SMC, Clouthier CM, Park J, Gagné D, Berghuis AM, Doucet N, Pelletier JN. Maintenance of native-like protein dynamics may not be required for engineering functional proteins. ACTA ACUST UNITED AC 2014; 21:1330-1340. [PMID: 25200606 DOI: 10.1016/j.chembiol.2014.07.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 06/27/2014] [Accepted: 07/09/2014] [Indexed: 12/16/2022]
Abstract
Proteins are dynamic systems, and understanding dynamics is critical for fully understanding protein function. Therefore, the question of whether laboratory engineering has an impact on protein dynamics is of general interest. Here, we demonstrate that two homologous, naturally evolved enzymes with high degrees of structural and functional conservation also exhibit conserved dynamics. Their similar set of slow timescale dynamics is highly restricted, consistent with evolutionary conservation of a functionally important feature. However, we also show that dynamics of a laboratory-engineered chimeric enzyme obtained by recombination of the two homologs exhibits striking difference on the millisecond timescale, despite function and high-resolution crystal structure (1.05 Å) being conserved. The laboratory-engineered chimera is thus functionally tolerant to modified dynamics on the timescale of catalytic turnover. Tolerance to dynamic variation implies that maintenance of native-like protein dynamics may not be required when engineering functional proteins.
Collapse
Affiliation(s)
- Sophie M C Gobeil
- PROTEO Network, Université Laval, Québec QC G1V 0A6, Canada; Département de Biochimie, Université de Montréal, Montréal QC H3T 1J4, Canada
| | - Christopher M Clouthier
- PROTEO Network, Université Laval, Québec QC G1V 0A6, Canada; Département de Chimie, Université de Montréal, Montréal QC H3T 1J4, Canada
| | - Jaeok Park
- PROTEO Network, Université Laval, Québec QC G1V 0A6, Canada; Department of Biochemistry and Department of Microbiology and Immunology, McGill University, Montreal QC H3G 1Y6, Canada; GRASP Network, McGill University, Montréal QC H3G 1Y6, Canada
| | - Donald Gagné
- PROTEO Network, Université Laval, Québec QC G1V 0A6, Canada; GRASP Network, McGill University, Montréal QC H3G 1Y6, Canada; INRS-Institut Armand-Frappier, Université du Québec, Laval QC H7V 1B7, Canada
| | - Albert M Berghuis
- PROTEO Network, Université Laval, Québec QC G1V 0A6, Canada; Department of Biochemistry and Department of Microbiology and Immunology, McGill University, Montreal QC H3G 1Y6, Canada; GRASP Network, McGill University, Montréal QC H3G 1Y6, Canada
| | - Nicolas Doucet
- PROTEO Network, Université Laval, Québec QC G1V 0A6, Canada; GRASP Network, McGill University, Montréal QC H3G 1Y6, Canada; INRS-Institut Armand-Frappier, Université du Québec, Laval QC H7V 1B7, Canada
| | - Joelle N Pelletier
- PROTEO Network, Université Laval, Québec QC G1V 0A6, Canada; Département de Biochimie, Université de Montréal, Montréal QC H3T 1J4, Canada; Département de Chimie, Université de Montréal, Montréal QC H3T 1J4, Canada; Center for Green Chemistry and Catalysis (CCVC), Montréal QC H3A 0B8, Canada.
| |
Collapse
|
33
|
Woo J, Robertson DL, Lovell SC. Constraints from protein structure and intra-molecular coevolution influence the fitness of HIV-1 recombinants. Virology 2014; 454-455:34-9. [PMID: 24725929 DOI: 10.1016/j.virol.2014.01.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 01/10/2014] [Accepted: 01/29/2014] [Indexed: 11/18/2022]
Abstract
A major challenge for developing effective treatments for HIV-1 is the viruses' ability to generate new variants. Inter-strain recombination is a major contributor to this high evolutionary rate, since at least 20% of viruses are observed to be recombinant. However, the patterns of recombination vary across the viral genome. A number of factors influence recombination, including sequence identity and secondary RNA structure. In addition the recombinant genome must code for a functional virus, and expressed proteins must fold to stable and functional structures. Any intragenic recombination that disrupts internal residue contacts may therefore produce an unfolded protein. Here we find that contact maps based on protein structures predict recombination breakpoints observed in the HIV-1 pandemic. Moreover, many pairs of contacting residues that are unlikely to be disrupted by recombination are coevolving. We conclude that purifying selection arising from protein structure and intramolecular coevolutionary changes shapes the observed patterns of recombination in HIV-1.
Collapse
Affiliation(s)
- Jeongmin Woo
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - David L Robertson
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| | - Simon C Lovell
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
34
|
Zaugg J, Gumulya Y, Gillam EMJ, Bodén M. Computational tools for directed evolution: a comparison of prospective and retrospective strategies. Methods Mol Biol 2014; 1179:315-333. [PMID: 25055787 DOI: 10.1007/978-1-4939-1053-3_21] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Directed evolution methods have proved to be highly effective in the design of novel proteins and in the generation of large libraries of diverse sequences. However, searching through the vast number of mutants produced during such experiments in order to find the best represents a daunting and difficult task. In recent years, a number of computational tools have been developed to provide guidance during this exploratory process. It can, however, be unclear as to which tool or tools best complement the chosen library design strategy. In this review, we describe and critically evaluate some of the more notable tools in this area, discussing the rationale behind each, the requirements for their implementation, and potential issues faced when using them. Some examples of their application in an experimental setting are also provided. The tools have been classified based on contrasting strategies as to how they function: prospective tools SCHEMA and OPTCOMB use extant sequence and structural data to predict optimal locations for crossover sites, whereas retrospective tools ProSAR and ASRA use property data from the mutant library to predict beneficial mutations and features. From our evaluation, we suggest that each tool can play a role in the design process; however this is largely dictated by the data available and the desired experimental strategy for the project.
Collapse
Affiliation(s)
- Julian Zaugg
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | | | | | | |
Collapse
|
35
|
Ho ML, Adler BA, Torre ML, Silberg JJ, Suh J. SCHEMA computational design of virus capsid chimeras: calibrating how genome packaging, protection, and transduction correlate with calculated structural disruption. ACS Synth Biol 2013; 2:724-33. [PMID: 23899192 DOI: 10.1021/sb400076r] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Adeno-associated virus (AAV) recombination can result in chimeric capsid protein subunits whose ability to assemble into an oligomeric capsid, package a genome, and transduce cells depends on the inheritance of sequence from different AAV parents. To develop quantitative design principles for guiding site-directed recombination of AAV capsids, we have examined how capsid structural perturbations predicted by the SCHEMA algorithm correlate with experimental measurements of disruption in seventeen chimeric capsid proteins. In our small chimera population, created by recombining AAV serotypes 2 and 4, we found that protection of viral genomes and cellular transduction were inversely related to calculated disruption of the capsid structure. Interestingly, however, we did not observe a correlation between genome packaging and calculated structural disruption; a majority of the chimeric capsid proteins formed at least partially assembled capsids and more than half packaged genomes, including those with the highest SCHEMA disruption. These results suggest that the sequence space accessed by recombination of divergent AAV serotypes is rich in capsid chimeras that assemble into 60-mer capsids and package viral genomes. Overall, the SCHEMA algorithm may be useful for delineating quantitative design principles to guide the creation of libraries enriched in genome-protecting virus nanoparticles that can effectively transduce cells. Such improvements to the virus design process may help advance not only gene therapy applications but also other bionanotechnologies dependent upon the development of viruses with new sequences and functions.
Collapse
Affiliation(s)
- Michelle L. Ho
- Department
of Bioengineering and ‡Department of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
| | - Benjamin A. Adler
- Department
of Bioengineering and ‡Department of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
| | - Michael L. Torre
- Department
of Bioengineering and ‡Department of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
| | - Jonathan J. Silberg
- Department
of Bioengineering and ‡Department of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
| | - Junghae Suh
- Department
of Bioengineering and ‡Department of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
| |
Collapse
|
36
|
Clouthier CM, Morin S, Gobeil SMC, Doucet N, Blanchet J, Nguyen E, Gagné SM, Pelletier JN. Chimeric β-lactamases: global conservation of parental function and fast time-scale dynamics with increased slow motions. PLoS One 2012; 7:e52283. [PMID: 23284969 PMCID: PMC3528772 DOI: 10.1371/journal.pone.0052283] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/15/2012] [Indexed: 11/18/2022] Open
Abstract
Enzyme engineering has been facilitated by recombination of close homologues, followed by functional screening. In one such effort, chimeras of two class-A β-lactamases – TEM-1 and PSE-4 – were created according to structure-guided protein recombination and selected for their capacity to promote bacterial proliferation in the presence of ampicillin (Voigt et al., Nat. Struct. Biol. 2002 9:553). To provide a more detailed assessment of the effects of protein recombination on the structure and function of the resulting chimeric enzymes, we characterized a series of functional TEM-1/PSE-4 chimeras possessing between 17 and 92 substitutions relative to TEM-1 β-lactamase. Circular dichroism and thermal scanning fluorimetry revealed that the chimeras were generally well folded. Despite harbouring important sequence variation relative to either of the two ‘parental’ β-lactamases, the chimeric β-lactamases displayed substrate recognition spectra and reactivity similar to their most closely-related parent. To gain further insight into the changes induced by chimerization, the chimera with 17 substitutions was investigated by NMR spin relaxation. While high order was conserved on the ps-ns timescale, a hallmark of class A β-lactamases, evidence of additional slow motions on the µs-ms timescale was extracted from model-free calculations. This is consistent with the greater number of resonances that could not be assigned in this chimera relative to the parental β-lactamases, and is consistent with this well-folded and functional chimeric β-lactamase displaying increased slow time-scale motions.
Collapse
Affiliation(s)
- Christopher M. Clouthier
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- Département de Chimie, Université de Montréal, Montréal, Québec, Canada
| | - Sébastien Morin
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- Département de Biochimie, Microbiologie et Bioinformatique, Université Laval, Laval Québec, Canada
| | - Sophie M. C. Gobeil
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- Département de Biochimie, Université de Montréal, Montréal, Québec, Canada
| | - Nicolas Doucet
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- INRS–Institut Armand-Frappier, Université du Québec, Laval, Québec, Canada
| | - Jonathan Blanchet
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- Département de Chimie, Université de Montréal, Montréal, Québec, Canada
| | - Elisabeth Nguyen
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- Département de Chimie, Université de Montréal, Montréal, Québec, Canada
| | - Stéphane M. Gagné
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- Département de Biochimie, Microbiologie et Bioinformatique, Université Laval, Laval Québec, Canada
| | - Joelle N. Pelletier
- PROTEO, the Québec Network for Research on Protein Structure, Function and Engineering, Université Laval, Laval, Québec, Canada
- Département de Chimie, Université de Montréal, Montréal, Québec, Canada
- Département de Biochimie, Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
37
|
Smith MA, Rentmeister A, Snow CD, Wu T, Farrow MF, Mingardon F, Arnold FH. A diverse set of family 48 bacterial glycoside hydrolase cellulases created by structure-guided recombination. FEBS J 2012; 279:4453-65. [DOI: 10.1111/febs.12032] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 10/10/2012] [Accepted: 10/12/2012] [Indexed: 12/01/2022]
Affiliation(s)
- Matthew A. Smith
- Division of Chemistry and Chemical Engineering; California Institute of Technology; Pasadena; CA; USA
| | | | | | - Timothy Wu
- Division of Chemistry and Chemical Engineering; California Institute of Technology; Pasadena; CA; USA
| | - Mary F. Farrow
- Division of Chemistry and Chemical Engineering; California Institute of Technology; Pasadena; CA; USA
| | - Florence Mingardon
- Division of Chemistry and Chemical Engineering; California Institute of Technology; Pasadena; CA; USA
| | - Frances H. Arnold
- Division of Chemistry and Chemical Engineering; California Institute of Technology; Pasadena; CA; USA
| |
Collapse
|
38
|
Steiner K, Schwab H. Recent advances in rational approaches for enzyme engineering. Comput Struct Biotechnol J 2012; 2:e201209010. [PMID: 24688651 PMCID: PMC3962183 DOI: 10.5936/csbj.201209010] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 11/29/2022] Open
Abstract
Enzymes are an attractive alternative in the asymmetric syntheses of chiral building blocks. To meet the requirements of industrial biotechnology and to introduce new functionalities, the enzymes need to be optimized by protein engineering. This article specifically reviews rational approaches for enzyme engineering and de novo enzyme design involving structure-based approaches developed in recent years for improvement of the enzymes’ performance, broadened substrate range, and creation of novel functionalities to obtain products with high added value for industrial applications.
Collapse
Affiliation(s)
- Kerstin Steiner
- ACIB GmbH, (Austrian Centre of Industrial Biotechnology), c/o TU Graz, 8010 Graz, Austria
| | - Helmut Schwab
- ACIB GmbH, (Austrian Centre of Industrial Biotechnology), c/o TU Graz, 8010 Graz, Austria ; Institute of Molecular Biotechnology, TU Graz, 8010 Graz, Austria
| |
Collapse
|
39
|
Romero PA, Arnold FH. Random field model reveals structure of the protein recombinational landscape. PLoS Comput Biol 2012; 8:e1002713. [PMID: 23055915 PMCID: PMC3464211 DOI: 10.1371/journal.pcbi.1002713] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/03/2012] [Indexed: 11/28/2022] Open
Abstract
We are interested in how intragenic recombination contributes to the evolution of proteins and how this mechanism complements and enhances the diversity generated by random mutation. Experiments have revealed that proteins are highly tolerant to recombination with homologous sequences (mutation by recombination is conservative); more surprisingly, they have also shown that homologous sequence fragments make largely additive contributions to biophysical properties such as stability. Here, we develop a random field model to describe the statistical features of the subset of protein space accessible by recombination, which we refer to as the recombinational landscape. This model shows quantitative agreement with experimental results compiled from eight libraries of proteins that were generated by recombining gene fragments from homologous proteins. The model reveals a recombinational landscape that is highly enriched in functional sequences, with properties dominated by a large-scale additive structure. It also quantifies the relative contributions of parent sequence identity, crossover locations, and protein fold to the tolerance of proteins to recombination. Intragenic recombination explores a unique subset of sequence space that promotes rapid molecular diversification and functional adaptation. Mutation and recombination are the primary sources of genetic variation in evolving populations. The relative benefit of these two diversification mechanisms and how they complement each other has been a long-standing question in evolutionary biology. While it is clear what types of genetic diversity these two mechanisms can create, a significant challenge is relating these sequence changes to changes in fitness. The fitness landscape, which describes this mapping from genotype to phenotype, is extraordinarily complex and defined over an incomprehensibly large space of sequences. Here, we develop a model of the landscape that relies not on the details of this mapping, but rather on the statistical relationships between sequences. By studying the expected values of landscape properties, we can gain insights into the structure of the landscape that are independent of the details of how genotype dictates phenotype. We use this random field model to understand how recombination explores a functionally enriched and diverse subset of protein sequence space.
Collapse
Affiliation(s)
| | - Frances H. Arnold
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Kumar A, Singh S. Directed evolution: tailoring biocatalysts for industrial applications. Crit Rev Biotechnol 2012; 33:365-78. [DOI: 10.3109/07388551.2012.716810] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
41
|
Goldsmith M, Tawfik DS. Directed enzyme evolution: beyond the low-hanging fruit. Curr Opin Struct Biol 2012; 22:406-12. [DOI: 10.1016/j.sbi.2012.03.010] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 03/14/2012] [Accepted: 03/14/2012] [Indexed: 12/26/2022]
|
42
|
Abstract
BACKGROUND DNA shuffling generates combinatorial libraries of chimeric genes by stochastically recombining parent genes. The resulting libraries are subjected to large-scale genetic selection or screening to identify those chimeras with favorable properties (e.g., enhanced stability or enzymatic activity). While DNA shuffling has been applied quite successfully, it is limited by its homology-dependent, stochastic nature. Consequently, it is used only with parents of sufficient overall sequence identity, and provides no control over the resulting chimeric library. RESULTS This paper presents efficient methods to extend the scope of DNA shuffling to handle significantly more diverse parents and to generate more predictable, optimized libraries. Our CODNS (cross-over optimization for DNA shuffling) approach employs polynomial-time dynamic programming algorithms to select codons for the parental amino acids, allowing for zero or a fixed number of conservative substitutions. We first present efficient algorithms to optimize the local sequence identity or the nearest-neighbor approximation of the change in free energy upon annealing, objectives that were previously optimized by computationally-expensive integer programming methods. We then present efficient algorithms for more powerful objectives that seek to localize and enhance the frequency of recombination by producing "runs" of common nucleotides either overall or according to the sequence diversity of the resulting chimeras. We demonstrate the effectiveness of CODNS in choosing codons and allocating substitutions to promote recombination between parents targeted in earlier studies: two GAR transformylases (41% amino acid sequence identity), two very distantly related DNA polymerases, Pol X and β (15%), and beta-lactamases of varying identity (26-47%). CONCLUSIONS Our methods provide the protein engineer with a new approach to DNA shuffling that supports substantially more diverse parents, is more deterministic, and generates more predictable and more diverse chimeric libraries.
Collapse
Affiliation(s)
- Lu He
- Dept of Computer Science, Dartmouth College, 6211 Sudikoff Laboratory, Hanover, NH 03755, USA
| | - Alan M Friedman
- Dept of Biological Sciences, Markey Center for Structural Biology, Purdue Cancer Center, and Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
| | - Chris Bailey-Kellogg
- Dept of Computer Science, Dartmouth College, 6211 Sudikoff Laboratory, Hanover, NH 03755, USA
| |
Collapse
|
43
|
Feng X, Sanchis J, Reetz MT, Rabitz H. Enhancing the efficiency of directed evolution in focused enzyme libraries by the adaptive substituent reordering algorithm. Chemistry 2012; 18:5646-54. [PMID: 22434591 DOI: 10.1002/chem.201103811] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Indexed: 11/11/2022]
Abstract
Directed evolution is a broadly successful strategy for protein engineering in the quest to enhance the stereoselectivity, activity, and thermostability of enzymes. To increase the efficiency of directed evolution based on iterative saturation mutagenesis, the adaptive substituent reordering algorithm (ASRA) is introduced here as an alternative to traditional quantitative structure-activity relationship (QSAR) methods for identifying potential protein mutants with desired properties from minimal sampling of focused libraries. The operation of ASRA depends on identifying the underlying regularity of the protein property landscape, allowing it to make predictions without explicit knowledge of the structure-property relationships. In a proof-of-principle study, ASRA identified all or most of the best enantioselective mutants among the synthesized epoxide hydrolase from Aspergillus niger, in the absence of peptide seeds with high E-values. ASRA even revealed a laboratory error from irregularities of the reordered E-value landscape alone.
Collapse
Affiliation(s)
- Xiaojiang Feng
- Department of Chemistry, Princeton University, New Jersey 08544, USA
| | | | | | | |
Collapse
|
44
|
He L, Friedman AM, Bailey-Kellogg C. A divide-and-conquer approach to determine the Pareto frontier for optimization of protein engineering experiments. Proteins 2012; 80:790-806. [PMID: 22180081 PMCID: PMC4939273 DOI: 10.1002/prot.23237] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 10/06/2011] [Accepted: 10/21/2011] [Indexed: 01/07/2023]
Abstract
In developing improved protein variants by site-directed mutagenesis or recombination, there are often competing objectives that must be considered in designing an experiment (selecting mutations or breakpoints): stability versus novelty, affinity versus specificity, activity versus immunogenicity, and so forth. Pareto optimal experimental designs make the best trade-offs between competing objectives. Such designs are not "dominated"; that is, no other design is better than a Pareto optimal design for one objective without being worse for another objective. Our goal is to produce all the Pareto optimal designs (the Pareto frontier), to characterize the trade-offs and suggest designs most worth considering, but to avoid explicitly considering the large number of dominated designs. To do so, we develop a divide-and-conquer algorithm, Protein Engineering Pareto FRontier (PEPFR), that hierarchically subdivides the objective space, using appropriate dynamic programming or integer programming methods to optimize designs in different regions. This divide-and-conquer approach is efficient in that the number of divisions (and thus calls to the optimizer) is directly proportional to the number of Pareto optimal designs. We demonstrate PEPFR with three protein engineering case studies: site-directed recombination for stability and diversity via dynamic programming, site-directed mutagenesis of interacting proteins for affinity and specificity via integer programming, and site-directed mutagenesis of a therapeutic protein for activity and immunogenicity via integer programming. We show that PEPFR is able to effectively produce all the Pareto optimal designs, discovering many more designs than previous methods. The characterization of the Pareto frontier provides additional insights into the local stability of design choices as well as global trends leading to trade-offs between competing criteria.
Collapse
Affiliation(s)
- Lu He
- Department of Computer Science, Dartmouth College, Hanover NH 03755
| | - Alan M. Friedman
- Department of Biological Sciences, Markey Center for Structural Biology, Purdue Cancer Center, and Bindley Bioscience Center, Purdue University
| | | |
Collapse
|
45
|
Parker AS, Griswold KE, Bailey-Kellogg C. Optimization of combinatorial mutagenesis. J Comput Biol 2011; 18:1743-56. [PMID: 21923411 DOI: 10.1089/cmb.2011.0152] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Protein engineering by combinatorial site-directed mutagenesis evaluates a portion of the sequence space near a target protein, seeking variants with improved properties (e.g., stability, activity, immunogenicity). In order to improve the hit-rate of beneficial variants in such mutagenesis libraries, we develop methods to select optimal positions and corresponding sets of the mutations that will be used, in all combinations, in constructing a library for experimental evaluation. Our approach, OCoM (Optimization of Combinatorial Mutagenesis), encompasses both degenerate oligonucleotides and specified point mutations, and can be directed accordingly by requirements of experimental cost and library size. It evaluates the quality of the resulting library by one- and two-body sequence potentials, averaged over the variants. To ensure that it is not simply recapitulating extant sequences, it balances the quality of a library with an explicit evaluation of the novelty of its members. We show that, despite dealing with a combinatorial set of variants, in our approach the resulting library optimization problem is actually isomorphic to single-variant optimization. By the same token, this means that the two-body sequence potential results in an NP-hard optimization problem. We present an efficient dynamic programming algorithm for the one-body case and a practically-efficient integer programming approach for the general two-body case. We demonstrate the effectiveness of our approach in designing libraries for three different case study proteins targeted by previous combinatorial libraries--a green fluorescent protein, a cytochrome P450, and a beta lactamase. We found that OCoM worked quite efficiently in practice, requiring only 1 hour even for the massive design problem of selecting 18 mutations to generate 10⁷ variants of a 443-residue P450. We demonstrate the general ability of OCoM in enabling the protein engineer to explore and evaluate trade-offs between quality and novelty as well as library construction technique, and identify optimal libraries for experimental evaluation.
Collapse
Affiliation(s)
- Andrew S Parker
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire, USA
| | | | | |
Collapse
|
46
|
Martin DP, Lefeuvre P, Varsani A, Hoareau M, Semegni JY, Dijoux B, Vincent C, Reynaud B, Lett JM. Complex recombination patterns arising during geminivirus coinfections preserve and demarcate biologically important intra-genome interaction networks. PLoS Pathog 2011; 7:e1002203. [PMID: 21949649 PMCID: PMC3174254 DOI: 10.1371/journal.ppat.1002203] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 06/24/2011] [Indexed: 02/05/2023] Open
Abstract
Genetic recombination is an important process during the evolution of many virus species and occurs particularly frequently amongst begomoviruses in the single stranded DNA virus family, Geminiviridae. As in many other recombining viruses it is apparent that non-random recombination breakpoint distributions observable within begomovirus genomes sampled from nature are the product of variations both in basal recombination rates across genomes and in the over-all viability of different recombinant genomes. Whereas factors influencing basal recombination rates might include local degrees of sequence similarity between recombining genomes, nucleic acid secondary structures and genomic sensitivity to nuclease attack or breakage, the viability of recombinant genomes could be influenced by the degree to which their co-evolved protein-protein and protein-nucleotide and nucleotide-nucleotide interactions are disreputable by recombination. Here we investigate patterns of recombination that occur over 120 day long experimental infections of tomato plants with the begomoviruses Tomato yellow leaf curl virus and Tomato leaf curl Comoros virus. We show that patterns of sequence exchange between these viruses can be extraordinarily complex and present clear evidence that factors such as local degrees of sequence similarity but not genomic secondary structure strongly influence where recombination breakpoints occur. It is also apparent from our experiment that over-all patterns of recombination are strongly influenced by selection against individual recombinants displaying disrupted intra-genomic interactions such as those required for proper protein and nucleic acid folding. Crucially, we find that selection favoring the preservation of co-evolved longer-range protein-protein and protein DNA interactions is so strong that its imprint can even be used to identify the exact sequence tracts involved in these interactions. Genetic recombination between viruses is a form of parasexual reproduction during which two parental viruses each contribute genetic information to an offspring, or recombinant, virus. Unlike with sexual reproduction, however, recombination in viruses can even involve the transfer of sequences between the members of distantly related species. When parental genomes are very distantly related, it is anticipated that recombination between them runs the risk of producing defective offspring. The reason for this is that the interactions between different parts of genomes and the proteins they encode (such as between different viral proteins or between viral proteins and the virus genomic DNA or RNA) often depend on particular co-evolved binding sites that recognize one another. When in a recombinant genome the partners in a binding site pair are each inherited from different parents there is a possibility that they will not interact with one another properly. Here we examine recombinant genomes arising during experimental mixed infections of two distantly related viruses to detect evidence that intra-genome interaction networks are broadly preserved in these genomes. We show this preservation is so strict that patterns of recombination in these viruses can even be used to identify the interacting regions within their genomes.
Collapse
MESH Headings
- Base Sequence
- Begomovirus/genetics
- Begomovirus/pathogenicity
- Coinfection
- DNA, Single-Stranded/chemistry
- DNA, Single-Stranded/genetics
- DNA, Single-Stranded/metabolism
- DNA, Viral/chemistry
- DNA, Viral/genetics
- DNA, Viral/metabolism
- Genome, Viral
- Solanum lycopersicum/virology
- Nucleic Acid Conformation
- Phylogeny
- Plant Diseases/virology
- Polymorphism, Genetic
- Protein Folding
- Recombination, Genetic
- Selection, Genetic
- Viral Proteins/chemistry
Collapse
Affiliation(s)
- Darren P Martin
- Computational Biology Group, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory, South Africa.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Alcolombri U, Elias M, Tawfik DS. Directed Evolution of Sulfotransferases and Paraoxonases by Ancestral Libraries. J Mol Biol 2011; 411:837-53. [DOI: 10.1016/j.jmb.2011.06.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/14/2011] [Accepted: 06/20/2011] [Indexed: 12/30/2022]
|
48
|
An Y, Chen L, Sun S, Lv A, Wu W. QuikChange shuffling: a convenient and robust method for site-directed mutagenesis and random recombination of homologous genes. N Biotechnol 2011; 28:320-5. [DOI: 10.1016/j.nbt.2011.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 02/12/2011] [Accepted: 03/02/2011] [Indexed: 11/25/2022]
|
49
|
Vuillaume F, Thébaud G, Urbino C, Forfert N, Granier M, Froissart R, Blanc S, Peterschmitt M. Distribution of the phenotypic effects of random homologous recombination between two virus species. PLoS Pathog 2011; 7:e1002028. [PMID: 21573141 PMCID: PMC3088723 DOI: 10.1371/journal.ppat.1002028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 02/28/2011] [Indexed: 11/24/2022] Open
Abstract
Recombination has an evident impact on virus evolution and emergence of new pathotypes, and has generated an immense literature. However, the distribution of phenotypic effects caused by genome-wide random homologous recombination has never been formally investigated. Previous data on the subject have promoted the implicit view that most viral recombinant genomes are likely to be deleterious or lethal if the nucleotide identity of parental sequences is below 90%. We decided to challenge this view by creating a bank of near-random recombinants between two viral species of the genus Begomovirus (Family Geminiviridae) exhibiting 82% nucleotide identity, and by testing infectivity and in planta accumulation of recombinant clones randomly extracted from this bank. The bank was created by DNA-shuffling-a technology initially applied to the random shuffling of individual genes, and here implemented for the first time to shuffle full-length viral genomes. Together with our previously described system allowing the direct cloning of full-length infectious geminivirus genomes, it provided a unique opportunity to generate hundreds of "mosaic" virus genomes, directly testable for infectivity. A subset of 47 randomly chosen recombinants was sequenced, individually inoculated into tomato plants, and compared with the parental viruses. Surprisingly, our results showed that all recombinants were infectious and accumulated at levels comparable or intermediate to that of the parental clones. This indicates that, in our experimental system, despite the fact that the parental genomes differ by nearly 20%, lethal and/or large deleterious effects of recombination are very rare, in striking contrast to the common view that has emerged from previous studies published on other viruses.
Collapse
Affiliation(s)
- Florence Vuillaume
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
| | - Gaël Thébaud
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
| | - Cica Urbino
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
| | - Nadège Forfert
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
| | - Martine Granier
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
| | - Rémy Froissart
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
- Laboratoire Maladies Infectieuses & Vecteurs: Écologie, Génétique, Évolution & Contrôle (MIVEGEC), CNRS-IRD-Université de Montpellier I, Agropolis, Montpellier, France
| | - Stéphane Blanc
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
| | - Michel Peterschmitt
- CIRAD, INRA, CNRS – Unité mixte de recherche Biologie & génétique des interactions plante-parasite (BGPI), Montpellier, France
| |
Collapse
|
50
|
Abstract
The best approach for creating libraries of functional proteins with large numbers of nondisruptive amino acid substitutions is protein recombination, in which structurally related polypeptides are swapped among homologous proteins. Unfortunately, as more distantly related proteins are recombined, the fraction of variants having a disrupted structure increases. One way to enrich the fraction of folded and potentially interesting chimeras in these libraries is to use computational algorithms to anticipate which structural elements can be swapped without disturbing the integrity of a protein's structure. Herein, we describe how the algorithm Schema uses the sequences and structures of the parent proteins recombined to predict the structural disruption of chimeras, and we outline how dynamic programming can be used to find libraries with a range of amino acid substitution levels that are enriched in variants with low Schema disruption.
Collapse
|