Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
2.
Nat Biomed Eng ; 8(3): 214-232, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37814006

ABSTRACT

Developing therapeutic antibodies is laborious and costly. Here we report a method for antibody discovery that leverages the Illumina HiSeq platform to, within 3 days, screen in the order of 108 antibody-antigen interactions. The method, which we named 'deep screening', involves the clustering and sequencing of antibody libraries, the conversion of the DNA clusters into complementary RNA clusters covalently linked to the instrument's flow-cell surface on the same location, the in situ translation of the clusters into antibodies tethered via ribosome display, and their screening via fluorescently labelled antigens. By using deep screening, we discovered low-nanomolar nanobodies to a model antigen using 4 × 106 unique variants from yeast-display-enriched libraries, and high-picomolar single-chain antibody fragment leads for human interleukin-7 directly from unselected synthetic repertoires. We also leveraged deep screening of a library of 2.4 × 105 sequences of the third complementarity-determining region of the heavy chain of an anti-human epidermal growth factor receptor 2 (HER2) antibody as input for a large language model that generated new single-chain antibody fragment sequences with higher affinity for HER2 than those in the original library.


Subject(s)
Antibodies , High-Throughput Nucleotide Sequencing , Humans , Antibodies/genetics , Antibodies/metabolism , Gene Library , Immunoglobulin Fragments , Ribosomes/genetics , Ribosomes/metabolism
3.
ChemSystemsChem ; 5(5)2023 Sep.
Article in English | MEDLINE | ID: mdl-38074198

ABSTRACT

The spontaneous emergence of function from diverse RNA sequence pools is widely considered an important transition in the origin of life. Here we show that diverse sequence pools are not a prerequisite for the emergence of function. Starting five independent selection experiments each from a single RNA seed sequence - comprising a central homopolymeric poly-A (or poly-U) segment flanked by different conserved primer binding sites - we observe transformation (continuous drift) of the seeds into low diversity sequence pools by mutation, truncation and recombination without ever reaching that of a random pool even after 24 rounds. Upon continuous error prone replication and selection for ATP binding we isolate specific ATP- or GTP-binding aptamers with low micromolar affinities. Our results have implications for early RNA evolution in the light of the high mutation rates associated with both non-enzymatic and enzymatic prebiotic RNA replication.

4.
Nature ; 623(7988): 842-852, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37853127

ABSTRACT

Optimum protein function and biochemical activity critically depends on water availability because solvent thermodynamics drive protein folding and macromolecular interactions1. Reciprocally, macromolecules restrict the movement of 'structured' water molecules within their hydration layers, reducing the available 'free' bulk solvent and therefore the total thermodynamic potential energy of water, or water potential. Here, within concentrated macromolecular solutions such as the cytosol, we found that modest changes in temperature greatly affect the water potential, and are counteracted by opposing changes in osmotic strength. This duality of temperature and osmotic strength enables simple manipulations of solvent thermodynamics to prevent cell death after extreme cold or heat shock. Physiologically, cells must sustain their activity against fluctuating temperature, pressure and osmotic strength, which impact water availability within seconds. Yet, established mechanisms of water homeostasis act over much slower timescales2,3; we therefore postulated the existence of a rapid compensatory response. We find that this function is performed by water potential-driven changes in macromolecular assembly, particularly biomolecular condensation of intrinsically disordered proteins. The formation and dissolution of biomolecular condensates liberates and captures free water, respectively, quickly counteracting thermal or osmotic perturbations of water potential, which is consequently robustly buffered in the cytoplasm. Our results indicate that biomolecular condensation constitutes an intrinsic biophysical feedback response that rapidly compensates for intracellular osmotic and thermal fluctuations. We suggest that preserving water availability within the concentrated cytosol is an overlooked evolutionary driver of protein (dis)order and function.


Subject(s)
Macromolecular Substances , Proteins , Solvents , Thermodynamics , Water , Cell Death , Cytosol/chemistry , Cytosol/metabolism , Homeostasis , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Osmolar Concentration , Pressure , Proteins/chemistry , Proteins/metabolism , Solvents/chemistry , Solvents/metabolism , Temperature , Time Factors , Water/chemistry , Water/metabolism
5.
Biochem J ; 478(17): 3319-3330, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34424335

ABSTRACT

Angiotensinogen fine-tunes the tightly controlled activity of the renin-angiotensin system by modulating the release of angiotensin peptides that control blood pressure. One mechanism by which this modulation is achieved is via angiotensinogen's Cys18-Cys138 disulfide bond that acts as a redox switch. Molecular dynamics simulations of each redox state of angiotensinogen reveal subtle dynamic differences between the reduced and oxidised forms, particularly at the N-terminus. Surface plasmon resonance data demonstrate that the two redox forms of angiotensinogen display different binding kinetics to an immobilised anti-angiotensinogen monoclonal antibody. Mass spectrometry mapped the epitope for the antibody to the N-terminal region of angiotensinogen. We therefore provide evidence that the different redox forms of angiotensinogen can be detected by an antibody-based detection method.


Subject(s)
Angiotensinogen/chemistry , Angiotensinogen/metabolism , Molecular Dynamics Simulation , Surface Plasmon Resonance/methods , Angiotensinogen/genetics , Angiotensinogen/immunology , Antibodies, Monoclonal/immunology , Blood Pressure/physiology , Cysteine/metabolism , Disulfides/metabolism , Epitopes/immunology , Humans , Kinetics , Oxidation-Reduction , Protein Binding , Protein Conformation, alpha-Helical , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Renin-Angiotensin System/physiology
6.
J Biol Chem ; 296: 100447, 2021.
Article in English | MEDLINE | ID: mdl-33617878

ABSTRACT

The fibronectin type III (FN3) monobody domain is a promising non-antibody scaffold, which features a less complex architecture than an antibody while maintaining analogous binding loops. We previously developed FN3Con, a hyperstable monobody derivative with diagnostic and therapeutic potential. Prestabilization of the scaffold mitigates the stability-function trade-off commonly associated with evolving a protein domain toward biological activity. Here, we aimed to examine if the FN3Con monobody could take on antibody-like binding to therapeutic targets, while retaining its extreme stability. We targeted the first of the Adnectin derivative of monobodies to reach clinical trials, which was engineered by directed evolution for binding to the therapeutic target VEGFR2; however, this function was gained at the expense of large losses in thermostability and increased oligomerization. In order to mitigate these losses, we grafted the binding loops from Adnectin-anti-VEGFR2 (CT-322) onto the prestabilized FN3Con scaffold to produce a domain that successfully bound with high affinity to the therapeutic target VEGFR2. This FN3Con-anti-VEGFR2 construct also maintains high thermostability, including remarkable long-term stability, retaining binding activity after 2 years of storage at 36 °C. Further investigations into buffer excipients doubled the presence of monomeric monobody in accelerated stability trials. These data suggest that loop grafting onto a prestabilized scaffold is a viable strategy for the development of monobody domains with desirable biophysical characteristics and that FN3Con is therefore well-suited to applications such as the evolution of multiple paratopes or shelf-stable diagnostics and therapeutics.


Subject(s)
Antibodies/metabolism , Fibronectin Type III Domain/genetics , Antibodies/immunology , Fibronectin Type III Domain/immunology , Fibronectins/genetics , Fibronectins/immunology , Fibronectins/metabolism , Genetic Engineering/methods , Humans , Matrix Attachment Regions , Mutation , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Binding/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Proc Natl Acad Sci U S A ; 117(36): 22341-22350, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32855302

ABSTRACT

Conformational diversity and self-cross-reactivity of antigens have been correlated with evasion from neutralizing antibody responses. We utilized single cell B cell sequencing, biolayer interferometry and X-ray crystallography to trace mutation selection pathways where the antibody response must resolve cross-reactivity between foreign and self-proteins bearing near-identical contact surfaces, but differing in conformational flexibility. Recurring antibody mutation trajectories mediate long-range rearrangements of framework (FW) and complementarity determining regions (CDRs) that increase binding site conformational diversity. These antibody mutations decrease affinity for self-antigen 19-fold and increase foreign affinity 67-fold, to yield a more than 1,250-fold increase in binding discrimination. These results demonstrate how conformational diversity in antigen and antibody does not act as a barrier, as previously suggested, but rather facilitates high affinity and high discrimination between foreign and self.


Subject(s)
Antibodies , Antibody Diversity/genetics , Autoantigens , Gene Rearrangement, B-Lymphocyte/genetics , Mutation/genetics , Animals , Antibodies/chemistry , Antibodies/genetics , Antibodies/metabolism , Antibody Affinity/genetics , Autoantibodies/chemistry , Autoantibodies/genetics , Autoantibodies/metabolism , Autoantigens/chemistry , Autoantigens/metabolism , Complementarity Determining Regions/genetics , Immunity, Humoral/genetics , Mice , Models, Molecular , Protein Conformation , Somatic Hypermutation, Immunoglobulin/genetics
8.
Nat Chem ; 12(8): 683-690, 2020 08.
Article in English | MEDLINE | ID: mdl-32690899

ABSTRACT

The ability of reverse transcriptases (RTs) to synthesize a complementary DNA from natural RNA and a range of unnatural xeno nucleic acid (XNA) template chemistries, underpins key methods in molecular and synthetic genetics. However, RTs have proven challenging to discover and engineer, in particular for the more divergent XNA chemistries. Here we describe a general strategy for the directed evolution of RT function for any template chemistry called compartmentalized bead labelling and demonstrate it by the directed evolution of efficient RTs for 2'-O-methyl RNA and hexitol nucleic acids and the discovery of RTs for the orphan XNA chemistries D-altritol nucleic acid and 2'-methoxyethyl RNA, for which previously no RTs existed. Finally, we describe the engineering of XNA RTs with active exonucleolytic proofreading as well as the directed evolution of RNA RTs with very high complementary DNA synthesis fidelities, even in the absence of proofreading.


Subject(s)
Evolution, Molecular , RNA-Directed DNA Polymerase/metabolism , RNA/metabolism , Gene Library , Leukemia Virus, Murine/enzymology , Mutagenesis, Site-Directed , Nucleic Acid Amplification Techniques , RNA-Directed DNA Polymerase/genetics
9.
Nat Immunol ; 20(10): 1299-1310, 2019 10.
Article in English | MEDLINE | ID: mdl-31534238

ABSTRACT

Resisting and tolerating microbes are alternative strategies to survive infection, but little is known about the evolutionary mechanisms controlling this balance. Here genomic analyses of anatomically modern humans, extinct Denisovan hominins and mice revealed a TNFAIP3 allelic series with alterations in the encoded immune response inhibitor A20. Each TNFAIP3 allele encoded substitutions at non-catalytic residues of the ubiquitin protease OTU domain that diminished IκB kinase-dependent phosphorylation and activation of A20. Two TNFAIP3 alleles encoding A20 proteins with partial phosphorylation deficits seemed to be beneficial by increasing immunity without causing spontaneous inflammatory disease: A20 T108A;I207L, originating in Denisovans and introgressed in modern humans throughout Oceania, and A20 I325N, from an N-ethyl-N-nitrosourea (ENU)-mutagenized mouse strain. By contrast, a rare human TNFAIP3 allele encoding an A20 protein with 95% loss of phosphorylation, C243Y, caused spontaneous inflammatory disease in humans and mice. Analysis of the partial-phosphorylation A20 I325N allele in mice revealed diminished tolerance of bacterial lipopolysaccharide and poxvirus inoculation as tradeoffs for enhanced immunity.


Subject(s)
Poxviridae Infections/immunology , Poxviridae/physiology , Protein Domains/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Alleles , Animals , Extinction, Biological , Humans , Immunity , Inflammation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense/genetics , Phosphorylation
10.
Nat Chem ; 11(6): 533-542, 2019 06.
Article in English | MEDLINE | ID: mdl-31011171

ABSTRACT

The physicochemical properties of nucleic acids are dominated by their highly charged phosphodiester backbone chemistry. This polyelectrolyte structure decouples information content (base sequence) from bulk properties, such as solubility, and has been proposed as a defining trait of all informational polymers. However, this conjecture has not been tested experimentally. Here, we describe the encoded synthesis of a genetic polymer with an uncharged backbone chemistry: alkyl phosphonate nucleic acids (phNAs) in which the canonical, negatively charged phosphodiester is replaced by an uncharged P-alkyl phosphonodiester backbone. Using synthetic chemistry and polymerase engineering, we describe the enzymatic, DNA-templated synthesis of P-methyl and P-ethyl phNAs, and the directed evolution of specific streptavidin-binding phNA aptamer ligands directly from random-sequence mixed P-methyl/P-ethyl phNA repertoires. Our results establish an example of the DNA-templated enzymatic synthesis and evolution of an uncharged genetic polymer and provide a foundational methodology for their exploration as a source of novel functional molecules.


Subject(s)
DNA/chemistry , Organophosphonates/chemistry , Aptamers, Nucleotide/chemistry , DNA/chemical synthesis , DNA/genetics , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , Directed Molecular Evolution/methods , Mutation , Nucleic Acid Conformation , Organophosphonates/chemical synthesis , Protein Engineering/methods , Streptavidin/chemistry , Thermococcaceae/enzymology , Thermococcales/enzymology
11.
Sci Rep ; 9(1): 3870, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30846766

ABSTRACT

Serine proteinase inhibitors (serpins), typically fold to a metastable native state and undergo a major conformational change in order to inhibit target proteases. However, conformational lability of the native serpin fold renders them susceptible to misfolding and aggregation, and underlies misfolding diseases such as α1-antitrypsin deficiency. Serpin specificity towards its protease target is dictated by its flexible and solvent exposed reactive centre loop (RCL), which forms the initial interaction with the target protease during inhibition. Previous studies have attempted to alter the specificity by mutating the RCL to that of a target serpin, but the rules governing specificity are not understood well enough yet to enable specificity to be engineered at will. In this paper, we use conserpin, a synthetic, thermostable serpin, as a model protein with which to investigate the determinants of serpin specificity by engineering its RCL. Replacing the RCL sequence with that from α1-antitrypsin fails to restore specificity against trypsin or human neutrophil elastase. Structural determination of the RCL-engineered conserpin and molecular dynamics simulations indicate that, although the RCL sequence may partially dictate specificity, local electrostatics and RCL dynamics may dictate the rate of insertion during protease inhibition, and thus whether it behaves as an inhibitor or a substrate. Engineering serpin specificity is therefore substantially more complex than solely manipulating the RCL sequence, and will require a more thorough understanding of how conformational dynamics achieves the delicate balance between stability, folding and function required by the exquisite serpin mechanism of action.


Subject(s)
Serpins/metabolism , Amino Acid Sequence , Escherichia coli , Humans , Leukocyte Elastase/metabolism , Molecular Dynamics Simulation , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Protein Binding , Protein Conformation , Protein Engineering , Protein Folding , Serpins/chemistry , Serpins/genetics , Static Electricity , Trypsin/metabolism
12.
Elife ; 72018 11 21.
Article in English | MEDLINE | ID: mdl-30461419

ABSTRACT

Recombination, the exchange of information between different genetic polymer strands, is of fundamental importance in biology for genome maintenance and genetic diversification and is mediated by dedicated recombinase enzymes. Here, we describe an innate capacity for non-enzymatic recombination (and ligation) in random-sequence genetic oligomer pools. Specifically, we examine random and semi-random eicosamer (N20) pools of RNA, DNA and the unnatural genetic polymers ANA (arabino-), HNA (hexitol-) and AtNA (altritol-nucleic acids). While DNA, ANA and HNA pools proved inert, RNA (and to a lesser extent AtNA) pools displayed diverse modes of spontaneous intermolecular recombination, connecting recombination mechanistically to the vicinal ring cis-diol configuration shared by RNA and AtNA. Thus, the chemical constitution that renders both susceptible to hydrolysis emerges as the fundamental determinant of an innate capacity for recombination, which is shown to promote a concomitant increase in compositional, informational and structural pool complexity and hence evolutionary potential.


Subject(s)
DNA/chemistry , Oligodeoxyribonucleotides/chemistry , Oligoribonucleotides/chemistry , RNA/chemistry , Recombination, Genetic , Base Pairing , Base Sequence , DNA/genetics , Kinetics , Models, Molecular , Nucleic Acid Conformation , Oligodeoxyribonucleotides/genetics , Oligoribonucleotides/genetics , Polysaccharides/chemistry , Polysaccharides/metabolism , RNA/genetics , Solutions , Sugar Alcohols/chemistry , Sugar Alcohols/metabolism , Thermodynamics
13.
ChemMedChem ; 13(23): 2504-2513, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30318749

ABSTRACT

The M1 metallo-aminopeptidase from Plasmodium falciparum, PfA-M1, is an attractive drug target for the design of new antimalarials. Bestatin, a broad-spectrum metalloprotease inhibitor, is a moderate inhibitor of PfA-M1, and has been used to provide structure-activity relationships to inform drug design. The crystal structure of PfA-M1 with bestatin bound within its active site has been determined; however, dynamics of the inhibitor and the association or dissociation pathway have yet to be characterized. Here we present an all-atom molecular dynamics study where we have generated a hidden Markov state model from 2.3 µs of molecular dynamics simulation. Our hidden Markov state model identifies five macrostates that clearly show the events involved in bestatin dissociation from the PfA-M1 active site. The results show for the first time that bestatin can escape the substrate specificity pockets of the enzyme, primarily due to weak interactions within the pockets. Our approach identifies relevant conformational sampling of the inhibitor inside the enzyme and the protein dynamics that could be exploited to produce potent and selective inhibitors that can differentiate between similar members of the M1 aminopeptidase superfamily.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Leucine/analogs & derivatives , Plasmodium falciparum/enzymology , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Catalytic Domain/drug effects , Drug Discovery , Humans , Leucine/pharmacology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Molecular Docking Simulation , Molecular Dynamics Simulation , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Protein Binding
14.
J Mol Biol ; 430(18 Pt B): 3200-3217, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30111491

ABSTRACT

Canonical mechanisms of protein evolution include the duplication and diversification of pre-existing folds through genetic alterations that include point mutations, insertions, deletions, and copy number amplifications, as well as post-translational modifications that modify processes such as folding efficiency and cellular localization. Following a survey of the human mutation database, we have identified an additional mechanism that we term "structural capacitance," which results in the de novo generation of microstructure in previously disordered regions. We suggest that the potential for structural capacitance confers select proteins with the capacity to evolve over rapid timescales, facilitating saltatory evolution as opposed to gradualistic canonical Darwinian mechanisms. Our results implicate the elements of protein microstructure generated by this distinct mechanism in the pathogenesis of a wide variety of human diseases. The benefits of rapidly furnishing the potential for evolutionary change conferred by structural capacitance are consequently counterbalanced by this accompanying risk. The phenomenon of structural capacitance has implications ranging from the ancestral diversification of protein folds to the engineering of synthetic proteins with enhanced evolvability.


Subject(s)
Disease Susceptibility , Evolution, Molecular , Proteins/chemistry , Humans , Models, Molecular , Mutation , Protein Conformation , Proteins/genetics , Proteins/metabolism , Structure-Activity Relationship
15.
Science ; 360(6385): 223-226, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29650674

ABSTRACT

Antibodies have the specificity to differentiate foreign antigens that mimic self antigens, but it remains unclear how such specificity is acquired. In a mouse model, we generated B cells displaying an antibody that cross-reacts with two related protein antigens expressed on self versus foreign cells. B cell anergy was imposed by self antigen but reversed upon challenge with high-density foreign antigen, leading to germinal center recruitment and antibody gene hypermutation. Single-cell analysis detected rapid selection for mutations that decrease self affinity and slower selection for epistatic mutations that specifically increase foreign affinity. Crystal structures revealed that these mutations exploited subtle topological differences to achieve 5000-fold preferential binding to foreign over self epitopes. Resolution of antigenic mimicry drove the optimal affinity maturation trajectory, highlighting the value of retaining self-reactive clones as substrates for protective antibody responses.


Subject(s)
Antibodies/genetics , Antibody Formation/genetics , Autoantigens/immunology , Germinal Center/immunology , Molecular Mimicry/genetics , Self Tolerance , Animals , Antibodies/chemistry , Antibodies/immunology , Antibody Affinity/genetics , B-Lymphocytes/immunology , Clonal Anergy , Cross Reactions , Crystallography, X-Ray , Mice , Mice, Mutant Strains , Mutation , Nucleoproteins/genetics , Nucleoproteins/immunology , Selection, Genetic , Single-Cell Analysis
16.
J Biomol Struct Dyn ; 36(10): 2595-2604, 2018 Aug.
Article in English | MEDLINE | ID: mdl-28782419

ABSTRACT

The M1 and M17 aminopeptidases are metallo-exopeptidases that rely on the presence of divalent cations, usually zinc, in their active site for proteolytic activity. They are from separate protease superfamilies, however, members often have overlapping substrate specificity. Inhibitors of one or both enzymes can be used to modulate hypertension, reduce proliferation of certain types of cancers and control malaria parasites. Current inhibitors act to chelate the zinc ions in the active site, locking the enzymes in an inactive transition state. We were interested in using a computational approach to understand the structure and dynamics of the M1 and M17 aminopeptidases, however, the presence of the essential metal ions in the proteases presents a challenge to classical molecular dynamics (MD) simulation. The zinc amber force field does not contain applicable descriptions of the zinc coordination environment present in either of these two protease families. To provide tools for the study of these two enzymes, we have used the metal centre parameter builder to generate new hybrid bonded/nonbonded force field (FF) parameters to correctly describe the active site architecture for each enzyme. The new parameters were evaluated by fitting the normal mode frequencies of molecular mechanics to the quantum mechanics frequencies and validated by performing short MD simulations. The new FF parameters now enable more accurate and reliable MD simulations for any member of the M1 or M17 aminopeptidase superfamilies.


Subject(s)
Aminopeptidases/chemistry , Molecular Dynamics Simulation , Plasmodium falciparum/enzymology , Zinc/chemistry , Time Factors
17.
Sci Signal ; 10(480)2017 May 23.
Article in English | MEDLINE | ID: mdl-28536301

ABSTRACT

Chemokines and their receptors collectively orchestrate the trafficking of leukocytes in normal immune function and inflammatory diseases. Different chemokines can induce distinct responses at the same receptor. In comparison to monocyte chemoattractant protein-1 (MCP-1; also known as CCL2), the chemokines MCP-2 (CCL8) and MCP-3 (CCL7) are partial agonists of their shared receptor CCR2, a key regulator of the trafficking of monocytes and macrophages that contribute to the pathology of atherosclerosis, obesity, and type 2 diabetes. Through experiments with chimeras of MCP-1 and MCP-3, we identified the chemokine amino-terminal region as being the primary determinant of both the binding and signaling selectivity of these two chemokines at CCR2. Analysis of CCR2 mutants showed that the chemokine amino terminus interacts with the major subpocket in the transmembrane helical bundle of CCR2, which is distinct from the interactions of some other chemokines with the minor subpockets of their receptors. These results suggest the major subpocket as a target for the development of small-molecule inhibitors of CCR2.


Subject(s)
Chemokines/chemistry , Chemokines/metabolism , Receptors, CCR2/chemistry , Receptors, CCR2/metabolism , Amino Acid Sequence , Chemokine CCL2/chemistry , Chemokine CCL2/metabolism , Chemokine CCL7/chemistry , Chemokine CCL7/metabolism , Humans , Models, Molecular , Protein Binding , Receptors, CCR2/genetics , Sequence Homology
18.
Proc Natl Acad Sci U S A ; 114(15): 3897-3902, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28356519

ABSTRACT

Ancestral protein reconstruction allows the resurrection and characterization of ancient proteins based on computational analyses of sequences of modern-day proteins. Unfortunately, many protein families are highly divergent and not suitable for sequence-based reconstruction approaches. This limitation is exemplified by the antigen receptors of jawed vertebrates (B- and T-cell receptors), heterodimers formed by pairs of Ig domains. These receptors are believed to have evolved from an extinct homodimeric ancestor through a process of gene duplication and diversification; however molecular evidence has so far remained elusive. Here, we use a structural approach and laboratory evolution to reconstruct such molecules and characterize their interaction with antigen. High-resolution crystal structures of reconstructed homodimeric receptors in complex with hen-egg white lysozyme demonstrate how nanomolar affinity binding of asymmetrical antigen is enabled through selective recruitment and structural plasticity within the receptor-binding site. Our results provide structural evidence in support of long-held theories concerning the evolution of antigen receptors, and provide a blueprint for the experimental reconstruction of protein ancestry in the absence of phylogenetic evidence.


Subject(s)
Evolution, Molecular , Phylogeny , Receptors, Polymeric Immunoglobulin/chemistry , Animals , Crystallography, X-Ray , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin kappa-Chains/chemistry , Immunoglobulin kappa-Chains/genetics , Muramidase/chemistry , Receptors, Polymeric Immunoglobulin/genetics , Vertebrates/genetics , Vertebrates/immunology
19.
Sci Rep ; 6: 35385, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27767076

ABSTRACT

The kallikrein-related peptidase (KLK) family of proteases is involved in many aspects of human health and disease. One member of this family, KLK4, has been implicated in cancer development and metastasis. Understanding mechanisms of inactivation are critical to developing selective KLK4 inhibitors. We have determined the X-ray crystal structures of KLK4 in complex with both sunflower trypsin inhibitor-1 (SFTI-1) and a rationally designed SFTI-1 derivative to atomic (~1 Å) resolution, as well as with bound nickel. These structures offer a structural rationalization for the potency and selectivity of these inhibitors, and together with MD simulation and computational analysis, reveal a dynamic pathway between the metal binding exosite and the active site, providing key details of a previously proposed allosteric mode of inhibition. Collectively, this work provides insight into both direct and indirect mechanisms of inhibition for KLK4 that have broad implications for the enzymology of the serine protease superfamily, and may potentially be exploited for the design of therapeutic inhibitors.


Subject(s)
Kallikreins/antagonists & inhibitors , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Gene Expression Regulation , Helianthus , Humans , Hydrogen Bonding , Metals/chemistry , Molecular Dynamics Simulation , Nickel/chemistry , Peptides, Cyclic/chemistry , Protein Binding , Protein Conformation , Protein Folding , Serine Proteases/chemistry , Trypsin/chemistry
20.
Sci Rep ; 6: 33958, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27667094

ABSTRACT

The rugged folding landscapes of functional proteins puts them at risk of misfolding and aggregation. Serine protease inhibitors, or serpins, are paradigms for this delicate balance between function and misfolding. Serpins exist in a metastable state that undergoes a major conformational change in order to inhibit proteases. However, conformational labiality of the native serpin fold renders them susceptible to misfolding, which underlies misfolding diseases such as α1-antitrypsin deficiency. To investigate how serpins balance function and folding, we used consensus design to create conserpin, a synthetic serpin that folds reversibly, is functional, thermostable, and polymerization resistant. Characterization of its structure, folding and dynamics suggest that consensus design has remodeled the folding landscape to reconcile competing requirements for stability and function. This approach may offer general benefits for engineering functional proteins that have risky folding landscapes, including the removal of aggregation-prone intermediates, and modifying scaffolds for use as protein therapeutics.

SELECTION OF CITATIONS
SEARCH DETAIL
...