1
|
Muñiz-Trejo R, Park Y, Thornton JW. Robustness of Ancestral Sequence Reconstruction to Among-site and Among-lineage Evolutionary Heterogeneity. Mol Biol Evol 2025; 42:msaf084. [PMID: 40203289 PMCID: PMC12046983 DOI: 10.1093/molbev/msaf084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Ancestral sequence reconstruction is typically performed using homogeneous evolutionary models, which assume that the same substitution propensities affect all sites and lineages. These assumptions are routinely violated: heterogeneous structural and functional constraints favor different amino acids at different sites, and these constraints often change among lineages as epistatic substitutions accrue at other sites. To evaluate how violations of the homogeneity assumption affect ancestral sequence reconstruction under realistic conditions, we developed site-specific substitution models and parameterized them using data from deep mutational scanning experiments on three protein families; we then used these models to perform ancestral sequence reconstruction on the empirical alignments and on alignments simulated under heterogeneous conditions derived from the experiments. Extensive among-site and -lineage heterogeneity is present in these datasets, but the sequences reconstructed from empirical alignments are almost identical when heterogeneous or homogeneous models are used for ancestral sequence reconstruction. Using models fit to deep mutational scanning data from distantly related proteins in which mutational effects are very different also has a minimal impact on ancestral sequence reconstruction. The rare differences occur primarily where phylogenetic signal is weak-at fast-evolving sites and nodes connected by long branches. When ancestral sequence reconstruction is performed on simulated data, errors in the reconstructed sequences become more likely as branch lengths increase, but incorporating heterogeneity into the model does not improve accuracy. These data establish that ancestral sequence reconstruction is robust to unincorporated realistic forms of evolutionary heterogeneity, because the primary determinant of ancestral sequence reconstruction is phylogenetic signal, not the substitution model. The best way to improve accuracy is therefore not to develop more elaborate models but to apply ancestral sequence reconstruction to densely sampled alignments that maximize phylogenetic signal at the nodes of interest.
Collapse
Affiliation(s)
- Ricardo Muñiz-Trejo
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | - Yeonwoo Park
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
| | - Joseph W Thornton
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Al-Nakhle H, Al-Shahrani R, Al-Ahmadi J, Al-Madani W, Al-Juhani R. Integrative In Silico Analysis to Identify Functional and Structural Impacts of nsSNPs on Programmed Cell Death Protein 1 (PD-1) Protein and UTRs: Potential Biomarkers for Cancer Susceptibility. Genes (Basel) 2025; 16:307. [PMID: 40149458 PMCID: PMC11942535 DOI: 10.3390/genes16030307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Programmed cell death protein 1 (PD-1), encoded by the PDCD1 gene, is critical in immune checkpoint regulation and cancer immune evasion. Variants in PDCD1 may alter its function, impacting cancer susceptibility and disease progression. Objectives: This study evaluates the structural, functional, and regulatory impacts of non-synonymous single-nucleotide polymorphisms (nsSNPs) in the PDCD1 gene, focusing on their pathogenic and oncogenic roles. Methods: Computational tools, including PredictSNP1.0, I-Mutant2.0, MUpro, HOPE, MutPred2, Cscape, Cscape-Somatic, GEPIA2, cBioPortal, and STRING, were used to analyze 695 nsSNPs in the PD1 protein. The analysis covered structural impacts, stability changes, regulatory effects, and oncogenic potential, focusing on conserved domains and protein-ligand interactions. Results: The analysis identified 84 deleterious variants, with 45 mapped to conserved regions like the Ig V-set domain essential for ligand-binding interactions. Stability analyses identified 78 destabilizing variants with significant protein instability (ΔΔG values). Ten nsSNPs were identified as potential cancer drivers. Expression profiling showed differential PDCD1 expression in tumor versus normal tissues, correlating with improved survival in skin melanoma but limited value in ovarian cancer. Regulatory SNPs disrupted miRNA-binding sites and transcriptional regulation, affecting PDCD1 expression. STRING analysis revealed key PD-1 protein partners within immune pathways, including PD-L1 and PD-L2. Conclusions: This study highlights the significance of PDCD1 nsSNPs as potential biomarkers for cancer susceptibility, advancing the understanding of PD-1 regulation. Experimental validation and multi-omics integration are crucial to refine these findings and enhance theraputic strategies.
Collapse
Affiliation(s)
- Hakeemah Al-Nakhle
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah 42353, Saudi Arabia
| | | | | | | | | |
Collapse
|
3
|
Diaz Arenas C, Alvarez M, Wilson RH, Shakhnovich EI, Ogbunugafor CB. Protein Quality Control is a Master Modulator of Molecular Evolution in Bacteria. Genome Biol Evol 2025; 17:evaf010. [PMID: 39837347 PMCID: PMC11789785 DOI: 10.1093/gbe/evaf010] [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: 05/31/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
The bacterial protein quality control (PQC) network comprises a set of genes that promote proteostasis (proteome homeostasis) through proper protein folding and function via chaperones, proteases, and protein translational machinery. It participates in vital cellular processes and influences organismal development and evolution. In this review, we examine the mechanistic bases for how the bacterial PQC network influences molecular evolution. We discuss the relevance of PQC components to contemporary issues in evolutionary biology including epistasis, evolvability, and the navigability of protein space. We examine other areas where proteostasis affects aspects of evolution and physiology, including host-parasite interactions. More generally, we demonstrate that the study of bacterial systems can aid in broader efforts to understand the relationship between genotype and phenotype across the biosphere.
Collapse
Affiliation(s)
- Carolina Diaz Arenas
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
| | - Maristella Alvarez
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
| | - Robert H Wilson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eugene I Shakhnovich
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - C Brandon Ogbunugafor
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| |
Collapse
|
4
|
Hermans P, Tsishyn M, Schwersensky M, Rooman M, Pucci F. Exploring Evolution to Uncover Insights Into Protein Mutational Stability. Mol Biol Evol 2025; 42:msae267. [PMID: 39786559 PMCID: PMC11721782 DOI: 10.1093/molbev/msae267] [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: 05/28/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/12/2025] Open
Abstract
Determining the impact of mutations on the thermodynamic stability of proteins is essential for a wide range of applications such as rational protein design and genetic variant interpretation. Since protein stability is a major driver of evolution, evolutionary data are often used to guide stability predictions. Many state-of-the-art stability predictors extract evolutionary information from multiple sequence alignments of proteins homologous to a query protein, and leverage it to predict the effects of mutations on protein stability. To evaluate the power and the limitations of such methods, we used the massive amount of stability data recently obtained by deep mutational scanning to study how best to construct multiple sequence alignments and optimally extract evolutionary information from them. We tested different evolutionary models and found that, unexpectedly, independent-site models achieve similar accuracy to more complex epistatic models. A detailed analysis of the latter models suggests that their inference often results in noisy couplings, which do not appear to add predictive power over the independent-site contribution, at least in the context of stability prediction. Interestingly, by combining any of the evolutionary features with a simple structural feature, the relative solvent accessibility of the mutated residue, we achieved similar prediction accuracy to supervised, machine learning-based, protein stability change predictors. Our results provide new insights into the relationship between protein evolution and stability, and show how evolutionary information can be exploited to improve the performance of mutational stability prediction.
Collapse
Affiliation(s)
- Pauline Hermans
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Matsvei Tsishyn
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Martin Schwersensky
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Marianne Rooman
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Fabrizio Pucci
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| |
Collapse
|
5
|
Muñiz-Trejo R, Park Y, Thornton JW. Robustness of ancestral sequence reconstruction to among-site evolutionary heterogeneity and epistasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629812. [PMID: 39763774 PMCID: PMC11702759 DOI: 10.1101/2024.12.20.629812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Ancestral sequence reconstruction (ASR) is typically performed using homogeneous evolutionary models, which assume that the same substitution propensities affect all sites and lineages. These assumptions are routinely violated: heterogeneous structural and functional constraints favor different amino acid states at different sites, and these constraints often change among lineages as epistatic substitutions accrue at other sites. To evaluate how realistic violations of the homogeneity assumption affect ASR, we developed site-specific substitution models and parameterized them using data from deep mutational scanning experiments on three protein families; we then used these models to perform ASR on the empirical alignments and on alignments simulated under heterogeneous conditions derived from the experiments. Extensive among-site and -lineage heterogeneity is present in these datasets, but the sequences reconstructed from empirical alignments are almost identical, irrespective of whether heterogeneous or homogeneous models are used for ASR. The rare differences occur primarily when phylogenetic signal is weak - at fast-evolving sites and nodes connected by long branches. When ASR is performed on simulated data, errors in the reconstructed sequences become more likely as branch lengths increase, but incorporating heterogeneity into the model does not improve accuracy. These data establish that ASR is robust to unincorporated realistic forms of evolutionary heterogeneity, because the primary determinant of ASR is phylogenetic signal, not the substitution model. The best way to improve accuracy is therefore not to develop more elaborate models but to apply ASR to densely sampled alignments that maximize phylogenetic signal at the nodes of interest.
Collapse
Affiliation(s)
- Ricardo Muñiz-Trejo
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
| | - Yeonwoo Park
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
| | - Joseph W Thornton
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| |
Collapse
|
6
|
Latrille T, Joseph J, Hartasánchez DA, Salamin N. Estimating the proportion of beneficial mutations that are not adaptive in mammals. PLoS Genet 2024; 20:e1011536. [PMID: 39724093 PMCID: PMC11709321 DOI: 10.1371/journal.pgen.1011536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/08/2025] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Mutations can be beneficial by bringing innovation to their bearer, allowing them to adapt to environmental change. These mutations are typically unpredictable since they respond to an unforeseen change in the environment. However, mutations can also be beneficial because they are simply restoring a state of higher fitness that was lost due to genetic drift in a stable environment. In contrast to adaptive mutations, these beneficial non-adaptive mutations can be predicted if the underlying fitness landscape is stable and known. The contribution of such non-adaptive mutations to molecular evolution has been widely neglected mainly because their detection is very challenging. We have here reconstructed protein-coding gene fitness landscapes shared between mammals, using mutation-selection models and a multi-species alignments across 87 mammals. These fitness landscapes have allowed us to predict the fitness effect of polymorphisms found in 28 mammalian populations. Using methods that quantify selection at the population level, we have confirmed that beneficial non-adaptive mutations are indeed positively selected in extant populations. Our work confirms that deleterious substitutions are accumulating in mammals and are being reverted, generating a balance in which genomes are damaged and restored simultaneously at different loci. We observe that beneficial non-adaptive mutations represent between 15% and 45% of all beneficial mutations in 24 of 28 populations analyzed, suggesting that a substantial part of ongoing positive selection is not driven solely by adaptation to environmental change in mammals.
Collapse
Affiliation(s)
- Thibault Latrille
- Department of Computational Biology, Université de Lausanne, Lausanne, Switzerland
| | - Julien Joseph
- Laboratoire de Biométrie et Biologie Evolutive, UMR5558, Université Lyon 1, Villeurbanne, France
| | | | - Nicolas Salamin
- Department of Computational Biology, Université de Lausanne, Lausanne, Switzerland
| |
Collapse
|
7
|
Afzal A, Jamshaid H, Badshah Y, Shabbir M, Trembley JH, Zafar S, Kamal GM, Afsar T, Husain FM, Razak S. Investigating the role of non-synonymous variant D67N of ADGRE2 in chronic myeloid leukemia. BMC Cancer 2024; 24:1354. [PMID: 39501172 PMCID: PMC11536965 DOI: 10.1186/s12885-024-13108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/24/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Chronic myeloid leukaemia (CML) is a type of blood cancer that begins in the hematopoietic stem cells. It is primarily characterized by a specific chromosomal aberration, the Philadelphia chromosome. While the fusion gene is a major contributor to CML, several other genes including ADGRE2, that are reported as highly expressed in hematopoietic stem cells and could be utilized as a therapeutic marker in leukemic patients are implicated in the disease's progression. Until recently, little research had been conducted to identify single nucleotide polymorphisms (SNPs) associated with CML. Therefore, this study aims to investigate the influence of non-synonymous variants on the structure and function of the gene encoding adhesion G protein-coupled receptor E2, ADGRE2, and to evaluate their association with CML and its clinical and pathological characteristics. METHODS Non-synonymous SNPs of ADGRE2 were retrieved from the ENSEMBL, COSMIC, and gnomAD genome browsers, and the pathogenicity of deleterious variants was assessed using several established computational tools, including SIFT, CADD, REVEL, PolyPhen, and MetaLR. RESULTS Various in silico analyses explored the impact of damaging SNP on the function, stability, and structure of EGF-like modules containing mucin-like hormone receptor-like2 (EMR2) protein encoded by the ADGRE2 gene. Genotype analysis was performed on collected blood samples, revealing that altered genotype TT of variant rs765071211 (C/T) was associated significantly with CML patients compared to the control. Further in vitro and in vivo analyses suggest that this SNP holds potential for clinical translation.
Collapse
Affiliation(s)
- Ayesha Afzal
- Department of Healthcare Biotechnology, Atta Ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Harooma Jamshaid
- Department of Healthcare Biotechnology, Atta Ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Yasmin Badshah
- Department of Healthcare Biotechnology, Atta Ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| | - Maria Shabbir
- Department of Healthcare Biotechnology, Atta Ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Janeen H Trembley
- Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Sameen Zafar
- Department of Healthcare Biotechnology, Atta Ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Ghulam Murtaza Kamal
- Department of Healthcare Biotechnology, Atta Ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
8
|
Vila JA. The origin of mutational epistasis. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:473-480. [PMID: 39443382 DOI: 10.1007/s00249-024-01725-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
The interconnected processes of protein folding, mutations, epistasis, and evolution have all been the subject of extensive analysis throughout the years due to their significance for structural and evolutionary biology. The origin (molecular basis) of epistasis-the non-additive interactions between mutations-is still, nonetheless, unknown. The existence of a new perspective on protein folding, a problem that needs to be conceived as an 'analytic whole', will enable us to shed light on the origin of mutational epistasis at the simplest level-within proteins-while also uncovering the reasons why the genetic background in which they occur, a key component of molecular evolution, could foster changes in epistasis effects. Additionally, because mutations are the source of epistasis, more research is needed to determine the impact of post-translational modifications, which can potentially increase the proteome's diversity by several orders of magnitude, on mutational epistasis and protein evolvability. Finally, a protein evolution thermodynamic-based analysis that does not consider specific mutational steps or epistasis effects will be briefly discussed. Our study explores the complex processes behind the evolution of proteins upon mutations, clearing up some previously unresolved issues, and providing direction for further research.
Collapse
Affiliation(s)
- Jorge A Vila
- IMASL-CONICET, Ejército de Los Andes 950, 5700, San Luis, Argentina.
| |
Collapse
|
9
|
Majid I, Sergeev YV. Linking Protein Stability to Pathogenicity: Predicting Clinical Significance of Single-Missense Mutations in Ocular Proteins Using Machine Learning. Int J Mol Sci 2024; 25:11649. [PMID: 39519200 PMCID: PMC11546782 DOI: 10.3390/ijms252111649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Understanding the effect of single-missense mutations on protein stability is crucial for clinical decision-making and therapeutic development. The impact of these mutations on protein stability and 3D structure remains underexplored. Here, we developed a program to investigate the relationship between pathogenic mutations with protein unfolding and compared seven machine learning (ML) models to predict the clinical significance of single-missense mutations with unknown impacts, based on protein stability parameters. We analyzed seven proteins associated with ocular disease-causing genes. The program revealed an R-squared value of 0.846 using Decision Tree Regression between pathogenic mutations and decreased protein stability, with 96.20% of pathogenic mutations in RPE65 leading to protein instability. Among the ML models, Random Forest achieved the highest AUC (0.922) and PR AUC (0.879) in predicting the clinical significance of mutations with unknown effects. Our findings indicate that most pathogenic mutations affecting protein stability occur in alpha-helices, beta-pleated sheets, and active sites. This study suggests that protein stability can serve as a valuable parameter for interpreting the clinical significance of single-missense mutations in ocular proteins.
Collapse
Affiliation(s)
| | - Yuri V. Sergeev
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institute of Health, Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Joseph J. Increased Positive Selection in Highly Recombining Genes Does not Necessarily Reflect an Evolutionary Advantage of Recombination. Mol Biol Evol 2024; 41:msae107. [PMID: 38829800 PMCID: PMC11173204 DOI: 10.1093/molbev/msae107] [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: 01/30/2024] [Revised: 04/08/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024] Open
Abstract
It is commonly thought that the long-term advantage of meiotic recombination is to dissipate genetic linkage, allowing natural selection to act independently on different loci. It is thus theoretically expected that genes with higher recombination rates evolve under more effective selection. On the other hand, recombination is often associated with GC-biased gene conversion (gBGC), which theoretically interferes with selection by promoting the fixation of deleterious GC alleles. To test these predictions, several studies assessed whether selection was more effective in highly recombining genes (due to dissipation of genetic linkage) or less effective (due to gBGC), assuming a fixed distribution of fitness effects (DFE) for all genes. In this study, I directly derive the DFE from a gene's evolutionary history (shaped by mutation, selection, drift, and gBGC) under empirical fitness landscapes. I show that genes that have experienced high levels of gBGC are less fit and thus have more opportunities for beneficial mutations. Only a small decrease in the genome-wide intensity of gBGC leads to the fixation of these beneficial mutations, particularly in highly recombining genes. This results in increased positive selection in highly recombining genes that is not caused by more effective selection. Additionally, I show that the death of a recombination hotspot can lead to a higher dN/dS than its birth, but with substitution patterns biased towards AT, and only at selected positions. This shows that controlling for a substitution bias towards GC is therefore not sufficient to rule out the contribution of gBGC to signatures of accelerated evolution. Finally, although gBGC does not affect the fixation probability of GC-conservative mutations, I show that by altering the DFE, gBGC can also significantly affect nonsynonymous GC-conservative substitution patterns.
Collapse
Affiliation(s)
- Julien Joseph
- Laboratoire de Biométrie et Biologie Evolutive, Université Lyon 1, CNRS, UMR 5558, Villeurbanne, France
| |
Collapse
|
11
|
Metzger BPH, Park Y, Starr TN, Thornton JW. Epistasis facilitates functional evolution in an ancient transcription factor. eLife 2024; 12:RP88737. [PMID: 38767330 PMCID: PMC11105156 DOI: 10.7554/elife.88737] [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: 05/22/2024] Open
Abstract
A protein's genetic architecture - the set of causal rules by which its sequence produces its functions - also determines its possible evolutionary trajectories. Prior research has proposed that the genetic architecture of proteins is very complex, with pervasive epistatic interactions that constrain evolution and make function difficult to predict from sequence. Most of this work has analyzed only the direct paths between two proteins of interest - excluding the vast majority of possible genotypes and evolutionary trajectories - and has considered only a single protein function, leaving unaddressed the genetic architecture of functional specificity and its impact on the evolution of new functions. Here, we develop a new method based on ordinal logistic regression to directly characterize the global genetic determinants of multiple protein functions from 20-state combinatorial deep mutational scanning (DMS) experiments. We use it to dissect the genetic architecture and evolution of a transcription factor's specificity for DNA, using data from a combinatorial DMS of an ancient steroid hormone receptor's capacity to activate transcription from two biologically relevant DNA elements. We show that the genetic architecture of DNA recognition consists of a dense set of main and pairwise effects that involve virtually every possible amino acid state in the protein-DNA interface, but higher-order epistasis plays only a tiny role. Pairwise interactions enlarge the set of functional sequences and are the primary determinants of specificity for different DNA elements. They also massively expand the number of opportunities for single-residue mutations to switch specificity from one DNA target to another. By bringing variants with different functions close together in sequence space, pairwise epistasis therefore facilitates rather than constrains the evolution of new functions.
Collapse
Affiliation(s)
- Brian PH Metzger
- Department of Ecology and Evolution, University of ChicagoChicagoUnited States
| | - Yeonwoo Park
- Program in Genetics, Genomics, and Systems Biology, University of ChicagoChicagoUnited States
| | - Tyler N Starr
- Department of Biochemistry and Molecular Biophysics, University of ChicagoChicagoUnited States
| | - Joseph W Thornton
- Department of Ecology and Evolution, University of ChicagoChicagoUnited States
- Department of Human Genetics, University of ChicagoChicagoUnited States
| |
Collapse
|
12
|
Araujo NA, Bubis J. Analysis of a Novel Peptide That Is Capable of Inhibiting the Enzymatic Activity of the Protein Kinase A Catalytic Subunit-Like Protein from Trypanosoma equiperdum. Protein J 2023; 42:709-727. [PMID: 37713008 DOI: 10.1007/s10930-023-10153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/16/2023]
Abstract
A 26-residue peptide possessing the αN-helix motif of the protein kinase A (PKA) regulatory subunit-like proteins from the Trypanozoom subgenera (VAP26, sequence = VAPYFEKSEDETALILKLLTYNVLFS), was shown to inhibit the enzymatic activity of the Trypanosoma equiperdum PKA catalytic subunit-like protein, in a similar manner that the mammalian heat-stable soluble PKA inhibitor known as PKI. However, VAP26 does not contain the PKI inhibitory sequence. Bioinformatics analyzes of the αN-helix motif from various Trypanozoon PKA regulatory subunit-like proteins suggested that the sequence could form favorable peptide-protein interactions of hydrophobic nature with the PKA catalytic subunit-like protein, which possibly may represent an alternative PKA inhibitory mechanism. The sequence of the αN-helix motif of the Trypanozoon proteins was shown to be highly homologous but significantly divergent from the corresponding αN-helix motifs of their Leishmania and mammalian counterparts. This sequence divergence contrasted with the proposed secondary structure of the αN-helix motif, which appeared conserved in every analyzed regulatory subunit-like protein. In silico mutation experiments at positions I234, L238 and F244 of the αN-helix motif from the Trypanozoon proteins destabilized both the specific motif and the protein. On the contrary, mutations at positions T239 and Y240 stabilized the motif and the protein. These results suggested that the αN-helix motif from the Trypanozoon proteins probably possessed a different evolutionary path than their Leishmania and mammalian counterparts. Moreover, finding stabilizing mutations indicated that new inhibitory peptides may be designed based on the αN-helix motif from the Trypanozoon PKA regulatory subunit-like proteins.
Collapse
Affiliation(s)
- Nelson A Araujo
- Escuela de Ciencias Agroalimentarias, Animales y Ambientales, Universidad de O'Higgins, Campus Colchagua, ruta I-90, Km 3, San Fernando, Chile.
| | - José Bubis
- Unidad de Polimorfismo Genético, Genómica y Proteómica, Dirección de Salud, Fundación Instituto de Estudios Avanzados IDEA, Caracas, 1015-A, Venezuela
- Unidad de Señalización Celular y Bioquímica de Parásitos, Dirección de Salud, Fundación Instituto de Estudios Avanzados IDEA, Caracas, 1015-A, Venezuela
- Departamento de Biología Celular, Universidad Simón Bolívar, Apartado 89.000, Caracas, 1081‑A, Venezuela
| |
Collapse
|
13
|
Haddox HK, Galloway JG, Dadonaite B, Bloom JD, Matsen IV FA, DeWitt WS. Jointly modeling deep mutational scans identifies shifted mutational effects among SARS-CoV-2 spike homologs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.31.551037. [PMID: 37577604 PMCID: PMC10418112 DOI: 10.1101/2023.07.31.551037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Deep mutational scanning (DMS) is a high-throughput experimental technique that measures the effects of thousands of mutations to a protein. These experiments can be performed on multiple homologs of a protein or on the same protein selected under multiple conditions. It is often of biological interest to identify mutations with shifted effects across homologs or conditions. However, it is challenging to determine if observed shifts arise from biological signal or experimental noise. Here, we describe a method for jointly inferring mutational effects across multiple DMS experiments while also identifying mutations that have shifted in their effects among experiments. A key aspect of our method is to regularize the inferred shifts, so that they are nonzero only when strongly supported by the data. We apply this method to DMS experiments that measure how mutations to spike proteins from SARS-CoV-2 variants (Delta, Omicron BA.1, and Omicron BA.2) affect cell entry. Most mutational effects are conserved between these spike homologs, but a fraction have markedly shifted. We experimentally validate a subset of the mutations inferred to have shifted effects, and confirm differences of > 1,000-fold in the impact of the same mutation on spike-mediated viral infection across spikes from different SARS-CoV-2 variants. Overall, our work establishes a general approach for comparing sets of DMS experiments to identify biologically important shifts in mutational effects.
Collapse
Affiliation(s)
- Hugh K. Haddox
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98102, USA
| | - Jared G. Galloway
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98102, USA
| | - Bernadeta Dadonaite
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jesse D. Bloom
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98102, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98109, USA
| | - Frederick A. Matsen IV
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98102, USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98109, USA
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| | - William S. DeWitt
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
14
|
Du Y, Wang M, Chen Y, Deng Y, Zhang L, Bai T, Ji M. Occurrence and mechanism of target-site resistance to bensulfuron-methyl in Monochoria korsakowii from China. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 191:105379. [PMID: 36963949 DOI: 10.1016/j.pestbp.2023.105379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/18/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Monochoria korsakowii is an increasingly significant threat to rice production across China, particularly in Liaoning province. Few studies have reported herbicide resistance in M. korsakowii, and resistance status and mechanisms are poorly understood. Here, thirty field populations of M. korsakowii were collected from 11 rice-growing regions of Liaoning, and 97% of populations had evolved resistance to bensulfuron-methyl (BM), with majority (24 of 28) showing high resistance levels (RI > 10). The first in-depth analysis of molecular features of AHAS1 and AHAS2 in BM-resistant populations showed that four Pro197 mutations (Pro197 to His, Ala, Leu or Ser) in AHAS1 and one mutation (Pro197Ser) in AHAS2 were identified. Notably, novel double Pro197Ser mutations co-occurred in both AHAS1 and AHAS2 in the most resistant line LN-20. Furthermore, resistant mutants were used to investigate the effect of Pro197 mutations on AHAS functionality, binding modes, gene expression and cross-resistance in M. korsakowii. All the detected Pro197 mutations considerably reduced in vitro AHAS sensitivity to BM by weakening hydrogen bonds and hydrophobic interactions in the predicted BM-AHAS complexes, especially the double Pro197Ser mutations. This novel resistance mutation combination slightly impacted the extractable AHAS activity, and increased the affinity and catalytic rate of pyruvate. Also, the AHAS expression level was significantly up-regulated. Moreover, all mutations provided resistance only to other sulfonylureas herbicides but not triazolopyrimidine or pyrimidinyl-benzoates herbicides. In conclusion, bensulfuron-methyl resistance in M. korsakowii was grim in Liaoning, China, and amino acid mutations on AHAS isozymes were the primary resistance mechanism. Double Pro197Ser mutations in both AHAS1 and AHAS2 confer higher herbicide resistance than single mutations in AHAS1. Thus, this work deepens our understanding of resistance status and mechanisms of M. korsakowii.
Collapse
Affiliation(s)
- Ying Du
- College of Plant Protection, Shenyang Agricultural University, Shenyang 110866, China
| | - Minlong Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yan Chen
- Institute of Plant Protection, Liaoning Academy of Agricultural Sciences, Shenyang 110161, China
| | - Yunyan Deng
- College of Plant Protection, Shenyang Agricultural University, Shenyang 110866, China
| | - Lulu Zhang
- College of Plant Protection, Shenyang Agricultural University, Shenyang 110866, China
| | - Tianlang Bai
- College of Plant Protection, Shenyang Agricultural University, Shenyang 110866, China
| | - Mingshan Ji
- College of Plant Protection, Shenyang Agricultural University, Shenyang 110866, China.
| |
Collapse
|
15
|
Biswas S, Bagchi A. Analysis of the structural dynamics of the mutations in the kinase domain of PINK1 protein associated with Parkinson's disease. Gene 2023; 857:147183. [PMID: 36623675 DOI: 10.1016/j.gene.2023.147183] [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: 11/11/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
Parkinson's disease (PD) is a very common neurodegenerative disorder and is considered to be one of the most severe disorders worldwide. Mutations in some PD causing genes are responsible for the early onset of the disease. Pathogenic variants in parkin, PINK1 and DJ1 genes can cause early-onset of PD. Many PINK1 gene mutations have been reported, but not all variants are pathogenic. The gene product of PINK1, also known as PINK1 protein, has 581 amino acid residues in it. Several different mutations are present throughout the kinase domain of PINK1 protein. In this work, we used in silico approaches to analyze the different types of mutations that are distributed in the kinase domain of the PINK1 protein. Based on our results, we categorized the mutations as high, moderate and low pathogenic variants. Furthermore, we performed molecular dynamics simulations of the pathogenic PINK1 variants to decipher their possible impacts on the structure and made a comparison with the wild type PINK1. In conclusion, we suggested the possible mechanistic roles of the pathogenic variants of PINK1 kinase domain that can affect its function. These pathogenic variants are the causative agents of early onset of PD called autosomal recessive Parkinson disease.
Collapse
Affiliation(s)
- Sima Biswas
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani - 741235, Nadia, West Bengal, India
| | - Angshuman Bagchi
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani - 741235, Nadia, West Bengal, India.
| |
Collapse
|
16
|
Johansson KE, Lindorff-Larsen K, Winther JR. Global Analysis of Multi-Mutants to Improve Protein Function. J Mol Biol 2023; 435:168034. [PMID: 36863661 DOI: 10.1016/j.jmb.2023.168034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023]
Abstract
The identification of amino acid substitutions that both enhance the stability and function of a protein is a key challenge in protein engineering. Technological advances have enabled assaying thousands of protein variants in a single high-throughput experiment, and more recent studies use such data in protein engineering. We present a Global Multi-Mutant Analysis (GMMA) that exploits the presence of multiply-substituted variants to identify individual amino acid substitutions that are beneficial for the stability and function across a large library of protein variants. We have applied GMMA to a previously published experiment reporting on >54,000 variants of green fluorescent protein (GFP), each with known fluorescence output, and each carrying 1-15 amino acid substitutions (Sarkisyan et al., 2016). The GMMA method achieves a good fit to this dataset while being analytically transparent. We show experimentally that the six top-ranking substitutions progressively enhance GFP. More broadly, using only a single experiment as input our analysis recovers nearly all the substitutions previously reported to be beneficial for GFP folding and function. In conclusion, we suggest that large libraries of multiply-substituted variants may provide a unique source of information for protein engineering.
Collapse
Affiliation(s)
- Kristoffer E Johansson
- Linderstrøm-Lang Centre for Protein Science, Section for Biomolecular Sciences, Department of Biology of (University of Copenhagen), Ole Maaloes Vej 5, DK-2200 Copenhagen N, Denmark.
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Section for Biomolecular Sciences, Department of Biology of (University of Copenhagen), Ole Maaloes Vej 5, DK-2200 Copenhagen N, Denmark.
| | - Jakob R Winther
- Linderstrøm-Lang Centre for Protein Science, Section for Biomolecular Sciences, Department of Biology of (University of Copenhagen), Ole Maaloes Vej 5, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
17
|
Ren L, Du W, Song D, Lu H, Hamblin MH, Wang C, Du C, Fan GC, Becker RC, Fan Y. Genetic ablation of diabetes-associated gene Ccdc92 reduces obesity and insulin resistance in mice. iScience 2023; 26:105769. [PMID: 36594018 PMCID: PMC9804112 DOI: 10.1016/j.isci.2022.105769] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple genome-wide association studies (GWAS) have identified specific genetic variants in the coiled-coil domain containing 92 (CCDC92) locus that is associated with obesity and type 2 diabetes in humans. However, the biological function of CCDC92 in obesity and insulin resistance remains to be explored. Utilizing wild-type (WT) and Ccdc92 whole-body knockout (KO) mice, we found that Ccdc92 KO reduced obesity and increased insulin sensitivity under high-fat diet (HFD) conditions. Ccdc92 KO inhibited macrophage infiltration and fibrosis in white adipose tissue (WAT), suggesting Ccdc92 ablation protects against adipose tissue dysfunction. Ccdc92 deletion also increased energy expenditure and further attenuated hepatic steatosis in mice on an HFD. Ccdc92 KO significantly inhibited the inflammatory response and suppressed the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome in WAT. Altogether, we demonstrated the critical role of CCDC92 in metabolism, constituting a potential target for treating obesity and insulin resistance.
Collapse
Affiliation(s)
- Lu Ren
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Wa Du
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Dan Song
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Haocheng Lu
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Milton H. Hamblin
- Tulane University Health Sciences Center, Tulane University, New Orleans, LA 70112, USA
- College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Chenran Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Chunying Du
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Richard C. Becker
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yanbo Fan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
18
|
Del Amparo R, González-Vázquez LD, Rodríguez-Moure L, Bastolla U, Arenas M. Consequences of Genetic Recombination on Protein Folding Stability. J Mol Evol 2023; 91:33-45. [PMID: 36463317 PMCID: PMC9849154 DOI: 10.1007/s00239-022-10080-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022]
Abstract
Genetic recombination is a common evolutionary mechanism that produces molecular diversity. However, its consequences on protein folding stability have not attracted the same attention as in the case of point mutations. Here, we studied the effects of homologous recombination on the computationally predicted protein folding stability for several protein families, finding less detrimental effects than we previously expected. Although recombination can affect multiple protein sites, we found that the fraction of recombined proteins that are eliminated by negative selection because of insufficient stability is not significantly larger than the corresponding fraction of proteins produced by mutation events. Indeed, although recombination disrupts epistatic interactions, the mean stability of recombinant proteins is not lower than that of their parents. On the other hand, the difference of stability between recombined proteins is amplified with respect to the parents, promoting phenotypic diversity. As a result, at least one third of recombined proteins present stability between those of their parents, and a substantial fraction have higher or lower stability than those of both parents. As expected, we found that parents with similar sequences tend to produce recombined proteins with stability close to that of the parents. Finally, the simulation of protein evolution along the ancestral recombination graph with empirical substitution models commonly used in phylogenetics, which ignore constraints on protein folding stability, showed that recombination favors the decrease of folding stability, supporting the convenience of adopting structurally constrained models when possible for inferences of protein evolutionary histories with recombination.
Collapse
Affiliation(s)
- Roberto Del Amparo
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain ,Departamento de Bioquímica, Genética e Inmunología, Universidade de Vigo, 36310 Vigo, Spain
| | - Luis Daniel González-Vázquez
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain ,Departamento de Bioquímica, Genética e Inmunología, Universidade de Vigo, 36310 Vigo, Spain
| | - Laura Rodríguez-Moure
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain ,Departamento de Bioquímica, Genética e Inmunología, Universidade de Vigo, 36310 Vigo, Spain
| | - Ugo Bastolla
- Centre for Molecular Biology Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | - Miguel Arenas
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain ,Departamento de Bioquímica, Genética e Inmunología, Universidade de Vigo, 36310 Vigo, Spain ,Galicia Sur Health Research Institute (IIS Galicia Sur), 36310 Vigo, Spain
| |
Collapse
|
19
|
Valanciute A, Nygaard L, Zschach H, Maglegaard Jepsen M, Lindorff-Larsen K, Stein A. Accurate protein stability predictions from homology models. Comput Struct Biotechnol J 2022; 21:66-73. [PMID: 36514339 PMCID: PMC9729920 DOI: 10.1016/j.csbj.2022.11.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Calculating changes in protein stability (ΔΔG) has been shown to be central for predicting the consequences of single amino acid substitutions in protein engineering as well as interpretation of genomic variants for disease risk. Structure-based calculations are considered most accurate, however the tools used to calculate ΔΔGs have been developed on experimentally resolved structures. Extending those calculations to homology models based on related proteins would greatly extend their applicability as large parts of e.g. the human proteome are not structurally resolved. In this study we aim to investigate the accuracy of ΔΔG values predicted on homology models compared to crystal structures. Specifically, we identified four proteins with a large number of experimentally tested ΔΔGs and templates for homology modeling across a broad range of sequence identities, and selected three methods for ΔΔG calculations to test. We find that ΔΔG-values predicted from homology models compare equally well to experimental ΔΔGs as those predicted on experimentally established crystal structures, as long as the sequence identity of the model template to the target protein is at least 40%. In particular, the Rosetta cartesian_ddg protocol is robust against the small perturbations in the structure which homology modeling introduces. In an independent assessment, we observe a similar trend when using ΔΔGs to categorize variants as low or wild-type-like abundance. Overall, our results show that stability calculations performed on homology models can substitute for those on crystal structures with acceptable accuracy as long as the model is built on a template with sequence identity of at least 40% to the target protein.
Collapse
Affiliation(s)
- Audrone Valanciute
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Nygaard
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henrike Zschach
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Maglegaard Jepsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark,Corresponding authors.
| | - Amelie Stein
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark,Corresponding authors.
| |
Collapse
|
20
|
Azbukina N, Zharikova A, Ramensky V. Intragenic compensation through the lens of deep mutational scanning. Biophys Rev 2022; 14:1161-1182. [PMID: 36345285 PMCID: PMC9636336 DOI: 10.1007/s12551-022-01005-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/26/2022] [Indexed: 12/20/2022] Open
Abstract
A significant fraction of mutations in proteins are deleterious and result in adverse consequences for protein function, stability, or interaction with other molecules. Intragenic compensation is a specific case of positive epistasis when a neutral missense mutation cancels effect of a deleterious mutation in the same protein. Permissive compensatory mutations facilitate protein evolution, since without them all sequences would be extremely conserved. Understanding compensatory mechanisms is an important scientific challenge at the intersection of protein biophysics and evolution. In human genetics, intragenic compensatory interactions are important since they may result in variable penetrance of pathogenic mutations or fixation of pathogenic human alleles in orthologous proteins from related species. The latter phenomenon complicates computational and clinical inference of an allele's pathogenicity. Deep mutational scanning is a relatively new technique that enables experimental studies of functional effects of thousands of mutations in proteins. We review the important aspects of the field and discuss existing limitations of current datasets. We reviewed ten published DMS datasets with quantified functional effects of single and double mutations and described rates and patterns of intragenic compensation in eight of them. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-022-01005-w.
Collapse
Affiliation(s)
- Nadezhda Azbukina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1-73, Leninskie Gory, 119991 Moscow, Russia
| | - Anastasia Zharikova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1-73, Leninskie Gory, 119991 Moscow, Russia
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigsky per., 10, Bld.3, 101000 Moscow, Russia
| | - Vasily Ramensky
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1-73, Leninskie Gory, 119991 Moscow, Russia
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigsky per., 10, Bld.3, 101000 Moscow, Russia
| |
Collapse
|
21
|
Samant N, Nachum G, Tsepal T, Bolon DNA. Sequence dependencies and biophysical features both govern cleavage of diverse cut-sites by HIV protease. Protein Sci 2022; 31:e4366. [PMID: 35762719 PMCID: PMC9207908 DOI: 10.1002/pro.4366] [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: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 11/12/2022]
Abstract
The infectivity of HIV-1 requires its protease (PR) cleave multiple cut-sites with low sequence similarity. The diversity of cleavage sites has made it challenging to investigate the underlying sequence properties that determine binding and turnover of substrates by PR. We engineered a mutational scanning approach utilizing yeast display, flow cytometry, and deep sequencing to systematically measure the impacts of all individual amino acid changes at 12 positions in three different cut-sites (MA/CA, NC/p1, and p1/p6). The resulting fitness landscapes revealed common physical features that underlie cutting of all three cut-sites at the amino acid positions closest to the scissile bond. In contrast, positions more than two amino acids away from the scissile bond exhibited a strong dependence on the sequence background of the rest of the cut-site. We observed multiple amino acid changes in cut-sites that led to faster cleavage rates, including a preference for negative charge five and six amino acids away from the scissile bond at locations where the surface of protease is positively charged. Analysis of individual cut sites using full-length matrix-capsid proteins indicate that long-distance sequence context can contribute to cutting efficiency such that analyses of peptides or shorter engineered constructs including those in this work should be considered carefully. This work provides a framework for understanding how diverse substrates interact with HIV-1 PR and can be extended to investigate other viral PRs with similar properties.
Collapse
Affiliation(s)
- Neha Samant
- Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Gily Nachum
- Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Tenzin Tsepal
- Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Daniel N. A. Bolon
- Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
22
|
Youssef N, Susko E, Roger AJ, Bielawski JP. Evolution of amino acid propensities under stability-mediated epistasis. Mol Biol Evol 2022; 39:6522130. [PMID: 35134997 PMCID: PMC8896634 DOI: 10.1093/molbev/msac030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Site-specific amino acid preferences are influenced by the genetic background of the protein. The preferences for resident amino acids are expected to, on average, increase over time because of replacements at other sites - a nonadaptive phenomenon referred to as the 'evolutionary Stokes shift'. Alternatively, decreases in resident amino acid propensity have recently been viewed as evidence of adaptations to external environmental changes. Using population genetics theory and thermodynamic stability-constraints, we show that nonadaptive evolution can lead to both positive and negative shifts in propensities following the fixation of an amino acid, emphasizing that the detection of negative shifts is not conclusive evidence of adaptation. Considering shifts in propensities over windows between substitutions at a focal site, we find that following ≈ 50% of substitutions the propensity for the new resident amino acid decreases over time, and both positive and negative shifts were comparable in magnitude. Preferences were often conserved via a significant negative autocorrelation in propensity changes-increases in propensities often followed by decreases, and vice versa. Lastly, we explore the underlying mechanisms that lead propensities to fluctuate. We observe that stabilizing replacements increase the mutational tolerance at a site and in doing so decrease the propensity for the resident amino acid. In contrast, destabilizing substitutions result in more rugged fitness landscapes that tend to favor the resident amino acid. In summary, our results characterize propensity trajectories under nonadaptive stability-constrained evolution against which evidence of adaptations should be calibrated.
Collapse
Affiliation(s)
- Noor Youssef
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Edward Susko
- Department of Mathematics and Statistics, Dalhousie University, Halifax, NS, Canada
| | - Andrew J Roger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Joseph P Bielawski
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada Department of Mathematics and Statistics, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
23
|
OUP accepted manuscript. Hum Mol Genet 2022; 31:2236-2261. [DOI: 10.1093/hmg/ddac029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/12/2022] Open
|
24
|
Youssef N, Susko E, Roger AJ, Bielawski JP. Shifts in amino acid preferences as proteins evolve: A synthesis of experimental and theoretical work. Protein Sci 2021; 30:2009-2028. [PMID: 34322924 PMCID: PMC8442975 DOI: 10.1002/pro.4161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 11/08/2022]
Abstract
Amino acid preferences vary across sites and time. While variation across sites is widely accepted, the extent and frequency of temporal shifts are contentious. Our understanding of the drivers of amino acid preference change is incomplete: To what extent are temporal shifts driven by adaptive versus nonadaptive evolutionary processes? We review phenomena that cause preferences to vary (e.g., evolutionary Stokes shift, contingency, and entrenchment) and clarify how they differ. To determine the extent and prevalence of shifted preferences, we review experimental and theoretical studies. Analyses of natural sequence alignments often detect decreases in homoplasy (convergence and reversions) rates, and variation in replacement rates with time-signals that are consistent with temporally changing preferences. While approaches inferring shifts in preferences from patterns in natural alignments are valuable, they are indirect since multiple mechanisms (both adaptive and nonadaptive) could lead to the observed signal. Alternatively, site-directed mutagenesis experiments allow for a more direct assessment of shifted preferences. They corroborate evidence from multiple sequence alignments, revealing that the preference for an amino acid at a site varies depending on the background sequence. However, shifts in preferences are usually minor in magnitude and sites with significantly shifted preferences are low in frequency. The small yet consistent perturbations in preferences could, nevertheless, jeopardize the accuracy of inference procedures, which assume constant preferences. We conclude by discussing if and how such shifts in preferences might influence widely used time-homogenous inference procedures and potential ways to mitigate such effects.
Collapse
Affiliation(s)
- Noor Youssef
- Department of BiologyDalhousie UniversityHalifaxNova ScotiaCanada
| | - Edward Susko
- Department of Mathematics and StatisticsDalhousie UniversityHalifaxNova ScotiaCanada
| | - Andrew J. Roger
- Department of Biochemistry and Molecular BiologyDalhousie UniversityHalifaxNova ScotiaCanada
| | - Joseph P. Bielawski
- Department of BiologyDalhousie UniversityHalifaxNova ScotiaCanada
- Department of Mathematics and StatisticsDalhousie UniversityHalifaxNova ScotiaCanada
| |
Collapse
|
25
|
Han AX, Felix Garza ZC, Welkers MRA, Vigeveno RM, Tran ND, Le TQM, Pham Quang T, Dang DT, Tran TNA, Ha MT, Nguyen TH, Le QT, Le TH, Hoang TBN, Chokephaibulkit K, Puthavathana P, Nguyen VVC, Nghiem MN, Nguyen VK, Dao TT, Tran TH, Wertheim HFL, Horby PW, Fox A, van Doorn HR, Eggink D, de Jong MD, Russell CA. Within-host evolutionary dynamics of seasonal and pandemic human influenza A viruses in young children. eLife 2021; 10:e68917. [PMID: 34342576 PMCID: PMC8382297 DOI: 10.7554/elife.68917] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/02/2021] [Indexed: 01/14/2023] Open
Abstract
The evolution of influenza viruses is fundamentally shaped by within-host processes. However, the within-host evolutionary dynamics of influenza viruses remain incompletely understood, in part because most studies have focused on infections in healthy adults based on single timepoint data. Here, we analyzed the within-host evolution of 82 longitudinally sampled individuals, mostly young children, infected with A/H1N1pdm09 or A/H3N2 viruses between 2007 and 2009. For A/H1N1pdm09 infections during the 2009 pandemic, nonsynonymous minority variants were more prevalent than synonymous ones. For A/H3N2 viruses in young children, early infection was dominated by purifying selection. As these infections progressed, nonsynonymous variants typically increased in frequency even when within-host virus titers decreased. Unlike the short-lived infections of adults where de novo within-host variants are rare, longer infections in young children allow for the maintenance of virus diversity via mutation-selection balance creating potentially important opportunities for within-host virus evolution.
Collapse
Affiliation(s)
- Alvin X Han
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Zandra C Felix Garza
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Matthijs RA Welkers
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - René M Vigeveno
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Nhu Duong Tran
- National Institute of Hygiene and EpidemiologyHanoiViet Nam
| | | | | | | | | | | | | | | | - Thanh Hai Le
- Vietnam National Children's HospitalHanoiViet Nam
| | | | | | | | | | | | | | | | - Tinh Hien Tran
- Siriraj Hospital, Mahidol UniversityBangkokThailand
- Oxford University Clinical Research UnitHo Chi Minh cityViet Nam
| | - Heiman FL Wertheim
- Oxford University Clinical Research UnitHo Chi Minh cityViet Nam
- Radboud Medical Centre, Radboud UniversityNijmegenNetherlands
- Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Peter W Horby
- Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Oxford University Clinical Research UnitHanoiViet Nam
| | - Annette Fox
- Oxford University Clinical Research UnitHanoiViet Nam
- Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
- WHO Collaborating Centre for Reference and Research on InfluenzaMelbourneAustralia
| | - H Rogier van Doorn
- Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Oxford University Clinical Research UnitHanoiViet Nam
| | - Dirk Eggink
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the EnvironmentBilthovenNetherlands
| | - Menno D de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Colin A Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| |
Collapse
|
26
|
Romero-Romero S, Costas M, Silva Manzano DA, Kordes S, Rojas-Ortega E, Tapia C, Guerra Y, Shanmugaratnam S, Rodríguez-Romero A, Baker D, Höcker B, Fernández-Velasco DA. The Stability Landscape of de novo TIM Barrels Explored by a Modular Design Approach. J Mol Biol 2021; 433:167153. [PMID: 34271011 PMCID: PMC8404036 DOI: 10.1016/j.jmb.2021.167153] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/18/2021] [Accepted: 07/06/2021] [Indexed: 11/25/2022]
Abstract
The TIM barrel is a versatile fold to understand structure-stability relationships. A collection of de novo TIM barrels with improved hydrophobic cores was designed. DeNovoTIMs are reversible in chemical and thermal unfolding, which is uncommon in TIM barrels. Epistatic effects play a central role in DeNovoTIMs stabilization. DeNovoTIMs navigate a previously uncharted region of the stability landscape.
The ability to design stable proteins with custom-made functions is a major goal in biochemistry with practical relevance for our environment and society. Understanding and manipulating protein stability provide crucial information on the molecular determinants that modulate structure and stability, and expand the applications of de novo proteins. Since the (β/⍺)8-barrel or TIM-barrel fold is one of the most common functional scaffolds, in this work we designed a collection of stable de novo TIM barrels (DeNovoTIMs), using a computational fixed-backbone and modular approach based on improved hydrophobic packing of sTIM11, the first validated de novo TIM barrel, and subjected them to a thorough folding analysis. DeNovoTIMs navigate a region of the stability landscape previously uncharted by natural TIM barrels, with variations spanning 60 degrees in melting temperature and 22 kcal per mol in conformational stability throughout the designs. Significant non-additive or epistatic effects were observed when stabilizing mutations from different regions of the barrel were combined. The molecular basis of epistasis in DeNovoTIMs appears to be related to the extension of the hydrophobic cores. This study is an important step towards the fine-tuned modulation of protein stability by design.
Collapse
Affiliation(s)
- Sergio Romero-Romero
- Laboratorio de Fisicoquímica e Ingeniería de Proteínas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico; Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany
| | - Miguel Costas
- Laboratorio de Biofisicoquímica, Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Daniel-Adriano Silva Manzano
- Department of Biochemistry, University of Washington, 98195 Seattle, USA; Institute for Protein Design, University of Washington, 98195 Seattle, USA
| | - Sina Kordes
- Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany
| | - Erendira Rojas-Ortega
- Laboratorio de Fisicoquímica e Ingeniería de Proteínas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Cinthya Tapia
- Laboratorio de Fisicoquímica e Ingeniería de Proteínas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Yasel Guerra
- Laboratorio de Fisicoquímica e Ingeniería de Proteínas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | | | - Adela Rodríguez-Romero
- Instituto de Química, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - David Baker
- Department of Biochemistry, University of Washington, 98195 Seattle, USA; Institute for Protein Design, University of Washington, 98195 Seattle, USA.
| | - Birte Höcker
- Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany.
| | - D Alejandro Fernández-Velasco
- Laboratorio de Fisicoquímica e Ingeniería de Proteínas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico.
| |
Collapse
|
27
|
Mahmood MS, Irshad S, Kalsoom U, Batool H, Batool S, Butt TA. In silico analysis of missense Single Nucleotide Variants (SNVs) in HBB gene associated with the β-thalassemia. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
28
|
Menghiu G, Ostafe V, Prodanović R, Fischer R, Ostafe R. A High-Throughput Screening System Based on Fluorescence-Activated Cell Sorting for the Directed Evolution of Chitinase A. Int J Mol Sci 2021; 22:ijms22063041. [PMID: 33809788 PMCID: PMC8002391 DOI: 10.3390/ijms22063041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
Chitinases catalyze the degradation of chitin, a polymer of N-acetylglucosamine found in crustacean shells, insect cuticles, and fungal cell walls. There is great interest in the development of improved chitinases to address the environmental burden of chitin waste from the food processing industry as well as the potential medical, agricultural, and industrial uses of partially deacetylated chitin (chitosan) and its products (chito-oligosaccharides). The depolymerization of chitin can be achieved using chemical and physical treatments, but an enzymatic process would be more environmentally friendly and more sustainable. However, chitinases are slow-acting enzymes, limiting their biotechnological exploitation, although this can be overcome by molecular evolution approaches to enhance the features required for specific applications. The two main goals of this study were the development of a high-throughput screening system for chitinase activity (which could be extrapolated to other hydrolytic enzymes), and the deployment of this new method to select improved chitinase variants. We therefore cloned and expressed the Bacillus licheniformis DSM8785 chitinase A (chiA) gene in Escherichia coli BL21 (DE3) cells and generated a mutant library by error-prone PCR. We then developed a screening method based on fluorescence-activated cell sorting (FACS) using the model substrate 4-methylumbelliferyl β-d-N,N′,N″-triacetyl chitotrioside to identify improved enzymes. We prevented cross-talk between emulsion compartments caused by the hydrophobicity of 4-methylumbelliferone, the fluorescent product of the enzymatic reaction, by incorporating cyclodextrins into the aqueous phases. We also addressed the toxicity of long-term chiA expression in E. coli by limiting the reaction time. We identified 12 mutants containing 2–8 mutations per gene resulting in up to twofold higher activity than wild-type ChiA.
Collapse
Affiliation(s)
- Gheorghita Menghiu
- Institute for Biology VII, Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (G.M.); (R.F.)
- Advanced Environmental Research Laboratories, Department of Biology–Chemistry, West University of Timisoara, Oituz 4, 300086 Timisoara, Romania;
| | - Vasile Ostafe
- Advanced Environmental Research Laboratories, Department of Biology–Chemistry, West University of Timisoara, Oituz 4, 300086 Timisoara, Romania;
| | - Radivoje Prodanović
- Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia;
| | - Rainer Fischer
- Institute for Biology VII, Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (G.M.); (R.F.)
- Departments of Biological Sciences and Chemistry, Purdue University, 207 S. Martin Jischke Dr., West Lafayette, IN 47907, USA
| | - Raluca Ostafe
- Institute for Biology VII, Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (G.M.); (R.F.)
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Molecular Evolution, Protein Engineering and Production, Purdue University, 207 S. Martin Jischke Dr., West Lafayette, IN 47907, USA
- Correspondence: ; Tel.: +1-317-765-496-4012
| |
Collapse
|
29
|
Liu R, Wang J, Xiong P, Chen Q, Liu H. De novo sequence redesign of a functional Ras-binding domain globally inverted the surface charge distribution and led to extreme thermostability. Biotechnol Bioeng 2021; 118:2031-2042. [PMID: 33590881 DOI: 10.1002/bit.27716] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/05/2021] [Accepted: 02/14/2021] [Indexed: 11/05/2022]
Abstract
To acquire extremely thermostable proteins of given functions is challenging for conventional protein engineering. Here we applied ABACUS, a statistical energy function we developed for de novo amino acid sequence design, to globally redesign a Ras-binding domain (RBD), and obtained an extremely thermostable RBD that unfolds reversibly at above 110°C, the redesigned RBD experimentally confirmed to have expected structure and Ras-binding interface. Directed evolution of the redesigned RBD improved its Ras-binding affinity to the native protein level without excessive loss of thermostability. The designed amino acid substitutions were mostly at the protein surface. For many substitutions, strong epistasis or significantly differentiated effects on thermostability in the native sequence context relative to the redesigned sequence context were observed, suggesting the globally redesigned sequence to be unreachable through combining beneficial mutations of the native sequence. Further analyses revealed that by replacing 38 of a total of 48 non-interfacial surface residues at once, ABACUS redesign was able to globally "invert" the protein's charge distribution pattern in an optimized way. Our study demonstrates that computational protein design provides powerful new tools to solve challenging protein engineering problems.
Collapse
Affiliation(s)
- Ruicun Liu
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Jichao Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Peng Xiong
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Quan Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiyan Liu
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, China.,School of Data Science, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
30
|
Hao Z, Jin DY, Chen X, Schurgers LJ, Stafford DW, Tie JK. γ-Glutamyl carboxylase mutations differentially affect the biological function of vitamin K-dependent proteins. Blood 2021; 137:533-543. [PMID: 33507293 PMCID: PMC7845004 DOI: 10.1182/blood.2020006329] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
γ-Glutamyl carboxylase (GGCX) is an integral membrane protein that catalyzes posttranslational carboxylation of a number of vitamin K-dependent (VKD) proteins involved in a wide variety of physiologic processes, including blood coagulation, vascular calcification, and bone metabolism. Naturally occurring GGCX mutations are associated with multiple distinct clinical phenotypes. However, the genotype-phenotype correlation of GGCX remains elusive. Here, we systematically examined the effect of all naturally occurring GGCX mutations on the carboxylation of 3 structure-function distinct VKD proteins in a cellular environment. GGCX mutations were transiently introduced into GGCX-deficient human embryonic kidney 293 cells stably expressing chimeric coagulation factor, matrix Gla protein (MGP), or osteocalcin as VKD reporter proteins, and then the carboxylation efficiency of these reporter proteins was evaluated. Our results show that GGCX mutations differentially affect the carboxylation of these reporter proteins and the efficiency of using vitamin K as a cofactor. Carboxylation of these reporter proteins by a C-terminal truncation mutation (R704X) implies that GGCX's C terminus plays a critical role in the binding of osteocalcin but not in the binding of coagulation factors and MGP. This has been confirmed by probing the protein-protein interaction between GGCX and its protein substrates in live cells using bimolecular fluorescence complementation and chemical cross-linking assays. Additionally, using a minigene splicing assay, we demonstrated that several GGCX missense mutations affect GGCX's pre-messenger RNA splicing rather than altering the corresponding amino acid residues. Results from this study interpreted the correlation of GGCX's genotype and its clinical phenotypes and clarified why vitamin K administration rectified bleeding disorders but not nonbleeding disorders.
Collapse
Affiliation(s)
- Zhenyu Hao
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| | - Da-Yun Jin
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| | - Xuejie Chen
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Darrel W Stafford
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| | - Jian-Ke Tie
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| |
Collapse
|
31
|
Rege NK, Liu M, Yang Y, Dhayalan B, Wickramasinghe NP, Chen YS, Rahimi L, Guo H, Haataja L, Sun J, Ismail-Beigi F, Phillips NB, Arvan P, Weiss MA. Evolution of insulin at the edge of foldability and its medical implications. Proc Natl Acad Sci U S A 2020; 117:29618-29628. [PMID: 33154160 PMCID: PMC7703552 DOI: 10.1073/pnas.2010908117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Proteins have evolved to be foldable, and yet determinants of foldability may be inapparent once the native state is reached. Insight has emerged from studies of diseases of protein misfolding, exemplified by monogenic diabetes mellitus due to mutations in proinsulin leading to endoplasmic reticulum stress and β-cell death. Cellular foldability of human proinsulin requires an invariant Phe within a conserved crevice at the receptor-binding surface (position B24). Any substitution, even related aromatic residue TyrB24, impairs insulin biosynthesis and secretion. As a seeming paradox, a monomeric TyrB24 insulin analog exhibits a native-like structure in solution with only a modest decrement in stability. Packing of TyrB24 is similar to that of PheB24, adjoining core cystine B19-A20 to seal the core; the analog also exhibits native self-assembly. Although affinity for the insulin receptor is decreased ∼20-fold, biological activities in cells and rats were within the range of natural variation. Together, our findings suggest that the invariance of PheB24 among vertebrate insulins and insulin-like growth factors reflects an essential role in enabling efficient protein folding, trafficking, and secretion, a function that is inapparent in native structures. In particular, we envision that the para-hydroxyl group of TyrB24 hinders pairing of cystine B19-A20 in an obligatory on-pathway folding intermediate. The absence of genetic variation at B24 and other conserved sites near this disulfide bridge-excluded due to β-cell dysfunction-suggests that insulin has evolved to the edge of foldability. Nonrobustness of a protein's fitness landscape underlies both a rare monogenic syndrome and "diabesity" as a pandemic disease of civilization.
Collapse
Affiliation(s)
- Nischay K Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 300052 Tianjin, China
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Yanwu Yang
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Balamurugan Dhayalan
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | | | - Yen-Shan Chen
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Leili Rahimi
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Huan Guo
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Jinhong Sun
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106
| | - Nelson B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Michael A Weiss
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106;
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
32
|
Kurahashi R, Tanaka SI, Takano K. Highly active enzymes produced by directed evolution with stability-based selection. Enzyme Microb Technol 2020; 140:109626. [DOI: 10.1016/j.enzmictec.2020.109626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
|
33
|
Youssef N, Susko E, Bielawski JP. Consequences of Stability-Induced Epistasis for Substitution Rates. Mol Biol Evol 2020; 37:3131-3148. [DOI: 10.1093/molbev/msaa151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
AbstractDo interactions between residues in a protein (i.e., epistasis) significantly alter evolutionary dynamics? If so, what consequences might they have on inference from traditional codon substitution models which assume site-independence for the sake of computational tractability? To investigate the effects of epistasis on substitution rates, we employed a mechanistic mutation-selection model in conjunction with a fitness framework derived from protein stability. We refer to this as the stability-informed site-dependent (S-SD) model and developed a new stability-informed site-independent (S-SI) model that captures the average effect of stability constraints on individual sites of a protein. Comparison of S-SI and S-SD offers a novel and direct method for investigating the consequences of stability-induced epistasis on protein evolution. We developed S-SI and S-SD models for three natural proteins and showed that they generate sequences consistent with real alignments. Our analyses revealed that epistasis tends to increase substitution rates compared with the rates under site-independent evolution. We then assessed the epistatic sensitivity of individual site and discovered a counterintuitive effect: Highly connected sites were less influenced by epistasis relative to exposed sites. Lastly, we show that, despite the unrealistic assumptions, traditional models perform comparably well in the presence and absence of epistasis and provide reasonable summaries of average selection intensities. We conclude that epistatic models are critical to understanding protein evolutionary dynamics, but epistasis might not be required for reasonable inference of selection pressure when averaging over time and sites.
Collapse
Affiliation(s)
- Noor Youssef
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Centre for Genomics and Evolutionary Bioinformatics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Edward Susko
- Centre for Genomics and Evolutionary Bioinformatics, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Mathematics and Statistics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joseph P Bielawski
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Centre for Genomics and Evolutionary Bioinformatics, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Mathematics and Statistics, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
34
|
Khanppnavar B, Roy A, Chandra K, Uversky VN, Maiti NC, Datta S. Deciphering the structural intricacy in virulence effectors for proton-motive force mediated unfolding in type-III protein secretion. Int J Biol Macromol 2020; 159:18-33. [DOI: 10.1016/j.ijbiomac.2020.04.266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 10/24/2022]
|
35
|
Mahase V, Sobitan A, Johnson C, Cooper F, Xie Y, Li L, Teng S. Computational analysis of hereditary spastic paraplegia mutations in the kinesin motor domains of KIF1A and KIF5A. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2020. [DOI: 10.1142/s0219633620410035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hereditary spastic paraplegias (HSPs) are a genetically heterogeneous collection of neurodegenerative disorders categorized by progressive lower-limb spasticity and frailty. The complex HSP forms are characterized by various neurological features including progressive spastic weakness, urinary sphincter dysfunction, extra pyramidal signs and intellectual disability (ID). The kinesin superfamily proteins (KIFs) are microtubule-dependent molecular motors involved in intracellular transport. Kinesins directionally transport membrane vesicles, protein complexes, and mRNAs along neurites, thus playing important roles in neuronal development and function. Recent genetic studies have identified kinesin mutations in patients with HSPs. In this study, we used the computational approaches to investigate the 40 missense mutations associated with HSP and ID in KIF1A and KIF5A. We performed homology modeling to construct the structures of kinesin–microtubule binding domain and kinesin–tubulin complex. We applied structure-based energy calculation methods to determine the effects of missense mutations on protein stability and protein–protein interaction. The results revealed that the most of disease-causing mutations could change the folding free energy of kinesin motor domain and the binding free energy of kinesin–tubulin complex. We found that E253K associated with ID in KIF1A decrease the protein stability of kinesin motor domains. We showed that the HSP mutations located in kinesin–tubulin complex interface, such as K253N and R280C in KIF5A, can destabilize the kinesin–tubulin complex. The computational analysis provides useful information for understanding the roles of kinesin mutations in the development of ID and HSPs.
Collapse
Affiliation(s)
- Vidhyanand Mahase
- Department of Biology, Howard University, Washington, D.C., 20059 USA
| | - Adebiyi Sobitan
- Department of Biology, Howard University, Washington, D.C., 20059 USA
| | - Christina Johnson
- Department of Biology, Howard University, Washington, D.C., 20059 USA
| | - Farion Cooper
- Department of Biology, Howard University, Washington, D.C., 20059 USA
| | - Yixin Xie
- Computational Science Program, University of Texas at El Paso, El Paso, Texas 79902, USA
| | - Lin Li
- Computational Science Program, University of Texas at El Paso, El Paso, Texas 79902, USA
- Department of Physics, University of Texas at El Paso, El Paso, Texas 79902, USA
| | - Shaolei Teng
- Department of Biology, Howard University, Washington, D.C., 20059 USA
| |
Collapse
|
36
|
Lambert AR, Hallinan JP, Werther R, Glöw D, Stoddard BL. Optimization of Protein Thermostability and Exploitation of Recognition Behavior to Engineer Altered Protein-DNA Recognition. Structure 2020; 28:760-775.e8. [PMID: 32359399 PMCID: PMC7347439 DOI: 10.1016/j.str.2020.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/17/2020] [Accepted: 04/11/2020] [Indexed: 01/07/2023]
Abstract
The redesign of a macromolecular binding interface and corresponding alteration of recognition specificity is a challenging endeavor that remains recalcitrant to computational approaches. This is particularly true for the redesign of DNA binding specificity, which is highly dependent upon bending, hydrogen bonds, electrostatic contacts, and the presence of solvent and counterions throughout the molecular interface. Thus, redesign of protein-DNA binding specificity generally requires iterative rounds of amino acid randomization coupled to selections. Here, we describe the importance of scaffold thermostability for protein engineering, coupled with a strategy that exploits the protein's specificity profile, to redesign the specificity of a pair of meganucleases toward three separate genomic targets. We determine and describe a series of changes in protein sequence, stability, structure, and activity that accumulate during the engineering process, culminating in fully retargeted endonucleases.
Collapse
Affiliation(s)
- Abigail R. Lambert
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N. Seattle WA 98109 USA
| | - Jazmine P. Hallinan
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N. Seattle WA 98109 USA
| | - Rachel Werther
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N. Seattle WA 98109 USA
| | - Dawid Glöw
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N. Seattle WA 98109 USA,Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Barry L. Stoddard
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N. Seattle WA 98109 USA,Corresponding Author and Lead Contact:
| |
Collapse
|
37
|
Gardner JM, Biler M, Risso VA, Sanchez-Ruiz JM, Kamerlin SCL. Manipulating Conformational Dynamics To Repurpose Ancient Proteins for Modern Catalytic Functions. ACS Catal 2020. [DOI: 10.1021/acscatal.0c00722] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jasmine M. Gardner
- Department of Chemistry - BMC, Uppsala University, Box 576, 751 23 Uppsala, Sweden
| | - Michal Biler
- Department of Chemistry - BMC, Uppsala University, Box 576, 751 23 Uppsala, Sweden
| | - Valeria A. Risso
- Departamento de Quı́mica Fisica, Facultad de Ciencias, Unidad de Excelencia de Quı́mica Aplicada a Biomedicina y Medioambiente (UEQ), Universidad de Granada, 18071 Granada, Spain
| | - Jose M. Sanchez-Ruiz
- Departamento de Quı́mica Fisica, Facultad de Ciencias, Unidad de Excelencia de Quı́mica Aplicada a Biomedicina y Medioambiente (UEQ), Universidad de Granada, 18071 Granada, Spain
| | - Shina C. L. Kamerlin
- Department of Chemistry - BMC, Uppsala University, Box 576, 751 23 Uppsala, Sweden
| |
Collapse
|
38
|
Evolution Rapidly Optimizes Stability and Aggregation in Lattice Proteins Despite Pervasive Landscape Valleys and Mazes. Genetics 2020; 214:1047-1057. [PMID: 32107278 DOI: 10.1534/genetics.120.302815] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/18/2020] [Indexed: 11/18/2022] Open
Abstract
The "fitness" landscapes of genetic sequences are characterized by high dimensionality and "ruggedness" due to sign epistasis. Ascending from low to high fitness on such landscapes can be difficult because adaptive trajectories get stuck at low-fitness local peaks. Compounding matters, recent theoretical arguments have proposed that extremely long, winding adaptive paths may be required to reach even local peaks: a "maze-like" landscape topography. The extent to which peaks and mazes shape the mode and tempo of evolution is poorly understood, due to empirical limitations and the abstractness of many landscape models. We explore the prevalence, scale, and evolutionary consequences of landscape mazes in a biophysically grounded computational model of protein evolution that captures the "frustration" between "stability" and aggregation propensity. Our stability-aggregation landscape exhibits extensive sign epistasis and local peaks galore. Although this frequently obstructs adaptive ascent to high fitness and virtually eliminates reproducibility of evolutionary outcomes, many adaptive paths do successfully complete the ascent from low to high fitness, with hydrophobicity a critical mediator of success. These successful paths exhibit maze-like properties on a global landscape scale, in which taking an indirect path helps to avoid low-fitness local peaks. This delicate balance of "hard but possible" adaptation could occur more broadly in other biological settings where competing interactions and frustration are important.
Collapse
|
39
|
Faber MS, Wrenbeck EE, Azouz LR, Steiner PJ, Whitehead TA. Impact of In Vivo Protein Folding Probability on Local Fitness Landscapes. Mol Biol Evol 2020; 36:2764-2777. [PMID: 31400199 DOI: 10.1093/molbev/msz184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is incompletely understood how biophysical properties like protein stability impact molecular evolution and epistasis. Epistasis is defined as specific when a mutation exclusively influences the phenotypic effect of another mutation, often at physically interacting residues. In contrast, nonspecific epistasis results when a mutation is influenced by a large number of nonlocal mutations. As most mutations are pleiotropic, the in vivo folding probability-governed by basal protein stability-is thought to determine activity-enhancing mutational tolerance, implying that nonspecific epistasis is dominant. However, evidence exists for both specific and nonspecific epistasis as the prevalent factor, with limited comprehensive data sets to support either claim. Here, we use deep mutational scanning to probe how in vivo enzyme folding probability impacts local fitness landscapes. We computationally designed two different variants of the amidase AmiE with statistically indistinguishable catalytic efficiencies but lower probabilities of folding in vivo compared with wild-type. Local fitness landscapes show slight alterations among variants, with essentially the same global distribution of fitness effects. However, specific epistasis was predominant for the subset of mutations exhibiting positive sign epistasis. These mutations mapped to spatially distinct locations on AmiE near the initial mutation or proximal to the active site. Intriguingly, the majority of specific epistatic mutations were codon dependent, with different synonymous codons resulting in fitness sign reversals. Together, these results offer a nuanced view of how protein folding probability impacts local fitness landscapes and suggest that transcriptional-translational effects are as important as stability in determining evolutionary outcomes.
Collapse
Affiliation(s)
- Matthew S Faber
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI
| | - Emily E Wrenbeck
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI
| | - Laura R Azouz
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI
| | - Paul J Steiner
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO
| | - Timothy A Whitehead
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI.,Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO.,E.E.W. Ginkgo Bioworks, L.R.A. McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX
| |
Collapse
|
40
|
Nutschel C, Fulton A, Zimmermann O, Schwaneberg U, Jaeger KE, Gohlke H. Systematically Scrutinizing the Impact of Substitution Sites on Thermostability and Detergent Tolerance for Bacillus subtilis Lipase A. J Chem Inf Model 2020; 60:1568-1584. [PMID: 31905288 DOI: 10.1021/acs.jcim.9b00954] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Improving an enzyme's (thermo-)stability or tolerance against solvents and detergents is highly relevant in protein engineering and biotechnology. Recent developments have tended toward data-driven approaches, where available knowledge about the protein is used to identify substitution sites with high potential to yield protein variants with improved stability, and subsequently, substitutions are engineered by site-directed or site-saturation (SSM) mutagenesis. However, the development and validation of algorithms for data-driven approaches have been hampered by the lack of availability of large-scale data measured in a uniform way and being unbiased with respect to substitution types and locations. Here, we extend our knowledge on guidelines for protein engineering following a data-driven approach by scrutinizing the impact of substitution sites on thermostability or/and detergent tolerance for Bacillus subtilis lipase A (BsLipA) at very large scale. We systematically analyze a complete experimental SSM library of BsLipA containing all 3439 possible single variants, which was evaluated as to thermostability and tolerances against four detergents under respectively uniform conditions. Our results provide systematic and unbiased reference data at unprecedented scale for a biotechnologically important protein, identify consistently defined hot spot types for evaluating the performance of data-driven protein-engineering approaches, and show that the rigidity theory and ensemble-based approach Constraint Network Analysis yields hot spot predictions with an up to ninefold gain in precision over random classification.
Collapse
Affiliation(s)
- Christina Nutschel
- John von Neumann Institute for Computing (NIC) and Institute for Complex Systems-Structural Biochemistry (ICS-6), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Alexander Fulton
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, 52425 Jülich, Germany
| | - Olav Zimmermann
- Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Ulrich Schwaneberg
- Institute of Biotechnology, RWTH Aachen University, 52074 Aachen, Germany.,DWI-Leibniz-Institute for Interactive Materials, 52056 Aachen, Germany
| | - Karl-Erich Jaeger
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, 52425 Jülich, Germany.,Institute of Bio- and Geosciences IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Holger Gohlke
- John von Neumann Institute for Computing (NIC) and Institute for Complex Systems-Structural Biochemistry (ICS-6), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
41
|
Ghosh DK, Kumar A, Ranjan A. T54R mutation destabilizes the dimer of superoxide dismutase 1T54R by inducing steric clashes at the dimer interface. RSC Adv 2020; 10:10776-10788. [PMID: 35492906 PMCID: PMC9050410 DOI: 10.1039/c9ra09870d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/28/2020] [Indexed: 11/29/2022] Open
Abstract
Mutations cause abnormalities in protein structure, function and oligomerization. Different mutations in the superoxide dismutase 1 (SOD1) protein cause its misfolding, loss of dimerization and aggravate its aggregation in the amyotrophic lateral sclerosis disease. In this study, we report the mechanistic details of how a threonine-to-arginine mutation at the 54th position (T54R) of SOD1 results in destabilization of the dimer interface of SOD1T54R. Using computational and experimental methods, we show that the T54R mutation increases fluctuation of the mutation-harboring loop (R54-loop) of SOD1T54R. Fluctuation of this loop causes steric clashes that involve arginine-54 (R54) and other residues of SOD1T54R, resulting in loss of inter-subunit contacts at the dimer interface. Since the T54 residue-containing loop is necessary for the dimerization of wild-type SOD1, fluctuation of the R54-loop, steric clashes involving R54 and loss of inter-subunit contacts give rise to the loss of SOD1T54R dimer stability. This correlates to energetically unfavorable tethering of the monomers of SOD1T54R. The outcome is gradual splitting of SOD1T54R dimers into monomers, thereby exposing the previously buried hydrophobic interface residues to the aqueous environment. This event finally leads to aggregation of SOD1T54R. T54R mutation has no effect in altering the relative positions of copper and zinc ion binding residues of SOD1T54R. The native SOD1 structure is stable, and there is no destabilizing effect at its dimer interface. Overall, our study reveals the intricate mechanism of T54R mutation-associated destabilization of the dimer of the SOD1T54R protein. T54R mutation destabilizes the dimer of SOD1T54R.![]()
Collapse
Affiliation(s)
- Debasish Kumar Ghosh
- Computational and Functional Genomics Group
- Centre for DNA Fingerprinting and Diagnostics
- Hyderabad 500039
- India
| | - Abhishek Kumar
- Computational and Functional Genomics Group
- Centre for DNA Fingerprinting and Diagnostics
- Hyderabad 500039
- India
- Graduate Studies
| | - Akash Ranjan
- Computational and Functional Genomics Group
- Centre for DNA Fingerprinting and Diagnostics
- Hyderabad 500039
- India
| |
Collapse
|
42
|
Ramos-Martín F, Annaval T, Buchoux S, Sarazin C, D'Amelio N. ADAPTABLE: a comprehensive web platform of antimicrobial peptides tailored to the user's research. Life Sci Alliance 2019; 2:e201900512. [PMID: 31740563 PMCID: PMC6864362 DOI: 10.26508/lsa.201900512] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 01/01/2023] Open
Abstract
Antimicrobial peptides (AMPs) are part of the innate immune response to pathogens in all of the kingdoms of life. They have received significant attention because of their extraordinary variety of activities, in particular, as candidate drugs against the threat of super-bacteria. A systematic study of the relation between the sequence and the mechanism of action is urgently needed, given the thousands of sequences already in multiple web resources. ADAPTABLE web platform (http://gec.u-picardie.fr/adaptable) introduces the concept of "property alignment" to create families of property and sequence-related peptides (SR families). This feature provides the researcher with a tool to select those AMPs meaningful to their research from among more than 40,000 nonredundant sequences. Selectable properties include the target organism and experimental activity concentration, allowing selection of peptides with multiple simultaneous actions. This is made possible by ADAPTABLE because it not only merges sequences of AMP databases but also merges their data, thereby standardizing values and handling non-proteinogenic amino acids. In this unified platform, SR families allow the creation of peptide scaffolds based on common traits in peptides with similar activity, independently of their source.
Collapse
Affiliation(s)
- Francisco Ramos-Martín
- Génie Enzymatique et Cellulaire, Unité Mixte de Recherche 7025, Centre National de la Recherche Scientifique, Université de Picardie Jules Verne, Amiens, France
| | - Thibault Annaval
- Génie Enzymatique et Cellulaire, Unité Mixte de Recherche 7025, Centre National de la Recherche Scientifique, Université de Picardie Jules Verne, Amiens, France
| | - Sébastien Buchoux
- Génie Enzymatique et Cellulaire, Unité Mixte de Recherche 7025, Centre National de la Recherche Scientifique, Université de Picardie Jules Verne, Amiens, France
| | - Catherine Sarazin
- Génie Enzymatique et Cellulaire, Unité Mixte de Recherche 7025, Centre National de la Recherche Scientifique, Université de Picardie Jules Verne, Amiens, France
| | - Nicola D'Amelio
- Génie Enzymatique et Cellulaire, Unité Mixte de Recherche 7025, Centre National de la Recherche Scientifique, Université de Picardie Jules Verne, Amiens, France
| |
Collapse
|
43
|
Activity-stability trade-off in random mutant proteins. J Biosci Bioeng 2019; 128:405-409. [DOI: 10.1016/j.jbiosc.2019.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/18/2019] [Accepted: 03/27/2019] [Indexed: 12/23/2022]
|
44
|
Tomala K, Zrebiec P, Hartl DL. Limits to Compensatory Mutations: Insights from Temperature-Sensitive Alleles. Mol Biol Evol 2019; 36:1874-1883. [PMID: 31058959 PMCID: PMC6735812 DOI: 10.1093/molbev/msz110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous experiments with temperature-sensitive mutants of the yeast enzyme orotidine 5'-phosphate decarboxylase (encoded in gene URA3) yielded the unexpected result that reversion occurs only through exact reversal of the original mutation (Jakubowska A, Korona R. 2009. Lack of evolutionary conservation at positions important for thermal stability in the yeast ODCase protein. Mol Biol Evol. 26(7):1431-1434.). We recreated a set of these mutations in which the codon had two nucleotide substitutions, making exact reversion much less likely. We screened these double mutants for reversion and obtained a number of compensatory mutations occurring at alternative sites in the molecule. None of these compensatory mutations fully restored protein performance. The mechanism of partial compensation is consistent with a model in which protein stabilization is additive, as the same secondary mutations can compensate different primary alternations. The distance between primary and compensatory residues precludes direct interaction between the sites. Instead, most of the compensatory mutants were clustered in proximity to the catalytic center. All of the second-site compensatory substitutions occurred at relatively conserved sites, and the amino acid replacements were to residues found at these sites in a multispecies alignment of the protein. Based on the estimated distribution of changes in Gibbs free energy among a large number of amino acid replacements, we estimate that, for most proteins, the probability that a second-site mutation would have a sufficiently large stabilizing effect to offset a temperature-sensitive mutation in the order of 10-4 or less. Hence compensation is likely to take place only for slightly destabilizing mutations because highly stabilizing mutations are exceeding rare.
Collapse
Affiliation(s)
- Katarzyna Tomala
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Piotr Zrebiec
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Daniel L Hartl
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA
| |
Collapse
|
45
|
Ferrada E. Gene Families, Epistasis and the Amino Acid Preferences of Protein Homologs. Evol Bioinform Online 2019; 15:1176934319870485. [PMID: 31452598 PMCID: PMC6698995 DOI: 10.1177/1176934319870485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 07/27/2019] [Indexed: 11/16/2022] Open
Abstract
In order to preserve structure and function, proteins tend to preferentially conserve amino acids at particular sites along the sequence. Because mutations can affect structure and function, the question arises whether the preference of a protein site for a particular amino acid varies between protein homologs, and to what extent that variation depends on sequence divergence. Answering these questions can help in the development of models of sequence evolution, as well as provide insights on the dependence of the fitness effects of mutations on the genetic background of sequences, a phenomenon known as epistasis. Here, I comment on recent computational work providing a systematic analysis of the extent to which the amino acid preferences of proteins depend on the background mutations of protein homologs.
Collapse
Affiliation(s)
- Evandro Ferrada
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Santiago, Chile
| |
Collapse
|
46
|
Wiel L, Baakman C, Gilissen D, Veltman JA, Vriend G, Gilissen C. MetaDome: Pathogenicity analysis of genetic variants through aggregation of homologous human protein domains. Hum Mutat 2019; 40:1030-1038. [PMID: 31116477 PMCID: PMC6772141 DOI: 10.1002/humu.23798] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/21/2019] [Accepted: 05/15/2019] [Indexed: 01/19/2023]
Abstract
The growing availability of human genetic variation has given rise to novel methods of measuring genetic tolerance that better interpret variants of unknown significance. We recently developed a concept based on protein domain homology in the human genome to improve variant interpretation. For this purpose, we mapped population variation from the Exome Aggregation Consortium (ExAC) and pathogenic mutations from the Human Gene Mutation Database (HGMD) onto Pfam protein domains. The aggregation of these variation data across homologous domains into meta-domains allowed us to generate amino acid resolution of genetic intolerance profiles for human protein domains. Here, we developed MetaDome, a fast and easy-to-use web server that visualizes meta-domain information and gene-wide profiles of genetic tolerance. We updated the underlying data of MetaDome to contain information from 56,319 human transcripts, 71,419 protein domains, 12,164,292 genetic variants from gnomAD, and 34,076 pathogenic mutations from ClinVar. MetaDome allows researchers to easily investigate their variants of interest for the presence or absence of variation at corresponding positions within homologous domains. We illustrate the added value of MetaDome by an example that highlights how it may help in the interpretation of variants of unknown significance. The MetaDome web server is freely accessible at https://stuart.radboudumc.nl/metadome.
Collapse
Affiliation(s)
- Laurens Wiel
- Department of Human Genetics, Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Coos Baakman
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Daan Gilissen
- Department of Human Genetics, Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Bio‐informaticaHAN University of Applied SciencesNijmegenThe Netherlands
| | - Joris A. Veltman
- Department of Human Genetics, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
- Institute of Genetic Medicine, International Centre for LifeNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Gerrit Vriend
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Christian Gilissen
- Department of Human Genetics, Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
47
|
Kinney JB, McCandlish DM. Massively Parallel Assays and Quantitative Sequence-Function Relationships. Annu Rev Genomics Hum Genet 2019; 20:99-127. [PMID: 31091417 DOI: 10.1146/annurev-genom-083118-014845] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Over the last decade, a rich variety of massively parallel assays have revolutionized our understanding of how biological sequences encode quantitative molecular phenotypes. These assays include deep mutational scanning, high-throughput SELEX, and massively parallel reporter assays. Here, we review these experimental methods and how the data they produce can be used to quantitatively model sequence-function relationships. In doing so, we touch on a diverse range of topics, including the identification of clinically relevant genomic variants, the modeling of transcription factor binding to DNA, the functional and evolutionary landscapes of proteins, and cis-regulatory mechanisms in both transcription and mRNA splicing. We further describe a unified conceptual framework and a core set of mathematical modeling strategies that studies in these diverse areas can make use of. Finally, we highlight key aspects of experimental design and mathematical modeling that are important for the results of such studies to be interpretable and reproducible.
Collapse
Affiliation(s)
- Justin B Kinney
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; ,
| | - David M McCandlish
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; ,
| |
Collapse
|
48
|
Ferrada E. The Site-Specific Amino Acid Preferences of Homologous Proteins Depend on Sequence Divergence. Genome Biol Evol 2019; 11:121-135. [PMID: 30496400 PMCID: PMC6326188 DOI: 10.1093/gbe/evy261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2018] [Indexed: 12/20/2022] Open
Abstract
The propensity of protein sites to be occupied by any of the 20 amino acids is known as site-specific amino acid preferences (SSAP). Under the assumption that SSAP are conserved among homologs, they can be used to parameterize evolutionary models for the reconstruction of accurate phylogenetic trees. However, simulations and experimental studies have not been able to fully assess the relative conservation of SSAP as a function of sequence divergence between protein homologs. Here, we implement a computational procedure to predict the SSAP of proteins based on the effect of changes in thermodynamic stability upon mutation. An advantage of this computational approach is that it allows us to interrogate a large and unbiased sample of homologous proteins, over the entire spectrum of sequence divergence, and under selection for the same molecular trait. We show that computational predictions have reproducibilities that resemble those obtained in experimental replicates, and can largely recapitulate the SSAP observed in a large-scale mutagenesis experiment. Our results support recent experimental reports on the conservation of SSAP of related homologs, with a slowly increasing fraction of up to 15% of different sites at sequence distances lower than 40%. However, even under the sole contribution of thermodynamic stability, our conservative approach identifies up to 30% of significant different sites between divergent homologs. We show that this relation holds for homologs of diverse sizes and structural classes. Analyses of residue contact networks suggest that an important determinant of these differences is the increasing accumulation of structural deviations that results from sequence divergence.
Collapse
Affiliation(s)
- Evandro Ferrada
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino La Pirámide 5750, Huechuraba, 8580745, Santiago, Chile
| |
Collapse
|
49
|
Exploiting correlated molecular-dynamics networks to counteract enzyme activity-stability trade-off. Proc Natl Acad Sci U S A 2018; 115:E12192-E12200. [PMID: 30530661 PMCID: PMC6310800 DOI: 10.1073/pnas.1812204115] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rigidifying flexible sites is a powerful method to improve enzyme stability. However, if the highly flexible regions form the active site, modifying them risks losing activity due to the activity–stability trade-off. We hypothesized here that regions outside the active site whose dynamics were highly correlated to flexible active sites, would provide good targets for stabilizing mutations. To test this hypothesis, six variants were constructed in the 3M variant of Escherichia coli transketolase. The best variant had a 10.8-fold improved half-life at 55 °C, and increased the Tm and Tagg by 3 °C and 4.3 °C, respectively. The variants even increased the activity, by up to threefold. This study highlights how protein engineering strategies could be potentially improved by considering long-range dynamics. The directed evolution of enzymes for improved activity or substrate specificity commonly leads to a trade-off in stability. We have identified an activity–stability trade-off and a loss in unfolding cooperativity for a variant (3M) of Escherichia coli transketolase (TK) engineered to accept aromatic substrates. Molecular dynamics simulations of 3M revealed increased flexibility in several interconnected active-site regions that also form part of the dimer interface. Mutating the newly flexible active-site residues to regain stability risked losing the new activity. We hypothesized that stabilizing mutations could be targeted to residues outside of the active site, whose dynamics were correlated with the newly flexible active-site residues. We previously stabilized WT TK by targeting mutations to highly flexible regions. These regions were much less flexible in 3M and would not have been selected a priori as targets using the same strategy based on flexibility alone. However, their dynamics were highly correlated with the newly flexible active-site regions of 3M. Introducing the previous mutations into 3M reestablished the WT level of stability and unfolding cooperativity, giving a 10.8-fold improved half-life at 55 °C, and increased midpoint and aggregation onset temperatures by 3 °C and 4.3 °C, respectively. Even the activity toward aromatic aldehydes increased up to threefold. Molecular dynamics simulations confirmed that the mutations rigidified the active-site via the correlated network. This work provides insights into the impact of rigidifying mutations within highly correlated dynamic networks that could also be useful for developing improved computational protein engineering strategies.
Collapse
|
50
|
Mosaad Z, Arafa A, Hussein HA, Shalaby MA. In silico thermodynamic stability of mammalian adaptation and virulence determinants in polymerase complex proteins of H9N2 virus. J Genet Eng Biotechnol 2018; 16:757-767. [PMID: 30733797 PMCID: PMC6353722 DOI: 10.1016/j.jgeb.2018.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/01/2018] [Accepted: 02/17/2018] [Indexed: 11/16/2022]
Abstract
The polymerase complex proteins (PB2, PB1, and PA) are responsible primarily for the replication of avian influenza virus and play an important role in virus virulence, mammalian adaptation, and interspecies transmission. In this study; eight Egyptian LPAI-H9N2 viruses isolated from apparent healthy chickens and quails from 2014 to 2016. Characterization of complete nucleotide sequences, phylogenetic and mutation analysis were carried out. The measurement of thermodynamic stability of the H9N2 polymerase protein in comparison to human H3N2 and H1N1 proteins was carried out using in silico method. Phylogenetic analysis of these viruses revealed a close relationship to viruses isolated from neighboring Middle Eastern countries with an average of 96-99% homology. They are sharing the common ancestor A/quail/Hong Kong/G1/1997 (G1-Like) without any evidence for genetic reassortment. In addition, eight markers related to virulence were identified, including the combination of 627V and 391E in the PB2 gene with full-length PB1-F2 and PA-X proteins were observed in all viruses and the substitution N66S in PB1-F2 which suggest increasing virus virulence. Moreover, six markers that may affect the virus replication and transmission in mammalian hosts were identified. Five mutations related to mammalian adaptation show a structural stabilizing effect on LPAI-H9N2 polymerase complex protein according to the free-energy change (ΔΔG). Three out of those six adaptive mutations shown to increase polymerase complex protein stability were found in Egyptian LPAI-H9N2 viruses similar to Human H3N2 and H1N1 (661 in PB2, 225 and 409 in PA genes). Our results suggested that the stabilizing mutations in the polymerase complex protein have likely affected the protein structure and induced favorable conditions for avian virus replication and transmission in mammalian hosts. Indeed, the study reports the mutational analysis of the circulating LPAI-H9N2 strains in Egypt.
Collapse
Affiliation(s)
- Zienab Mosaad
- National Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, P.O. Box 264-Dokki, Giza 12618, Egypt
| | - Abdelsatar Arafa
- National Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, P.O. Box 264-Dokki, Giza 12618, Egypt
| | - Hussein A. Hussein
- VirologyDept, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Mohamed A. Shalaby
- VirologyDept, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|