Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Crit Rev Biotechnol ; 41(5): 649-668, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33563072

ABSTRACT

The lignocellulosic sugar d-xylose has recently gained prominence as an inexpensive alternative substrate for the production of value-added compounds using genetically modified organisms. Among the prokaryotes, Escherichia coli has become the de facto host for the development of engineered microbial cell factories. The favored status of E. coli resulted from a century of scientific explorations leading to a deep understanding of its systems. However, there are limited literature reviews that discuss engineered E. coli as a platform for the conversion of d-xylose to any target compounds. Additionally, available critical review articles tend to focus on products rather than the host itself. This review aims to provide relevant and current information about significant advances in the metabolic engineering of d-xylose metabolism in E. coli. This focusses on unconventional and synthetic d-xylose metabolic pathways as several review articles have already discussed the engineering of native d-xylose metabolism. This paper, in particular, is essential to those who are working on engineering of d-xylose metabolism using E. coli as the host.


Subject(s)
Escherichia coli , Xylose , Escherichia coli/genetics , Metabolic Engineering , Metabolic Networks and Pathways/genetics
2.
Appl Microbiol Biotechnol ; 105(13): 5309-5324, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34215905

ABSTRACT

The xylose oxidative pathway (XOP) has been engineered in microorganisms for the production of a wide range of industrially relevant compounds. However, the performance of metabolically engineered XOP-utilizing microorganisms is typically hindered by D-xylonic acid accumulation. It acidifies the media and perturbs cell growth due to toxicity, thus curtailing enzymatic activity and target product formation. Fortunately, from the growing portfolio of genetic tools, several strategies that can be adapted for the generation of efficient microbial cell factories have been implemented to address D-xylonic acid accumulation. This review centers its discussion on the causes of D-xylonic acid accumulation and how to address it through different engineering and synthetic biology techniques with emphasis given on bacterial strains. In the first part of this review, the ability of certain microorganisms to produce and tolerate D-xylonic acid is also tackled as an important aspect in developing efficient microbial cell factories. Overall, this review could shed some insights and clarity to those working on XOP in bacteria and its engineering for the development of industrially applicable product-specialist strains. KEY POINTS: D-Xylonic acid accumulation is attributed to the overexpression of xylose dehydrogenase concomitant with basal or inefficient expression of enzymes involved in D-xylonic acid assimilation. Redox imbalance and insufficient cofactors contribute to D-xylonic acid accumulation. Overcoming D-xylonic acid accumulation can increase product formation among engineered strains. Engineering strategies involving enzyme engineering, evolutionary engineering, coutilization of different sugar substrates, and synergy of different pathways could potentially address D-xylonic acid accumulation.


Subject(s)
Metabolic Engineering , Xylose , Bacteria/genetics , Culture Media , Xylose/analogs & derivatives
3.
Bioprocess Biosyst Eng ; 44(6): 1081-1091, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33527231

ABSTRACT

Microbial biorefinery is a promising route toward sustainable production of glycolic acid (GA), a valuable raw material for various industries. However, inherent microbial GA production has limited substrate consumption using either D-xylose or D-glucose as carbon catabolite repression (CCR) averts their co-utilization. To bypass CCR, a GA-producing strain using D-xylose via Dahms pathway was engineered to allow cellobiose uptake. Unlike glucose, cellobiose was assimilated and intracellularly degraded without repressing D-xylose uptake. The final GA-producing E. coli strain (CLGA8) has an overexpressed cellobiose phosphorylase (cep94A) from Saccharophagus degradans 2-40 and an activated glyoxylate shunt pathway. Expression of cep94A improved GA production reaching the maximum theoretical yield (0.51 g GA g-1 xylose), whereas activation of glyoxylate shunt pathway enabled GA production from cellobiose, which further increased the GA titer (2.25 g GA L-1). To date, this is the highest reported GA yield from D-xylose through Dahms pathway in an engineered E. coli with cellobiose as co-substrate.


Subject(s)
Cellobiose/metabolism , Escherichia coli , Glycolates/metabolism , Metabolic Engineering , Microorganisms, Genetically-Modified , Xylose/metabolism , Cellobiose/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/metabolism , Xylose/genetics
4.
Appl Microbiol Biotechnol ; 104(5): 2097-2108, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31900554

ABSTRACT

The xylose oxidative pathway (XOP) is continuously gaining prominence as an alternative for the traditional pentose assimilative pathways in prokaryotes. It begins with the oxidation of D-xylose to D-xylonic acid, which is further converted to α-ketoglutarate or pyruvate + glycolaldehyde through a series of enzyme reactions. The persistent drawback of XOP is the accumulation of D-xylonic acid intermediate that causes culture media acidification. This study addresses this issue through the development of a novel pH-responsive synthetic genetic controller that uses a modified transmembrane transcription factor called CadCΔ. This genetic circuit was tested for its ability to detect extracellular pH and to control the buildup of D-xylonic acid in the culture media. Results showed that the pH-responsive genetic sensor confers dynamic regulation of D-xylonic acid accumulation, which adjusts with the perturbation of culture media pH. This is the first report demonstrating the use of a pH-responsive transmembrane transcription factor as a transducer in a synthetic genetic circuit that was designed for XOP. This may serve as a benchmark for the development of other genetic controllers for similar pathways that involve acidic intermediates.


Subject(s)
Culture Media/chemistry , Escherichia coli/metabolism , Xylose/analogs & derivatives , Xylose/metabolism , Culture Media/metabolism , Escherichia coli/genetics , Hydrogen-Ion Concentration , Oxidation-Reduction
5.
Appl Microbiol Biotechnol ; 104(5): 2273-2274, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31950218

ABSTRACT

In the published version, the y-axis data of Fig. 3c was incorrectly inserted (OD600 instead of D-xylonate (g L-1) and the x-axes of Figs. 3b, 3d, 3e and 3f ended at 48 h instead of 72 h. See the correct Fig. 3 below.

6.
Biotechnol Lett ; 42(11): 2231-2238, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32519168

ABSTRACT

OBJECTIVE: To identify and characterize a new ß-agarase from Cellulophaga omnivescoria W5C capable of producing biologically-active neoagarooligosaccharides from agar. RESULTS: The ß-agarase, Aga1, has signal peptides on both N- and C-terminals, which are involved in the type IX secretion system. It shares 75% protein sequence identity with AgaD from Zobellia galactanivorans and has a molecular weight of 54 kDa. Biochemical characterization reveals optimum agarolytic activities at pH 7-8 and temperature 30-45 °C. Aga1 retains at least 33% activity at temperatures lower than the sol-gel transition state of agarose. Metal ions are generally not essential, but calcium and potassium enhance its activity whereas iron and zinc are inhibitory. Finally, hydrolysis of agarose with Aga1 yields neoagarotetraose, neoagarohexaose, and neoagarooctaose. CONCLUSIONS: Aga1 displays unique traits such as moderate psychrophilicity, stability, and synergy with other agarases, which makes it an excellent candidate for biosynthetic production of neoagarooligosaccharides from agar.


Subject(s)
Flavobacteriaceae/enzymology , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Sequence Analysis, DNA/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cloning, Molecular , Flavobacteriaceae/genetics , Gene Expression , Glycoside Hydrolases/chemistry , Hot Temperature , Hydrogen-Ion Concentration , Hydrolysis , Molecular Weight , Protein Sorting Signals , Sepharose/chemistry
7.
Appl Microbiol Biotechnol ; 103(19): 8063-8074, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31482281

ABSTRACT

The capability of Escherichia coli to catabolize D-xylonate is a crucial component for building and optimizing the Dahms pathway. It relies on the inherent dehydratase and keto-acid aldolase activities of E. coli. Although the biochemical characteristics of these enzymes are known, their inherent expression regulation remains unclear. This knowledge is vital for the optimization of D-xylonate assimilation, especially in addressing the problem of D-xylonate accumulation, which hampers both cell growth and target product formation. In this report, molecular biology techniques and synthetic biology tools were combined to build a simple genetic switch controller for D-xylonate. First, quantitative and relative expression analysis of the gene clusters involved in D-xylonate catabolism were performed, revealing two D-xylonate-inducible operons, yagEF and yjhIHG. The 5'-flanking DNA sequence of these operons were then subjected to reporter gene assays which showed PyjhI to have low background activity and wide response range to D-xylonate. A PyjhI-driven synthetic genetic switch was then constructed containing feedback control to autoregulate D-xylonate accumulation and to activate the expression of the genes for 1,2,4-butanetriol (BTO) production. The genetic switch effectively reduced D-xylonate accumulation, which led to 31% BTO molar yield, the highest for direct microbial fermentation systems thus far. This genetic switch can be further modified and employed in the production of other compounds from D-xylose through the xylose oxidative pathway.


Subject(s)
Butanols/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/drug effects , Metabolic Engineering/methods , Promoter Regions, Genetic/drug effects , Xylose/analogs & derivatives , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Artificial Gene Fusion , Gene Expression Profiling , Genes, Reporter , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Xylose/metabolism
8.
J Ind Microbiol Biotechnol ; 46(2): 159-169, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30554290

ABSTRACT

The non-conventional D-xylose metabolism called the Dahms pathway which only requires the expression of at least three enzymes to produce pyruvate and glycolaldehyde has been previously engineered in Escherichia coli. Strains that rely on this pathway exhibit lower growth rates which were initially attributed to the perturbed redox homeostasis as evidenced by the lower intracellular NADPH concentrations during exponential growth phase. NADPH-regenerating systems were then tested to restore the redox homeostasis. The membrane-bound pyridine nucleotide transhydrogenase, PntAB, was overexpressed and resulted to a significant increase in biomass and glycolic acid titer and yield. Furthermore, expression of PntAB in an optimized glycolic acid-producing strain improved the growth and product titer significantly. This work demonstrated that compensating for the NADPH demand can be achieved by overexpression of PntAB in E. coli strains assimilating D-xylose through the Dahms pathway. Consequently, increase in biomass accumulation and product concentration was also observed.


Subject(s)
Escherichia coli/metabolism , Glycolates/metabolism , NADP Transhydrogenases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , NADP/genetics , NADP/metabolism , NADP Transhydrogenases/genetics , Xylose/metabolism
9.
Appl Microbiol Biotechnol ; 102(18): 7703-7716, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30003296

ABSTRACT

The D-xylose oxidative pathway (XOP) has recently been employed in several recombinant microorganisms for growth or for the production of several valuable compounds. The XOP is initiated by D-xylose oxidation to D-xylonolactone, which is then hydrolyzed into D-xylonic acid. D-Xylonic acid is then dehydrated to form 2-keto-3-deoxy-D-xylonic acid, which may be further dehydrated then oxidized into α-ketoglutarate or undergo aldol cleavage to form pyruvate and glycolaldehyde. This review introduces a brief discussion about XOP and its discovery in bacteria and archaea, such as Caulobacter crescentus and Haloferax volcanii. Furthermore, the current advances in the metabolic engineering of recombinant strains employing the XOP are discussed. This includes utilization of XOP for the production of diols, triols, and short-chain organic acids in Escherichia coli, Saccharomyces cerevisiae, and Corynebacterium glutamicum. Improving the D-xylose uptake, growth yields, and product titer through several metabolic engineering techniques bring some of these recombinant strains close to industrial viability. However, more developments are still needed to optimize the XOP pathway in the host strains, particularly in the minimization of by-product formation.


Subject(s)
Archaea/metabolism , Bacteria/metabolism , Metabolic Engineering , Recombination, Genetic , Xylose/metabolism , Yeasts/metabolism , Archaea/genetics , Bacteria/genetics , Oxidation-Reduction , Yeasts/genetics
10.
Appl Microbiol Biotechnol ; 102(5): 2179-2189, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29392388

ABSTRACT

Glycolic acid (GA) is an ⍺-hydroxy acid used in cosmetics, packaging, and medical industries due to its excellent properties, especially in its polymeric form. In this study, Escherichia coli was engineered to produce GA from D-xylose by linking the Dahms pathway, the glyoxylate bypass, and the partial reverse glyoxylate pathway (RGP). Initially, a GA-producing strain was constructed by disrupting the xylAB and glcD genes in the E. coli genome and overexpressing the xdh(Cc) from Caulobacter crescentus. This strain was further improved through modular optimization of the Dahms pathway and the glyoxylate bypass. Results for module 1 showed that the rate-limiting step of the Dahms pathway was the xylonate dehydratase reaction, and the overexpression of yagF was sufficient to overcome this bottleneck. Furthermore, the appropriate aldolase gene for module 1 was proven to be yagE. The results also show that overexpression of the lactaldehyde dehydrogenase gene, aldA, is needed to increase the GA production while the overexpression of glyoxylate reductase gene, ycdW, was only essential when the glyoxylate bypass was active. On the other hand, the module 2 enzymes AceA and AceK were vital in activating the glyoxylate bypass, while the RGP enzymes were dispensable. The final strain (GA19) produced 4.57 g/L GA with a yield of 0.46 g/g from D-xylose. So far, this is the highest value achieved for GA production in engineered E. coli through the Dahms pathway.


Subject(s)
Escherichia coli/metabolism , Glycolates/metabolism , Glyoxylates/metabolism , Metabolic Engineering , Xylose/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism
11.
Curr Microbiol ; 75(7): 925-933, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29536113

ABSTRACT

The continued research in the isolation of novel bacterial strains is inspired by the fact that native microorganisms possess certain desired phenotypes necessary for recombinant microorganisms in the biotech industry. Most studies have focused on the isolation and characterization of strains from marine ecosystems as they present a higher microbial diversity than other sources. In this study, a marine bacterium, W5C, was isolated from red seaweed collected from Yeosu, South Korea. The isolate can utilize several natural polysaccharides such as agar, alginate, carrageenan, and chitin. Genome sequence and comparative genomics analyses suggest that strain W5C belongs to a novel species of the Cellulophaga genus, from which the name Cellulophaga omnivescoria sp. nov. is proposed. Its genome harbors 3,083 coding sequences and 146 carbohydrate-active enzymes (CAZymes). Compared to other reported Cellulophaga species, the genome of W5C contained a higher proportion of CAZymes (4.7%). Polysaccharide utilization loci (PUL) for agar, alginate, and carrageenan were identified in the genome, along with other several putative PULs. These PULs are excellent sources for discovering novel hydrolytic enzymes and pathways with unique characteristics required for biorefinery applications, particularly in the utilization of marine renewable biomass. The type strain is JCM 32108T (= KCTC 13157BPT).


Subject(s)
Flavobacteriaceae/metabolism , Genome, Bacterial , Polysaccharides/metabolism , Seawater/microbiology , Sepharose/metabolism , Biodegradation, Environmental , Flavobacteriaceae/classification , Flavobacteriaceae/genetics , Flavobacteriaceae/isolation & purification , Phylogeny , Republic of Korea , Seawater/chemistry
12.
Article in English | MEDLINE | ID: mdl-29035626

ABSTRACT

The feasibility of open-pore polyurethane (PU) foam as packing material for wet chemical scrubber was tested for NH3 and H2S removals. The foam is inexpensive, light-weight, highly porous (low pressure drop) and provides large surface area per unit volume, which are desirable properties for enhanced gas/liquid mass transfer. Conventional HCl/HOCl (for NH3) and NaOH/NaOCl (for H2S) scrubbing solutions were used to absorb and oxidize the gases. Assessment of the wet chemical scrubbers reveals that pH and ORP levels are important to maintain the gas removal efficiencies >95%. A higher re-circulation rate of scrubbing solutions also proved to enhance the performance of the NH3 and H2S columns. Accumulation of salts was confirmed by the gradual increase in total dissolved solids and conductivity values of scrubbing solutions. The critical elimination capacities at >95% gas removals were found to be 5.24 g NH3-N/m3-h and 17.2 g H2S-S/m3-h at an empty bed gas residence time of 23.6 s. Negligible pressure drops (< 4 mm H2O) after continuous operation demonstrate the suitability of PU as a practical packing material in wet chemical scrubbers for NH3 and H2S removals from high-volume dilute emissions.


Subject(s)
Air Pollutants/chemistry , Air Pollution/prevention & control , Ammonia/chemistry , Gases/chemistry , Hydrogen Sulfide/chemistry , Polyurethanes/metabolism , Adsorption , Air Pollutants/metabolism , Ammonia/metabolism , Filtration , Gases/metabolism , Humans , Hydrogen Sulfide/metabolism , Odorants/prevention & control , Polyurethanes/chemistry
13.
Tumour Biol ; 37(4): 4323-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26493999

ABSTRACT

Pancreatic cancer is one of the most lethal cancers and remains a major unsolved health problem. Less than 20 % of patients are surgical candidates, and the median survival for non-resected patients is approximately 3 to 4 months. Despite the existence of many conventional cancer therapies, few targeted therapies have been developed for pancreatic cancer. Combination therapy using erlotinib and gemcitabine is an approved standard chemotherapy for advanced pancreatic cancer, but it has marginal therapeutic benefit. To try to improve the therapeutic outlook, we studied the efficacy of another combination treatment and the relevance to E-cadherin in human pancreatic cancer cells. We treated two human pancreatic cancer cell lines with the histone deacetylase inhibitor (HDACi) SAHA. Interestingly, in these Panc-1 and Capan1 cells, we observed that the expression levels of E-cadherin and phosphorylated EGFR were gradually upregulated after treatment with SAHA. Furthermore, these cells underwent induced cell death after exposure to the combination treatment of SAHA and erlotinib. In Panc-1 cells, overexpression of E-cadherin activated the phosphorylation of EGFR and increased the cell sensitivity to erlotinib. In Capan1 cells, knocking down E-cadherin decreased the expression of phosphorylated EGFR, and these cells did not respond to erlotinib. Therefore, we demonstrated the efficacy of the combined treatment with SAHA and erlotinib in human pancreatic cancer cells, and we determined that the increased efficacy was due, at least in part, to the effects of SAHA on the expression of E-cadherin. Our studies suggest that E-cadherin may be a potent biomarker for pancreatic cancer.


Subject(s)
Cadherins/genetics , ErbB Receptors/biosynthesis , Erlotinib Hydrochloride/administration & dosage , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Pancreatic Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cadherins/biosynthesis , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Vorinostat , Gemcitabine
14.
Bioprocess Biosyst Eng ; 38(9): 1761-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26048478

ABSTRACT

Biosynthetic pathways for the production of biofuels often rely on inherent aldehyde reductases (ALRs) of the microbial host. These native ALRs play vital roles in the success of the microbial production of 1,3-propanediol, 1,4-butanediol, and isobutanol. In the present study, the main ALR for 1,2,4-butanetriol (BT) production in Escherichia coli was identified. Results of real-time PCR analysis for ALRs in EWBT305 revealed the increased expression of adhP, fucO, adhE, and yqhD genes during BT production. The highest increase of expression was observed up to four times in yqhD. Singular deletion of adhP, fucO, or adhE gene showed marginal differences in BT production compared to that of the parent strain, EWBT305. Remarkably, yqhD gene deletion (KBTA4 strain) almost completely abolished BT production while its re-introduction (wild-type gene with its native promoter) on a low copy plasmid restored 75 % of BT production (KBTA4-2 strain). This suggests that yqhD gene is the main ALR of the BT pathway. In addition, KBTA4 showed almost no NADPH-dependent ALR activity, but was also restored upon re-introduction of the yqhD gene (KBTA4-2 strain). Therefore, the required ALR activity to complete the BT pathway was mainly contributed by YqhD. Increased gene expression and promiscuity of YqhD were both found essential factors to render YqhD as the key ALR for the BT pathway.


Subject(s)
Aldehyde Reductase/physiology , Biofuels/microbiology , Butanols/metabolism , Escherichia coli/physiology , Genetic Enhancement/methods , Xylose/metabolism , Butanols/isolation & purification , Catalysis , Enzyme Activation , Substrate Specificity
15.
Bioprocess Biosyst Eng ; 37(3): 383-91, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23820824

ABSTRACT

D-galactose is an attractive substrate for bioconversion. Herein, Escherichia coli was metabolically engineered to convert D-galactose into D-galactonate, a valuable compound in the polymer and cosmetic industries. D-galactonate productions by engineered E. coli strains were observed in shake flask cultivations containing 2 g L(-1) D-galactose. Engineered E. coli expressing gld coding for galactose dehydrogenase from Pseudomonas syringae was able to produce 0.17 g L(-1) D-galactonate. Inherent metabolic pathways for assimilating both D-galactose and D-galactonate were blocked to enhance the production of D-galactonate. This approach finally led to a 7.3-fold increase with D-galactonate concentration of 1.24 g L(-1) and yield of 62.0 %. Batch fermentation in 20 g L(-1) D-galactose of E. coli ∆galK∆dgoK mutant expressing the gld resulted in 17.6 g L(-1) of D-galactonate accumulation and highest yield of 88.1 %. Metabolic engineering strategy developed in this study could be useful for industrial production of D-galactonate.


Subject(s)
Escherichia coli/metabolism , Sugar Acids/metabolism , Base Sequence , Cloning, Molecular , Culture Media , DNA Primers , Escherichia coli/genetics , Galactose Dehydrogenases/genetics , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Molecular Structure , Pseudomonas syringae/enzymology , Sugar Acids/chemistry
16.
Bioprocess Biosyst Eng ; 37(12): 2505-13, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24928200

ABSTRACT

An engineered Escherichia coli strain was developed for enhanced isoprene production using D-galactose as substrate. Isoprene is a valuable compound that can be biosynthetically produced from pyruvate and glyceraldehyde-3-phosphate (G3P) through the methylerythritol phosphate pathway (MEP). The Leloir and De Ley-Doudoroff (DD) pathways are known existing routes in E. coli that can supply the MEP precursors from D-galactose. The DD pathway was selected as it is capable of supplying equimolar amounts of pyruvate and G3P simultaneously. To exclusively direct D-galactose toward the DD pathway, an E. coli ΔgalK strain with blocked Leloir pathway was used as the host. To obtain a fully functional DD pathway, a dehydrogenase encoding gene (gld) was recruited from Pseudomonas syringae to catalyze D-galactose conversion to D-galactonate. Overexpressions of endogenous genes known as MEP bottlenecks, and a heterologous gene, were conducted to enhance and enable isoprene production, respectively. Growth test confirmed a functional DD pathway concomitant with equimolar generation of pyruvate and G3P, in contrast to the wild-type strain where G3P was limiting. Finally, the engineered strain with combined DD-MEP pathway exhibited the highest isoprene production. This suggests that the equimolar pyruvate and G3P pools resulted in a more efficient carbon flux toward isoprene production. This strategy provides a new platform for developing improved isoprenoid producing strains through the combined DD-MEP pathway.


Subject(s)
Biotechnology/methods , Erythritol/analogs & derivatives , Escherichia coli/metabolism , Galactose/chemistry , Hemiterpenes/biosynthesis , Sugar Phosphates/chemistry , Butadienes/chemistry , Carbon/chemistry , Catalysis , DNA/chemistry , DNA Primers/chemistry , Erythritol/chemistry , Glyceraldehyde 3-Phosphate/chemistry , Hemiterpenes/chemistry , Pentanes/chemistry , Phosphates/metabolism , Plasmids/metabolism , Pseudomonas syringae/enzymology , Pyruvic Acid/chemistry
17.
Appl Microbiol Biotechnol ; 97(8): 3409-17, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23233208

ABSTRACT

Ethylene glycol (EG) is an important platform chemical with steadily expanding global demand. Its commercial production is currently limited to fossil resources; no biosynthesis route has been delineated. Herein, a biosynthesis route for EG production from D-xylose is reported. This route consists of four steps: D-xylose → D-xylonate → 2-dehydro-3-deoxy-D-pentonate → glycoaldehyde → EG. Respective enzymes, D-xylose dehydrogenase, D-xylonate dehydratase, 2-dehydro-3-deoxy-D-pentonate aldolase, and glycoaldehyde reductase, were assembled. The route was implemented in a metabolically engineered Escherichia coli, in which the D-xylose → D-xylulose reaction was prevented by disrupting the D-xylose isomerase gene. The most efficient construct produced 11.7 g L(-1) of EG from 40.0 g L(-1) of D-xylose. Glycolate is a carbon-competing by-product during EG production in E. coli; blockage of glycoaldehyde → glycolate reaction was also performed by disrupting the gene encoding aldehyde dehydrogenase, but from this approach, EG productivity was not improved but rather led to D-xylonate accumulation. To channel more carbon flux towards EG than the glycolate pathway, further systematic metabolic engineering and fermentation optimization studies are still required to improve EG productivity.


Subject(s)
Biosynthetic Pathways/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Ethylene Glycol/metabolism , Metabolic Engineering/methods , Xylose/metabolism
18.
Apoptosis ; 14(7): 913-22, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19526344

ABSTRACT

Sulindac is a non-steroidal anti-inflammatory agent with anti-tumor activities that include the induction of apoptosis in various cancer cells and the inhibition malignant transformation. However, the molecular mechanisms underlying these effects are unclear. Recently, it has been shown that sulindac can inhibit NF-kappaB activation. Here, we demonstrate that sulindac induces apoptotic cell death in susceptible human breast cancer cells through, at least in part, inhibition of IKKbeta activity. More specifically, when we compared two different human breast cancer cell lines, Hs578T, which has relatively low basal IKKbeta activity, and MDA-MB231, which has relatively high basal IKKbeta activity, we found that MDA-MB231 was markedly more sensitive to sulindac-induced apoptosis than Hs578T. This was associated with greater caspase-3 and -9 activity in sulindac-treated MDA-MB231 cells. Using a combination of chemical kinase inhibitors and siRNA-mediated knockdown of specific kinases, we found that sulindac inhibits IKKbeta, which, in turn, leads to the p38 MAPK-dependent activation of JNK1. Together, these findings suggest that sulindac induces apoptosis in susceptible human breast cancer cells through, at least in part, the inhibition of IKKbeta and the subsequent p38 MAPK-dependent activation of JNK1.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Breast Neoplasms , Cell Line, Tumor/drug effects , I-kappa B Kinase/antagonists & inhibitors , Sulindac/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor/metabolism , Drug Resistance, Neoplasm/drug effects , Enzyme Activation , Female , Humans , Mitogen-Activated Protein Kinase 8/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Int J Oncol ; 32(2): 435-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202766

ABSTRACT

Recent studies have suggested that Skp2, an SCF-type ubiquitin ligase, positively regulates cell cycle through degradation of p27, which is an inhibitor of cyclin-dependent kinase 2 (CDK2), which drives cells from the G1 to S phase of cell cycles. In the present study, we examined key regulatory proteins involved in serum starvation-induced cell cycle arrest in human ovarian cancer cells, SK-OV-3. Cell cycle analysis showed that cells were arrested at the G1 phase after serum starvation. Western blot analysis showed that the protein levels of CDK4 and CDK2 were significantly decreased in SK-OV-3 cells. Consistently, Roscovitine, an inhibitor of CDK2, induced cell cycle arrest in normally proliferating cells and a chemical inhibitor of CDK4, 3-ATA [3-Amino-9-thio(10H)-acridone], was found to induce growth arrest. We also found that the protein level of Skp2 was dramatically decreased in response to serum starvation. Moreover, CDK2 protein, which allows cell cycle transit from the G1 to the S phase, was decreased when the Skp2 expression was inhibited by specific siRNA of Skp2, but CDK4 was not decreased. Therefore, these results suggest that serum starvation induces G1 arrest through suppression of Skp2-dependent CDK2 activity and Skp2-independent CDK4 activity in human SK-OV-3 ovarian cancer cells.


Subject(s)
Culture Media, Serum-Free/pharmacology , Cyclin-Dependent Kinase 2/biosynthesis , Cyclin-Dependent Kinase 4/biosynthesis , G1 Phase , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/metabolism , S-Phase Kinase-Associated Proteins/biosynthesis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Humans , Ovarian Neoplasms/drug therapy , RNA Interference , RNA, Small Interfering/metabolism , Time Factors
20.
N Biotechnol ; 40(Pt B): 261-267, 2018 Jan 25.
Article in English | MEDLINE | ID: mdl-28962879

ABSTRACT

Research on the enzymatic breakdown of seaweed-derived agar has recently gained attention due to the progress in green technologies for marine biomass utilization. The enzymes known as agarases catalyze the cleavage of glycosidic bonds within the polysaccharide. In this study, a new ß-agarase, Aga2, was identified from Cellulophaga omnivescoria W5C. Aga2 is one of four putative agarases from the W5C genome, and it belongs to the glycoside hydrolase 16 family. It was shown to be exclusive to the Cellulophaga genus. Agarase activity assays showed that Aga2 is an endolytic-type ß-agarase that produces tetrameric and hexameric neoagaro-oligosaccharides, with optimum activity at 45°C and pH 8.0. Zinc ions slightly enhanced its activity while manganese ions had inhibitory effects even at very low concentrations. Aga2 has a Km of 2.59mgmL-1 and Vmax of 275.48Umg-1. The Kcat is 1.73×102s-1, while the Kcat/Km is 8.04×106s-1M-1. Aga2 also showed good thermostability at 45°C and above, and retained >90% of its activity after repeated freeze-thaw cycles. Bioinformatic analysis of its amino acid sequence revealed that intrinsic properties of the protein (e.g. presence of certain dipeptides and the relative volume occupied by aliphatic amino acids) and tertiary structural elements (e.g. presence of salt bridges, hydrophobic interactions and H-bonding) contributed to its thermostability.


Subject(s)
Flavobacteriaceae/enzymology , Glycoside Hydrolases/metabolism , Temperature , Computational Biology , Dose-Response Relationship, Drug , Enzyme Stability , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/chemistry , Manganese/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL