Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Microb Cell Fact ; 23(1): 173, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867236

RESUMEN

BACKGROUND: The microbial chiral product (R)-3-hydroxybutyrate (3-HB) is a gateway to several industrial and medical compounds. Acetyl-CoA is the key precursor for 3-HB, and several native pathways compete with 3-HB production. The principal competing pathway in wild-type Escherichia coli for acetyl-CoA is mediated by citrate synthase (coded by gltA), which directs over 60% of the acetyl-CoA into the tricarboxylic acid cycle. Eliminating citrate synthase activity (deletion of gltA) prevents growth on glucose as the sole carbon source. In this study, an alternative approach is used to generate an increased yield of 3-HB: citrate synthase activity is reduced but not eliminated by targeted substitutions in the chromosomally expressed enzyme. RESULTS: Five E. coli GltA variants were examined for 3-HB production via heterologous overexpression of a thiolase (phaA) and NADPH-dependent acetoacetyl-CoA reductase (phaB) from Cupriavidus necator. In shake flask studies, four variants showed nearly 5-fold greater 3-HB yield compared to the wild-type, although pyruvate accumulated. Overexpression of either native thioesterases TesB or YciA eliminated pyruvate formation, but diverted acetyl-CoA towards acetate formation. Overexpression of pantothenate kinase similarly decreased pyruvate formation but did not improve 3-HB yield. Controlled batch studies at the 1.25 L scale demonstrated that the GltA[A267T] variant produced the greatest 3-HB titer of 4.9 g/L with a yield of 0.17 g/g. In a phosphate-starved repeated batch process, E. coli ldhA poxB pta-ackA gltA::gltA[A267T] generated 15.9 g/L 3-HB (effective concentration of 21.3 g/L with dilution) with yield of 0.16 g/g from glucose as the sole carbon source. CONCLUSIONS: This study demonstrates that GltA variants offer a means to affect the generation of acetyl-CoA derived products. This approach should benefit a wide range of acetyl-CoA derived biochemical products in E. coli and other microbes. Enhancing substrate affinity of the introduced pathway genes like thiolase towards acetyl-CoA will likely further increase the flux towards 3-HB while reducing pyruvate and acetate accumulation.


Asunto(s)
Ácido 3-Hidroxibutírico , Acetilcoenzima A , Citrato (si)-Sintasa , Escherichia coli , Escherichia coli/genética , Escherichia coli/metabolismo , Acetilcoenzima A/metabolismo , Citrato (si)-Sintasa/metabolismo , Citrato (si)-Sintasa/genética , Ácido 3-Hidroxibutírico/metabolismo , Ácido 3-Hidroxibutírico/biosíntesis , Ingeniería Metabólica/métodos , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Cetona Oxidorreductasas/metabolismo , Cetona Oxidorreductasas/genética , Oxidorreductasas de Alcohol
2.
Eng Life Sci ; 23(3): e2200054, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36874610

RESUMEN

Several chromosomally expressed AceE variants were constructed in Escherichia coli ΔldhA ΔpoxB ΔppsA and compared using glucose as the sole carbon source. These variants were examined in shake flask cultures for growth rate, pyruvate accumulation, and acetoin production via heterologous expression of the budA and budB genes from Enterobacter cloacae ssp. dissolvens. The best acetoin-producing strains were subsequently studied in controlled batch culture at the one-liter scale. PDH variant strains attained up to four-fold greater acetoin than the strain expressing the wild-type PDH. In a repeated batch process, the H106V PDH variant strain attained over 43 g/L of pyruvate-derived products, acetoin (38.5 g/L) and 2R,3R-butanediol (5.0 g/L), corresponding to an effective concentration of 59 g/L considering the dilution. The acetoin yield from glucose was 0.29 g/g with a volumetric productivity of 0.9 g/L·h (0.34 g/g and 1.0 g/L·h total products). The results demonstrate a new tool in pathway engineering, the modification of a key metabolic enzyme to improve the formation of a product via a kinetically slow, introduced pathway. Direct modification of the pathway enzyme offers an alternative to promoter engineering in cases where the promoter is involved in a complex regulatory network.

3.
Microorganisms ; 10(11)2022 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-36363817

RESUMEN

Limiting an essential nutrient has a profound impact on microbial growth. The notion of growth under limited conditions was first described using simple Monod kinetics proposed in the 1940s. Different operational modes (chemostat, fed-batch processes) were soon developed to address questions related to microbial physiology and cell maintenance and to enhance product formation. With more recent developments of metabolic engineering and systems biology, as well as high-throughput approaches, the focus of current engineers and applied microbiologists has shifted from these fundamental biochemical processes. This review draws attention again to nutrient-limited processes. Indeed, the sophisticated gene editing tools not available to pioneers offer the prospect of metabolic engineering strategies which leverage nutrient limited processes. Thus, nutrient- limited processes continue to be very relevant to generate microbially derived biochemicals.

4.
Appl Environ Microbiol ; 88(4): e0186821, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-34910566

RESUMEN

Bacteria were isolated from wastewater and soil containing charred wood remnants based on their ability to use levoglucosan as a sole carbon source and on their levoglucosan dehydrogenase (LGDH) activity. On the basis of their 16S rRNA gene sequences, these bacteria represented the diverse genera Microbacterium, Paenibacillus, Shinella, and Klebsiella. Genomic sequencing of the isolates verified that two isolates represented novel species, Paenibacillus athensensis MEC069T and Shinella sumterensis MEC087T, while the remaining isolates were closely related to Microbacterium lacusdiani or Klebsiella pneumoniae. The genetic sequence of LGDH, lgdA, was found in the genomes of these four isolates as well as Pseudarthrobacter phenanthrenivorans Sphe3. The identity of the P. phenanthrenivorans LGDH was experimentally verified following recombinant expression in Escherichia coli. Comparison of the putative genes surrounding lgdA in the isolate genomes indicated that several other gene products facilitate the bacterial catabolism of levoglucosan, including a putative sugar isomerase and several transport proteins. IMPORTANCE Levoglucosan is the most prevalent soluble carbohydrate remaining after high-temperature pyrolysis of lignocellulosic biomass, but it is not fermented by typical production microbes such as Escherichia coli and Saccharomyces cerevisiae. A few fungi metabolize levoglucosan via the enzyme levoglucosan kinase, while several bacteria metabolize levoglucosan via levoglucosan dehydrogenase. This study describes the isolation and characterization of four bacterial species that degrade levoglucosan. Each isolate is shown to contain several genes within an operon involved in levoglucosan degradation, furthering our understanding of bacteria that metabolize levoglucosan.


Asunto(s)
Glucosa , Paenibacillus , Biomasa , Glucosa/análogos & derivados , Glucosa/metabolismo , Paenibacillus/genética , ARN Ribosómico 16S/genética
5.
Appl Environ Microbiol ; 87(13): e0048721, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33863707

RESUMEN

Altering metabolic flux at a key branch point in metabolism has commonly been accomplished through gene knockouts or by modulating gene expression. An alternative approach to direct metabolic flux preferentially toward a product is decreasing the activity of a key enzyme through protein engineering. In Escherichia coli, pyruvate can accumulate from glucose when carbon flux through the pyruvate dehydrogenase complex is suppressed. Based on this principle, 16 chromosomally expressed AceE variants were constructed in E. coli C and compared for growth rate and pyruvate accumulation using glucose as the sole carbon source. To prevent conversion of pyruvate to other products, the strains also contained deletions in two nonessential pathways: lactate dehydrogenase (ldhA) and pyruvate oxidase (poxB). The effect of deleting phosphoenolpyruvate synthase (ppsA) on pyruvate assimilation was also examined. The best pyruvate-accumulating strains were examined in controlled batch and continuous processes. In a nitrogen-limited chemostat process at steady-state growth rates of 0.15 to 0.28 h-1, an engineered strain expressing the AceE[H106V] variant accumulated pyruvate at a yield of 0.59 to 0.66 g pyruvate/g glucose with a specific productivity of 0.78 to 0.92 g pyruvate/g cells·h. These results provide proof of concept that pyruvate dehydrogenase complex variants can effectively shift carbon flux away from central carbon metabolism to allow pyruvate accumulation. This approach can potentially be applied to other key enzymes in metabolism to direct carbon toward a biochemical product. IMPORTANCE Microbial production of biochemicals from renewable resources has become an efficient and cost-effective alternative to traditional chemical synthesis methods. Metabolic engineering tools are important for optimizing a process to perform at an economically feasible level. This study describes an additional tool to modify central metabolism and direct metabolic flux to a product. We have shown that variants of the pyruvate dehydrogenase complex can direct metabolic flux away from cell growth to increase pyruvate production in Escherichia coli. This approach could be paired with existing strategies to optimize metabolism and create industrially relevant and economically feasible processes.


Asunto(s)
Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Complejo Piruvato Deshidrogenasa/genética , Complejo Piruvato Deshidrogenasa/metabolismo , Ácido Pirúvico/metabolismo , Acetilcoenzima A/metabolismo , Escherichia coli/genética , L-Lactato Deshidrogenasa/genética , Ingeniería Metabólica , Mutación , Fosfotransferasas (Aceptores Pareados)/genética , Piruvato Oxidasa/genética
6.
Biotechnol Bioeng ; 117(12): 3785-3798, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32716047

RESUMEN

To maximize the productivity of engineered metabolic pathway, in silico model is an established means to provide features of enzyme reaction dynamics. In our previous study, Escherichia coli engineered with acrylate pathway yielded low propionic acid titer. To understand the bottleneck behind this low productivity, a kinetic model was developed that incorporates the enzymatic reactions of the acrylate pathway. The resulting model was capable of simulating the fluxes reported under in vitro studies with good agreement, suggesting repression of propionyl-CoA transferase (Pct) by carboxylate metabolites as the main limiting factor for propionate production. Furthermore, the predicted flux control coefficients of the pathway enzymes under steady state conditions revealed that the control of flux is shared between Pct and lactoyl-CoA dehydratase. Increase in lactate concentration showed gradual decrease in flux control coefficients of Pct that in turn confirmed the control exerted by the carboxylate substrate. To interpret these in silico predictions under in vivo system, an organized study was conducted with a lactic acid bacteria strain engineered with acrylate pathway. Analysis reported a decreased product formation rate on attainment of inhibitory titer by suspected metabolites and supported the model.


Asunto(s)
Acrilatos/metabolismo , Simulación por Computador , Lactococcus lactis , Ingeniería Metabólica , Modelos Biológicos , Lactococcus lactis/genética , Lactococcus lactis/metabolismo
7.
Metab Eng ; 61: 171-180, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32569710

RESUMEN

Metabolic engineering is used to improve titers, yields and generation rates for biochemical products in host microbes such as Escherichia coli. A wide range of biochemicals are derived from the central carbon metabolite acetyl-CoA, and the largest native drain of acetyl-CoA in most microbes including E. coli is entry into the tricarboxylic acid (TCA) cycle via citrate synthase (coded by the gltA gene). Since the pathway to any biochemical derived from acetyl-CoA must ultimately compete with citrate synthase, a reduction in citrate synthase activity should facilitate the increased formation of products derived from acetyl-CoA. To test this hypothesis, we integrated into E. coli C ΔpoxB twenty-eight citrate synthase variants having specific point mutations that were anticipated to reduce citrate synthase activity. These variants were assessed in shake flasks for growth and the production of acetate, a model product derived from acetyl-CoA. Mutations in citrate synthase at residues W260, A267 and V361 resulted in the greatest acetate yields (approximately 0.24 g/g glucose) compared to the native citrate synthase (0.05 g/g). These variants were further examined in controlled batch and continuous processes. The results provide important insights on improving the production of compounds derived from acetyl-CoA.


Asunto(s)
Acetatos/metabolismo , Citrato (si)-Sintasa , Proteínas de Escherichia coli , Escherichia coli , Mutación Puntual , Ingeniería de Proteínas , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Citrato (si)-Sintasa/genética , Citrato (si)-Sintasa/metabolismo , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo
8.
Biotechnol Bioeng ; 117(9): 2781-2790, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32497258

RESUMEN

The microbial product citramalic acid (citramalate) serves as a five-carbon precursor for the chemical synthesis of methacrylic acid. This biochemical is synthesized in Escherichia coli directly by the condensation of pyruvate and acetyl-CoA via the enzyme citramalate synthase. The principal competing enzyme with citramalate synthase is citrate synthase, which mediates the condensation reaction of oxaloacetate and acetyl-CoA to form citrate and begin the tricarboxylic acid cycle. A deletion in the gltA gene coding citrate synthase prevents acetyl-CoA flux into the tricarboxylic acid cycle, and thus necessitates the addition of glutamate. In this study the E. coli citrate synthase was engineered to contain point mutations intended to reduce the enzyme's affinity for acetyl-CoA, but not eliminate its activity. Cell growth, enzyme activity and citramalate production were compared in several variants in shake flasks and controlled fermenters. Citrate synthase GltA[F383M] not only facilitated cell growth without the presence of glutamate, but also improved the citramalate production by 125% compared with the control strain containing the native citrate synthase in batch fermentation. An exponential feeding strategy was employed in a fed-batch process using MEC626/pZE12-cimA harboring the GltA[F383M] variant, which generated over 60 g/L citramalate with a yield of 0.53 g citramalate/g glucose in 132 hr. These results demonstrate protein engineering can be used as an effective tool to redirect carbon flux by reducing enzyme activity and improve the microbial production of traditional commodity chemicals.


Asunto(s)
Citrato (si)-Sintasa , Escherichia coli , Malatos/metabolismo , Ingeniería Metabólica/métodos , Técnicas de Cultivo Celular por Lotes , Vías Biosintéticas , Citrato (si)-Sintasa/genética , Citrato (si)-Sintasa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Metacrilatos/metabolismo , Mutación Puntual/genética
9.
Appl Biochem Biotechnol ; 192(1): 243-256, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32372381

RESUMEN

The biorefinery concept makes use of renewable lignocellulosic biomass to produce commodities sustainably. A synthetic microbial consortium can enable the simultaneous utilization of sugars such as glucose and xylose to produce biochemicals, where each consortium member converts one sugar into the target product. In this study, woody biomass was used to generate glucose and xylose after pretreatment with 20% (w/v) sulfuric acid and 60-min reaction time. We compared several strategies for detoxification with charcoal and sodium borohydride treatments to improve the fermentability of this hydrolysate in a defined medium for the production of the growth-associated product pyruvate. In shake flask culture, the highest pyruvate yield on xylose of 0.8 g/g was found using pH 6 charcoal-treated hydrolysate. In bioreactor studies, a consortium of two engineered E. coli strains converted the mixture of glucose and xylose in batch studies to 12.8 ± 2.7 g/L pyruvate in 13 h. These results demonstrate that lignocellulosic biomass as the sole carbon source can be used to produce growth-related products after employing suitable detoxification strategies.


Asunto(s)
Escherichia coli/metabolismo , Lignina/química , Ingeniería Metabólica , Piruvatos/metabolismo , Madera , Biomasa , Reactores Biológicos , Borohidruros/química , Fermentación , Glucosa/química , Concentración de Iones de Hidrógeno , Hidrólisis , Consorcios Microbianos , Microorganismos Modificados Genéticamente , Ácido Pirúvico/química , Saccharomyces cerevisiae/metabolismo , Ácidos Sulfúricos/química , Temperatura , Xilosa/química
10.
Appl Biochem Biotechnol ; 190(3): 918-930, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31605303

RESUMEN

The genes involved in the aerobic bacterial metabolism of furfural and 5-hydroxymethylfurfural (HMF) have been characterized in two species, Pseudomonas putida Fu1 and Cupriavidus basilensis HMF14. A third furan-metabolizing strain, Pseudomonas putida ALS1267, was recently identified that grows robustly on both furfural and HMF as sole carbon sources, with a growth rate of 0.250 h-1 on furfural and 0.311 h-1 on HMF, and we have characterized the genes involved in furfural and HMF metabolism in this bacterium. Unlike C. basilensis HMF14, which contains separate furfural and HMF operons, P. putida ALS1267 contains one contiguous 18.1-kb operon, which harbors all of the furfural- and HMF-metabolizing genes except for one, the hmfH gene that encodes HMF/furfural oxidoreductase and has both HMF acid oxidase and furfural/HMF dehydrogenase activity. The 18.1-kb operon was cloned into P. putida KT2440, which cannot metabolize furans, enabling growth on furfural as the sole carbon source with a growth rate of 0.340 h-1. The clone did not allow P. putida KT2440 to metabolize HMF, most likely due to the lack of the hmfH gene. No hmfH homolog was identified in P. putida ALS1267, suggesting that another gene in the ALS1267 genome provides this function.


Asunto(s)
Furaldehído/análogos & derivados , Furaldehído/metabolismo , Pseudomonas putida/metabolismo , Genes Bacterianos , Inactivación Metabólica , Redes y Vías Metabólicas , Operón , Pseudomonas putida/genética
11.
Biotechnol Biofuels ; 12: 91, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31044004

RESUMEN

BACKGROUND: Lignocellulosic biomass is an attractive, inexpensive source of potentially fermentable sugars. However, hydrolysis of lignocellulose results in a complex mixture containing microbial inhibitors at variable composition. A single microbial species is unable to detoxify or even tolerate these non-sugar components while converting the sugar mixtures effectively to a product of interest. Often multiple substrates are metabolized sequentially because of microbial regulatory mechanisms. To overcome these problems, we engineered strains of Acinetobacter baylyi ADP1 to comprise a consortium able to degrade benzoate and 4-hydroxybenzoate simultaneously under batch and continuous conditions in the presence of sugars. We furthermore used a thermotolerant yeast, Kluyveromyces marxianus, to convert the glucose remaining after detoxification to ethanol. RESULTS: The two engineered strains, one unable to metabolize benzoate and another unable to metabolize 4-hydroxybenzoate, when grown together removed these two inhibitors simultaneously under batch conditions. Under continuous conditions, a single strain with a deletion in the gcd gene metabolized both inhibitors in the presence of sugars. After this batch detoxification using ADP1-derived mutants, K. marxianus generated 36.6 g/L ethanol. CONCLUSIONS: We demonstrated approaches for the simultaneous removal of two aromatic inhibitors from a simulated lignocellulosic hydrolysate. A two-stage batch process converted the residual sugar into a non-growth-associated product, ethanol. Such a two-stage process with bacteria (A. baylyi) and yeast (K. marxianus) is advantageous, because the yeast fermentation occurs at a higher temperature which prevents growth and ethanol consumption of A. baylyi. Conceptually, the process can be extended to other inhibitors or sugars found in real hydrolysates. That is, additional strains which degrade components of lignocellulosic hydrolysates could be made substrate-selective and targeted for use with specific complex mixtures found in a hydrolysate.

12.
Eng Life Sci ; 19(11): 770-780, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32624970

RESUMEN

Acetate formation is a disadvantage in the use of Escherichia coli for recombinant protein production, and many studies have focused on optimizing fermentation processes or altering metabolism to eliminate acetate accumulation. In this study, E. coli MEC697 (MG1655 nadR nudC mazG) maintained a larger pool of NAD(H) compared to the wild-type control, and also accumulated lower concentrations of acetate when grown in batch culture on glucose. In steady-state cultures, the elevated total NAD(H) found in MEC697 delayed the threshold dilution rate for acetate formation to a growth rate of 0.27 h-1. Batch and fed-batch processes using MEC697 were examined for the production of ß-galactosidase as a model recombinant protein. Fed-batch culture of MEC697/pTrc99A-lacZ compared to MG1655/pTrc99A-lacZ at a growth rate of 0.22 h-1 showed only a modest increase of protein formation. However, 1 L batch growth of MEC697/pTrc99A-lacZ resulted in 50% lower acetate formation compared to MG1655/pTrc99A-lacZ and a two-fold increase in recombinant protein production.

13.
J Ind Microbiol Biotechnol ; 45(11): 939-950, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30159648

RESUMEN

The NAD+/NADH ratio and the total NAD(H) play important roles for whole-cell biochemical redox transformations. After the carbon source is exhausted, the degradation of NAD(H) could contribute to a decline in the rate of a desired conversion. In this study, methods to slow the native rate of NAD(H) degradation were examined using whole-cell Escherichia coli with two model oxidative NAD+-dependent biotransformations. A high phosphate concentration (50 mM) was observed to slow NAD(H) degradation. We also constructed E. coli strains with deletions in genes coding several enzymes involved in NAD+ degradation. In shake-flask experiments, the total NAD(H) concentration positively correlated with conversion of xylitol to L-xylulose by xylitol 4-dehydrogenase, and the greatest conversion (80%) was observed using MG1655 nadR nudC mazG/pZE12-xdh/pCS27-nox. Controlled 1-L batch processes comparing E. coli nadR nudC mazG with a wild-type background strain demonstrated a 30% increase in final L-xylulose concentration (5.6 vs. 7.9 g/L) and a 25% increase in conversion (0.53 vs. 0.66 g/g). MG1655 nadR nudC mazG was also examined for the conversion of galactitol to L-tagatose by galactitol 2-dehydrogenase. A batch process using 15 g/L glycerol and 10 g/L galactitol generated over 9.4 g/L L-tagatose, corresponding to 90% conversion and a yield of 0.95 g L-tagatose/g galactitol consumed. The results demonstrate the value of minimizing NAD(H) degradation as a means to improve NAD+-dependent biotransformations.


Asunto(s)
D-Xilulosa Reductasa/genética , Escherichia coli/metabolismo , NAD/metabolismo , Fermentación , Glicerol/metabolismo , Microbiología Industrial , Cinética , Oxidación-Reducción , Fosforilación Oxidativa , Xilitol/metabolismo , Xilulosa/metabolismo
14.
Proc Natl Acad Sci U S A ; 115(27): 7105-7110, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29915086

RESUMEN

Experimental evolution is a critical tool in many disciplines, including metabolic engineering and synthetic biology. However, current methods rely on the chance occurrence of a key step that can dramatically accelerate evolution in natural systems, namely increased gene dosage. Our studies sought to induce the targeted amplification of chromosomal segments to facilitate rapid evolution. Since increased gene dosage confers novel phenotypes and genetic redundancy, we developed a method, Evolution by Amplification and Synthetic Biology (EASy), to create tandem arrays of chromosomal regions. In Acinetobacter baylyi, EASy was demonstrated on an important bioenergy problem, the catabolism of lignin-derived aromatic compounds. The initial focus on guaiacol (2-methoxyphenol), a common lignin degradation product, led to the discovery of Amycolatopsis genes (gcoAB) encoding a cytochrome P450 enzyme that converts guaiacol to catechol. However, chromosomal integration of gcoAB in Pseudomonas putida or A. baylyi did not enable guaiacol to be used as the sole carbon source despite catechol being a growth substrate. In ∼1,000 generations, EASy yielded alleles that in single chromosomal copy confer growth on guaiacol. Different variants emerged, including fusions between GcoA and CatA (catechol 1,2-dioxygenase). This study illustrates the power of harnessing chromosomal gene amplification to accelerate the evolution of desirable traits.


Asunto(s)
Proteínas Bacterianas/genética , Cromosomas Bacterianos/genética , Evolución Molecular , Dosificación de Gen , Genes Bacterianos , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/enzimología
15.
Microbiology (Reading) ; 164(2): 163-172, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29393018

RESUMEN

Escherichia coli Δglk ΔmanZ ΔptsG glucose- strains that lack the glucose phosphotransferase system (PTS) and the mannose PTS as well as glucokinase have been widely used by researchers studying the PTS. In this study we show that both fast- and slow-growing spontaneous glucose+ revertants can be readily obtained from Δglk ΔmanZ ΔptsG glucose- strains. All of the fast-growing revertants either altered the N-acetylglucosamine PTS or caused its overproduction by inactivating the NagC repressor protein, which regulates the N-acetylglucosamine PTS, and these revertants could utilize either glucose or N-acetylglucosamine as a sole carbon source. When a ΔnagE deletion, which abolishes the N-acetylglucosamine PTS, was introduced into the Δglk ΔmanZ ΔptsG glucose- strains, fast-growing revertants could no longer be isolated. Based on our results and other studies, it is clear that the N-acetylglucosamine PTS is the most easily adaptable PTS for transporting and phosphorylating glucose, other than the glucose PTS and mannose PTS, which are the primary glucose transport systems. While the slow-growing glucose+revertants were not characterized, they were likely mutations that other researchers have observed before and affect other PTSs or sugar kinases.


Asunto(s)
Acetilglucosamina/metabolismo , Proteínas de Escherichia coli/genética , Escherichia coli/metabolismo , Glucosa/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Transporte Biológico/genética , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/metabolismo , Eliminación de Gen , Expresión Génica , Genes Bacterianos/genética , Glucoquinasa/genética , Manosa/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Proteínas Represoras/genética , Especificidad por Sustrato
16.
Eng Life Sci ; 18(1): 40-47, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32624859

RESUMEN

Two strains of Escherichia coli were engineered to accumulate pyruvic acid from two sugars found in lignocellulosic hydrolysates by knockouts in the aceE, ppsA, poxB, and ldhA genes. Additionally, since glucose and xylose are typically consumed sequentially due to carbon catabolite repression in E. coli, one strain (MEC590) was engineered to grow only on glucose while a second strain (MEC589) grew only on xylose. On a single substrate, each strain generated pyruvate at a yield of about 0.60 g/g in both continuous culture and batch culture. In a glucose-xylose mixture under continuous culture, a consortium of both strains maintained a pyruvate yield greater than 0.60 g/g when three different concentrations of glucose and xylose were sequentially fed into the system. In a fed-batch process, both sugars in a glucose-xylose mixture were consumed simultaneously to accumulate 39 g/L pyruvate in less than 24 h at a yield of 0.59 g/g.

17.
Enzyme Microb Technol ; 106: 106-113, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28859803

RESUMEN

Escherichia coli expressing NAD-dependent xylitol-4-dehydrogenase (XDH) from Pantoea ananatis and growing on glucose or glycerol converts xylitol to the rare sugar l-xylulose. Although blocking potential l-xylulose consumption (l-xylulosekinase, lyxK) or co-expression of the glycerol facilitator (glpF) did not significantly affect l-xylulose formation, co-expressing XDH with water-forming NADH oxidase (NOX) from Streptococcus pneumoniae increased l-xylulose formation in shake flasks when glycerol was the carbon source. Controlled batch processes at the 1L scale demonstrated that the final equilibrium l-xylulose/xylitol ratio was correlated to the intracellular NAD+/NADH ratio, with 69% conversion of xylitol to l-xylulose and a yield of 0.88g l-xylulose/g xylitol consumed attained for MG1655/pZE12-xdh/pCS27-nox growing on glycerol. NADH oxidase was less effective at improving l-xylulose formation in the bioreactor than in shake flasks, likely as a result of an intrinsic maximum NAD+/NADH and l-xylulose/xylitol equilibrium ratio being attained. Intermittently feeding carbon source was ineffective at increasing the final l-xylulose concentration because introduction of carbon source was accompanied by a reduction in NAD+/NADH ratio. A batch process using 12g/L glycerol and 22g/L xylitol generated over 14g/L l-xylulose after 80h, corresponding to 65% conversion and a yield of 0.89g l-xylulose/g xylitol consumed.


Asunto(s)
D-Xilulosa Reductasa/metabolismo , Escherichia coli/metabolismo , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Xilulosa/biosíntesis , Acuaporinas/genética , Acuaporinas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Reactores Biológicos/microbiología , D-Xilulosa Reductasa/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fermentación , Glucosa/metabolismo , Glicerol/metabolismo , Complejos Multienzimáticos/genética , NADH NADPH Oxidorreductasas/genética , Pantoea/enzimología , Pantoea/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptococcus pneumoniae/enzimología , Streptococcus pneumoniae/genética , Xilitol/metabolismo
18.
J Ind Microbiol Biotechnol ; 44(10): 1483-1490, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28744578

RESUMEN

Citramalic acid (citramalate) serves as a five-carbon precursor for the chemical synthesis of methacrylic acid. We compared citramalate and acetate accumulation from glycerol using Escherichia coli strains expressing a modified citramalate synthase gene cimA from Methanococcus jannaschii. These studies revealed that gltA coding citrate synthase, leuC coding 3-isopropylmalate dehydratase, and acetate pathway genes play important roles in elevating citramalate and minimizing acetate formation. Controlled 1.0 L batch experiments confirmed that deletions in all three acetate-production genes (poxB, ackA, and pta) were necessary to reduce acetate formation to less than 1 g/L during citramalate production from 30 g/L glycerol. Fed-batch processes using MEC568/pZE12-cimA (gltA leuC ackA-pta poxB) generated over 31 g/L citramalate and less than 2 g/L acetate from either purified or crude glycerol at yields exceeding 0.50 g citramalate/g glycerol in 132 h. These results hold promise for the viable formation of citramalate from unrefined glycerol.


Asunto(s)
Escherichia coli/genética , Escherichia coli/metabolismo , Glicerol/metabolismo , Malatos/metabolismo , Ingeniería Metabólica , Acetatos/metabolismo , Acetiltransferasas/metabolismo , Técnicas de Cultivo Celular por Lotes , Biocombustibles , Hidroliasas/genética , Hidroliasas/metabolismo , Methanocaldococcus/enzimología , Methanocaldococcus/genética
19.
Microb Cell Fact ; 16(1): 114, 2017 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-28637476

RESUMEN

BACKGROUND: Citramalate, a chemical precursor to the industrially important methacrylic acid (MAA), can be synthesized using Escherichia coli overexpressing citramalate synthase (cimA gene). Deletion of gltA encoding citrate synthase and leuC encoding 3-isopropylmalate dehydratase were critical to achieving high citramalate yields. Acetate is an undesirable by-product potentially formed from pyruvate and acetyl-CoA, the precursors of citramalate during aerobic growth of E. coli. This study investigated strategies to minimize acetate and maximize citramalate production in E. coli mutants expressing the cimA gene. RESULTS: Key knockouts that minimized acetate formation included acetate kinase (ackA), phosphotransacetylase (pta), and in particular pyruvate oxidase (poxB). Deletion of glucose 6-phosphate dehydrogenase (zwf) and ATP synthase (atpFH) aimed at improving glycolytic flux negatively impacted cell growth and citramalate accumulation in shake flasks. In a repetitive fed-batch process, E. coli gltA leuC ackA-pta poxB overexpressing cimA generated 54.1 g/L citramalate with a yield of 0.64 g/g glucose (78% of theoretical maximum yield), and only 1.4 g/L acetate in 87 h. CONCLUSIONS: This study identified the gene deletions critical to reducing acetate accumulation during aerobic growth and citramalate production in metabolically engineered E. coli strains. The citramalate yield and final titer relative to acetate at the end of the fed-batch process are the highest reported to date (a mass ratio of citramalate to acetate of nearly 40) without being detrimental to citramalate productivity, significantly improving a potential process for the production of this five-carbon chemical.


Asunto(s)
Acetatos/metabolismo , Escherichia coli/metabolismo , Malatos/metabolismo , Ingeniería Metabólica , Acetilcoenzima A/metabolismo , Aerobiosis , Técnicas de Cultivo Celular por Lotes , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Eliminación de Gen , Genes Bacterianos , Mutación , Ácido Pirúvico/metabolismo
20.
Microbiology (Reading) ; 163(6): 866-877, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28640743

RESUMEN

Escherichia coli lacking the glucose phosphotransferase system (PTS), mannose PTS and glucokinase are supposedly unable to grow on glucose as the sole carbon source (Curtis SJ, Epstein W. J Bacteriol 1975;122:1189-1199). We report that W ptsG manZ glk (ALS1406) grows slowly on glucose in media containing glucose with a second carbon source: ALS1406 metabolizes glucose after that other carbon source, including arabinose, fructose, glycerol, succinate or xylose, is exhausted. Galactose is an exception to this rule, as ALS1406 simultaneously consumes both galactose and glucose. The ability of ALS1406 to metabolize glucose in a xylose-glucose mixture was unchanged by an additional knockout in any single gene involved in carbohydrate transport and utilization, including agp (periplasmic glucose-1-phosphatase), galP (galactose permease), xylA (xylose isomerase), alsK (allose kinase), crr (glucose PTS enzyme IIA), galK (galactose kinase), mak (mannokinase), malE (maltose transporter), malX (maltose PTS enzyme IIBC), mglB (methyl-galactose transporter subunit), nagE (N-acetyl glucosamine PTS enzyme IICBA), nanK (N-acetyl mannosamine kinase) or pgm (phosphoglucose mutase). Glucose metabolism was only blocked by the deletion of two metabolic genes, pgi (phosphoglucose isomerase) and zwf (glucose-6-phosphate 1-dehydrogenase), which prevents the entry of glucose-6-phosphate into the pentose phosphate and Embden-Meyerhof-Parnas pathways. Carbon-limited steady-state studies demonstrated that xylose must be sub-saturating for glucose to be metabolized, while nitrogen-limited studies showed that xylose is partly converted to glucose when xylose is in excess. Under transient conditions, ALS1406 converts almost 25 % (mass) xylose into glucose as a result of reversible transketolase and transaldolase and the re-entry of carbon into the pentose phosphate pathway via glucose-6-phosphate 1-dehydrogenase.


Asunto(s)
Proteínas de Escherichia coli/genética , Escherichia coli/metabolismo , Glucosa/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Arabinosa/metabolismo , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fermentación , Fructosa/metabolismo , Glucólisis , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Mutación , Vía de Pentosa Fosfato , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Ácido Succínico/metabolismo , Xilosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...