Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 5218, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890276

RESUMO

Technologies that generate precise combinatorial genome modifications are well suited to dissect the polygenic basis of complex phenotypes and engineer synthetic genomes. Genome modifications with engineered nucleases can lead to undesirable repair outcomes through imprecise homology-directed repair, requiring non-cleavable gene editing strategies. Eukaryotic multiplex genome engineering (eMAGE) generates precise combinatorial genome modifications in Saccharomyces cerevisiae without generating DNA breaks or using engineered nucleases. Here, we systematically optimize eMAGE to achieve 90% editing frequency, reduce workflow time, and extend editing distance to 20 kb. We further engineer an inducible dominant negative mismatch repair system, allowing for high-efficiency editing via eMAGE while suppressing the elevated background mutation rate 17-fold resulting from mismatch repair inactivation. We apply these advances to construct a library of cancer-associated mutations in the ligand-binding domains of human estrogen receptor alpha and progesterone receptor to understand their impact on ligand-independent autoactivation. We validate that this yeast model captures autoactivation mutations characterized in human breast cancer models and further leads to the discovery of several previously uncharacterized autoactivating mutations. This work demonstrates the development and optimization of a cleavage-free method of genome editing well suited for applications requiring efficient multiplex editing with minimal background mutations.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Mutação , Saccharomyces cerevisiae , Edição de Genes/métodos , Saccharomyces cerevisiae/genética , Humanos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Receptores de Progesterona/genética , Reparo de Erro de Pareamento de DNA/genética , Neoplasias da Mama/genética , Feminino
2.
Nucleic Acids Res ; 51(22): 12522-12536, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-37941137

RESUMO

The widespread Pseudomonas genus comprises a collection of related species with remarkable abilities to degrade plastics and polluted wastes and to produce a broad set of valuable compounds, ranging from bulk chemicals to pharmaceuticals. Pseudomonas possess characteristics of tolerance and stress resistance making them valuable hosts for industrial and environmental biotechnology. However, efficient and high-throughput genetic engineering tools have limited metabolic engineering efforts and applications. To improve their genome editing capabilities, we first employed a computational biology workflow to generate a genus-specific library of potential single-stranded DNA-annealing proteins (SSAPs). Assessment of the library was performed in different Pseudomonas using a high-throughput pooled recombinase screen followed by Oxford Nanopore NGS analysis. Among different active variants with variable levels of allelic replacement frequency (ARF), efficient SSAPs were found and characterized for mediating recombineering in the four tested species. New variants yielded higher ARFs than existing ones in Pseudomonas putida and Pseudomonas aeruginosa, and expanded the field of recombineering in Pseudomonas taiwanensisand Pseudomonas fluorescens. These findings will enhance the mutagenesis capabilities of these members of the Pseudomonas genus, increasing the possibilities for biotransformation and enhancing their potential for synthetic biology applications. .


Assuntos
Edição de Genes , Pseudomonas , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Edição de Genes/métodos , Engenharia Metabólica , Pseudomonas/genética , Pseudomonas putida/genética
3.
Sci Adv ; 9(17): eade8934, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37115918

RESUMO

Fitness landscapes are models of the sequence space of a genetic element that map how each sequence corresponds to its activity and can be used to guide laboratory evolution. The ribosome is a macromolecular machine that is essential for protein synthesis in all organisms. Because of the prevalence of dominant lethal mutations, a comprehensive fitness landscape of the ribosomal peptidyl transfer center (PTC) has not yet been attained. Here, we develop a method to functionally map an orthogonal tethered ribosome (oRiboT), which permits complete mutagenesis of nucleotides located in the PTC and the resulting epistatic interactions. We found that most nucleotides studied showed flexibility to mutation, and identified epistatic interactions between them, which compensate for deleterious mutations. This work provides a basis for a deeper understanding of ribosome function and malleability and could be used to inform design of engineered ribosomes with applications to synthesize next-generation biomaterials and therapeutics.


Assuntos
Biossíntese de Proteínas , Ribossomos , Ribossomos/genética , Ribossomos/metabolismo , Mutação , Nucleotídeos/metabolismo
4.
iScience ; 25(7): 104562, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35789833

RESUMO

Introducing heterologous pathways into host cells constitutes a promising strategy for synthesizing nonstandard amino acids (nsAAs) to enable the production of proteins with expanded chemistries. However, this strategy has proven challenging, as the expression of heterologous pathways can disrupt cellular homeostasis of the host cell. Here, we sought to optimize the heterologous production of the nsAA para-aminophenylalanine (pAF) in Escherichia coli. First, we incorporated a heterologous pAF biosynthesis pathway into a genome-scale model of E. coli metabolism and computationally identified metabolic interventions in the host's native metabolism to improve pAF production. Next, we explored different approaches of imposing these flux interventions experimentally and found that the upregulation of flux in the chorismate biosynthesis pathway through the elimination of feedback inhibition mechanisms could significantly raise pAF titers (∼20-fold) while maintaining a reasonable pAF production-growth rate trade-off. Overall, this study provides a promising strategy for the biosynthesis of nsAAs in engineered cells.

5.
Cell ; 185(9): 1487-1505.e14, 2022 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-35366417

RESUMO

Small molecules encoded by biosynthetic pathways mediate cross-species interactions and harbor untapped potential, which has provided valuable compounds for medicine and biotechnology. Since studying biosynthetic gene clusters in their native context is often difficult, alternative efforts rely on heterologous expression, which is limited by host-specific metabolic capacity and regulation. Here, we describe a computational-experimental technology to redesign genes and their regulatory regions with hybrid elements for cross-species expression in Gram-negative and -positive bacteria and eukaryotes, decoupling biosynthetic capacity from host-range constraints to activate silenced pathways. These synthetic genetic elements enabled the discovery of a class of microbiome-derived nucleotide metabolites-tyrocitabines-from Lactobacillus iners. Tyrocitabines feature a remarkable orthoester-phosphate, inhibit translational activity, and invoke unexpected biosynthetic machinery, including a class of "Amadori synthases" and "abortive" tRNA synthetases. Our approach establishes a general strategy for the redesign, expression, mobilization, and characterization of genetic elements in diverse organisms and communities.


Assuntos
Vias Biossintéticas , Interações entre Hospedeiro e Microrganismos , Microbiota , Biologia Sintética/métodos , Bactérias/classificação , Bactérias/genética , Bactérias/metabolismo , Eucariotos/genética , Eucariotos/metabolismo , Engenharia Genética , Humanos , Metabolômica
6.
ACS Synth Biol ; 11(2): 522-527, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35176864

RESUMO

The ability to construct, synthesize, and edit genes and genomes at scale and with speed enables, in synergy with other tools of engineering biology, breakthrough applications with far-reaching implications for society. As SARS-CoV-2 spread around the world in early spring of 2020, researchers rapidly mobilized, using these tools in the development of diagnostics, therapeutics, and vaccines for COVID-19. The sharing of knowledge was crucial to making rapid progress. Several publications described the use of reverse genetics for the de novo construction of SARS-CoV-2 in the laboratory, one in the form of a protocol. Given the demonstrable harm caused by the virus, the unequal distribution of mitigating vaccines and therapeutics, their unknown efficacy against variants, and the interest in this research by laboratories unaccustomed to working with highly transmissible pandemic pathogens, there are risks associated with such publications, particularly as protocols. We describe considerations and offer suggestions for enhancing security in the publication of synthetic biology research and techniques. We recommend: (1) that protocol manuscripts for the de novo synthesis of certain pathogenic viruses undergo a mandatory safety and security review; (2) that if published, such papers include descriptions of the discussions or review processes that occurred regarding security considerations in the main text; and (3) the development of a governance framework for the inclusion of basic security screening during the publication process of engineering biology/synthetic biology manuscripts to build and support a safe and secure research enterprise that is able to maximize its positive impacts and minimize any negative outcomes.


Assuntos
Bioengenharia , Editoração , Medidas de Segurança/organização & administração , Genes Virais , SARS-CoV-2/genética , Biologia Sintética
7.
Nat Commun ; 13(1): 829, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149672

RESUMO

Advances in synthetic biology permit the genetic encoding of synthetic chemistries at monomeric precision, enabling the synthesis of programmable proteins with tunable properties. Bacterial pili serve as an attractive biomaterial for the development of engineered protein materials due to their ability to self-assemble into mechanically robust filaments. However, most biomaterials lack electronic functionality and atomic structures of putative conductive proteins are not known. Here, we engineer high electronic conductivity in pili produced by a genomically-recoded E. coli strain. Incorporation of tryptophan into pili increased conductivity of individual filaments >80-fold. Computationally-guided ordering of the pili into nanostructures increased conductivity 5-fold compared to unordered pili networks. Site-specific conjugation of pili with gold nanoparticles, facilitated by incorporating the nonstandard amino acid propargyloxy-phenylalanine, increased filament conductivity ~170-fold. This work demonstrates the sequence-defined production of highly-conductive protein nanowires and hybrid organic-inorganic biomaterials with genetically-programmable electronic functionalities not accessible in nature or through chemical-based synthesis.


Assuntos
Condutividade Elétrica , Nanopartículas Metálicas/química , Nanofios , Proteínas/metabolismo , Fenômenos Químicos , Escherichia coli/genética , Proteínas de Fímbrias , Fímbrias Bacterianas/metabolismo , Ouro/química , Nanoestruturas , Nanofios/química , Fenilalanina/metabolismo , Engenharia de Proteínas , Triptofano/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35046019

RESUMO

The use of biologics in the treatment of numerous diseases has increased steadily over the past decade due to their high specificities, low toxicity, and limited side effects. Despite this success, peptide- and protein-based drugs are limited by short half-lives and immunogenicity. To address these challenges, we use a genomically recoded organism to produce genetically encoded elastin-like polypeptide-protein fusions containing multiple instances of para-azidophenylalanine (pAzF). Precise lipidation of these pAzF residues generated a set of sequence-defined synthetic biopolymers with programmable binding affinity to albumin without ablating the activity of model fusion proteins, and with tunable blood serum half-lives spanning 5 to 94% of albumin's half-life in a mouse model. Our findings present a proof of concept for the use of genetically encoded bioorthogonal conjugation sites for multisite lipidation to tune protein stability in mouse serum. This work establishes a programmable approach to extend and tune the half-life of protein or peptide therapeutics and a technical foundation to produce functionalized biopolymers endowed with programmable chemical and biophysical properties with broad applications in medicine, materials science, and biotechnology.


Assuntos
Biopolímeros/química , Lipídeos/química , Peptídeos/química , Proteínas/química , Aminoácidos , Animais , Meia-Vida , Camundongos , Engenharia de Proteínas/métodos , Biologia Sintética/métodos
9.
Nat Commun ; 13(1): 180, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013328

RESUMO

Genome editing technologies introduce targeted chromosomal modifications in organisms yet are constrained by the inability to selectively modify repetitive genetic elements. Here we describe filtered editing, a genome editing method that embeds group 1 self-splicing introns into repetitive genetic elements to construct unique genetic addresses that can be selectively modified. We introduce intron-containing ribosomes into the E. coli genome and perform targeted modifications of these ribosomes using CRISPR/Cas9 and multiplex automated genome engineering. Self-splicing of introns post-transcription yields scarless RNA molecules, generating a complex library of targeted combinatorial variants. We use filtered editing to co-evolve the 16S rRNA to tune the ribosome's translational efficiency and the 23S rRNA to isolate antibiotic-resistant ribosome variants without interfering with native translation. This work sets the stage to engineer mutant ribosomes that polymerize abiological monomers with diverse chemistries and expands the scope of genome engineering for precise editing and evolution of repetitive DNA sequences.


Assuntos
Escherichia coli/genética , Edição de Genes/métodos , Genoma Bacteriano , Mutagênese Sítio-Dirigida/métodos , Splicing de RNA , Ribossomos/genética , Antibacterianos/farmacologia , Sistemas CRISPR-Cas , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Éxons , Engenharia Genética , Íntrons , Polímeros/química , Biossíntese de Proteínas , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , RNA Ribossômico 23S/genética , RNA Ribossômico 23S/metabolismo , Sequências Repetitivas de Ácido Nucleico , Ribossomos/metabolismo
10.
Cell Chem Biol ; 29(6): 1046-1052.e4, 2022 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34965380

RESUMO

The site-specific incorporation of nonstandard amino acids (nsAAs) during translation has expanded the chemistry and function of proteins. The nsAA para-azido-phenylalanine (pAzF) encodes a biorthogonal chemical moiety that facilitates "click" reactions to attach diverse chemical groups for protein functionalization. However, the azide moiety is unstable in physiological conditions and is reduced to para-amino-phenylalanine (pAF). Azide reduction decreases the yield of pAzF residues in proteins to 50%-60% per azide and limits protein functionalization by click reactions. Here, we describe the use of a pH-tunable diazotransfer reaction that converts pAF to pAzF at >95% efficiency in proteins. The method selectively restores pAzF at multiple sites per protein without introducing off-target modifications. This work addresses a key limitation in the production of pAzF-containing proteins by restoring azides for multi-site functionalization with diverse chemical moieties, setting the stage for the production of genetically encoded biomaterials with broad applications in biotherapeutics, materials science, and biotechnology.


Assuntos
Azidas , Fenilalanina , Aminoácidos , Azidas/química , Materiais Biocompatíveis , Química Click/métodos , Fenilalanina/química , Proteínas/química
11.
Cell Rep ; 36(3): 109416, 2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34289367

RESUMO

Advances in genetic code expansion have enabled the production of proteins containing site-specific, authentic post-translational modifications. Here, we use a recoded bacterial strain with an expanded genetic code to encode phosphoserine into a human kinase protein. We directly encode phosphoserine into WNK1 (with-no-lysine [K] 1) or WNK4 kinases at multiple, distinct sites, which produced activated, phosphorylated WNK that phosphorylated and activated SPAK/OSR kinases, thereby synthetically activating this human kinase network in recoded bacteria. We used this approach to identify biochemical properties of WNK kinases, a motif for SPAK substrates, and small-molecule kinase inhibitors for phosphorylated SPAK. We show that the kinase inhibitors modulate SPAK substrates in cells, alter cell volume, and reduce migration of glioblastoma cells. Our work establishes a protein-engineering platform technology that demonstrates that synthetically active WNK kinase networks can accurately model cellular systems and can be used more broadly to target networks of phosphorylated proteins for research and discovery.


Assuntos
Escherichia coli/metabolismo , Transdução de Sinais , Proteína Quinase 1 Deficiente de Lisina WNK/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Glioblastoma/patologia , Células HEK293 , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Especificidade por Substrato
12.
ACS Synth Biol ; 10(5): 907-910, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-33977723

RESUMO

Engineering biology is being applied toward solving or mitigating some of the greatest challenges facing society. As with many other rapidly advancing technologies, the development of these powerful tools must be considered in the context of ethical uses for personal, societal, and/or environmental advancement. Researchers have a responsibility to consider the diverse outcomes that may result from the knowledge and innovation they contribute to the field. Together, we developed a Statement of Ethics in Engineering Biology Research to guide researchers as they incorporate the consideration of long-term ethical implications of their work into every phase of the research lifecycle. Herein, we present and contextualize this Statement of Ethics and its six guiding principles. Our goal is to facilitate ongoing reflection and collaboration among technical researchers, social scientists, policy makers, and other stakeholders to support best outcomes in engineering biology innovation and development.


Assuntos
Bioengenharia/ética , Pesquisa Biomédica/ética , Invenções/ética , Pessoal Administrativo/ética , Comunicação , Saúde Ambiental , Humanos , Pessoal de Laboratório Médico/ética , Saúde Pública , Projetos de Pesquisa , Pesquisadores/ética , Responsabilidade Social
13.
Nat Chem Biol ; 17(6): 724-731, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33820990

RESUMO

Genetically modified microorganisms (GMMs) can enable a wide range of important applications including environmental sensing and responsive engineered living materials. However, containment of GMMs to prevent environmental escape and satisfy regulatory requirements is a bottleneck for real-world use. While current biochemical strategies restrict unwanted growth of GMMs in the environment, there is a need for deployable physical containment technologies to achieve redundant, multi-layered and robust containment. We developed a hydrogel-based encapsulation system that incorporates a biocompatible multilayer tough shell and an alginate-based core. This deployable physical containment strategy (DEPCOS) allows no detectable GMM escape, bacteria to be protected against environmental insults including antibiotics and low pH, controllable lifespan and easy retrieval of genomically recoded bacteria. To highlight the versatility of DEPCOS, we demonstrated that robustly encapsulated cells can execute useful functions, including performing cell-cell communication with other encapsulated bacteria and sensing heavy metals in water samples from the Charles River.


Assuntos
Bactérias/efeitos dos fármacos , Hidrogéis/farmacologia , Alginatos/química , Antibacterianos/farmacologia , Bactérias/genética , Materiais Biocompatíveis , Bioengenharia , DNA Bacteriano/química , DNA Bacteriano/genética , Monitoramento Ambiental , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Heme/química , Metais Pesados/química , Organismos Geneticamente Modificados , Percepção de Quorum , Rios , Poluentes da Água/química
14.
Science ; 372(6541): 460-461, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33926938
15.
Artigo em Inglês | MEDLINE | ID: mdl-35540496

RESUMO

Recombination-mediated genetic engineering, also known as recombineering, is the genomic incorporation of homologous single-stranded or double-stranded DNA into bacterial genomes. Recombineering and its derivative methods have radically improved genome engineering capabilities, perhaps none more so than multiplex automated genome engineering (MAGE). MAGE is representative of a set of highly multiplexed single-stranded DNA-mediated technologies. First described in Escherichia coli, both MAGE and recombineering are being rapidly translated into diverse prokaryotes and even into eukaryotic cells. Together, this modern set of tools offers the promise of radically improving the scope and throughput of experimental biology by providing powerful new methods to ease the genetic manipulation of model and non-model organisms. In this Primer, we describe recombineering and MAGE, their optimal use, their diverse applications and methods for pairing them with other genetic editing tools. We then look forward to the future of genetic engineering.

16.
Nano Lett ; 20(12): 8890-8896, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33164530

RESUMO

Fluorescence microscopy has been one of the most discovery-rich methods in biology. In the digital age, the discipline is becoming increasingly quantitative. Virtually all biological laboratories have access to fluorescence microscopes, but abilities to quantify biomolecule copy numbers are limited by the complexity and sophistication associated with current quantification methods. Here, we present DNA-origami-based fluorescence brightness standards for counting 5-300 copies of proteins in bacterial and mammalian cells, tagged with fluorescent proteins or membrane-permeable organic dyes. Compared to conventional quantification techniques, our brightness standards are robust, straightforward to use, and compatible with nearly all fluorescence imaging applications, thereby providing a practical and versatile tool to quantify biomolecules via fluorescence microscopy.


Assuntos
DNA , Corantes Fluorescentes , Animais , Microscopia de Fluorescência , Proteínas
17.
Life (Basel) ; 9(3)2019 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-31284384

RESUMO

The genetic code defines how information in the genome is translated into protein. Aside from a handful of isolated exceptions, this code is universal. Researchers have developed techniques to artificially expand the genetic code, repurposing codons and translational machinery to incorporate nonstandard amino acids (nsAAs) into proteins. A key challenge for robust genetic code expansion is orthogonality; the engineered machinery used to introduce nsAAs into proteins must co-exist with native translation and gene expression without cross-reactivity or pleiotropy. The issue of orthogonality manifests at several levels, including those of codons, ribosomes, aminoacyl-tRNA synthetases, tRNAs, and elongation factors. In this concept paper, we describe advances in genome recoding, translational engineering and associated challenges rooted in establishing orthogonality needed to expand the genetic code.

18.
Nano Lett ; 19(1): 247-254, 2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30540482

RESUMO

Polypeptides are promising carriers for chemotherapeutics: they have minimal toxicity, can be recombinantly synthesized with precise control over molecular weight, and enhance drug pharmacokinetics as self-assembled nanoparticles. Polypeptide-based systems also provide the ability to achieve active targeting with genetically encoded targeting ligands. While passive targeting promotes accumulation of nanocarriers in solid tumors, active targeting provides an additional layer of tunable control and widens the therapeutic window. However, fusion of most targeting proteins to polypeptide carriers exposes the limitations of this approach: the residues that are used for drug attachment are also promiscuously distributed on protein surfaces. We present here a universal methodology to solve this problem by the site-specific attachment of extrinsic moieties to polypeptide drug delivery systems without cross-reactivity to fused targeting domains. We incorporate an unnatural amino acid, p-acetylphenylalanine, to provide a biorthogonal ketone for attachment of doxorubicin in the presence of reactive amino acids in a nanobody-targeted, elastin-like polypeptide nanoparticle. These nanoparticles exhibit significantly greater cytotoxicity than nontargeted controls in multiple cancer cell lines.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Peptídeos/química , Animais , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacologia , Elastina/química , Elastina/farmacologia , Humanos , Ligantes , Micelas , Nanopartículas/administração & dosagem , Peptídeos/farmacologia , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/farmacologia
19.
Elife ; 72018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30375330

RESUMO

Organisms possessing genetic codes with unassigned codons raise the question of how cellular machinery resolves such codons and how this could impact horizontal gene transfer. Here, we use a genomically recoded Escherichia coli to examine how organisms address translation at unassigned UAG codons, which obstruct propagation of UAG-containing viruses and plasmids. Using mass spectrometry, we show that recoded organisms resolve translation at unassigned UAG codons via near-cognate suppression, dramatic frameshifting from at least -3 to +19 nucleotides, and rescue by ssrA-encoded tmRNA, ArfA, and ArfB. We then demonstrate that deleting tmRNA restores expression of UAG-ending proteins and propagation of UAG-containing viruses and plasmids in the recoded strain, indicating that tmRNA rescue and nascent peptide degradation is the cause of impaired virus and plasmid propagation. The ubiquity of tmRNA homologs suggests that genomic recoding is a promising path for impairing horizontal gene transfer and conferring genetic isolation in diverse organisms.


Assuntos
Códon de Terminação/genética , Proteínas de Escherichia coli/genética , Transferência Genética Horizontal/genética , Código Genético/genética , Proteínas de Ligação a RNA/genética , Escherichia coli/genética , Mudança da Fase de Leitura do Gene Ribossômico/genética , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano/genética , Plasmídeos/genética , RNA Bacteriano/genética , Vírus/genética
20.
Curr Opin Chem Biol ; 46: 203-211, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30072242

RESUMO

Engineering of the translation apparatus has permitted the site-specific incorporation of nonstandard amino acids (nsAAs) into proteins, thereby expanding the genetic code of organisms. Conventional approaches have focused on porting tRNAs and aminoacyl-tRNA synthetases (aaRS) from archaea into bacterial and eukaryotic systems where they have been engineered to site-specifically encode nsAAs. More recent work in genome engineering has opened up the possibilities of whole genome recoding, in which organisms with alternative genetic codes have been constructed whereby codons removed from the genetic code can be repurposed as new sense codons dedicated for incorporation of nsAAs. These advances, together with the advent of engineered ribosomes and new molecular evolution methods, enable multisite incorporation of nsAAs and nonstandard monomers (nsM) paving the way for the template-directed production of functionalized proteins, new classes of polymers, and genetically encoded materials.


Assuntos
Código Genético , Engenharia Genética/métodos , Genoma , Aminoácidos/genética , Animais , Evolução Molecular Direcionada/métodos , Edição de Genes/métodos , Genômica/métodos , Humanos , Ribossomos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...