Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Cell ; 157(7): 1644-1656, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24949974

RESUMO

Because apoptosis of infected cells can limit virus production and spread, some viruses have co-opted prosurvival genes from the host. This includes the Epstein-Barr virus (EBV) gene BHRF1, a homolog of human Bcl-2 proteins that block apoptosis and are associated with cancer. Computational design and experimental optimization were used to generate a novel protein called BINDI that binds BHRF1 with picomolar affinity. BINDI recognizes the hydrophobic cleft of BHRF1 in a manner similar to other Bcl-2 protein interactions but makes many additional contacts to achieve exceptional affinity and specificity. BINDI induces apoptosis in EBV-infected cancer lines, and when delivered with an antibody-targeted intracellular delivery carrier, BINDI suppressed tumor growth and extended survival in a xenograft disease model of EBV-positive human lymphoma. High-specificity-designed proteins that selectively kill target cells may provide an advantage over the toxic compounds used in current generation antibody-drug conjugates.


Assuntos
Herpesvirus Humano 4/química , Engenharia de Proteínas , Proteínas/farmacologia , Proteínas Virais/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Biologia Computacional , Cristalografia por Raios X , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Herpesvirus Humano 4/fisiologia , Xenoenxertos , Humanos , Linfoma de Células B/tratamento farmacológico , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Transplante de Neoplasias , Proteínas/química , Proteínas/metabolismo , Alinhamento de Sequência , Proteínas Virais/química
2.
Nat Chem Biol ; 20(7): 906-915, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38831036

RESUMO

Natural photosystems couple light harvesting to charge separation using a 'special pair' of chlorophyll molecules that accepts excitation energy from the antenna and initiates an electron-transfer cascade. To investigate the photophysics of special pairs independently of the complexities of native photosynthetic proteins, and as a first step toward creating synthetic photosystems for new energy conversion technologies, we designed C2-symmetric proteins that hold two chlorophyll molecules in closely juxtaposed arrangements. X-ray crystallography confirmed that one designed protein binds two chlorophylls in the same orientation as native special pairs, whereas a second designed protein positions them in a previously unseen geometry. Spectroscopy revealed that the chlorophylls are excitonically coupled, and fluorescence lifetime imaging demonstrated energy transfer. The cryo-electron microscopy structure of a designed 24-chlorophyll octahedral nanocage with a special pair on each edge closely matched the design model. The results suggest that the de novo design of artificial photosynthetic systems is within reach of current computational methods.


Assuntos
Clorofila , Clorofila/química , Clorofila/metabolismo , Cristalografia por Raios X , Modelos Moleculares , Fotossíntese , Transferência de Energia , Microscopia Crioeletrônica , Conformação Proteica , Complexos de Proteínas Captadores de Luz/química , Complexos de Proteínas Captadores de Luz/metabolismo
3.
Nucleic Acids Res ; 51(9): 4467-4487, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-36987874

RESUMO

Type IIS restriction endonucleases contain separate DNA recognition and catalytic domains and cleave their substrates at well-defined distances outside their target sequences. They are employed in biotechnology for a variety of purposes, including the creation of gene-targeting zinc finger and TAL effector nucleases and DNA synthesis applications such as Golden Gate assembly. The most thoroughly studied Type IIS enzyme, FokI, has been shown to require multimerization and engagement with multiple DNA targets for optimal cleavage activity; however, details of how it or similar enzymes forms a DNA-bound reaction complex have not been described at atomic resolution. Here we describe biochemical analyses of DNA cleavage by the Type IIS PaqCI restriction endonuclease and a series of molecular structures in the presence and absence of multiple bound DNA targets. The enzyme displays a similar tetrameric organization of target recognition domains in the absence or presence of bound substrate, with a significant repositioning of endonuclease domains in a trapped DNA-bound complex that is poised to deliver the first of a series of double-strand breaks. PaqCI and FokI share similar structural mechanisms of DNA cleavage, but considerable differences in their domain organization and quaternary architecture, facilitating comparisons between distinct Type IIS enzymes.


Assuntos
DNA , Desoxirribonucleases de Sítio Específico do Tipo II , Desoxirribonucleases de Sítio Específico do Tipo II/química , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , DNA/química , DNA/genética , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Especificidade por Substrato
4.
Nucleic Acids Res ; 51(8): 3513-3528, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-36794719

RESUMO

Bacteriophage exclusion ('BREX') systems are multi-protein complexes encoded by a variety of bacteria and archaea that restrict phage by an unknown mechanism. One BREX factor, termed BrxL, has been noted to display sequence similarity to various AAA+ protein factors including Lon protease. In this study we describe multiple CryoEM structures of BrxL that demonstrate it to be a chambered, ATP-dependent DNA binding protein. The largest BrxL assemblage corresponds to a dimer of heptamers in the absence of bound DNA, versus a dimer of hexamers when DNA is bound in its central pore. The protein displays DNA-dependent ATPase activity, and ATP binding promotes assembly of the complex on DNA. Point mutations within several regions of the protein-DNA complex alter one or more in vitro behaviors and activities, including ATPase activity and ATP-dependent association with DNA. However, only the disruption of the ATPase active site fully eliminates phage restriction, indicating that other mutations can still complement BrxL function within the context of an otherwise intact BREX system. BrxL displays significant structural homology to MCM subunits (the replicative helicase in archaea and eukaryotes), implying that it and other BREX factors may collaborate to disrupt initiation of phage DNA replication.


Assuntos
Acinetobacter , Protease La , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Archaea/genética , Bacteriófagos/genética , Bacteriófagos/metabolismo , DNA/metabolismo , DNA Helicases/metabolismo , Ligação Proteica , Acinetobacter/enzimologia , Acinetobacter/virologia , Protease La/ultraestrutura
5.
PLoS Biol ; 19(10): e3001428, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34644300

RESUMO

To overcome CRISPR-Cas defense systems, many phages and mobile genetic elements (MGEs) encode CRISPR-Cas inhibitors called anti-CRISPRs (Acrs). Nearly all characterized Acrs directly bind Cas proteins to inactivate CRISPR immunity. Here, using functional metagenomic selection, we describe AcrIIA22, an unconventional Acr found in hypervariable genomic regions of clostridial bacteria and their prophages from human gut microbiomes. AcrIIA22 does not bind strongly to SpyCas9 but nonetheless potently inhibits its activity against plasmids. To gain insight into its mechanism, we obtained an X-ray crystal structure of AcrIIA22, which revealed homology to PC4-like nucleic acid-binding proteins. Based on mutational analyses and functional assays, we deduced that acrIIA22 encodes a DNA nickase that relieves torsional stress in supercoiled plasmids. This may render them less susceptible to SpyCas9, which uses free energy from negative supercoils to form stable R-loops. Modifying DNA topology may provide an additional route to CRISPR-Cas resistance in phages and MGEs.


Assuntos
Proteínas de Bactérias/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , DNA/metabolismo , Proteínas de Bactérias/química , Mapeamento de Sequências Contíguas , DNA Super-Helicoidal/metabolismo , Genoma Bacteriano , Metagenômica , Plasmídeos , Prófagos/genética , Multimerização Proteica
6.
Nature ; 561(7724): 485-491, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30209393

RESUMO

The regular arrangements of ß-strands around a central axis in ß-barrels and of α-helices in coiled coils contrast with the irregular tertiary structures of most globular proteins, and have fascinated structural biologists since they were first discovered. Simple parametric models have been used to design a wide range of α-helical coiled-coil structures, but to date there has been no success with ß-barrels. Here we show that accurate de novo design of ß-barrels requires considerable symmetry-breaking to achieve continuous hydrogen-bond connectivity and eliminate backbone strain. We then build ensembles of ß-barrel backbone models with cavity shapes that match the fluorogenic compound DFHBI, and use a hierarchical grid-based search method to simultaneously optimize the rigid-body placement of DFHBI in these cavities and the identities of the surrounding amino acids to achieve high shape and chemical complementarity. The designs have high structural accuracy and bind and fluorescently activate DFHBI in vitro and in Escherichia coli, yeast and mammalian cells. This de novo design of small-molecule binding activity, using backbones custom-built to bind the ligand, should enable the design of increasingly sophisticated ligand-binding proteins, sensors and catalysts that are not limited by the backbone geometries available in known protein structures.


Assuntos
Compostos de Benzil/química , Fluorescência , Imidazolinas/química , Proteínas/química , Animais , Compostos de Benzil/análise , Células COS , Chlorocebus aethiops , Escherichia coli , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Ligação de Hidrogênio , Imidazolinas/análise , Ligantes , Ligação Proteica , Domínios Proteicos , Dobramento de Proteína , Estabilidade Proteica , Estrutura Secundária de Proteína , Reprodutibilidade dos Testes , Leveduras
7.
Nucleic Acids Res ; 50(9): 5171-5190, 2022 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-35511079

RESUMO

Bacteriophage exclusion ('BREX') phage restriction systems are found in a wide range of bacteria. Various BREX systems encode unique combinations of proteins that usually include a site-specific methyltransferase; none appear to contain a nuclease. Here we describe the identification and characterization of a Type I BREX system from Acinetobacter and the effect of deleting each BREX ORF on growth, methylation, and restriction. We identified a previously uncharacterized gene in the BREX operon that is dispensable for methylation but involved in restriction. Biochemical and crystallographic analyses of this factor, which we term BrxR ('BREX Regulator'), demonstrate that it forms a homodimer and specifically binds a DNA target site upstream of its transcription start site. Deletion of the BrxR gene causes cell toxicity, reduces restriction, and significantly increases the expression of BrxC. In contrast, the introduction of a premature stop codon into the BrxR gene, or a point mutation blocking its DNA binding ability, has little effect on restriction, implying that the BrxR coding sequence and BrxR protein play independent functional roles. We speculate that elements within the BrxR coding sequence are involved in cis regulation of anti-phage activity, while the BrxR protein itself plays an additional regulatory role, perhaps during horizontal transfer.


Assuntos
Acinetobacter/fisiologia , Fatores de Restrição Antivirais , Bacteriófagos , Acinetobacter/genética , Acinetobacter/virologia , Fatores de Restrição Antivirais/genética , Bacteriófagos/fisiologia , DNA/metabolismo , Metiltransferases/genética , Óperon
8.
Nature ; 528(7583): 585-8, 2015 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-26675735

RESUMO

Tandem repeat proteins, which are formed by repetition of modular units of protein sequence and structure, play important biological roles as macromolecular binding and scaffolding domains, enzymes, and building blocks for the assembly of fibrous materials. The modular nature of repeat proteins enables the rapid construction and diversification of extended binding surfaces by duplication and recombination of simple building blocks. The overall architecture of tandem repeat protein structures--which is dictated by the internal geometry and local packing of the repeat building blocks--is highly diverse, ranging from extended, super-helical folds that bind peptide, DNA, and RNA partners, to closed and compact conformations with internal cavities suitable for small molecule binding and catalysis. Here we report the development and validation of computational methods for de novo design of tandem repeat protein architectures driven purely by geometric criteria defining the inter-repeat geometry, without reference to the sequences and structures of existing repeat protein families. We have applied these methods to design a series of closed α-solenoid repeat structures (α-toroids) in which the inter-repeat packing geometry is constrained so as to juxtapose the amino (N) and carboxy (C) termini; several of these designed structures have been validated by X-ray crystallography. Unlike previous approaches to tandem repeat protein engineering, our design procedure does not rely on template sequence or structural information taken from natural repeat proteins and hence can produce structures unlike those seen in nature. As an example, we have successfully designed and validated closed α-solenoid repeats with a left-handed helical architecture that--to our knowledge--is not yet present in the protein structure database.


Assuntos
Motivos de Aminoácidos , Bioengenharia , Simulação por Computador , Estrutura Secundária de Proteína , Proteínas/química , Cristalografia por Raios X , Bases de Dados de Proteínas , Modelos Moleculares , Reprodutibilidade dos Testes
9.
Nature ; 518(7537): 55-60, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25607366

RESUMO

Genetically modified organisms (GMOs) are increasingly deployed at large scales and in open environments. Genetic biocontainment strategies are needed to prevent unintended proliferation of GMOs in natural ecosystems. Existing biocontainment methods are insufficient because they impose evolutionary pressure on the organism to eject the safeguard by spontaneous mutagenesis or horizontal gene transfer, or because they can be circumvented by environmentally available compounds. Here we computationally redesign essential enzymes in the first organism possessing an altered genetic code (Escherichia coli strain C321.ΔA) to confer metabolic dependence on non-standard amino acids for survival. The resulting GMOs cannot metabolically bypass their biocontainment mechanisms using known environmental compounds, and they exhibit unprecedented resistance to evolutionary escape through mutagenesis and horizontal gene transfer. This work provides a foundation for safer GMOs that are isolated from natural ecosystems by a reliance on synthetic metabolites.


Assuntos
Aminoácidos/química , Aminoácidos/metabolismo , Contenção de Riscos Biológicos/métodos , Proteínas de Escherichia coli/biossíntese , Escherichia coli/genética , Escherichia coli/metabolismo , Organismos Geneticamente Modificados/genética , Biologia Sintética/métodos , Evolução Biológica , Códon/genética , Ecossistema , Escherichia coli/enzimologia , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Transferência Genética Horizontal/genética , Genes Essenciais/genética , Código Genético/genética , Engenharia Genética/métodos , Viabilidade Microbiana/genética , Mutação/genética , Organismos Geneticamente Modificados/metabolismo , Segurança , Seleção Genética
12.
Nucleic Acids Res ; 47(1): 450-467, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30395313

RESUMO

BbvCI, a Type IIT restriction endonuclease, recognizes and cleaves the seven base pair sequence 5'-CCTCAGC-3', generating 3-base, 5'-overhangs. BbvCI is composed of two protein subunits, each containing one catalytic site. Either site can be inactivated by mutation resulting in enzyme variants that nick DNA in a strand-specific manner. Here we demonstrate that the holoenzyme is labile, with the R1 subunit dissociating at low pH. Crystallization of the R2 subunit under such conditions revealed an elongated dimer with the two catalytic sites located on opposite sides. Subsequent crystallization at physiological pH revealed a tetramer comprising two copies of each subunit, with a pair of deep clefts each containing two catalytic sites appropriately positioned and oriented for DNA cleavage. This domain organization was further validated with single-chain protein constructs in which the two enzyme subunits were tethered via peptide linkers of variable length. We were unable to crystallize a DNA-bound complex; however, structural similarity to previously crystallized restriction endonucleases facilitated creation of an energy-minimized model bound to DNA, and identification of candidate residues responsible for target recognition. Mutation of residues predicted to recognize the central C:G base pair resulted in an altered enzyme that recognizes and cleaves CCTNAGC (N = any base).


Assuntos
Clivagem do DNA , Enzimas de Restrição do DNA/química , Holoenzimas/química , Subunidades Proteicas/química , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Domínio Catalítico , Enzimas de Restrição do DNA/genética , Enzimas de Restrição do DNA/isolamento & purificação , Escherichia coli/enzimologia , Holoenzimas/genética , Holoenzimas/isolamento & purificação , Mutação , Peptídeos/química , Multimerização Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/isolamento & purificação
13.
Nucleic Acids Res ; 46(10): 4845-4871, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29718463

RESUMO

Protein engineering is used to generate novel protein folds and assemblages, to impart new properties and functions onto existing proteins, and to enhance our understanding of principles that govern protein structure. While such approaches can be employed to reprogram protein-protein interactions, modifying protein-DNA interactions is more difficult. This may be related to the structural features of protein-DNA interfaces, which display more charged groups, directional hydrogen bonds, ordered solvent molecules and counterions than comparable protein interfaces. Nevertheless, progress has been made in the redesign of protein-DNA specificity, much of it driven by the development of engineered enzymes for genome modification. Here, we summarize the creation of novel DNA specificities for zinc finger proteins, meganucleases, TAL effectors, recombinases and restriction endonucleases. The ease of re-engineering each system is related both to the modularity of the protein and the extent to which the proteins have evolved to be capable of readily modifying their recognition specificities in response to natural selection. The development of engineered DNA binding proteins that display an ideal combination of activity, specificity, deliverability, and outcomes is not a fully solved problem, however each of the current platforms offers unique advantages, offset by behaviors and properties requiring further study and development.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Engenharia de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Pareamento de Bases , DNA/química , Clivagem do DNA , Enzimas de Restrição do DNA/química , Enzimas de Restrição do DNA/genética , Enzimas de Restrição do DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Desoxirribonucleases/química , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Edição de Genes , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Recombinases/química , Recombinases/genética , Recombinases/metabolismo , Efetores Semelhantes a Ativadores de Transcrição/química , Efetores Semelhantes a Ativadores de Transcrição/genética , Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Dedos de Zinco
14.
Nature ; 501(7466): 212-216, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-24005320

RESUMO

The ability to design proteins with high affinity and selectivity for any given small molecule is a rigorous test of our understanding of the physiochemical principles that govern molecular recognition. Attempts to rationally design ligand-binding proteins have met with little success, however, and the computational design of protein-small-molecule interfaces remains an unsolved problem. Current approaches for designing ligand-binding proteins for medical and biotechnological uses rely on raising antibodies against a target antigen in immunized animals and/or performing laboratory-directed evolution of proteins with an existing low affinity for the desired ligand, neither of which allows complete control over the interactions involved in binding. Here we describe a general computational method for designing pre-organized and shape complementary small-molecule-binding sites, and use it to generate protein binders to the steroid digoxigenin (DIG). Of seventeen experimentally characterized designs, two bind DIG; the model of the higher affinity binder has the most energetically favourable and pre-organized interface in the design set. A comprehensive binding-fitness landscape of this design, generated by library selections and deep sequencing, was used to optimize its binding affinity to a picomolar level, and X-ray co-crystal structures of two variants show atomic-level agreement with the corresponding computational models. The optimized binder is selective for DIG over the related steroids digitoxigenin, progesterone and ß-oestradiol, and this steroid binding preference can be reprogrammed by manipulation of explicitly designed hydrogen-bonding interactions. The computational design method presented here should enable the development of a new generation of biosensors, therapeutics and diagnostics.


Assuntos
Simulação por Computador , Digoxigenina/metabolismo , Desenho de Fármacos , Proteínas/química , Proteínas/metabolismo , Sítios de Ligação , Biotecnologia , Cristalografia por Raios X , Digoxigenina/química , Estradiol/química , Estradiol/metabolismo , Ligantes , Modelos Moleculares , Progesterona/química , Progesterona/metabolismo , Ligação Proteica , Reprodutibilidade dos Testes , Especificidade por Substrato
15.
Nucleic Acids Res ; 45(3): 1516-1528, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28180307

RESUMO

R.SwaI, a Type IIP restriction endonuclease, recognizes a palindromic eight base pair (bp) symmetric sequence, 5΄-ATTTAAAT-3΄, and cleaves that target at its center to generate blunt-ended DNA fragments. Here, we report three crystal structures of SwaI: unbound enzyme, a DNA-bound complex with calcium ions; and a DNA-bound, fully cleaved complex with magnesium ions. We compare these structures to two structurally similar 'PD-D/ExK' restriction endonucleases (EcoRV and HincII) that also generate blunt-ended products, and to a structurally distinct enzyme (the HNH endonuclease PacI) that also recognizes an 8-bp target site consisting solely of A:T base pairs. Binding by SwaI induces an extreme bend in the target sequence accompanied by un-pairing and re-ordering of its central A:T base pairs. This result is reminiscent of a more dramatic target deformation previously described for PacI, implying that long A:T-rich target sites might display structural or dynamic behaviors that play a significant role in endonuclease recognition and cleavage.


Assuntos
DNA/química , DNA/metabolismo , Desoxirribonucleases de Sítio Específico do Tipo II/química , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Sequência Rica em At , Sequência de Aminoácidos , Pareamento de Bases , Sítios de Ligação , Cristalografia por Raios X , DNA/genética , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Conformação Proteica , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Homologia Estrutural de Proteína , Especificidade por Substrato
16.
Nucleic Acids Res ; 45(11): 6960-6970, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28460076

RESUMO

Transcription activator-like effectors (TALEs) recognize their DNA targets via tandem repeats, each specifying a single nucleotide base in a one-to-one sequential arrangement. Due to this modularity and their ability to bind long DNA sequences with high specificity, TALEs have been used in many applications. Contributions of individual repeat-nucleotide associations to affinity and specificity have been characterized. Here, using in vitro binding assays, we examined the relationship between the number of repeats in a TALE and its affinity, for both target and non-target DNA. Each additional repeat provides extra binding energy for the target DNA, with the gain decaying exponentially such that binding energy saturates. Affinity for non-target DNA also increases non-linearly with the number of repeats, but with a slower decay of gain. The difference between the effect of length on affinity for target versus non-target DNA manifests in specificity increasing then diminishing with increasing TALE length, peaking between 15 and 19 repeats. Modeling across different hypothetical saturation levels and rates of gain decay, reflecting different repeat compositions, yielded a similar range of specificity optima. This range encompasses the mean and median length of native TALEs, suggesting that these proteins as a group have evolved for maximum specificity.


Assuntos
Proteínas de Bactérias/química , Efetores Semelhantes a Ativadores de Transcrição/química , Proteínas de Bactérias/fisiologia , Sequência de Bases , Sítios de Ligação , DNA Bacteriano/química , Ligação Proteica , Sequências de Repetição em Tandem , Termodinâmica , Efetores Semelhantes a Ativadores de Transcrição/fisiologia , Xanthomonas
17.
Nucleic Acids Res ; 45(14): 8621-8634, 2017 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-28637173

RESUMO

The retargeting of protein-DNA specificity, outside of extremely modular DNA binding proteins such as TAL effectors, has generally proved to be quite challenging. Here, we describe structural analyses of five different extensively retargeted variants of a single homing endonuclease, that have been shown to function efficiently in ex vivo and in vivo applications. The redesigned proteins harbor mutations at up to 53 residues (18%) of their amino acid sequence, primarily distributed across the DNA binding surface, making them among the most significantly reengineered ligand-binding proteins to date. Specificity is derived from the combined contributions of DNA-contacting residues and of neighboring residues that influence local structural organization. Changes in specificity are facilitated by the ability of all those residues to readily exchange both form and function. The fidelity of recognition is not precisely correlated with the fraction or total number of residues in the protein-DNA interface that are actually involved in DNA contacts, including directional hydrogen bonds. The plasticity of the DNA-recognition surface of this protein, which allows substantial retargeting of recognition specificity without requiring significant alteration of the surrounding protein architecture, reflects the ability of the corresponding genetic elements to maintain mobility and persistence in the face of genetic drift within potential host target sites.


Assuntos
DNA/química , DNA/metabolismo , Endodesoxirribonucleases/química , Endodesoxirribonucleases/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sequência de Bases , Sítios de Ligação/genética , Cristalografia , Culicidae/enzimologia , Culicidae/genética , DNA/genética , Endodesoxirribonucleases/genética , Modelos Moleculares , Mutação , Conformação de Ácido Nucleico , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Especificidade por Substrato
18.
Proc Natl Acad Sci U S A ; 112(12): 3704-9, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25775555

RESUMO

We describe a computationally designed enzyme, formolase (FLS), which catalyzes the carboligation of three one-carbon formaldehyde molecules into one three-carbon dihydroxyacetone molecule. The existence of FLS enables the design of a new carbon fixation pathway, the formolase pathway, consisting of a small number of thermodynamically favorable chemical transformations that convert formate into a three-carbon sugar in central metabolism. The formolase pathway is predicted to use carbon more efficiently and with less backward flux than any naturally occurring one-carbon assimilation pathway. When supplemented with enzymes carrying out the other steps in the pathway, FLS converts formate into dihydroxyacetone phosphate and other central metabolites in vitro. These results demonstrate how modern protein engineering and design tools can facilitate the construction of a completely new biosynthetic pathway.


Assuntos
Carbono/química , Engenharia de Proteínas/métodos , Proteínas/química , Biomassa , Vias Biossintéticas , Ciclo do Carbono , Catálise , Clonagem Molecular , Escherichia coli/enzimologia , Formaldeído/química , Formiatos/química , Espectroscopia de Ressonância Magnética , Reação em Cadeia da Polimerase , Software , Termodinâmica
19.
Proc Natl Acad Sci U S A ; 111(11): 4061-6, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24591643

RESUMO

LAGLIDADG homing endonucleases (meganucleases) are sequence-specific DNA cleavage enzymes used for genome engineering. Recently, meganucleases fused to transcription activator-like effectors have been demonstrated to efficiently introduce targeted genome modifications. However, retargeting meganucleases to genomic sequences of interest remains challenging because it usually requires extensive alteration of a large number of amino acid residues that are situated in and near the DNA interface. Here we describe an effective strategy to extensively redesign such an extensive biomolecular interface. Well-characterized meganucleases are computationally screened to identify the best candidate enzyme to target a genomic region; that protein is then redesigned using iterative rounds of in vitro selections within compartmentalized aqueous droplets, which enable screening of extremely large numbers of protein variants at each step. The utility of this approach is illustrated by engineering three different meganucleases to cleave three human genomic sites (found in two exons and one flanking intron in two clinically relevant genes) and a fourth endonuclease that discriminates between single-nucleotide polymorphism variants of one of those targets. Fusion with transcription activator-like effector DNA binding domains significantly enhances targeted modification induced by meganucleases engineered in this study. Simultaneous expression of two such fusion endonucleases results in efficient excision of a defined genomic region.


Assuntos
DNA/química , Endodesoxirribonucleases/química , Endodesoxirribonucleases/metabolismo , Engenharia de Proteínas/métodos , Sequência de Bases , Western Blotting , Clonagem Molecular , DNA/metabolismo , Clivagem do DNA , Terapia Genética/métodos , Ensaios de Triagem em Larga Escala , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Análise de Sequência de DNA , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa