Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Bioorg Med Chem Lett ; 101: 129653, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38360420

RESUMO

Gene-encoded aldehyde tag technology has been widely utilized in protein bioorthogonal chemistry and biotechnological application. Herein, we report utilization of the promiscuous rSAM cyclophane synthase SjiB involved in triceptide biosynthesis as a dedicated and highly efficient formylglycine synthase. The new aldehyde tag sequence in this system, YQSSI, is biosynthetically orthogonal to the known aldehyde tag (C/S)x(P/A)xR. The potential use of SjiB/YQSSI aldehyde tag system was further validated in fluorescent labelling of model proteins.


Assuntos
Aldeídos , Ciclofanos , Proteínas
2.
Nucleic Acids Res ; 50(5): 2973-2985, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35191490

RESUMO

Serine integrases are emerging as one of the most powerful biological tools for synthetic biology. They have been widely used across genome engineering and genetic circuit design. However, developing serine integrase-based tools for directly/precisely manipulating synthetic biobricks is still missing. Here, we report SYMBIOSIS, a versatile method that can robustly manipulate DNA parts in vivo and in vitro. First, we propose a 'keys match locks' model to demonstrate that three orthogonal serine integrases are able to irreversibly and stably switch on seven synthetic biobricks with high accuracy in vivo. Then, we demonstrate that purified integrases can facilitate the assembly of 'donor' and 'acceptor' plasmids in vitro to construct composite plasmids. Finally, we use SYMBIOSIS to assemble different chromoprotein genes and create novel colored Escherichia coli. We anticipate that our SYMBIOSIS strategy will accelerate synthetic biobrick manipulation, genetic circuit design and multiple plasmid assembly for synthetic biology with broad potential applications.


Assuntos
Integrases , Serina , Biologia Sintética/métodos , Escherichia coli/genética , Integrases/genética , Plasmídeos/genética , Serina/genética
3.
Biotechnol Bioeng ; 120(3): 793-802, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36510694

RESUMO

Nonribosomal peptides (NRPs) are a large family of secondary metabolites with notable bioactivities, which distribute widely in natural resources across microbes and plants. To obtain these molecules, heterologous production of NRPs in robust surrogate hosts like Escherichia coli represent a feasible approach. However, reconstitution of the full biosynthetic pathway in a host often leads to low productivity, which is at least in part due to the low efficiency of enzyme interaction in vivo except for the well-known reasons of metabolic burden (e.g., expression of large NRP synthetases-NRPSs with molecular weights of >100 kDa) and cellular toxicity on host cells. To enhance the catalytic efficiency of large NRPSs in vivo, here we propose to staple NRPS enzymes by using short peptide/protein pairs (e.g., SpyTag/SpyCatcher) for enhanced NRP production. We achieve this goal by introducing a stapled NRPS system for the biosynthesis of the antibiotic NRP valinomycin in E. coli. The results indicate that stapled valinomycin synthetase (Vlm1 and Vlm2) enables higher product accumulation than those two free enzymes (e.g., the maximum improvement is nearly fourfold). After further optimization by strain and bioprocess engineering, the final valinomycin titer maximally reaches about 2800 µg/L, which is 73 times higher than the initial titer of 38 µg/L. We expect that stapling NRPS enzymes will be a promising catalytic strategy for high-level biosynthesis of NRP natural products.


Assuntos
Antibacterianos , Escherichia coli , Valinomicina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Vias Biossintéticas , Peptídeo Sintases/genética , Peptídeo Sintases/metabolismo , Peptídeos/metabolismo
4.
Angew Chem Int Ed Engl ; 62(52): e202312906, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-37966024

RESUMO

In this study, we addressed the limitations of conventional enzyme-polymer-conjugate-based Pickering emulsions for interfacial biocatalysis, which traditionally suffer from nonspecific and uncontrollable conjugation positions that can impede catalytic performance. By introducing a non-canonical amino acid (ncAA) at a specific site on target enzymes, we enabled precise polymer-enzyme conjugation. These engineered conjugates then acted as biocatalytically active emulsifiers to stabilize Pickering emulsions, while encapsulating a cell-free protein synthesis (CFPS) system in the aqueous phase for targeted enzyme expression. The resulting cascade reaction system leveraged enzymes expressed in the aqueous phase and on the emulsion interface for optimized chemical biosynthesis. The use of the cell-free system eliminated the need for intact whole cells or purified enzymes, representing a significant advancement in biocatalysis. Remarkably, the integration of Pickering emulsion, precise enzyme-polymer conjugation, and CFPS resulted in a fivefold enhancement in catalytic performance as compared to traditional single-phase reactions. Therefore, our approach harnesses the combined strengths of advanced biochemical engineering techniques, offering an efficient and practical solution for the synthesis of value-added chemicals in various biocatalysis and biotransformation applications.


Assuntos
Polímeros , Emulsões/química , Biocatálise , Catálise , Biotransformação
5.
Metab Eng ; 60: 37-44, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32224263

RESUMO

Natural products are important because of their significant pharmaceutical properties such as antiviral, antimicrobial, and anticancer activity. Recent breakthroughs in DNA sequencing reveal that a great number of cryptic natural product biosynthetic gene clusters are encoded in microbial genomes, for example, those of Streptomyces species. However, it is still challenging to access compounds from these clusters because many source organisms are uncultivable or the genes are silent during laboratory cultivation. To address this challenge, we develop an efficient cell-free platform for the rapid, in vitro total biosynthesis of the nonribosomal peptide valinomycin as a model. We achieve this goal in two ways. First, we used a cell-free protein synthesis (CFPS) system to express the entire valinomycin biosynthetic gene cluster (>19 kb) in a single-pot reaction, giving rise to approximately 37 µg/L of valinomycin after optimization. Second, we coupled CFPS with cell-free metabolic engineering system by mixing two enzyme-enriched cell lysates to perform a two-stage biosynthesis. This strategy improved valinomycin production ~5000-fold to nearly 30 mg/L. We expect that cell-free biosynthetic systems will provide a new avenue to express, discover, and characterize natural product gene clusters of interest in vitro.


Assuntos
Antibacterianos/biossíntese , Valinomicina/biossíntese , Bioengenharia , Sistema Livre de Células , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Bacterianos , Engenharia Metabólica/métodos , Família Multigênica , Streptomyces/genética , Streptomyces/metabolismo
6.
Biotechnol Bioeng ; 117(12): 4001-4008, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32827317

RESUMO

Biocatalytic cascade reactions have become increasingly important and useful for chemical synthesis. However, biocatalysts are often incompatible with organic solvents, which prohibits many cascade reactions involving nonpolar substrates. In this study, we used cell-free protein synthesis (CFPS) to express enzymes in an aqueous-organic biphasic system for the construction of an artificial enzymatic pathway. CFPS-expressed enzymes without purification performed efficiently to convert styrene (below 20 mM) to (S)-1-phenyl-1,2-ethanediol (two steps in one pot) with 100% conversion. In addition, our CFPS system showed great tolerance to different organic solvents, and, importantly, the entire biocatalytic system can be consistently scaled up without a reduction of the substrate conversion rate. We, therefore, anticipate that our cell-free approach will make a possible cost-effective, high-yielding synthesis of valuable chemicals.


Assuntos
Escherichia coli/química , Proteínas de Fluorescência Verde/biossíntese , Biossíntese de Proteínas , Biotransformação , Sistema Livre de Células , Escherichia coli/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(13): 3521-6, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26979951

RESUMO

Production of ribosomally synthesized and posttranslationally modified peptides (RiPPs) has rarely been reported in fungi, even though organisms of this kingdom have a long history as a prolific source of natural products. Here we report an investigation of the phomopsins, antimitotic mycotoxins. We show that phomopsin is a fungal RiPP and demonstrate the widespread presence of a pathway for the biosynthesis of a family of fungal cyclic RiPPs, which we term dikaritins. We characterize PhomM as an S-adenosylmethionine-dependent α-N-methyltransferase that converts phomopsin A to an N,N-dimethylated congener (phomopsin E), and show that the methyltransferases involved in dikaritin biosynthesis have evolved differently and likely have broad substrate specificities. Genome mining studies identified eight previously unknown dikaritins in different strains, highlighting the untapped capacity of RiPP biosynthesis in fungi and setting the stage for investigating the biological activities and unknown biosynthetic transformations of this family of fungal natural products.


Assuntos
Ascomicetos/metabolismo , Micotoxinas/biossíntese , Sequência de Aminoácidos , Antimitóticos/química , Antimitóticos/metabolismo , Ascomicetos/genética , Produtos Biológicos/química , Produtos Biológicos/metabolismo , Vias Biossintéticas , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Genes Fúngicos , Dados de Sequência Molecular , Família Multigênica , Micotoxinas/química , Micotoxinas/genética , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Sequências Repetitivas de Aminoácidos , Ribossomos/metabolismo
8.
Angew Chem Int Ed Engl ; 58(19): 6235-6238, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30884058

RESUMO

HemN is a radical S-adenosyl-l-methionine (SAM) enzyme that catalyzes the oxidative decarboxylation of coproporphyrinogen III to produce protoporphyrinogen IX, an intermediate in heme biosynthesis. HemN binds two SAM molecules in the active site, but how these two SAMs are utilized for the sequential decarboxylation of the two propionate groups of coproporphyrinogen III remains largely elusive. Provided here is evidence showing that in HemN catalysis a SAM serves as a hydrogen relay which mediates a radical-based hydrogen transfer from the propionate to the 5'-deoxyadenosyl (dAdo) radical generated from another SAM in the active site. Also observed was an unexpected shunt product resulting from trapping of the SAM-based methylene radical by the vinyl moiety of the mono-decarboxylated intermediate, harderoporphyrinogen. These results suggest a major revision of the HemN mechanism and reveal a new paradigm of the radical-mediated hydrogen transfer in radical SAM enzymology.


Assuntos
Proteínas de Bactérias/metabolismo , Coproporfirinogênio Oxidase/metabolismo , Biocatálise , Domínio Catalítico , Coproporfirinogênios/metabolismo , Escherichia coli/metabolismo , Hidrogênio/química , Hidrogênio/metabolismo , Metano/análogos & derivados , Metano/química , Ligação Proteica , Protoporfirinas/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo
9.
J Am Chem Soc ; 140(4): 1365-1371, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29300094

RESUMO

Regiospecific dehydration of vicinal diols by enzymes is a difficult reaction that usually requires activation by dedicated organic cofactors. The enzymatic use of radical-based chemistry is an effective but challenging alternative as radical intermediates are difficult to control. Here we report the X-ray structure of the radical S-adenosyl-l-methionine (SAM) dehydratase AprD4 involved in the biosynthesis of the aminoglycoside (AG) antibiotic apramycin. Using in vitro characterizations and theoretical calculations based on our crystal structure, we have been able to propose a detailed mechanism of AprD4 catalysis, which involves a complex partially substrate-induced proton relay network in the enzyme active site and highlights the key role of the protein matrix in driving high-energy intermediates.


Assuntos
Álcoois/metabolismo , Hidroliases/metabolismo , Prótons , S-Adenosilmetionina/metabolismo , Álcoois/química , Biocatálise , Cristalografia por Raios X , Desidratação , Radicais Livres/química , Radicais Livres/metabolismo , Hidroliases/química , Modelos Moleculares , Teoria Quântica , S-Adenosilmetionina/química , Streptomyces/enzimologia , Especificidade por Substrato
10.
J Org Chem ; 83(13): 7271-7275, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29357665

RESUMO

A chemoenzymatic approach for the synthesis of teixobactin analogues has been established by using the tandem thioesterase (TE) of the nonribosomal peptide synthase (NRPS) Txo2. We show that, unlike the closely related counterparts involved in lysobactin biosynthesis (in which the N-terminal TE is solely responsible for the lactonization reaction), the two teixobactin TE domains are functionally exchangeable and likely act synergistically, representing an unprecedented off-loading mechanism in NRPS enzymology. The substrate specificity of this tandem TE was also investigated in this study.


Assuntos
Depsipeptídeos/síntese química , Esterases/metabolismo , Sequência de Aminoácidos , Cromatografia Líquida de Alta Pressão/métodos , Depsipeptídeos/química , Espectrometria de Massas/métodos , Conformação Proteica , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
11.
Biotechnol J ; 19(4): e2400114, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38622790

RESUMO

Molecular cloning facilitates the assembly of heterologous DNA fragments with vectors, resulting in the generation of plasmids that can steadily replicate in host cells. To efficiently and accurately screen out the expected plasmid candidates, various methods, such as blue-white screening, have been developed for visualization. However, these methods typically require additional genetic manipulations and costs. To simplify the process of visualized molecular cloning, here we report Rainbow Screening, a method that combines Gibson Assembly with chromoproteins to distinguish Escherichia coli (E. coli) colonies by naked eyes, eliminating the need for additional genetic manipulations or costs. To illustrate the design, we select both E. coli 16s rRNA and sfGFP expression module as two inserted fragments. Using Rainbow Screening, false positive colonies can be easily distinguished on LB-agar plates. Moreover, both the assembly efficiency and the construct accuracy can exceed 80%. We anticipate that Rainbow Screening will enrich the molecular cloning methodology and expand the application of chromoproteins in biotechnology and synthetic biology.


Assuntos
DNA , Escherichia coli , Escherichia coli/genética , RNA Ribossômico 16S , Clonagem Molecular , Plasmídeos , DNA/genética , Vetores Genéticos
12.
Biotechnol J ; 19(1): e2300327, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37800393

RESUMO

Escherichia coli Nissle 1917 (EcN) is a probiotic microbe that has the potential to be developed as a promising chassis for synthetic biology applications. However, the molecular tools and techniques for utilizing EcN remain to be further explored. To address this opportunity, the EcN-based toolbox was systematically expanded, enabling EcN as a powerful platform for more applications. First, two EcN cryptic plasmids and other compatible plasmids were genetically engineered to enrich the manipulable plasmid toolbox for multiple gene coexpression. Next, two EcN-based technologies were developed, including the conjugation strategy for DNA transfer, and quantification of protein expression capability. Finally, the EcN-based applications were further expanded by developing EcN native integrase-mediated genetic engineering and establishing an in vitro cell-free protein synthesis (CFPS) system. Overall, this study expanded the toolbox for manipulating and making full use of EcN as a commonly used probiotic chassis, providing several simplified, dependable, and predictable strategies for researchers working in synthetic biology fields.


Assuntos
Escherichia coli , Probióticos , Escherichia coli/genética , Escherichia coli/metabolismo , Biologia Sintética , Engenharia Genética/métodos , Plasmídeos/genética
13.
ACS Synth Biol ; 13(5): 1434-1441, 2024 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-38695987

RESUMO

Enzymatic cascades have become a green and sustainable approach for the synthesis of valuable chemicals and pharmaceuticals. Using sequential enzymes to construct a multienzyme complex is an effective way to enhance the overall performance of biosynthetic routes. Here we report the design of an efficient in vitro hybrid biocatalytic system by assembling three enzymes that can convert styrene to (S)-1-phenyl-1,2-ethanediol. Specifically, we prepared the three enzymes in different ways, which were cell surface-displayed, purified, and cell-free expressed. To assemble them, we fused two orthogonal peptide-protein pairs (i.e., SpyTag/SpyCatcher and SnoopTag/SnoopCatcher) to the three enzymes, allowing their spatial organization by covalent assembly. By doing this, we constructed a multienzyme complex, which could enhance the production of (S)-1-phenyl-1,2-ethanediol by 3 times compared to the free-floating enzyme system without assembly. After optimization of the reaction system, the final product yield reached 234.6 µM with a substrate conversion rate of 46.9% (based on 0.5 mM styrene). Taken together, our strategy integrates the merits of advanced biochemical engineering techniques, including cellular surface display, spatial enzyme organization, and cell-free expression, which offers a new solution for chemical biosynthesis by enzymatic cascade biotransformation. We, therefore, anticipate that our approach will hold great potential for designing and constructing highly efficient systems to synthesize chemicals of agricultural, industrial, and pharmaceutical significance.


Assuntos
Biocatálise , Sistema Livre de Células , Estireno/metabolismo , Estireno/química , Escherichia coli/genética , Escherichia coli/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo
14.
Nat Commun ; 15(1): 4336, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773100

RESUMO

Ribosomally synthesized and post-translationally modified peptides (RiPPs) are a major class of natural products with diverse chemical structures and potent biological activities. A vast majority of RiPP gene clusters remain unexplored in microbial genomes, which is partially due to the lack of rapid and efficient heterologous expression systems for RiPP characterization and biosynthesis. Here, we report a unified biocatalysis (UniBioCat) system based on cell-free gene expression for rapid biosynthesis and engineering of RiPPs. We demonstrate UniBioCat by reconstituting a full biosynthetic pathway for de novo biosynthesis of salivaricin B, a lanthipeptide RiPP. Next, we delete several protease/peptidase genes from the source strain to enhance the performance of UniBioCat, which then can synthesize and screen salivaricin B variants with enhanced antimicrobial activity. Finally, we show that UniBioCat is generalizable by synthesizing and evaluating the bioactivity of ten uncharacterized lanthipeptides. We expect UniBioCat to accelerate the discovery, characterization, and synthesis of RiPPs.


Assuntos
Sistema Livre de Células , Processamento de Proteína Pós-Traducional , Ribossomos , Ribossomos/metabolismo , Ribossomos/genética , Peptídeos/metabolismo , Peptídeos/genética , Peptídeos/química , Vias Biossintéticas/genética , Família Multigênica , Biocatálise
15.
Phytochemistry ; 217: 113918, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37952710

RESUMO

Four undescribed sesquiterpene-shikimates (1-4), eight undescribed monoterpene-shikimates (5-12), together with two known ones were isolated and identified from the 95% ethanol extract of the plant endophytic fungus Phyllosticta capitalensis cultured in rice medium. Capitalensis A (1) was identified as the first sesquiterpene-shikimate-conjugated spirocyclic meroterpenoid degradation product, while capitalensis B (2) is a sesquiterpene-shikimate-conjugated spirocyclic meroterpenoid with a unique D-ring formed by a C-2-O-C-9' connection. The structures of these previously undescribed compounds were elucidated by multiple techniques, including IR, HR-ESI-MS, and NMR analysis. Furthermore, their absolute configurations were established through the comprehensive approach that involved the calculations of ECD spectra, optical rotation values, and single-crystal X-ray analysis. Moreover, the anti-inflammatory activity of all isolated compounds was evaluated using a lipopolysaccharide (LPS)-induced inflammation model in BV2 microglial cells. Meanwhile, these compounds exhibited activity in inhibiting NO production. Four compounds, capitalensis C (3), capitalensis D (4), 15-hydroxyl tricycloalternarene 5b (13) and guignarenone A (14) showed strong inhibitory effects with IC50 values of 21.6 ± 1.33, 12.2 ± 1.08, 18.6 ± 1.27, and 15.8 ± 1.20 µM, respectively. In addition, the structure-activity relationship of the anti-inflammatory activity of the compounds was discussed.


Assuntos
Sesquiterpenos , Ácido Chiquímico , Estrutura Molecular , Anti-Inflamatórios/química , Sesquiterpenos/química
16.
Synth Syst Biotechnol ; 8(4): 610-617, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37781172

RESUMO

Plant-originated natural products are important drug sources. However, total biosynthesis of these compounds is often not achievable due to their uncharacterized, lengthy biosynthetic pathways. In nature, phenethylisoquinoline alkaloids (PIAs) such as colchicine are biosynthesized via a common precursor 6,7-dihydroxy-1-(4-hydroxyphenylethyl)-1,2,3,4-tetrahydroisoquinoline (i.e., phenethylisoquinoline scaffold, PIAS). PIAS is naturally synthesized in plants by using two upstream substrates (l-phenylalanine and l-tyrosine) catalyzed by eight enzymes. To shorten this native pathway, here we designed an artificial route to synthesize PIAS with two enzymatic steps from two alternative substrates of 3-(4-hydroxyphenyl) propanol (4-HPP) and dopamine. In the two-step bioconversion, an alcohol dehydrogenase selected from yeast (i.e., ADH7) was able to oxidize its non-native alcohol substrate 4-HPP to form the corresponding aldehyde product, which was then condensed with dopamine by the (S)-norcoclaurine synthase (NCS) to synthesize PIAS. After optimization, the final enzymatic reaction system was successfully scaled up by 200 times from 50 µL to 10 mL, generating 5.4 mM of PIAS. We envision that this study will provide an easy and sustainable approach to produce PIAS and thus lay the foundation for large-scale production of PIAS-derived natural products.

17.
J Agric Food Chem ; 71(22): 8551-8557, 2023 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-37216486

RESUMO

Cell-free expression systems have emerged as a potent and promising platform for the biosynthesis of chemicals by reconstituting in vitro expressed enzymes. Here, we report cell-free biosynthesis of cinnamyl alcohol (cinOH) with enhanced productivity by using the Plackett-Burman experimental design for multifactor optimization. Initially, four enzymes were individually expressed in vitro and directly mixed to reconstitute a biosynthetic route for the synthesis of cinOH. Then, the Plackett-Burman experimental design was used to screen multiple reaction factors and found three crucial parameters (i.e., reaction temperature, reaction volume, and carboxylic acid reductase) for the cinOH production. With the optimum reaction conditions, approximately 300 µM of cinOH was synthesized after 10 h of cell-free biosynthesis. Extending the production time to 24 h also increased the production to a maximum yield of 807 µM, which is nearly 10 times higher than the initial yield without optimization. This study demonstrates that cell-free biosynthesis can be combined with other powerful optimization methodologies such as the Plackett-Burman experimental design for enhanced production of valuable chemicals.


Assuntos
Sistema Livre de Células , Propanóis
18.
Metab Eng Commun ; 16: e00217, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36578475

RESUMO

Fatty acid-derived products such as alkanes, fatty aldehydes, and fatty alcohols have many applications in the chemical industry. These products are predominately produced from fossil resources, but their production processes are often not environmentally friendly. While microbes like Escherichia coli have been engineered to convert fatty acids to corresponding products, the design and optimization of metabolic pathways in cells for high productivity is challenging due to low mass transfer, heavy metabolic burden, and intermediate/product toxicity. Here, we describe an E. coli-based cell-free protein synthesis (CFPS) platform for in vitro conversion of long-chain fatty acids to value-added chemicals with product selectivity, which can also avoid the above issues when using microbial production systems. We achieve the selective biotransformation by cell-free expression of different enzymes and the use of different conditions (e.g., light and heating) to drive the biocatalysis toward different final products. Specifically, in response to blue light, cell-free expressed fatty acid photodecarboxylase (CvFAP, a photoenzyme) was able to convert fatty acids to alkanes with approximately 90% conversion. When the expressed enzyme was switched to carboxylic acid reductase (CAR), fatty acids were reduced to corresponding fatty aldehydes, which, however, could be further reduced to fatty alcohols by endogenous reductases in the cell-free system. By using a thermostable CAR and a heating treatment, the endogenous reductases were deactivated and fatty aldehydes could be selectively accumulated (>97% in the product mixture) without over-reduction to alcohols. Overall, our cell-free platform provides a new strategy to convert fatty acids to valuable chemicals with notable properties of operation flexibility, reaction controllability, and product selectivity.

19.
ACS Synth Biol ; 12(4): 1349-1357, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37040607

RESUMO

Crude extract-based cell-free expression systems have been used to produce natural products by reconstitution of their biosynthetic pathways in vitro. However, the chemical scope of cell-free synthesized natural compounds is still limited, which is partially due to the length of biosynthetic gene clusters. To expand the product scope, here, we report cell-free biosynthesis of several lysine-derived unnatural amino acids with functional moieties such as chloro, alkene, and alkyne groups. Specifically, five related enzymes (i.e., halogenase, oxidase, lyase, ligase, and hydroxylase) involved in ß-ethynylserine biosynthesis are selected for cell-free expression. These enzymes can be expressed in single, in pairs, or in trios to synthesize different compounds, including, for instance, 4-Cl-l-lysine, 4-Cl-allyl-l-glycine, and l-propargylglycine. The final product of γ-l-glutamyl-l-ß-ethynylserine (a dipeptide with an alkyne group) can also be synthesized by cell-free expression of the full biosynthetic pathway (i.e., five enzymes). Our results demonstrate the flexibility of cell-free systems, enabling easy regulation and rational optimization for target compound formation. Overall, this work expands not only the type of enzymes (e.g., halogenase) but also the scope of natural products (e.g., terminal-alkyne amino acid) that can be rapidly produced in cell-free systems. With the development of cell-free biotechnology, we envision that cell-free strategies will create a new frontier for natural product biosynthesis.


Assuntos
Aminoácidos , Produtos Biológicos , Alcenos , Alcinos/metabolismo , Aminoácidos/química , Lisina/metabolismo , Sistema Livre de Células
20.
Adv Sci (Weinh) ; 10(14): e2207008, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36938858

RESUMO

Erythritol, one of the natural sugar alcohols, is widely used as a sugar substitute sweetener in food industries. Humans themselves are not able to catabolize erythritol and their gut microbes lack related catabolic pathways either to metabolize erythritol. Here, Escherichia coli (E. coli) is engineered to utilize erythritol as sole carbon source aiming for defined applications. First, the erythritol metabolic gene cluster is isolated and the erythritol-binding transcriptional repressor and its DNA-binding site are experimentally characterized. Transcriptome analysis suggests that carbohydrate metabolism-related genes in the engineered E. coli are overall upregulated. In particular, the enzymes of transaldolase (talA and talB) and transketolase (tktA and tktB) are notably overexpressed (e.g., the expression of tktB is improved by nearly sixfold). By overexpression of the four genes, cell growth can be increased as high as three times compared to the cell cultivation without overexpression. Finally, engineered E. coli strains can be used as a living detector to distinguish erythritol-containing soda soft drinks and can grow in the simulated intestinal fluid supplemented with erythritol. This work is expected to inspire the engineering of more hosts to respond and utilize erythritol for broad applications in metabolic engineering, synthetic biology, and biomedical engineering.


Assuntos
Eritritol , Escherichia coli , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Eritritol/metabolismo , Carbono , Fatores de Transcrição/genética , Engenharia Metabólica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa