Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
ACS Synth Biol ; 13(4): 1290-1302, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38526141

RESUMO

The important roles that protein glycosylation plays in modulating the activities and efficacies of protein therapeutics have motivated the development of synthetic glycosylation systems in living bacteria and in vitro. A key challenge is the lack of glycosyltransferases that can efficiently and site-specifically glycosylate desired target proteins without the need to alter primary amino acid sequences at the acceptor site. Here, we report an efficient and systematic method to screen a library of glycosyltransferases capable of modifying comprehensive sets of acceptor peptide sequences in parallel. This approach is enabled by cell-free protein synthesis and mass spectrometry of self-assembled monolayers and is used to engineer a recently discovered prokaryotic N-glycosyltransferase (NGT). We screened 26 pools of site-saturated NGT libraries to identify relevant residues that determine polypeptide specificity and then characterized 122 NGT mutants, using 1052 unique peptides and 52,894 unique reaction conditions. We define a panel of 14 NGTs that can modify 93% of all sequences within the canonical X-1-N-X+1-S/T eukaryotic glycosylation sequences as well as another panel for many noncanonical sequences (with 10 of 17 non-S/T amino acids at the X+2 position). We then successfully applied our panel of NGTs to increase the efficiency of glycosylation for three protein therapeutics. Our work promises to significantly expand the substrates amenable to in vitro and bacterial glycoengineering.


Assuntos
Proteínas de Bactérias , Glicosiltransferases , Glicosilação , Glicosiltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Glucosiltransferases/metabolismo , Peptídeos/metabolismo , Bactérias/metabolismo
2.
Nat Commun ; 14(1): 3897, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37400446

RESUMO

Antibody discovery is bottlenecked by the individual expression and evaluation of antigen-specific hits. Here, we address this bottleneck by developing a workflow combining cell-free DNA template generation, cell-free protein synthesis, and binding measurements of antibody fragments in a process that takes hours rather than weeks. We apply this workflow to evaluate 135 previously published antibodies targeting the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), including all 8 antibodies previously granted emergency use authorization for coronavirus disease 2019 (COVID-19), and demonstrate identification of the most potent antibodies. We also evaluate 119 anti-SARS-CoV-2 antibodies from a mouse immunized with the SARS-CoV-2 spike protein and identify neutralizing antibody candidates, including the antibody SC2-3, which binds the SARS-CoV-2 spike protein of all tested variants of concern. We expect that our cell-free workflow will accelerate the discovery and characterization of antibodies for future pandemics and for research, diagnostic, and therapeutic applications more broadly.


Assuntos
COVID-19 , Animais , Humanos , Camundongos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Anticorpos Neutralizantes , Anticorpos Antivirais
3.
ACS Synth Biol ; 12(4): 1216-1226, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-36940255

RESUMO

In resource-limited settings, it can be difficult to safely deliver sensitive biologic medicines to patients due to cold chain and infrastructure constraints. Point-of-care drug manufacturing could circumvent these challenges since medicines could be produced locally and used on-demand. Toward this vision, we combine cell-free protein synthesis (CFPS) and a 2-in-1 affinity purification and enzymatic cleavage scheme to develop a platform for point-of-care drug manufacturing. As a model, we use this platform to synthesize a panel of peptide hormones, an important class of medications that can be used to treat a wide variety of diseases including diabetes, osteoporosis, and growth disorders. With this approach, temperature-stable lyophilized CFPS reaction components can be rehydrated with DNA encoding a SUMOylated peptide hormone of interest when needed. Strep-Tactin affinity purification and on-bead SUMO protease cleavage yield peptide hormones in their native form that are recognized by ELISA antibodies and that can bind their respective receptors. With further development to ensure proper biologic activity and patient safety, we envision that this platform could be used to manufacture valuable peptide hormone drugs in a decentralized way.


Assuntos
Hormônios Peptídicos , Humanos , Sistemas Automatizados de Assistência Junto ao Leito , Biossíntese de Proteínas
4.
Nat Commun ; 13(1): 6257, 2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36270990

RESUMO

Mucin-type-O-glycosylation on proteins is integrally involved in human health and disease and is coordinated by an enzyme family of 20 N-acetylgalactosaminyltransferases (GalNAc-Ts). Detailed knowledge on the biological effects of site-specific O-glycosylation is limited due to lack of information on specific glycosylation enzyme activities and O-glycosylation site-occupancies. Here we present a systematic analysis of the isoform-specific targets of all GalNAc-Ts expressed within a tissue-forming human skin cell line, and demonstrate biologically significant effects of O-glycan initiation on epithelial formation. We find over 300 unique glycosylation sites across a diverse set of proteins specifically regulated by one of the GalNAc-T isoforms, consistent with their impact on the tissue phenotypes. Notably, we discover a high variability in the O-glycosylation site-occupancy of 70 glycosylated regions of secreted proteins. These findings revisit the relevance of individual O-glycosylation sites in the proteome, and provide an approach to establish which sites drive biological functions.


Assuntos
N-Acetilgalactosaminiltransferases , Proteoma , Humanos , Glicosilação , Proteoma/metabolismo , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Linhagem Celular , Mucinas/metabolismo , Polissacarídeos
5.
AIChE J ; 67(12)2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36329688

RESUMO

Bacterial biofilms are associated with chronic infectious diseases and are highly resistant to conventional antibiotics. Antimicrobial bacteriocins are alternatives to conventional antibiotics and are characterized by unique cell-killing mechanisms, including pore formation on cell membranes, nuclease activity, and cell wall synthesis inhibition. Here, we used cell-free protein synthesis to rapidly evaluate the anti-biofilm activities of colicins E1, E2, and E3. We found that E2 (with DNase activity) most effectively killed target biofilm cells (i.e., the K361 strain) while leaving non-targeted biofilms intact. We then engineered probiotic Escherichia coli microorganisms with genetic circuits to controllably synthesize and secrete colicin E2, which successfully inhibited biofilms and killed pre-formed indicator biofilms. Our findings suggest that colicins rapidly and selectively kill target biofilm cells in multispecies biofilms and demonstrate the potential of using microorganisms engineered to produce antimicrobial colicin proteins as live therapeutic strategies to treat biofilm-associated infections.

6.
J Ind Microbiol Biotechnol ; 47(11): 977-991, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33090335

RESUMO

Protein glycosylation, the enzymatic modification of amino acid sidechains with sugar moieties, plays critical roles in cellular function, human health, and biotechnology. However, studying and producing defined glycoproteins remains challenging. Cell-free glycoprotein synthesis systems, in which protein synthesis and glycosylation are performed in crude cell extracts, offer new approaches to address these challenges. Here, we review versatile, state-of-the-art systems for biomanufacturing glycoproteins in prokaryotic and eukaryotic cell-free systems with natural and synthetic N-linked glycosylation pathways. We discuss existing challenges and future opportunities in the use of cell-free systems for the design, manufacture, and study of glycoprotein biomedicines.


Assuntos
Sistema Livre de Células , Glicoproteínas , Glicosilação , Biotecnologia , Glicoproteínas/genética , Humanos , Células Procarióticas , Biossíntese de Proteínas
7.
ACS Synth Biol ; 9(7): 1534-1562, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32526139

RESUMO

Protein glycosylation, the attachment of sugars to amino acid side chains, can endow proteins with a wide variety of properties of great interest to the engineering biology community. However, natural glycosylation systems are limited in the diversity of glycoproteins they can synthesize, the scale at which they can be harnessed for biotechnology, and the homogeneity of glycoprotein structures they can produce. Here we provide an overview of the emerging field of synthetic glycobiology, the application of synthetic biology tools and design principles to better understand and engineer glycosylation. Specifically, we focus on how the biosynthetic and analytical tools of synthetic biology have been used to redesign glycosylation systems to obtain defined glycosylation structures on proteins for diverse applications in medicine, materials, and diagnostics. We review the key biological parts available to synthetic biologists interested in engineering glycoproteins to solve compelling problems in glycoscience, describe recent efforts to construct synthetic glycoprotein synthesis systems, and outline exemplary applications as well as new opportunities in this emerging space.


Assuntos
Proteínas/metabolismo , Biologia Sintética/métodos , Animais , Bactérias/metabolismo , Biotecnologia , Fungos/metabolismo , Glicosilação , Glicosiltransferases/metabolismo , Plantas/metabolismo , Proteínas/química
8.
ACS Cent Sci ; 6(2): 144-154, 2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-32123732

RESUMO

Protein glycosylation is a common post-translational modification that influences the functions and properties of proteins. Despite advances in methods to produce defined glycoproteins by chemoenzymatic elaboration of monosaccharides, the understanding and engineering of glycoproteins remain challenging, in part, due to the difficulty of site-specifically controlling glycosylation at each of several positions within a protein. Here, we address this limitation by discovering and exploiting the unique, conditionally orthogonal peptide acceptor specificities of N-glycosyltransferases (NGTs). We used cell-free protein synthesis and mass spectrometry of self-assembled monolayers to rapidly screen 41 putative NGTs and rigorously characterize the unique acceptor sequence preferences of four NGT variants using 1254 acceptor peptides and 8306 reaction conditions. We then used the optimized NGT-acceptor sequence pairs to sequentially install monosaccharides at four sites within one target protein. This strategy to site-specifically control the installation of N-linked monosaccharides for elaboration to a variety of functional N-glycans overcomes a major limitation in synthesizing defined glycoproteins for research and therapeutic applications.

9.
Anal Chem ; 92(2): 1963-1971, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31854989

RESUMO

High-throughput quantification of the post-translational modification of many individual protein samples is challenging with current label-based methods. This paper demonstrates an efficient method that addresses this gap by combining Escherichia coli-based cell-free protein synthesis (CFPS) and self-assembled monolayers for matrix-assisted laser desorption/ionization mass spectrometry (SAMDI-MS) to analyze intact proteins. This high-throughput approach begins with polyhistidine-tagged protein substrates expressed from linear DNA templates by CFPS. Here, we synthesized an 87-member library of the E. coli Immunity Protein 7 (Im7) containing an acceptor sequence optimized for glycosylation by the Actinobacillus pleuropneumoniae N-glycosyltransferase (NGT) at every possible position along the protein backbone. These protein substrates were individually treated with NGT and then selectively immobilized to self-assembled monolayers presenting nickel-nitrilotriacetic acid (Ni-NTA) complexes before final analysis by SAMDI-MS to quantify the conversion of substrate to glycoprotein. This method offers new opportunities for rapid synthesis and quantitative evaluation of intact glycoproteins.


Assuntos
Proteínas de Transporte/análise , Proteínas de Escherichia coli/análise , Glicoproteínas/análise , Ensaios de Triagem em Larga Escala/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Actinobacillus pleuropneumoniae/enzimologia , Proteínas de Transporte/síntese química , Proteínas de Transporte/genética , Escherichia coli/química , Proteínas de Escherichia coli/síntese química , Proteínas de Escherichia coli/genética , Glicoproteínas/síntese química , Glicoproteínas/genética , Glicosilação , Glicosiltransferases/química , Mutação , Biblioteca de Peptídeos , Estudo de Prova de Conceito , Proteínas Recombinantes/análise , Proteínas Recombinantes/síntese química , Proteínas Recombinantes/genética
10.
Nat Commun ; 10(1): 5404, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31776339

RESUMO

Glycosylation plays important roles in cellular function and endows protein therapeutics with beneficial properties. However, constructing biosynthetic pathways to study and engineer precise glycan structures on proteins remains a bottleneck. Here, we report a modular, versatile cell-free platform for glycosylation pathway assembly by rapid in vitro mixing and expression (GlycoPRIME). In GlycoPRIME, glycosylation pathways are assembled by mixing-and-matching cell-free synthesized glycosyltransferases that can elaborate a glucose primer installed onto protein targets by an N-glycosyltransferase. We demonstrate GlycoPRIME by constructing 37 putative protein glycosylation pathways, creating 23 unique glycan motifs, 18 of which have not yet been synthesized on proteins. We use selected pathways to synthesize a protein vaccine candidate with an α-galactose adjuvant motif in a one-pot cell-free system and human antibody constant regions with minimal sialic acid motifs in glycoengineered Escherichia coli. We anticipate that these methods and pathways will facilitate glycoscience and make possible new glycoengineering applications.


Assuntos
Sistema Livre de Células/metabolismo , Engenharia de Proteínas/métodos , Proteínas/metabolismo , Antígenos CD/metabolismo , Escherichia coli/genética , Glicoproteínas/biossíntese , Glicosilação , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Humanos , Redes e Vias Metabólicas , Oligossacarídeos/metabolismo , Polissacarídeos/metabolismo , Proteínas/genética , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Sialiltransferases/metabolismo
11.
Methods Protoc ; 2(2)2019 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-36358105

RESUMO

Colicins are antimicrobial proteins produced by Escherichia coli that hold great promise as viable complements or alternatives to antibiotics. Cell-free protein synthesis (CFPS) is a useful production platform for toxic proteins because it eliminates the need to maintain cell viability, a common problem in cell-based production. Previously, we demonstrated that colicins produced by CFPS based on crude Escherichia coli lysates are effective in eradicating antibiotic-tolerant bacteria known as persisters. However, we also found that some colicins have poor solubility or low cell-killing activity. In this study, we improved the solubility of colicin M from 16% to nearly 100% by producing it in chaperone-enriched E. coli extracts, resulting in enhanced cell-killing activity. We also improved the cytotoxicity of colicin E3 by adding or co-expressing the E3 immunity protein during the CFPS reaction, suggesting that the E3 immunity protein enhances colicin E3 activity in addition to protecting the host strain. Finally, we confirmed our previous finding that active colicins can be rapidly synthesized by observing colicin E1 production over time in CFPS. Within three hours of CFPS incubation, colicin E1 reached its maximum production yield and maintained high cytotoxicity during longer incubations up to 20 h. Taken together, our findings indicate that colicin production can be easily optimized for improved solubility and activity using the CFPS platform.

12.
Nat Chem Biol ; 14(6): 627-635, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29736039

RESUMO

Glycosylation is an abundant post-translational modification that is important in disease and biotechnology. Current methods to understand and engineer glycosylation cannot sufficiently explore the vast experimental landscapes required to accurately predict and design glycosylation sites modified by glycosyltransferases. Here we describe a systematic platform for glycosylation sequence characterization and optimization by rapid expression and screening (GlycoSCORES), which combines cell-free protein synthesis and mass spectrometry of self-assembled monolayers. We produced six N- and O-linked polypeptide-modifying glycosyltransferases from bacteria and humans in vitro and rigorously determined their substrate specificities using 3,480 unique peptides and 13,903 unique reaction conditions. We then used GlycoSCORES to optimize and design small glycosylation sequence motifs that directed efficient N-linked glycosylation in vitro and in the Escherichia coli cytoplasm for three heterologous proteins, including the human immunoglobulin Fc domain. We find that GlycoSCORES is a broadly applicable method to facilitate fundamental understanding of glycosyltransferases and engineer synthetic glycoproteins.


Assuntos
Glicosiltransferases/química , Peptídeos/química , Proteínas de Bactérias/química , Sítios de Ligação , Sistema Livre de Células , Citoplasma/metabolismo , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Glicoproteínas/química , Glicosilação , Humanos , Cinética , Espectrometria de Massas , Domínios Proteicos , Engenharia de Proteínas/métodos , Processamento de Proteína Pós-Traducional , Especificidade por Substrato
13.
Synth Biol (Oxf) ; 3(1): ysy004, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-32995513

RESUMO

Colicins are antimicrobial proteins produced by Escherichia coli, which, upon secretion from the host, kill non-host E. coli strains by forming pores in the inner membrane and degrading internal cellular components such as DNA and RNA. Due to their unique cell-killing activities, colicins are considered viable alternatives to conventional antibiotics. Recombinant production of colicins requires co-production of immunity proteins to protect host cells; otherwise, the colicins are lethal to the host. In this study, we used cell-free protein synthesis (CFPS) to produce active colicins without the need for protein purification and co-production of immunity proteins. Cell-free synthesized colicins were active in killing model E. coli cells with different modes of cytotoxicity. Pore-forming colicins E1 and nuclease colicin E2 killed actively growing cells in a nutrient-rich medium, but the cytotoxicity of colicin Ia was low compared to E1 and E2. Moreover, colicin E1 effectively killed cells in a nutrient-free solution, while the activity of E2 was decreased compared to nutrient-rich conditions. Both colicins E1 and E2 decreased the level of persister cells (metabolically dormant cell populations that are insensitive to antibiotics) by up to six orders of magnitude compared to that of the rifampin pretreated persister cells. This study finds that colicins can eradicate non-growing cells including persisters, and that CFPS is a promising platform for rapid production and characterization of toxic proteins.

14.
Biotechnol Bioeng ; 115(3): 739-750, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29178580

RESUMO

Protein glycosylation, or the attachment of sugar moieties (glycans) to proteins, is important for protein stability, activity, and immunogenicity. However, understanding the roles and regulations of site-specific glycosylation events remains a significant challenge due to several technological limitations. These limitations include a lack of available tools for biochemical characterization of enzymes involved in glycosylation. A particular challenge is the synthesis of oligosaccharyltransferases (OSTs), which catalyze the attachment of glycans to specific amino acid residues in target proteins. The difficulty arises from the fact that canonical OSTs are large (>70 kDa) and possess multiple transmembrane helices, making them difficult to overexpress in living cells. Here, we address this challenge by establishing a bacterial cell-free protein synthesis platform that enables rapid production of a variety of OSTs in their active conformations. Specifically, by using lipid nanodiscs as cellular membrane mimics, we obtained yields of up to 420 µg/ml for the single-subunit OST enzyme, "Protein glycosylation B" (PglB) from Campylobacter jejuni, as well as for three additional PglB homologs from Campylobacter coli, Campylobacter lari, and Desulfovibrio gigas. Importantly, all of these enzymes catalyzed N-glycosylation reactions in vitro with no purification or processing needed. Furthermore, we demonstrate the ability of cell-free synthesized OSTs to glycosylate multiple target proteins with varying N-glycosylation acceptor sequons. We anticipate that this broadly applicable production method will advance glycoengineering efforts by enabling preparative expression of membrane-embedded OSTs from all kingdoms of life.


Assuntos
Proteínas de Bactérias/biossíntese , Campylobacter/enzimologia , Desulfovibrio/enzimologia , Glicosiltransferases/biossíntese , Proteínas de Bactérias/genética , Campylobacter/genética , Sistema Livre de Células/metabolismo , Desulfovibrio/genética , Glicosilação
15.
Electron. j. biotechnol ; 17(1): 3-3, Jan. 2014. ilus
Artigo em Inglês | LILACS | ID: lil-706517

RESUMO

Background: Algae offer many advantages as biofuel sources including: high growth rates, high lipid content, the ability to grow on non-agricultural land, and the genetic versatility to improve strains rapidly and produce co-products. Research is ongoing to make algae biofuels a more financially attractive energy option; however, it is becoming evident that the economic viability of algae-based fuels may hinge upon high-value co-products. This work evaluated the feasibility of using a co-product, algae extract, as a nutrient source in cell culture media. Results: Algae extract prepared from autolysed Chlamydomonas reinhardtii was found to contain 3.0% protein, 9.2% total carbohydrate, and 3.9% free α-amino acid which is similar to the nutrient content of commercially available yeast extract. The effects of algae extract on the growth and metabolism of laboratory strains of Escherichia coli and Saccharomyces cerevisiae were tested by substituting algae extract for yeast extract in LB and YPAD growth media recipes. Complex laboratory media supplemented with algae extract instead of yeast extract showed markedly improved effects on the growth and metabolism of common laboratory microorganisms in all cases except ethanol production rates in yeast. Conclusions: This study showed that algae extract derived from C. reinhardtii is similar, if not superior, to commercially available yeast extract in nutrient content and effects on the growth and metabolism of E. coli and S. cerevisiae. Bacto™ yeast extract is valued at USD $0.15-0.35 per gram, if algae extract was sold at similar prices, it would serve as a high-value co-product in algae-based fuel processes.


Assuntos
Extratos Vegetais/metabolismo , Chlamydomonas reinhardtii/metabolismo , Leveduras , Extratos Vegetais/química , Chlamydomonas reinhardtii/química , Meios de Cultura , Etanol , Biocombustíveis , Microalgas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...