Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 83(11): 1936-1952.e7, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267908

ABSTRACT

Non-native conformations drive protein-misfolding diseases, complicate bioengineering efforts, and fuel molecular evolution. No current experimental technique is well suited for elucidating them and their phenotypic effects. Especially intractable are the transient conformations populated by intrinsically disordered proteins. We describe an approach to systematically discover, stabilize, and purify native and non-native conformations, generated in vitro or in vivo, and directly link conformations to molecular, organismal, or evolutionary phenotypes. This approach involves high-throughput disulfide scanning (HTDS) of the entire protein. To reveal which disulfides trap which chromatographically resolvable conformers, we devised a deep-sequencing method for double-Cys variant libraries of proteins that precisely and simultaneously locates both Cys residues within each polypeptide. HTDS of the abundant E. coli periplasmic chaperone HdeA revealed distinct classes of disordered hydrophobic conformers with variable cytotoxicity depending on where the backbone was cross-linked. HTDS can bridge conformational and phenotypic landscapes for many proteins that function in disulfide-permissive environments.


Subject(s)
Escherichia coli Proteins , Protein Folding , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Conformation , Disulfides/metabolism , High-Throughput Nucleotide Sequencing , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 121(23): e2314518121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38820002

ABSTRACT

SARS-CoV-2 employs its spike protein's receptor binding domain (RBD) to enter host cells. The RBD is constantly subjected to immune responses, while requiring efficient binding to host cell receptors for successful infection. However, our understanding of how RBD's biophysical properties contribute to SARS-CoV-2's epidemiological fitness remains largely incomplete. Through a comprehensive approach, comprising large-scale sequence analysis of SARS-CoV-2 variants and the identification of a fitness function based on binding thermodynamics, we unravel the relationship between the biophysical properties of RBD variants and their contribution to viral fitness. We developed a biophysical model that uses statistical mechanics to map the molecular phenotype space, characterized by dissociation constants of RBD to ACE2, LY-CoV016, LY-CoV555, REGN10987, and S309, onto an epistatic fitness landscape. We validate our findings through experimentally measured and machine learning (ML) estimated binding affinities, coupled with infectivity data derived from population-level sequencing. Our analysis reveals that this model effectively predicts the fitness of novel RBD variants and can account for the epistatic interactions among mutations, including explaining the later reversal of Q493R. Our study sheds light on the impact of specific mutations on viral fitness and delivers a tool for predicting the future epidemiological trajectory of previously unseen or emerging low-frequency variants. These insights offer not only greater understanding of viral evolution but also potentially aid in guiding public health decisions in the battle against COVID-19 and future pandemics.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Humans , COVID-19/virology , COVID-19/epidemiology , COVID-19/genetics , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/chemistry , Protein Binding , Thermodynamics , Mutation , Machine Learning
3.
Mol Cell ; 70(5): 894-905.e5, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29883608

ABSTRACT

Despite considerable efforts, no physical mechanism has been shown to explain N-terminal codon bias in prokaryotic genomes. Using a systematic study of synonymous substitutions in two endogenous E. coli genes, we show that interactions between the coding region and the upstream Shine-Dalgarno (SD) sequence modulate the efficiency of translation initiation, affecting both intracellular mRNA and protein levels due to the inherent coupling of transcription and translation in E. coli. We further demonstrate that far-downstream mutations can also modulate mRNA levels by occluding the SD sequence through the formation of non-equilibrium secondary structures. By contrast, a non-endogenous RNA polymerase that decouples transcription and translation largely alleviates the effects of synonymous substitutions on mRNA levels. Finally, a complementary statistical analysis of the E. coli genome specifically implicates avoidance of intra-molecular base pairing with the SD sequence. Our results provide general physical insights into the coding-level features that optimize protein expression in prokaryotes.


Subject(s)
Codon, Initiator , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Nucleotide Motifs , RNA, Bacterial/genetics , RNA, Messenger/genetics , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/biosynthesis , Gene Expression Regulation, Bacterial , Genome, Bacterial , Nucleic Acid Conformation , Protein Biosynthesis , RNA Stability , RNA, Bacterial/biosynthesis , RNA, Messenger/biosynthesis , Structure-Activity Relationship , Transcription, Genetic
4.
Proc Natl Acad Sci U S A ; 120(18): e2219855120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37094144

ABSTRACT

Enzymes play a vital role in life processes; they control chemical reactions and allow functional cycles to be synchronized. Many enzymes harness large-scale motions of their domains to achieve tremendous catalytic prowess and high selectivity for specific substrates. One outstanding example is provided by the three-domain enzyme adenylate kinase (AK), which catalyzes phosphotransfer between ATP to AMP. Here we study the phenomenon of substrate inhibition by AMP and its correlation with domain motions. Using single-molecule FRET spectroscopy, we show that AMP does not block access to the ATP binding site, neither by competitive binding to the ATP cognate site nor by directly closing the LID domain. Instead, inhibitory concentrations of AMP lead to a faster and more cooperative domain closure by ATP, leading in turn to an increased population of the closed state. The effect of AMP binding can be modulated through mutations throughout the structure of the enzyme, as shown by the screening of an extensive AK mutant library. The mutation of multiple conserved residues reduces substrate inhibition, suggesting that substrate inhibition is an evolutionary well conserved feature in AK. Combining these insights, we developed a model that explains the complex activity of AK, particularly substrate inhibition, based on the experimentally observed opening and closing rates. Notably, the model indicates that the catalytic power is affected by the microsecond balance between the open and closed states of the enzyme. Our findings highlight the crucial role of protein motions in enzymatic activity.


Subject(s)
Adenosine Triphosphate , Adenylate Kinase , Adenylate Kinase/metabolism , Ligands , Binding Sites , Protein Domains , Adenosine Triphosphate/metabolism
5.
Biophys J ; 122(16): 3238-3253, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37422697

ABSTRACT

Many secreted proteins, including viral proteins, contain multiple disulfide bonds. How disulfide formation is coupled to protein folding in the cell remains poorly understood at the molecular level. Here, we combine experiment and simulation to address this question as it pertains to the SARS-CoV-2 receptor binding domain (RBD). We show that the RBD can only refold reversibly if its native disulfides are present before folding. But in their absence, the RBD spontaneously misfolds into a nonnative, molten-globule-like state that is structurally incompatible with complete disulfide formation and that is highly prone to aggregation. Thus, the RBD native structure represents a metastable state on the protein's energy landscape with reduced disulfides, indicating that nonequilibrium mechanisms are needed to ensure native disulfides form before folding. Our atomistic simulations suggest that this may be achieved via co-translational folding during RBD secretion into the endoplasmic reticulum. Namely, at intermediate translation lengths, native disulfide pairs are predicted to come together with high probability, and thus, under suitable kinetic conditions, this process may lock the protein into its native state and circumvent highly aggregation-prone nonnative intermediates. This detailed molecular picture of the RBD folding landscape may shed light on SARS-CoV-2 pathology and molecular constraints governing SARS-CoV-2 evolution.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Disulfides/chemistry , Proteins/chemistry , Protein Folding
6.
Mol Syst Biol ; 17(6): e10200, 2021 06.
Article in English | MEDLINE | ID: mdl-34180142

ABSTRACT

The relationship between sequence variation and phenotype is poorly understood. Here, we use metabolomic analysis to elucidate the molecular mechanism underlying the filamentous phenotype of E. coli strains that carry destabilizing mutations in dihydrofolate reductase (DHFR). We find that partial loss of DHFR activity causes reversible filamentation despite SOS response indicative of DNA damage, in contrast to thymineless death (TLD) achieved by complete inhibition of DHFR activity by high concentrations of antibiotic trimethoprim. This phenotype is triggered by a disproportionate drop in intracellular dTTP, which could not be explained by drop in dTMP based on the Michaelis-Menten-like in vitro activity curve of thymidylate kinase (Tmk), a downstream enzyme that phosphorylates dTMP to dTDP. Instead, we show that a highly cooperative (Hill coefficient 2.5) in vivo activity of Tmk is the cause of suboptimal dTTP levels. dTMP supplementation rescues filamentation and restores in vivo Tmk kinetics to Michaelis-Menten. Overall, this study highlights the important role of cellular environment in sculpting enzymatic kinetics with system-level implications for bacterial phenotype.


Subject(s)
Escherichia coli , Point Mutation , Escherichia coli/genetics , Phenotype
7.
Proc Natl Acad Sci U S A ; 116(23): 11265-11274, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31097595

ABSTRACT

Proteins are only moderately stable. It has long been debated whether this narrow range of stabilities is solely a result of neutral drift toward lower stability or purifying selection against excess stability-for which no experimental evidence was found so far-is also at work. Here, we show that mutations outside the active site in the essential Escherichia coli enzyme adenylate kinase (Adk) result in a stability-dependent increase in substrate inhibition by AMP, thereby impairing overall enzyme activity at high stability. Such inhibition caused substantial fitness defects not only in the presence of excess substrate but also under physiological conditions. In the latter case, substrate inhibition caused differential accumulation of AMP in the stationary phase for the inhibition-prone mutants. Furthermore, we show that changes in flux through Adk could accurately describe the variation in fitness effects. Taken together, these data suggest that selection against substrate inhibition and hence excess stability may be an important factor determining stability observed for modern-day Adk.


Subject(s)
Adenylate Kinase/metabolism , Enzyme Stability/physiology , Adenylate Kinase/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Kinetics , Mutation/genetics , Protein Stability , Thermodynamics
8.
Biophys J ; 120(12): 2413-2424, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33932438

ABSTRACT

Every amino acid residue can influence a protein's overall stability, making stability highly susceptible to change throughout evolution. We consider the distribution of protein stabilities evolutionarily permittable under two previously reported protein fitness functions: flux dynamics and misfolding avoidance. We develop an evolutionary dynamics theory and find that it agrees better with an extensive protein stability data set for dihydrofolate reductase orthologs under the misfolding avoidance fitness function rather than the flux dynamics fitness function. Further investigation with ribonuclease H data demonstrates that not any misfolded state is avoided; rather, it is only the unfolded state. At the end, we discuss how our work pertains to the universal protein abundance-evolutionary rate correlation seen across organisms' proteomes. We derive a closed-form expression relating protein abundance to evolutionary rate that captures Escherichia coli, Saccharomyces cerevisiae, and Homo sapiens experimental trends without fitted parameters.


Subject(s)
Evolution, Molecular , Saccharomyces cerevisiae , Humans , Protein Folding , Protein Stability , Protein Unfolding , Proteome
9.
Biophys J ; 120(21): 4738-4750, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34571014

ABSTRACT

To what degree are individual structural elements within proteins modular such that similar structures from unrelated proteins can be interchanged? We study subdomain modularity by creating 20 chimeras of an enzyme, Escherichia coli dihydrofolate reductase (DHFR), in which a catalytically important, 10-residue α-helical sequence is replaced by α-helical sequences from a diverse set of proteins. The chimeras stably fold but have a range of diminished thermal stabilities and catalytic activities. Evolutionary coupling analysis indicates that the residues of this α-helix are under selection pressure to maintain catalytic activity in DHFR. Reversion to phenylalanine at key position 31 was found to partially restore catalytic activity, which could be explained by evolutionary coupling values. We performed molecular dynamics simulations using replica exchange with solute tempering. Chimeras with low catalytic activity exhibit nonhelical conformations that block the binding site and disrupt the positioning of the catalytically essential residue D27. Simulation observables and in vitro measurements of thermal stability and substrate-binding affinity are strongly correlated. Several E. coli strains with chromosomally integrated chimeric DHFRs can grow, with growth rates that follow predictions from a kinetic flux model that depends on the intracellular abundance and catalytic activity of DHFR. Our findings show that although α-helices are not universally substitutable, the molecular and fitness effects of modular segments can be predicted by the biophysical compatibility of the replacement segment.


Subject(s)
Escherichia coli , Tetrahydrofolate Dehydrogenase , Catalytic Domain , Escherichia coli/genetics , Escherichia coli/metabolism , Kinetics , Protein Conformation , Tetrahydrofolate Dehydrogenase/genetics
10.
Mol Cell ; 49(1): 133-44, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23219534

ABSTRACT

What are the molecular properties of proteins that fall on the radar of protein quality control (PQC)? Here we mutate the E. coli's gene encoding dihydrofolate reductase (DHFR) and replace it with bacterial orthologous genes to determine how components of PQC modulate fitness effects of these genetic changes. We find that chaperonins GroEL/ES and protease Lon compete for binding to molten globule intermediate of DHFR, resulting in a peculiar symmetry in their action: overexpression of GroEL/ES and deletion of Lon both restore growth of deleterious DHFR mutants and most of the slow-growing orthologous DHFR strains. Kinetic steady-state modeling predicts and experimentation verifies that mutations affect fitness by shifting the flux balance in cellular milieu between protein production, folding, and degradation orchestrated by PQC through the interaction with folding intermediates.


Subject(s)
Escherichia coli/genetics , Mutation, Missense , Protein Folding , Tetrahydrofolate Dehydrogenase/metabolism , Algorithms , Amino Acid Motifs , Base Sequence , Chaperonin 10/genetics , Chaperonin 10/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Escherichia coli/enzymology , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Gene Knockout Techniques , Genetic Fitness , Homeostasis , Kinetics , Microbial Viability , Models, Biological , Molecular Sequence Data , Protease La/genetics , Protease La/metabolism , Protein Biosynthesis , Proteolysis , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/genetics
11.
Biophys J ; 118(12): 2872-2878, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32416078

ABSTRACT

The distribution of protein stability effects is known to be well approximated by a Gaussian distribution from previous empirical fits. Starting from first-principles statistical mechanics, we more rigorously motivate this empirical observation by deriving per-residue-position protein stability effects to be Gaussian. Our derivation requires the number of amino acids to be large, which is satisfied by the standard set of 20 amino acids found in nature. No assumption is needed on the number of residues in close proximity in space, in contrast to previous applications of the central limit theorem to protein energetics. We support our derivation results with computational and experimental data on mutant protein stabilities across all types of protein residues.


Subject(s)
Amino Acids , Proteins , Mutation , Normal Distribution , Protein Stability , Proteins/genetics
12.
Biophys J ; 119(6): 1123-1134, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32857962

ABSTRACT

Cotranslational folding depends on the folding speed and stability of the nascent protein. It remains difficult, however, to predict which proteins cotranslationally fold. Here, we simulate evolution of model proteins to investigate how native structure influences evolution of cotranslational folding. We developed a model that connects protein folding during and after translation to cellular fitness. Model proteins evolved improved folding speed and stability, with proteins adopting one of two strategies for folding quickly. Low contact order proteins evolve to fold cotranslationally. Such proteins adopt native conformations early on during the translation process, with each subsequently translated residue establishing additional native contacts. On the other hand, high contact order proteins tend not to be stable in their native conformations until the full chain is nearly extruded. We also simulated evolution of slowly translating codons, finding that slower translation speeds at certain positions enhances cotranslational folding. Finally, we investigated real protein structures using a previously published data set that identified evolutionarily conserved rare codons in Escherichia coli genes and associated such codons with cotranslational folding intermediates. We found that protein substructures preceding conserved rare codons tend to have lower contact orders, in line with our finding that lower contact order proteins are more likely to fold cotranslationally. Our work shows how evolutionary selection pressure can cause proteins with local contact topologies to evolve cotranslational folding.


Subject(s)
Protein Biosynthesis , Protein Folding , Codon , Escherichia coli/genetics , Escherichia coli/metabolism , Proteins/genetics , Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 114(43): 11434-11439, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073068

ABSTRACT

Recent experiments and simulations have demonstrated that proteins can fold on the ribosome. However, the extent and generality of fitness effects resulting from cotranslational folding remain open questions. Here we report a genome-wide analysis that uncovers evidence of evolutionary selection for cotranslational folding. We describe a robust statistical approach to identify loci within genes that are both significantly enriched in slowly translated codons and evolutionarily conserved. Surprisingly, we find that domain boundaries can explain only a small fraction of these conserved loci. Instead, we propose that regions enriched in slowly translated codons are associated with cotranslational folding intermediates, which may be smaller than a single domain. We show that the intermediates predicted by a native-centric model of cotranslational folding account for the majority of these loci across more than 500 Escherichia coli proteins. By making a direct connection to protein folding, this analysis provides strong evidence that many synonymous substitutions have been selected to optimize translation rates at specific locations within genes. More generally, our results indicate that kinetics, and not just thermodynamics, can significantly alter the efficiency of self-assembly in a biological context.


Subject(s)
Bacteria/genetics , Bacterial Proteins/chemistry , Evolution, Molecular , Selection, Genetic , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Computer Simulation , Conserved Sequence , Gene Expression Regulation, Bacterial/physiology , Genome-Wide Association Study , Models, Biological , Protein Biosynthesis , Protein Folding
14.
J Biol Chem ; 293(46): 17997-18009, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30242128

ABSTRACT

Increased light scattering in the eye lens due to aggregation of the long-lived lens proteins, crystallins, is the cause of cataract disease. Several mutations in the gene encoding human γD-crystallin (HγD) cause misfolding and aggregation. Cataract-associated substitutions at Trp42 cause the protein to aggregate in vitro from a partially unfolded intermediate locked by an internal disulfide bridge, and proteomic evidence suggests a similar aggregation precursor is involved in age-onset cataract. Surprisingly, WT HγD can promote aggregation of the W42Q variant while itself remaining soluble. Here, a search for a biochemical mechanism for this interaction has revealed a previously unknown oxidoreductase activity in HγD. Using in vitro oxidation, mutational analysis, cysteine labeling, and MS, we have assigned this activity to a redox-active internal disulfide bond that is dynamically exchanged among HγD molecules. The W42Q variant acts as a disulfide sink, reducing oxidized WT and forming a distinct internal disulfide that kinetically traps the aggregation-prone intermediate. Our findings suggest a redox "hot potato" competition among WT and mutant or modified polypeptides wherein variants with the lowest kinetic stability are trapped in aggregation-prone intermediate states upon accepting disulfides from more stable variants. Such reactions may occur in other long-lived proteins that function in oxidizing environments. In these cases, aggregation may be forestalled by inhibiting disulfide flow toward mutant or damaged polypeptides.


Subject(s)
Disulfides/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , gamma-Crystallins/metabolism , Amino Acid Substitution , Cysteine/chemistry , Disulfides/chemistry , Escherichia coli , Humans , Mutagenesis, Site-Directed , Mutation , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Oxidoreductases Acting on Sulfur Group Donors/genetics , Protein Domains , Protein Multimerization , Protein Unfolding , Proteomics , gamma-Crystallins/chemistry , gamma-Crystallins/genetics
15.
Mol Biol Evol ; 35(10): 2390-2400, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29955873

ABSTRACT

Viral evolutionary pathways are determined by the fitness landscape, which maps viral genotype to fitness. However, a quantitative description of the landscape and the evolutionary forces on it remain elusive. Here, we apply a biophysical fitness model based on capsid folding stability and antibody binding affinity to predict the evolutionary pathway of norovirus escaping a neutralizing antibody. The model is validated by experimental evolution in bulk culture and in a drop-based microfluidics that propagates millions of independent small viral subpopulations. We demonstrate that along the axis of binding affinity, selection for escape variants and drift due to random mutations have the same direction, an atypical case in evolution. However, along folding stability, selection and drift are opposing forces whose balance is tuned by viral population size. Our results demonstrate that predictable epistatic tradeoffs between molecular traits of viral proteins shape viral evolution.


Subject(s)
Antibody Affinity , Biological Evolution , Genetic Fitness , Models, Genetic , Norovirus/genetics , Animals , Antibodies, Neutralizing , Capsid Proteins/physiology , Epistasis, Genetic , Mice , Protein Folding , Protein Stability , Selection, Genetic
16.
Bioinformatics ; 34(20): 3557-3565, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29741573

ABSTRACT

Motivation: Protein evolution spans time scales and its effects span the length of an organism. A web app named ProteomeVis is developed to provide a comprehensive view of protein evolution in the Saccharomyces cerevisiae and Escherichia coli proteomes. ProteomeVis interactively creates protein chain graphs, where edges between nodes represent structure and sequence similarities within user-defined ranges, to study the long time scale effects of protein structure evolution. The short time scale effects of protein sequence evolution are studied by sequence evolutionary rate (ER) correlation analyses with protein properties that span from the molecular to the organismal level. Results: We demonstrate the utility and versatility of ProteomeVis by investigating the distribution of edges per node in organismal protein chain universe graphs (oPCUGs) and putative ER determinants. S.cerevisiae and E.coli oPCUGs are scale-free with scaling constants of 1.79 and 1.56, respectively. Both scaling constants can be explained by a previously reported theoretical model describing protein structure evolution. Protein abundance most strongly correlates with ER among properties in ProteomeVis, with Spearman correlations of -0.49 (P-value < 10-10) and -0.46 (P-value < 10-10) for S.cerevisiae and E.coli, respectively. This result is consistent with previous reports that found protein expression to be the most important ER determinant. Availability and implementation: ProteomeVis is freely accessible at http://proteomevis.chem.harvard.edu. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Proteome/analysis , Software , Amino Acid Sequence , Carrier Proteins/analysis , Escherichia coli/chemistry , Escherichia coli Proteins/analysis , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/analysis
17.
Proc Natl Acad Sci U S A ; 113(11): E1470-8, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929328

ABSTRACT

Fitness landscapes of drug resistance constitute powerful tools to elucidate mutational pathways of antibiotic escape. Here, we developed a predictive biophysics-based fitness landscape of trimethoprim (TMP) resistance for Escherichia coli dihydrofolate reductase (DHFR). We investigated the activity, binding, folding stability, and intracellular abundance for a complete set of combinatorial DHFR mutants made out of three key resistance mutations and extended this analysis to DHFR originated from Chlamydia muridarum and Listeria grayi We found that the acquisition of TMP resistance via decreased drug affinity is limited by a trade-off in catalytic efficiency. Protein stability is concurrently affected by the resistant mutants, which precludes a precise description of fitness from a single molecular trait. Application of the kinetic flux theory provided an accurate model to predict resistance phenotypes (IC50) quantitatively from a unique combination of the in vitro protein molecular properties. Further, we found that a controlled modulation of the GroEL/ES chaperonins and Lon protease levels affects the intracellular steady-state concentration of DHFR in a mutation-specific manner, whereas IC50 is changed proportionally, as indeed predicted by the model. This unveils a molecular rationale for the pleiotropic role of the protein quality control machinery on the evolution of antibiotic resistance, which, as we illustrate here, may drastically confound the evolutionary outcome. These results provide a comprehensive quantitative genotype-phenotype map for the essential enzyme that serves as an important target of antibiotic and anticancer therapies.


Subject(s)
Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Tetrahydrofolate Dehydrogenase/genetics , Trimethoprim/pharmacology , Amino Acid Sequence , Biophysics/methods , Chlamydia muridarum/genetics , Directed Molecular Evolution , Enzyme Stability/genetics , Epistasis, Genetic , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Inhibitory Concentration 50 , Listeria/genetics , Molecular Sequence Data , Mutation , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Trimethoprim/metabolism
18.
Proc Biol Sci ; 285(1872)2018 02 14.
Article in English | MEDLINE | ID: mdl-29445020

ABSTRACT

Mutations in a microbial population can increase the frequency of a genotype not only by increasing its exponential growth rate, but also by decreasing its lag time or adjusting the yield (resource efficiency). The contribution of multiple life-history traits to selection is a critical question for evolutionary biology as we seek to predict the evolutionary fates of mutations. Here we use a model of microbial growth to show that there are two distinct components of selection corresponding to the growth and lag phases, while the yield modulates their relative importance. The model predicts rich population dynamics when there are trade-offs between phases: multiple strains can coexist or exhibit bistability due to frequency-dependent selection, and strains can engage in rock-paper-scissors interactions due to non-transitive selection. We characterize the environmental conditions and patterns of traits necessary to realize these phenomena, which we show to be readily accessible to experiments. Our results provide a theoretical framework for analysing high-throughput measurements of microbial growth traits, especially interpreting the pleiotropy and correlations between traits across mutants. This work also highlights the need for more comprehensive measurements of selection in simple microbial systems, where the concept of an ordinary fitness landscape breaks down.


Subject(s)
Bacteria/growth & development , Biological Evolution , Fungi/growth & development , Selection, Genetic , Bacteria/genetics , Fungi/genetics , Models, Genetic , Phenotype , Population Dynamics
19.
PLoS Genet ; 11(10): e1005612, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26484862

ABSTRACT

Horizontal gene transfer (HGT) plays a central role in bacterial evolution, yet the molecular and cellular constraints on functional integration of the foreign genes are poorly understood. Here we performed inter-species replacement of the chromosomal folA gene, encoding an essential metabolic enzyme dihydrofolate reductase (DHFR), with orthologs from 35 other mesophilic bacteria. The orthologous inter-species replacements caused a marked drop (in the range 10-90%) in bacterial growth rate despite the fact that most orthologous DHFRs are as stable as E.coli DHFR at 37°C and are more catalytically active than E. coli DHFR. Although phylogenetic distance between E. coli and orthologous DHFRs as well as their individual molecular properties correlate poorly with growth rates, the product of the intracellular DHFR abundance and catalytic activity (kcat/KM), correlates strongly with growth rates, indicating that the drop in DHFR abundance constitutes the major fitness barrier to HGT. Serial propagation of the orthologous strains for ~600 generations dramatically improved growth rates by largely alleviating the fitness barriers. Whole genome sequencing and global proteome quantification revealed that the evolved strains with the largest fitness improvements have accumulated mutations that inactivated the ATP-dependent Lon protease, causing an increase in the intracellular DHFR abundance. In one case DHFR abundance increased further due to mutations accumulated in folA promoter, but only after the lon inactivating mutations were fixed in the population. Thus, by apparently distinguishing between self and non-self proteins, protein homeostasis imposes an immediate and global barrier to the functional integration of foreign genes by decreasing the intracellular abundance of their products. Once this barrier is alleviated, more fine-tuned evolution occurs to adjust the function/expression of the transferred proteins to the constraints imposed by the intracellular environment of the host organism.


Subject(s)
Evolution, Molecular , Gene Transfer, Horizontal/genetics , Phylogeny , Tetrahydrofolate Dehydrogenase/genetics , Amino Acid Sequence/genetics , Escherichia coli/genetics , High-Throughput Nucleotide Sequencing , Homeostasis/genetics , Mutation , Species Specificity
20.
Biophys J ; 112(7): 1350-1365, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28402878

ABSTRACT

Homology modeling is a powerful tool for predicting a protein's structure. This approach is successful because proteins whose sequences are only 30% identical still adopt the same structure, while structure similarity rapidly deteriorates beyond the 30% threshold. By studying the divergence of protein structure as sequence evolves in real proteins and in evolutionary simulations, we show that this nonlinear sequence-structure relationship emerges as a result of selection for protein folding stability in divergent evolution. Fitness constraints prevent the emergence of unstable protein evolutionary intermediates, thereby enforcing evolutionary paths that preserve protein structure despite broad sequence divergence. However, on longer timescales, evolution is punctuated by rare events where the fitness barriers obstructing structure evolution are overcome and discovery of new structures occurs. We outline biophysical and evolutionary rationale for broad variation in protein family sizes, prevalence of compact structures among ancient proteins, and more rapid structure evolution of proteins with lower packing density.


Subject(s)
Evolution, Molecular , Proteins/chemistry , Selection, Genetic , Computer Simulation , Models, Molecular , Monte Carlo Method , Protein Domains , Protein Stability , Protein Structure, Secondary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL