Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Cell ; 160(1-2): 339-50, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25533786

RESUMEN

Eukaryotic cells execute complex transcriptional programs in which specific loci throughout the genome are regulated in distinct ways by targeted regulatory assemblies. We have applied this principle to generate synthetic CRISPR-based transcriptional programs in yeast and human cells. By extending guide RNAs to include effector protein recruitment sites, we construct modular scaffold RNAs that encode both target locus and regulatory action. Sets of scaffold RNAs can be used to generate synthetic multigene transcriptional programs in which some genes are activated and others are repressed. We apply this approach to flexibly redirect flux through a complex branched metabolic pathway in yeast. Moreover, these programs can be executed by inducing expression of the dCas9 protein, which acts as a single master regulatory control point. CRISPR-associated RNA scaffolds provide a powerful way to construct synthetic gene expression programs for a wide range of applications, including rewiring cell fates or engineering metabolic pathways.


Asunto(s)
Sistemas CRISPR-Cas , Expresión Génica , Técnicas Genéticas , Células HEK293 , Humanos , Ingeniería Metabólica , ARN Guía de Kinetoplastida/genética , Saccharomyces cerevisiae/genética , Streptococcus pyogenes/genética
2.
Nature ; 579(7798): E8, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32094663

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nucleic Acids Res ; 52(7): 4098-4107, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38499480

RESUMEN

Long single-stranded DNA (ssDNA) is a versatile molecular reagent with applications including RNA-guided genome engineering and DNA nanotechnology, yet its production is typically resource-intensive. We introduce a novel method utilizing an engineered Escherichia coli 'helper' strain and phagemid system that simplifies long ssDNA generation to a straightforward transformation and purification procedure. Our method obviates the need for helper plasmids and their associated contamination by integrating M13mp18 genes directly into the E. coli chromosome. We achieved ssDNA lengths ranging from 504 to 20 724 nt with titers up to 250 µg/l following alkaline lysis purification. The efficacy of our system was confirmed through its application in primary T-cell genome modifications and DNA origami folding. The reliability, scalability and ease of our approach promise to unlock new experimental applications requiring large quantities of long ssDNA.


Asunto(s)
ADN de Cadena Simple , Escherichia coli , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Ingeniería Genética/métodos , Plásmidos/genética
4.
Nature ; 572(7768): 265-269, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31341280

RESUMEN

De novo-designed proteins1-3 hold great promise as building blocks for synthetic circuits, and can complement the use of engineered variants of natural proteins4-7. One such designer protein-degronLOCKR, which is based on 'latching orthogonal cage-key proteins' (LOCKR) technology8-is a switch that degrades a protein of interest in vivo upon induction by a genetically encoded small peptide. Here we leverage the plug-and-play nature of degronLOCKR to implement feedback control of endogenous signalling pathways and synthetic gene circuits. We first generate synthetic negative and positive feedback in the yeast mating pathway by fusing degronLOCKR to endogenous signalling molecules, illustrating the ease with which this strategy can be used to rewire complex endogenous pathways. We next evaluate feedback control mediated by degronLOCKR on a synthetic gene circuit9, to quantify the feedback capabilities and operational range of the feedback control circuit. The designed nature of degronLOCKR proteins enables simple and rational modifications to tune feedback behaviour in both the synthetic circuit and the mating pathway. The ability to engineer feedback control into living cells represents an important milestone in achieving the full potential of synthetic biology10,11,12. More broadly, this work demonstrates the large and untapped potential of de novo design of proteins for generating tools that implement complex synthetic functionalities in cells for biotechnological and therapeutic applications.


Asunto(s)
Retroalimentación Fisiológica , Redes Reguladoras de Genes , Genes del Tipo Sexual de los Hongos/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Transducción de Señal , Biología Sintética/métodos , Ingeniería Celular , Redes Reguladoras de Genes/genética , Genes del Tipo Sexual de los Hongos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal/genética
5.
Nature ; 572(7768): 205-210, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31341284

RESUMEN

Allosteric regulation of protein function is widespread in biology, but is challenging for de novo protein design as it requires the explicit design of multiple states with comparable free energies. Here we explore the possibility of designing switchable protein systems de novo, through the modulation of competing inter- and intramolecular interactions. We design a static, five-helix 'cage' with a single interface that can interact either intramolecularly with a terminal 'latch' helix or intermolecularly with a peptide 'key'. Encoded on the latch are functional motifs for binding, degradation or nuclear export that function only when the key displaces the latch from the cage. We describe orthogonal cage-key systems that function in vitro, in yeast and in mammalian cells with up to 40-fold activation of function by key. The ability to design switchable protein functions that are controlled by induced conformational change is a milestone for de novo protein design, and opens up new avenues for synthetic biology and cell engineering.


Asunto(s)
Regulación Alostérica , Ingeniería de Proteínas/métodos , Proteínas/química , Proteínas/síntesis química , Proteína 11 Similar a Bcl2/metabolismo , Núcleo Celular/metabolismo , Supervivencia Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Unión Proteica , Transporte de Proteínas , Proteínas/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Biología Sintética
6.
Nature ; 560(7717): 248-252, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30069054

RESUMEN

The capacity to diversify genetic codes advances our ability to understand and engineer biological systems1,2. A method for continuously diversifying user-defined regions of a genome would enable forward genetic approaches in systems that are not amenable to efficient homology-directed oligonucleotide integration. It would also facilitate the rapid evolution of biotechnologically useful phenotypes through accelerated and parallelized rounds of mutagenesis and selection, as well as cell-lineage tracking through barcode mutagenesis. Here we present EvolvR, a system that can continuously diversify all nucleotides within a tunable window length at user-defined loci. This is achieved by directly generating mutations using engineered DNA polymerases targeted to loci via CRISPR-guided nickases. We identified nickase and polymerase variants that offer a range of targeted mutation rates that are up to 7,770,000-fold greater than rates seen in wild-type cells, and editing windows with lengths of up to 350 nucleotides. We used EvolvR to identify novel ribosomal mutations that confer resistance to the antibiotic spectinomycin. Our results demonstrate that CRISPR-guided DNA polymerases enable multiplexed and continuous diversification of user-defined genomic loci, which will be useful for a broad range of basic and biotechnological applications.


Asunto(s)
Sistemas CRISPR-Cas/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Evolución Molecular Dirigida/métodos , Edición Génica/métodos , Mutagénesis Sitio-Dirigida/métodos , Nucleótidos/genética , ADN Polimerasa Dirigida por ADN/genética , Farmacorresistencia Microbiana/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Mutación , Tasa de Mutación , Nucleótidos/metabolismo , Proteínas Ribosómicas/genética , Espectinomicina/farmacología
7.
Nat Chem Biol ; 17(1): 96-103, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33046851

RESUMEN

Eukaryotic cells compartmentalize metabolic pathways in organelles to achieve optimal reaction conditions and avoid crosstalk with cytosolic factors. We found that cytosolic expression of norcoclaurine synthase (NCS), the enzyme that catalyzes the first committed reaction in benzylisoquinoline alkaloid biosynthesis, is toxic in Saccharomyces cerevisiae and, consequently, restricts (S)-reticuline production. We developed a compartmentalization strategy that alleviates NCS toxicity while promoting increased (S)-reticuline titer. This strategy is achieved through efficient targeting of toxic NCS to the peroxisome while, crucially, taking advantage of the free flow of metabolite substrates and products across the peroxisome membrane. We demonstrate that expression of engineered transcription factors can mimic the oleate response for larger peroxisomes, further increasing benzylisoquinoline alkaloid titer without the requirement for peroxisome induction with fatty acids. This work specifically addresses the challenges associated with toxic NCS expression and, more broadly, highlights the potential for engineering organelles with desired characteristics for metabolic engineering.


Asunto(s)
Bencilisoquinolinas/metabolismo , Ligasas de Carbono-Nitrógeno/genética , Regulación Fúngica de la Expresión Génica , Peroxisomas/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transporte Biológico , Ligasas de Carbono-Nitrógeno/metabolismo , Compartimento Celular , Citosol/metabolismo , Genes Reporteros , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ingeniería Metabólica/métodos , Redes y Vías Metabólicas , Ácido Oléico/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Fluorescente Roja
8.
Nat Chem Biol ; 14(3): 256-261, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29309053

RESUMEN

Indigo is an ancient dye uniquely capable of producing the signature tones in blue denim; however, the dyeing process requires chemical steps that are environmentally damaging. We describe a sustainable dyeing strategy that not only circumvents the use of toxic reagents for indigo chemical synthesis but also removes the need for a reducing agent for dye solubilization. This strategy utilizes a glucose moiety as a biochemical protecting group to stabilize the reactive indigo precursor indoxyl to form indican, preventing spontaneous oxidation to crystalline indigo during microbial fermentation. Application of a ß-glucosidase removes the protecting group from indican, resulting in indigo crystal formation in the cotton fibers. We identified the gene coding for the glucosyltransferase PtUGT1 from the indigo plant Polygonum tinctorium and solved the structure of PtUGT1. Heterologous expression of PtUGT1 in Escherichia coli supported high indican conversion, and biosynthesized indican was used to dye cotton swatches and a garment.


Asunto(s)
Color , Glucósidos/química , Glucosiltransferasas/química , Carmin de Índigo/química , Polygonum/enzimología , beta-Glucosidasa/química , Reactores Biológicos , Dominio Catalítico , Cristalografía por Rayos X , ADN Complementario/metabolismo , Dimerización , Escherichia coli , Fermentación , Perfilación de la Expresión Génica , Biblioteca de Genes , Indoles/química , Hojas de la Planta/enzimología , Proteínas de Plantas/química , Polygonum/genética , Proteínas Recombinantes/química , Textiles , Transcriptoma
9.
Metab Eng ; 45: 180-188, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29247865

RESUMEN

Betalains are a family of natural pigments found exclusively in the plant order Caryophyllales. All members of this chemical family are biosynthesized through the common intermediate betalamic acid, which is capable of spontaneously condensing with various primary and secondary amines to produce betalains. Of particular interest is the red-violet betanin, most commonly obtained from Beta vulgaris (beet) as a natural food dye. We demonstrate the first complete microbial production of betanin in Saccharomyces cerevisiae from glucose, an early step towards a fermentation process enabling rapid, on-demand production of this natural dye. A titer of 17mg/L was achieved, corresponding to a color intensity obtained from 10g/L of beetroot extract. Further, we expanded the spectrum of betalain colors by condensing betalamic acid with various amines fed to an engineered strain of S. cerevisiae. Our work establishes a platform for microbial production of betalains of various colors as a potential alternative to land- and resource-intensive agricultural production.


Asunto(s)
Beta vulgaris/genética , Betacianinas/biosíntesis , Betalaínas/biosíntesis , Ingeniería Metabólica , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
10.
Nat Chem Biol ; 11(7): 465-71, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25984720

RESUMEN

Benzylisoquinoline alkaloids (BIAs) are a diverse family of plant-specialized metabolites that include the pharmaceuticals codeine and morphine and their derivatives. Microbial synthesis of BIAs holds promise as an alternative to traditional crop-based manufacturing. Here we demonstrate the production of the key BIA intermediate (S)-reticuline from glucose in Saccharomyces cerevisiae. To aid in this effort, we developed an enzyme-coupled biosensor for the upstream intermediate L-3,4-dihydroxyphenylalanine (L-DOPA). Using this sensor, we identified an active tyrosine hydroxylase and improved its L-DOPA yields by 2.8-fold via PCR mutagenesis. Coexpression of DOPA decarboxylase enabled what is to our knowledge the first demonstration of dopamine production from glucose in yeast, with a 7.4-fold improvement in titer obtained for our best mutant enzyme. We extended this pathway to fully reconstitute the seven-enzyme pathway from L-tyrosine to (S)-reticuline. Future work to improve titers and connect these steps with downstream pathway branches, already demonstrated in S. cerevisiae, will enable low-cost production of many high-value BIAs.


Asunto(s)
Alcaloides/biosíntesis , Bencilisoquinolinas/metabolismo , Técnicas Biosensibles , Dihidroxifenilalanina/análisis , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Saccharomyces cerevisiae/metabolismo , Dihidroxifenilalanina/metabolismo , Dopa-Decarboxilasa/genética , Dopa-Decarboxilasa/metabolismo , Dopamina/biosíntesis , Proteínas Fúngicas/genética , Glucosa/metabolismo , Ingeniería Metabólica , Mutagénesis Sitio-Dirigida , Narcóticos/metabolismo , Saccharomyces cerevisiae/genética , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
11.
Biotechnol Bioeng ; 114(6): 1301-1309, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165133

RESUMEN

A common challenge in metabolic engineering is rapidly identifying rate-controlling enzymes in heterologous pathways for subsequent production improvement. We demonstrate a workflow to address this challenge and apply it to improving xylose utilization in Saccharomyces cerevisiae. For eight reactions required for conversion of xylose to ethanol, we screened enzymes for functional expression in S. cerevisiae, followed by a combinatorial expression analysis to achieve pathway flux balancing and identification of limiting enzymatic activities. In the next round of strain engineering, we increased the copy number of these limiting enzymes and again tested the eight-enzyme combinatorial expression library in this new background. This workflow yielded a strain that has a ∼70% increase in biomass yield and ∼240% increase in xylose utilization. Finally, we chromosomally integrated the expression library. This library enriched for strains with multiple integrations of the pathway, which likely were the result of tandem integrations mediated by promoter homology. Biotechnol. Bioeng. 2017;114: 1301-1309. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Mejoramiento Genético/métodos , Ingeniería Metabólica/métodos , Análisis de Flujos Metabólicos/métodos , Complejos Multienzimáticos/genética , Saccharomyces cerevisiae/fisiología , Xilosa/metabolismo , Técnicas Químicas Combinatorias , Simulación por Computador , Metabolismo , Modelos Biológicos , Complejos Multienzimáticos/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
12.
Angew Chem Int Ed Engl ; 55(39): 11824-8, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27570932

RESUMEN

We describe herein formal syntheses of the indole alkaloids cis-trikentrin A and herbindole B from a common meso-hydroquinone intermediate prepared by a ruthenium-catalyzed [2+2+1+1] cycloaddition that has not been used previously in natural product synthesis. Key steps include a sterically demanding Buchwald-Hartwig amination as well as a unique C(sp(3) )-H amination/indole formation. Studies toward a selective desymmetrization of the meso-hydroquinone are also reported.

13.
Nucleic Acids Res ; 41(22): 10668-78, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24038353

RESUMEN

Engineered metabolic pathways often suffer from flux imbalances that can overburden the cell and accumulate intermediate metabolites, resulting in reduced product titers. One way to alleviate such imbalances is to adjust the expression levels of the constituent enzymes using a combinatorial expression library. Typically, this approach requires high-throughput assays, which are unfortunately unavailable for the vast majority of desirable target compounds. To address this, we applied regression modeling to enable expression optimization using only a small number of measurements. We characterized a set of constitutive promoters in Saccharomyces cerevisiae that spanned a wide range of expression and maintained their relative strengths irrespective of the coding sequence. We used a standardized assembly strategy to construct a combinatorial library and express for the first time in yeast the five-enzyme violacein biosynthetic pathway. We trained a regression model on a random sample comprising 3% of the total library, and then used that model to predict genotypes that would preferentially produce each of the products in this highly branched pathway. This generalizable method should prove useful in engineering new pathways for the sustainable production of small molecules.


Asunto(s)
Vías Biosintéticas/genética , Ingeniería Metabólica/métodos , Saccharomyces cerevisiae/genética , Regulación de la Expresión Génica , Biblioteca de Genes , Técnicas de Genotipaje , Indoles/metabolismo , Modelos Lineales , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Saccharomyces cerevisiae/metabolismo
14.
Proc Natl Acad Sci U S A ; 109(44): 18090-5, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23071327

RESUMEN

Synthetic biology applies engineering principles to facilitate the predictable design of biological systems. Biological systems composed of modular parts with clearly defined interactions are generally easier to manipulate than complex systems exhibiting a large number of subtle interactions. However, recreating the function of a naturally complex system with simple modular parts can increase fragility. Here, inspired by scaffold-directed signaling in higher organisms, we modularize prokaryotic signal transduction to allow programmable redirection of phosphate flux from a histidine kinase to response regulators based on targeting by eukaryotic protein-protein interaction domains. Although scaffold-directed colocalization alone was sufficient to direct signaling between components, this minimal system suffered from high sensitivity to changing expression levels of each component. To address this fragility, we demonstrate how to engineer autoinhibition into the kinase so that phosphotransfer is possible only upon binding to the scaffold. This system, in which scaffold performs the dual functions of activating this autoinhibited kinase and directing flux to the cotargeted response regulator, was significantly more robust to varying component concentrations. Thus, we demonstrate that design principles inspired by the complex signal-transduction pathways of eukaryotes may be generalized, abstracted, and applied to prokaryotes using well-characterized parts.


Asunto(s)
Biología Sintética , Fósforo/metabolismo
15.
Metab Eng ; 25: 20-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24930894

RESUMEN

Fermentation of xylose, a major constituent of lignocellulose, will be important for expanding sustainable biofuel production. We sought to better understand the effects of intrinsic (genotypic) and extrinsic (growth conditions) variables on optimal gene expression of the Scheffersomyces stipitis xylose utilization pathway in Saccharomyces cerevisiae by using a set of five promoters to simultaneously regulate each gene. Three-gene (xylose reductase, xylitol dehydrogenase (XDH), and xylulokinase) and eight-gene (expanded with non-oxidative pentose phosphate pathway enzymes and pyruvate kinase) promoter libraries were enriched under aerobic and anaerobic conditions or with a mutant XDH with altered cofactor usage. Through characterization of enriched strains, we observed (1) differences in promoter enrichment for the three-gene library depending on whether the pentose phosphate pathway genes were included during the aerobic enrichment; (2) the importance of selection conditions, where some aerobically-enriched strains underperform in anaerobic conditions compared to anaerobically-enriched strains; (3) improved growth rather than improved fermentation product yields for optimized strains carrying the mutant XDH compared to the wild-type XDH.


Asunto(s)
Técnicas Químicas Combinatorias/métodos , Perfilación de la Expresión Génica/métodos , Biblioteca de Genes , Mejoramiento Genético/métodos , Complejos Multienzimáticos/fisiología , Saccharomyces cerevisiae/fisiología , Xilosa/metabolismo , Proliferación Celular/fisiología , Ingeniería Metabólica/métodos , Regiones Promotoras Genéticas/genética
16.
Nature ; 454(7207): 1005-8, 2008 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-18650806

RESUMEN

Enterohaemorrhagic Escherichia coli attaches to the intestine through actin pedestals that are formed when the bacterium injects its protein EspF(U) (also known as TccP) into host cells. EspF(U) potently activates the host WASP (Wiskott-Aldrich syndrome protein) family of actin-nucleating factors, which are normally activated by the GTPase CDC42, among other signalling molecules. Apart from its amino-terminal type III secretion signal, EspF(U) consists of five-and-a-half 47-amino-acid repeats. Here we show that a 17-residue motif within this EspF(U) repeat is sufficient for interaction with N-WASP (also known as WASL). Unlike most pathogen proteins that interface with the cytoskeletal machinery, this motif does not mimic natural upstream activators: instead of mimicking an activated state of CDC42, EspF(U) mimics an autoinhibitory element found within N-WASP. Thus, EspF(U) activates N-WASP by competitively disrupting the autoinhibited state. By mimicking an internal regulatory element and not the natural activator, EspF(U) selectively activates only a precise subset of CDC42-activated processes. Although one repeat is able to stimulate actin polymerization, we show that multiple-repeat fragments have notably increased potency. The activities of these EspF(U) fragments correlate with their ability to coordinate activation of at least two N-WASP proteins. Thus, this pathogen has used a simple autoinhibitory fragment as a component to build a highly effective actin polymerization machine.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Escherichia coli Enterohemorrágica/metabolismo , Proteínas de Escherichia coli/metabolismo , Imitación Molecular , Actinas/química , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Escherichia coli Enterohemorrágica/patogenicidad , Proteínas de Escherichia coli/química , Péptidos y Proteínas de Señalización Intracelular , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Células 3T3 NIH , Estructura Terciaria de Proteína , Secuencias Repetitivas de Ácidos Nucleicos , Transducción de Señal/fisiología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/química , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo
17.
Nucleic Acids Res ; 40(4): 1879-89, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22021385

RESUMEN

Synthetic scaffolds that permit spatial and temporal organization of enzymes in living cells are a promising post-translational strategy for controlling the flow of information in both metabolic and signaling pathways. Here, we describe the use of plasmid DNA as a stable, robust and configurable scaffold for arranging biosynthetic enzymes in the cytoplasm of Escherichia coli. This involved conversion of individual enzymes into custom DNA-binding proteins by genetic fusion to zinc-finger domains that specifically bind unique DNA sequences. When expressed in cells that carried a rationally designed DNA scaffold comprising corresponding zinc finger binding sites, the titers of diverse metabolic products, including resveratrol, 1,2-propanediol and mevalonate were increased as a function of the scaffold architecture. These results highlight the utility of DNA scaffolds for assembling biosynthetic enzymes into functional metabolic structures. Beyond metabolism, we anticipate that DNA scaffolds may be useful in sequestering different types of enzymes for specifying the output of biological signaling pathways or for coordinating other assembly-line processes such as protein folding, degradation and post-translational modifications.


Asunto(s)
Vías Biosintéticas , ADN/química , Ingeniería Metabólica , Sitios de Unión , Biocatálisis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Enzimas/genética , Enzimas/metabolismo , Escherichia coli/metabolismo , Ácido Mevalónico/metabolismo , Plásmidos/genética , Propilenglicol/metabolismo , Resveratrol , Estilbenos/metabolismo , Dedos de Zinc
18.
bioRxiv ; 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38464036

RESUMEN

Long single-stranded DNA (ssDNA) is a versatile molecular reagent with applications including RNA-guided genome engineering and DNA nanotechnology, yet its production is typically resource-intensive. We introduce a novel method utilizing an engineered E. coli "helper" strain and phagemid system that simplifies long ssDNA generation to a straightforward transformation and purification procedure. Our method obviates the need for helper plasmids and their associated contamination by integrating M13mp18 genes directly into the E. coli chromosome. We achieved ssDNA lengths ranging from 504 to 20,724 nucleotides with titers up to 250 µg/L following alkaline-lysis purification. The efficacy of our system was confirmed through its application in primary T cell genome modifications and DNA origami folding. The reliability, scalability, and ease of our approach promises to unlock new experimental applications requiring large quantities of long ssDNA.

19.
Metab Eng ; 14(3): 242-51, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21946160

RESUMEN

As synthetic pathways built from exogenous enzymes become more complicated, the probability of encountering undesired interactions with host organisms increases, thereby lowering product titer. An emerging strategy to combat this problem is to spatially organize pathway enzymes into multi-protein complexes, where high local concentrations of enzymes and metabolites may enhance flux and limit problematic interactions with the cellular milieu. Co-localizing enzymes using synthetic scaffolds has improved titers for multiple pathways. While lacking physical diffusion barriers, scaffolded systems could concentrate intermediates locally through a mechanism analogous to naturally occurring microdomains. A more direct strategy for compartmentalizing pathway components would be to encapsulate them within protein shells. Several classes of shells have been loaded with exogenous proteins and expressed successfully in industrial hosts. A critical challenge for achieving ideal pathway compartmentalization with protein shells will likely be evolving pores to selectively limit intermediate diffusion. Eventually, these tools should enhance our ability to rationally design metabolic pathways.


Asunto(s)
Enzimas Inmovilizadas , Ingeniería Metabólica/métodos , Complejos Multienzimáticos , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/genética , Enzimas Inmovilizadas/metabolismo , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo
20.
Metab Eng ; 12(3): 298-305, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20117231

RESUMEN

The field of metabolic engineering has the potential to produce a wide variety of chemicals in both an inexpensive and ecologically-friendly manner. Heterologous expression of novel combinations of enzymes promises to provide new or improved synthetic routes towards a substantially increased diversity of small molecules. Recently, we constructed a synthetic pathway to produce d-glucaric acid, a molecule that has been deemed a "top-value added chemical" from biomass, starting from glucose. Limiting flux through the pathway is the second recombinant step, catalyzed by myo-inositol oxygenase (MIOX), whose activity is strongly influenced by the concentration of the myo-inositol substrate. To synthetically increase the effective concentration of myo-inositol, polypeptide scaffolds were built from protein-protein interaction domains to co-localize all three pathway enzymes in a designable complex as previously described (Dueber et al., 2009). Glucaric acid titer was found to be strongly affected by the number of scaffold interaction domains targeting upstream Ino1 enzymes, whereas the effect of increased numbers of MIOX-targeted domains was much less significant. We determined that the scaffolds directly increased the specific MIOX activity and that glucaric acid titers were strongly correlated with MIOX activity. Overall, we observed an approximately 5-fold improvement in product titers over the non-scaffolded control, and a 50% improvement over the previously reported highest titers. These results further validate the utility of these synthetic scaffolds as a tool for metabolic engineering.


Asunto(s)
Escherichia coli/metabolismo , Ácido Glucárico/metabolismo , Animales , Escherichia coli/enzimología , Escherichia coli/genética , Glucosa/genética , Glucosa/metabolismo , Inositol/genética , Inositol/metabolismo , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Dominios y Motivos de Interacción de Proteínas , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA