Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
ACS Synth Biol ; 13(6): 1866-1878, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38836566

ABSTRACT

3-Fucosyllactose (3-FL) is an important fucosylated human milk oligosaccharide (HMO) with biological functions such as promoting immunity and brain development. Therefore, the construction of microbial cell factories is a promising approach to synthesizing 3-FL from renewable feedstocks. In this study, a combinatorial engineering strategy was used to achieve efficient de novo 3-FL production in Escherichia coli. α-1,3-Fucosyltransferase (futM2) from Bacteroides gallinaceum was introduced into E. coli and optimized to create a 3-FL-producing chassis strain. Subsequently, the 3-FL titer increased to 5.2 g/L by improving the utilization of the precursor lactose and down-regulating the endogenous competitive pathways. Furthermore, a synthetic membraneless organelle system based on intrinsically disordered proteins was designed to spatially regulate the pathway enzymes, producing 7.3 g/L 3-FL. The supply of the cofactors NADPH and GTP was also enhanced, after which the 3-FL titer of engineered strain E26 was improved to 8.2 g/L in a shake flask and 10.8 g/L in a 3 L fermenter. In this study, we developed a valuable approach for constructing an efficient 3-FL-producing cell factory and provided a versatile workflow for other chassis cells and HMOs.


Subject(s)
Escherichia coli , Fucosyltransferases , Metabolic Engineering , Trisaccharides , Escherichia coli/genetics , Escherichia coli/metabolism , Trisaccharides/metabolism , Trisaccharides/biosynthesis , Metabolic Engineering/methods , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Lactose/metabolism , Bacteroides/genetics , Bacteroides/metabolism , Fermentation , Oligosaccharides
2.
J Agric Food Chem ; 72(25): 14191-14198, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38878091

ABSTRACT

3-Fucosyllactose (3-FL), an important fucosylated human milk oligosaccharide in breast milk, offers numerous health benefits to infants. Previously, we metabolically engineered Escherichia coli BL21(DE3) for the in vivo biosynthesis of 3-FL. In this study, we initially optimized culture conditions to double 3-FL production. Competing pathway genes involved in in vivo guanosine 5'-diphosphate-fucose biosynthesis were subsequently inactivated to redirect fluxes toward 3-FL biosynthesis. Next, three promising transporters were evaluated using plasmid-based or chromosomally integrated expression to maximize extracellular 3-FL production. Additionally, through analysis of α1,3-fucosyltransferase (FutM2) structure, we identified Q126 residues as a highly mutable residue in the active site. After site-saturation mutation, the best-performing mutant, FutM2-Q126A, was obtained. Structural analysis and molecular dynamics simulations revealed that small residue replacement positively influenced helical structure generation. Finally, the best strain BD3-A produced 6.91 and 52.1 g/L of 3-FL in a shake-flask and fed-batch cultivations, respectively, highlighting its potential for large-scale industrial applications.


Subject(s)
Escherichia coli , Fucosyltransferases , Metabolic Engineering , Trisaccharides , Escherichia coli/genetics , Escherichia coli/metabolism , Trisaccharides/metabolism , Trisaccharides/biosynthesis , Trisaccharides/chemistry , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Humans , Oligosaccharides
3.
Mar Drugs ; 22(5)2024 May 18.
Article in English | MEDLINE | ID: mdl-38786621

ABSTRACT

Alginate oligosaccharides (AOS), products of alginate degradation by endotype alginate lyases, possess favorable biological activities and have broad applications. Although many have been reported, alginate lyases with homogeneous AOS products and secretory production by an engineered host are scarce. Herein, the alginate lyase AlyC7 from Vibrio sp. C42 was characterized as a trisaccharide-producing lyase exhibiting high activity and broad substrate specificity. With PelB as the signal peptide and 500 mM glycine as the additive, the extracellular production of AlyC7 in Escherichia coli reached 1122.8 U/mL after 27 h cultivation in Luria-Bertani medium. The yield of trisaccharides from sodium alginate degradation by the produced AlyC7 reached 758.6 mg/g, with a purity of 85.1%. The prepared AOS at 20 µg/mL increased the root length of lettuce, tomato, wheat, and maize by 27.5%, 25.7%, 9.7%, and 11.1%, respectively. This study establishes a robust foundation for the industrial and agricultural applications of AlyC7.


Subject(s)
Escherichia coli , Polysaccharide-Lyases , Trisaccharides , Vibrio , Polysaccharide-Lyases/metabolism , Trisaccharides/biosynthesis , Vibrio/enzymology , Substrate Specificity , Alginates , Zea mays , Oligosaccharides
4.
Biotechnol Bioeng ; 120(2): 524-535, 2023 02.
Article in English | MEDLINE | ID: mdl-36326175

ABSTRACT

As one of the most abundant components in human milk oligosaccharides, 2'-fucosyllactose (2'-FL) possesses versatile beneficial health effects. Although most studies focused on overexpressing or fine-tuning the expression of pathway enzymes and achieved a striking increase of 2'-FL production, directly facilitating the metabolic flux toward the key intermediate GDP-l-fucose seems to be ignored. Here, multienzyme complexes consisting of sequential pathway enzymes were constructed by using specific peptide interaction motifs in recombinant Escherichia coli to achieve a higher titer of 2'-FL. Specifically, we first fine-tuned the expression level of pathway enzymes and balanced the metabolic flux toward 2'-FL synthesis. Then, two key enzymes (GDP-mannose 4,6-dehydratase and GDP- l-fucose synthase) were self-assembled into enzyme complexes in vivo via a short peptide interaction pair RIAD-RIDD (RI anchoring disruptor-RI dimer D/D domains), resulting in noticeable improvement of 2'-FL production. Next, to further strengthen the metabolic flux toward 2'-FL, three pathway enzymes were further aggregated into multienzyme assemblies by using another orthogonal protein interaction motif (Spycatcher-SpyTag or PDZ-PDZlig). Intracellular multienzyme assemblies remarkably enlarged the flux toward 2'-FL biosynthesis and showed a 2.1-fold increase of 2'-FL production compared with a strain expressing free-floating and unassembled enzymes. The optimally engineered strain EZJ23 accumulated 4.8 g/L 2'-FL in shake flask fermentation and was capable of producing 25.1 g/L 2'-FL by fed-batch cultivation. This work provides novel approaches for further improvement and large-scale production of 2'-FL and demonstrates the effectiveness of spatial assembly of pathway enzymes to improve the production of valuable products in the engineered host strain.


Subject(s)
Escherichia coli , Fucose , Trisaccharides , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Fucose/metabolism , Guanosine Diphosphate Fucose/metabolism , Metabolic Engineering/methods , Multienzyme Complexes/metabolism , Peptides/metabolism , Trisaccharides/biosynthesis
5.
Nucleic Acids Res ; 50(11): 6587-6600, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35670665

ABSTRACT

Dynamic regulation is an effective strategy for control of gene expression in microbial cell factories. In some pathway contexts, several metabolic modules must be controlled in a time dependent or ordered manner to maximize production, while the creation of genetic circuits with ordered regulation capacity still remains a great challenge. In this work, we develop a pathway independent and programmable system that enables multi-modular ordered control of metabolism in Bacillus subtilis. First, a series of thermosensors were created and engineered to expand their thresholds. Then we designed single-input-multi-output circuits for ordered control based on the use of thermosensors with different transition points. Meanwhile, a repression circuit was constructed by combining CRISPRi-based NOT gates. As a proof-of-concept, these genetic circuits were applied for multi-modular ordered control of 2'-fucosyllactose (2'-FL) biosynthesis, resulting in a production of 1839.7 mg/l in shake flask, which is 5.16-times that of the parental strain. In a 5-l bioreactor, the 2'-FL titer reached 28.2 g/l with down-regulation of autolysis. Taken together, this work provides programmable and versatile thermosensitive genetic toolkits for dynamic regulation in B. subtilis and a multi-modular ordered control framework that can be used to improve metabolic modules in other chassis cells and for other compounds.


Subject(s)
Bacillus subtilis , Clustered Regularly Interspaced Short Palindromic Repeats , Metabolic Engineering , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Gene Regulatory Networks , Metabolic Engineering/methods , Temperature , Trisaccharides/biosynthesis
6.
Microb Cell Fact ; 20(1): 165, 2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34425826

ABSTRACT

BACKGROUND: 2'-fucosyllactose (2'-FL) is one of the most abundant oligosaccharides in human milk. It constitutes an authorized functional additive to improve infant nutrition and health in manufactured infant formulations. As a result, a cost-effective method for mass production of 2'-FL is highly desirable. RESULTS: A microbial cell factory for 2'-FL production was constructed in Saccharomyces cerevisiae by expressing a putative α-1, 2-fucosyltransferase from Bacillus cereus (FutBc) and enhancing the de novo GDP-L-fucose biosynthesis. When enabled lactose uptake, this system produced 2.54 g/L of 2'-FL with a batch flask cultivation using galactose as inducer and carbon source, representing a 1.8-fold increase compared with the commonly used α-1, 2-fucosyltransferase from Helicobacter pylori (FutC). The production of 2'-FL was further increased to 3.45 g/L by fortifying GDP-mannose synthesis. Further deleting gal80 enabled the engineered strain to produce 26.63 g/L of 2'-FL with a yield of 0.85 mol/mol from lactose with sucrose as a carbon source in a fed-batch fermentation. CONCLUSION: FutBc combined with the other reported engineering strategies holds great potential for developing commercial scale processes for economic 2'-FL production using a food-grade microbial cell factory.


Subject(s)
Bacillus cereus/enzymology , Fucosyltransferases/genetics , Metabolic Engineering/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Trisaccharides/biosynthesis , Bacillus cereus/genetics , Batch Cell Culture Techniques , Fermentation , Fucosyltransferases/classification , Trisaccharides/genetics
7.
ACS Synth Biol ; 10(10): 2488-2498, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34415729

ABSTRACT

2'-Fucosyllactose (2'-FL) has been widely used as a nutritional additive in infant formula due to its multifarious nutraceutical and pharmaceutical functions in neonate health. As such, it is essential to develop an efficient and extensive microbial fermentation platform to cater to the needs of the 2'-FL market. In this study, a spatial synthetic biology strategy was employed to promote 2'-FL biosynthesis in recombinant Escherichia coli. First, the salvage pathway for 2'-FL production from l-fucose and lactose was constructed by introducing a bifunctional enzyme l-fucokinase/GDP-l-fucose pyrophosphorylase (Fkp) derived from Bacteroides fragilis and an α-1,2-fucosyltransferase (FutC) derived from Helicobacter pylori into engineered E. coli BL21(DE3). Next, the endogenous genes involved in the degradation and shunting of the substrate and key intermediate were inactivated to improve the availability of precursors for 2'-FL biosynthesis. Moreover, to further improve the yield and titer of 2'-FL, a short peptide pair (RIAD-RIDD) was used to form self-assembling multienzyme complexes in vivo. The spatial localization of peptides and stoichiometry of enzyme assemblies were subsequently optimized to further improve 2'-FL production. Finally, cofactor regeneration was also considered to alleviate the potential cofactor deficiency and redox flux imbalance in the biocatalysis process. Fed-batch fermentation of the final WLS20 strain accumulated 30.5 g/L extracellular 2'-FL with the yield and productivity of 0.661 mol/mol fucose and 0.48 g/L/h, respectively. This research has demonstrated that the application of spatial synthetic biology and metabolic engineering strategies can dramatically enlarge the titer and yield of 2'-FL biosynthesis in engineered E. coli.


Subject(s)
Dietary Supplements , Escherichia coli/genetics , Fucose/metabolism , Metabolic Engineering , Multienzyme Complexes/metabolism , Trisaccharides/biosynthesis , Genome, Bacterial
8.
J Biotechnol ; 340: 30-38, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34450187

ABSTRACT

2'-Fucosyllactose (2'-FL), a major oligosaccharide of human breast milk, and is currently supplemented into infant formula. For the overproduction of 2'-FL via fucosylation of lactose, conventional approaches have focused on the episomal overexpression of de novo or salvage GDP-L-fucose biosynthetic pathway and α-1,2-fucosyltransferase (FucT2) through T7 RNA polymerase expression system in engineered E. coli. However, these approaches have drawbacks of metabolic burden, plasmid instability, and inclusion body formation. In this study, a deletion mutant of waaF coding for ADP-heptose:LPS heptosyltransferase II was employed for 2'-FL production. As the waaF deletion induces accumulation of colanic acid, additional deletion of wcaJ coding for UDP-glucose-1-phosphate transferase in the waaF deletion mutant resulted in enhanced accumulation of GDP-L-fucose. Besides, 2'-FL yields and titers were drastically improved when T7 promoter was replaced with Trc promoter for α-1,2 fucosyltransferase expressions in the waaF and wcaJ deleted strain. As a result, when FucT2 was expressed under Trc promoter in the E. coli JM109(DE3) ΔwaaFΔwcaJ, 14.7 g/L of 2'-FL was produced with a productivity of 0.31 g/L/h in a fed-batch fermentation. We envision that the deletion-based metabolic design and decreased promoter strength for fucosyltransferase expression can resolve the drawbacks of T7 RNA polymerase-based expression design for 2'-FL production in E. coli.


Subject(s)
Escherichia coli , Fucosyltransferases , Trisaccharides/biosynthesis , Escherichia coli/genetics , Fucosyltransferases/genetics , Guanosine Diphosphate Fucose , Phosphotransferases (Phosphate Group Acceptor)/genetics
9.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299187

ABSTRACT

By culturing microorganisms under standard laboratory conditions, most biosynthetic gene clusters (BGCs) are not expressed, and thus, the products are not produced. To explore this biosynthetic potential, we developed a novel "semi-targeted" approach focusing on activating "silent" BGCs by concurrently introducing a group of regulator genes into streptomycetes of the Tübingen strain collection. We constructed integrative plasmids containing two classes of regulatory genes under the control of the constitutive promoter ermE*p (cluster situated regulators (CSR) and Streptomyces antibiotic regulatory proteins (SARPs)). These plasmids were introduced into Streptomyces sp. TÜ17, Streptomyces sp. TÜ10 and Streptomyces sp. TÜ102. Introduction of the CSRs-plasmid into strain S. sp. TÜ17 activated the production of mayamycin A. By using the individual regulator genes, we proved that Aur1P, was responsible for the activation. In strain S. sp. TÜ102, the introduction of the SARP-plasmid triggered the production of a chartreusin-like compound. Insertion of the CSRs-plasmid into strain S. sp. TÜ10 resulted in activating the warkmycin-BGC. In both recombinants, activation of the BGCs was only possible through the simultaneous expression of aur1PR3 and griR in S. sp. TÜ102 and aur1P and pntR in of S. sp. TÜ10.


Subject(s)
Bacterial Proteins/genetics , Benz(a)Anthracenes/metabolism , Multigene Family , Recombinant Proteins/genetics , Streptomyces/genetics , Bacterial Proteins/metabolism , Benzopyrans , Gene Expression Regulation, Bacterial , Glycosides/biosynthesis , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Streptomyces/growth & development , Streptomyces/metabolism , Transcription Factors/metabolism , Trisaccharides/biosynthesis
10.
J Ind Microbiol Biotechnol ; 48(5-6)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34137896

ABSTRACT

Fructooligosaccharides (FOSs)-fructose-based oligosaccharides-are typical prebiotics with health-promoting effects in humans and animals. The trisaccharide 1-kestotriose is the most attractive inulin-type FOS. We previously reported a recombinant sucrose:sucrose 1-fructosyltransferase (1-SST, EC 2.4.1.99) from Schedonorus arundinaceus (Sa) that efficiently converts sucrose into 1-kestotriose. In this study, Pichia pastoris PGFT6x-308 constitutively expressing nine copies of the Sa1-SST gene displayed fructosyltransferase activity in undisrupted biomass (49.8 U/ml) and culture supernatant (120.7 U/ml) in fed-batch fermentation (72 hr) with sugarcane molasses. Toluene permeabilization increased 2.3-fold the Sa1-SSTrec activity of whole cells entrapped in calcium-alginate beads. The reaction with refined or raw sugar (600 g/l) yielded 1-kestotriose and 1,1-kestotetraose in a ratio of 8:2 with their sum representing above 55% (wt/wt) of total carbohydrates. The FOSs yield decreased to 45% (wt/wt) when sugarcane syrup and molasses were used as cheaper sucrose sources. The beads retained 80% residual Sa1-SSTrec activity after a 30-day batchwise operation with refined cane sugar at 30°C and pH 5.5. The immobilized biocatalyst is attractive for the continuous production of short-chain FOSs, most particularly 1-kestotriose.


Subject(s)
Hexosyltransferases/metabolism , Oligosaccharides/metabolism , Pichia/metabolism , Alginates/chemistry , Carbohydrates/analysis , Cell Membrane Permeability/drug effects , Cells, Immobilized , Fermentation , Hexosyltransferases/genetics , Humans , Industrial Microbiology , Inulin/metabolism , Molasses , Pichia/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomycetales , Sucrose , Toluene/pharmacology , Trisaccharides/biosynthesis
11.
ACS Synth Biol ; 10(3): 447-458, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33687208

ABSTRACT

Human milk oligosaccharide (HMO) is a key component of human milk carbohydrates and is closely related to the nutrition and health benefits of breastfeeding in infants. 2'-Fucosyllactose (2'-FL) is the most abundant fucosylated HMO, which has remarkable value in nutrition and medicine, such as suppressing pathogen infection, regulating intestinal flora, and boosting immunity. However, 2'-FL production via the method of extraction or chemical synthesis cannot meet its large demand, and as a result, environmentally friendly and efficient biotechnological approaches, including in vitro enzymatic synthesis and microbial cell factory production, have been developed and applied to its commercialized production. This review introduces, summarizes, and discusses the recent advances in the biotechnological production of 2'-FL. Furthermore, future research directions for the biotechnological production of 2'-FL as well as the strategies to further improve its concentration are highlighted and discussed.


Subject(s)
Biotechnology , Milk, Human/metabolism , Trisaccharides/biosynthesis , Fucosyltransferases/chemistry , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Humans , Metabolic Engineering , Saccharomyces cerevisiae/metabolism , alpha-L-Fucosidase/chemistry , alpha-L-Fucosidase/genetics , alpha-L-Fucosidase/metabolism
12.
J Agric Food Chem ; 69(5): 1567-1577, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33499605

ABSTRACT

2'-Fucosyllactose (2'-FL), one of the most valuable oligosaccharides in human milk, is used as an emerging food ingredient in the nutraceutical and food industries due to its numerous health benefits. Herein, the de novo and salvage pathways for GDP-fucose synthesis were engineered and optimized in Escherichia coli BL21 (DE3) to improve the production of 2'-FL. The de novo pathway genes encoding phosphomannomutase (ManB), mannose-1-phosphate guanyltransferase (ManC), GDP-d-mannose-4,6-dehydratase (Gmd), and GDP-l-fucose synthase (WcaG) combined with the gene from the salvage pathway encoding fucose kinase/fucose-1-phosphate guanylyltransferase (Fkp) were reconstructed in two vectors to evaluate the GDP-fucose biosynthesis. Then, the fucT2 gene, encoding α1,2-fucosyltransferase, was introduced into the GDP-fucose-overproducing strains to realize 2'-FL biosynthesis. Furthermore, the genes in bypass pathways, including lacZ, fucI, fucK, and wcaJ, were inactivated to improve 2'-FL production. In addition, the two GDP-fucose synthesis pathways, along with fucT2, were transcriptionally fine-tuned to efficiently increase 2'-FL production. The final metabolically engineered E. coli produced 2.62 and 14.1 g/L in shake-flask and fed-batch cultivations, respectively.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Trisaccharides/biosynthesis , Biosynthetic Pathways , Escherichia coli/growth & development , Fucose/metabolism , Lactose/metabolism , Mannose/metabolism , Metabolic Engineering
13.
ACS Synth Biol ; 9(10): 2784-2796, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32966739

ABSTRACT

Human milk oligosaccharides (HMOs) are unique components of human breast milk. Their large-scale production by fermentation allows infant formulas to be fortified with HMOs, but current fermentation processes require lactose as a starting material, increasing the costs, bioburden, and environmental impact of manufacturing. Here we report the development of an Escherichia coli strain that produces 2'-fucosyllactose (2'-FL), the most abundant HMO, de novo using sucrose as the sole carbon source. Strain engineering required the expression of a novel glucose-accepting galactosyltransferase, overexpression of the de novo UDP-d-galactose and GDP-l-fucose pathways, the engineering of an intracellular pool of free glucose, and overexpression of a suitable α(1,2)-fucosyltransferase. The export of 2'-FL was facilitated using a sugar efflux transporter. The final production strain achieved 2'-FL yields exceeding 60 g/L after fermentation for 84 h. This efficient strategy facilitates the lactose-independent production of HMOs by fermentation, which will improve product quality and reduce the costs of manufacturing.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Metabolic Engineering/methods , Milk, Human/chemistry , Sucrose/metabolism , Trisaccharides/biosynthesis , Batch Cell Culture Techniques , Carbon/metabolism , Fermentation , Food Quality , Fucose/metabolism , Fucosyltransferases/metabolism , Galactose/metabolism , Galactosyltransferases/metabolism , Humans , Infant Formula/chemistry , Lactose/metabolism
14.
Food Chem ; 332: 127438, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32645671

ABSTRACT

ß-N-acetylhexosaminidases have attracted much attention in recent years due to their potential application in oligosaccharide production, in particular lacto-N-triose II (LNT2) and lacto-N-neotetraose (LNnT) synthesis, which can be further used as backbone precursors for human milk oligosaccharides. A novel ß-N-acetylhexosaminidase gene from Tyzzerella nexilis (TnHex189) was heterologously expressed in Bacillus subtilis. The highest ß-N-acetylhexosaminidase activity of 14.5 U mL-1 was obtained in a 5-L fermentor by fed-batch fermentation for 27 h. TnHex189 was optimally active at pH 5.0 and 45 °C. It efficiently synthesized LNT2 with a conversion ratio of 57.2% (4.7 g L-1). The synthesized LNT2 was further converted to LNnT by a reported ß-galactosidase (BgaD-D) in 8 h, with a conversion ratio of 17.3% (6.1 g L-1). These unique synthesis activities may make this enzyme a good candidate for the food industry.


Subject(s)
Bacterial Proteins/metabolism , Clostridiales/enzymology , Trisaccharides/biosynthesis , beta-N-Acetylhexosaminidases/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Clostridiales/genetics , Enzyme Stability , Fermentation , Gene Expression , Hydrogen-Ion Concentration , Oligosaccharides/metabolism , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/genetics
15.
Appl Microbiol Biotechnol ; 104(13): 5813-5826, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32388762

ABSTRACT

Fucosyllactoses have gained much attention owing to their multiple functions, including prebiotic, immune, gut, and cognition benefits. In this study, human milk oligosaccharide (HMO) 2'-fucosyllactose (α-L-Fuc-(1,2)-D-Galß-1,4-Glu, 2'FL) and its isomer 3'-fucosyllactose (α-L-Fuc-(1,3)-D-Galß-1,4-Glu, 3'FL) with potential prebiotic effect were synthesized efficiently by a novel recombinant α-L-fucosidase. An α-L-fucosidase gene (PbFuc) from Pedobacter sp. CAU209 was successfully cloned and expressed in Escherichia coli (E. coli). The deduced amino acid sequence shared the highest identity of 36.8% with the amino sequences of other reported α-L-fucosidases. The purified α-L-fucosidase (PbFuc) had a molecular mass of 50 kDa. The enzyme exhibited specific activity (26.3 U/mg) towards 4-nitrophenyl-α-L-fucopyranoside (pNP-FUC), 3'FL (8.9 U/mg), and 2'FL (3.4 U/mg). It showed the highest activity at pH 5.0 and 35 °C, respectively. PbFuc catalyzed the synthesis of 3'FL and 2'FL through a transglycosylation reaction using pNP-FUC as donor and lactose as acceptor, and total conversion ratio was up to 85% at the optimized reaction conditions. The synthesized mixture of 2'FL and 3'FL promoted the growth of Lactobacillus delbrueckii subsp. bulgaricus NRRL B-548, L. casei subsp. casei NRRL B-1922, L. casei subsp. casei AS 1.2435, and Bifidobacterium longum NRRL B-41409. However, the growths of E. coli ATCC 11775, S. enterica AS 1.1552, L. monocytogenes CICC 21635, and S. aureus AS 1.1861 were not stimulated by the mixture of 2'FL and 3'FL. Overall, our findings suggest that PbFuc possesses a great potential for the specific synthesis of fucosylated compounds.Key Points• A novel α-L-fucosidase (PbFuc) from Pedobacter sp. was cloned and expressed.• PbFuc showed the highest hydrolysis activity at pH 5.0 and 35 °C, respectively.• It was used for synthesis of 3'-fucosyllactose (3'FL) and 2'-fucosyllactose (2'FL).• The mixture of 3'FL and 2'FL promoted the growth of some Lactobacillus sp. and Bifidobacteria sp.


Subject(s)
Bacterial Proteins/metabolism , Oligosaccharides/biosynthesis , Pedobacter/enzymology , Trisaccharides/biosynthesis , alpha-L-Fucosidase/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Glycosides/metabolism , Glycosylation , Hydrogen-Ion Concentration , Hydrolysis , Lactose/metabolism , Molecular Weight , Pedobacter/genetics , Prebiotics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity , Temperature , alpha-L-Fucosidase/chemistry , alpha-L-Fucosidase/genetics , alpha-L-Fucosidase/isolation & purification
16.
Chem Commun (Camb) ; 56(47): 6408-6411, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32390019

ABSTRACT

The first synthesis of 3-deoxy-3-fluoro-l-fucose is presented, which employs a d- to l-sugar translation strategy, and involves an enzymatic oxidation of 3-deoxy-3-fluoro-l-fucitol. Enzymatic activation (FKP) and glycosylation using an α-1,2 and an α-1,3 fucosyltransferase to obtain two fluorinated trisaccharides demonstrates its potential as a novel versatile chemical probe in glycobiology.


Subject(s)
Fucosyltransferases/metabolism , Glycoconjugates/biosynthesis , Trisaccharides/biosynthesis , Fucosyltransferases/chemistry , Glycoconjugates/chemistry , Glycosylation , Halogenation , Molecular Conformation , Oxidation-Reduction , Trisaccharides/chemistry
17.
Carbohydr Polym ; 237: 116121, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32241400

ABSTRACT

Here, we proposed an effective strategy to enhance a novel endoxylanase (Taxy11) activity and elucidated an efficient catalysis mechanism to produce xylooligosaccharides (XOSs). Codon optimization and recruitment of natural propeptide in Pichia pastoris resulted in achievement of Taxy11 activity to 1405.65 ±â€¯51.24 U/mL. Analysis of action mode reveals that Taxy11 requires at least three xylose (xylotriose) residues for hydrolysis to yield xylobiose. Results of site-directed mutagenesis indicate that residues Glu119, Glu210, and Asp53 of Taxy11 are key catalytic sites, while Asp203 plays an auxiliary role. The novel mechanism whereby Taxy11 catalyzes conversion of xylan or XOSs into major product xylobiose involves transglycosylation of xylose to xylotriose or xylotetraose as substrate, to form xylotetraose or xylopentaose intermediate, respectively. Taxy11 displayed highly hydrolytic activity toward corncob xylan, producing 50.44 % of xylobiose within 0.5 h. This work provides a cost-effective and sustainable way to produce value-added biomolecules XOSs (xylobiose-enriched) from agricultural waste.


Subject(s)
Disaccharides/biosynthesis , Endo-1,4-beta Xylanases/metabolism , Xylan Endo-1,3-beta-Xylosidase/metabolism , Xylans/metabolism , Cloning, Molecular , Hydrolysis , Kinetics , Pichia/genetics , Substrate Specificity , Trichoderma/enzymology , Trisaccharides/biosynthesis , Xylose/metabolism
18.
Plant J ; 100(5): 879-891, 2019 12.
Article in English | MEDLINE | ID: mdl-31400245

ABSTRACT

Type 2 diabetes (T2D) affects over 320 million people worldwide. Healthy lifestyles, improved drugs and effective nutraceuticals are different components of a response against the growing T2D epidemic. The specialized metabolite montbretin A (MbA) is being developed for treatment of T2D and obesity due to its unique pharmacological activity as a highly effective and selective inhibitor of the human pancreatic α-amylase. MbA is an acylated flavonol glycoside found in small amounts in montbretia (Crocosmia × crocosmiiflora) corms. MbA cannot be obtained in sufficient quantities for drug development from its natural source or by chemical synthesis. To overcome these limitations through metabolic engineering, we are investigating the genes and enzymes of MbA biosynthesis. We previously reported the first three steps of MbA biosynthesis from myricetin to myricetin 3-O-(6'-O-caffeoyl)-glucosyl rhamnoside (mini-MbA). Here, we describe the sequence of reactions from mini-MbA to MbA, and the discovery and characterization of the gene and enzyme responsible for the glucosylation of mini-MbA. The UDP-dependent glucosyltransferase CcUGT3 (UGT703E1) catalyzes the 1,2-glucosylation of mini-MbA to produce myricetin 3-O-(glucosyl-6'-O-caffeoyl)-glucosyl rhamnoside. Co-expression of CcUGT3 with genes for myricetin and mini-MbA biosynthesis in Nicotiana benthamiana validated its biological function and expanded the set of genes available for metabolic engineering of MbA.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Flavones/biosynthesis , Glucosyltransferases/metabolism , Hypoglycemic Agents/metabolism , Metabolic Engineering/methods , Trisaccharides/biosynthesis , Caffeic Acids/chemistry , Caffeic Acids/metabolism , Flavones/chemistry , Flavones/pharmacology , Flavones/therapeutic use , Flavonoids/chemistry , Flavonoids/metabolism , Flavonols/chemistry , Flavonols/metabolism , Gene Expression Profiling , Gene Expression Regulation, Plant/genetics , Glucose/chemistry , Glucose/metabolism , Glycosides/chemistry , Glycosides/metabolism , Glycosylation , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Iridaceae/chemistry , Iridaceae/enzymology , Phylogeny , Plant Proteins/metabolism , Plant Stems/chemistry , Plant Stems/metabolism , Plants, Genetically Modified , Rhamnose/chemistry , Rhamnose/metabolism , Secondary Metabolism , Synthetic Biology/methods , Nicotiana/metabolism , Transcriptome/genetics , Trisaccharides/chemistry , Trisaccharides/pharmacology , Trisaccharides/therapeutic use , Xylose/chemistry , Xylose/metabolism
19.
Biotechnol Bioeng ; 116(9): 2412-2417, 2019 09.
Article in English | MEDLINE | ID: mdl-31145478

ABSTRACT

2'-Fucosyllactose (2-FL), one of the most abundant oligosaccharides in human milk, has been spotlighted for its neutraceutical and pharmaceutical potentials. Microbial production of 2-FL is promising since it is efficient as compared to other production methods. In 2-FL microbial production via the salvage pathway for biosynthesis of guanosine 5'-diphosphate (GDP)-l-fucose from fucose, the conversion yield from fucose is important because of the high price of fucose. In this study, deletion of the genes (araA and rhaA) coding for arabinose isomerase (AraA) and rhamnose isomerase (RhaA) was attempted in engineered Escherichia coli for improving 2-FL production by using fucose, lactose, and glycerol. The engineered E. coli constructed previously is able to express fucokinase/GDP-l-fucose pyrophosphorylase (Fkp) from Bacteroides fragilis and the α-1,2-fucosyltransferase (FucT2) from Helicobacter pylori and deficient in ß-galactosidase (LacZ), fucose isomerase (FucI), and fuculose kinase (FucK). The additional double-deletion of the araA and rhaA genes in the engineered E. coli enhanced the product yield of 2-FL to 0.52 mole 2-FL/mole fucose, and hence the concentration of 2-FL reached to 47.0 g/L, which are 44% and two-fold higher than those (23.1 g/L and 0.36 mole 2-FL/mole fucose) of the control strain in fed-batch fermentation. Elimination of sugar isomerases exhibiting promiscuous activities with fucose might be critical in the microbial production of 2-FL through the salvage pathway of GDP-l-fucose.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Fucose/metabolism , Gene Deletion , Metabolic Engineering , Trisaccharides/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fucose/genetics , Trisaccharides/genetics
20.
Microbiol Res ; 222: 35-42, 2019 May.
Article in English | MEDLINE | ID: mdl-30928028

ABSTRACT

2'-Fucosyllactose (2'-FL) is the most abundant human milk oligosaccharide and is important for infant nutrition and health. Because 2'-FL has potential as a functional ingredient in advanced infant formula and as a prebiotic in various foods, a cost-effective method for 2'-FL production is desirable. α1,2-Fucosyltransferase (α1,2-FT) is one of the key enzymes enabling the microbial biosynthesis of this complex sugar. However, the α1,2-FTs reported so far for the whole-cell biosynthesis of 2'-FL originate from pathogens, posing a potential hurdle for approval as a food production method depending on countries. In this study, 10 α1,2-FT genes from bacteria of biosafety level one were identified, and the main features of the deduced amino acid sequences were characterized. Four codon-optimized α1,2-FT genes were synthesized and introduced into Escherichia coli ΔL M15 strain containing the plasmid pBCGW encoding guanosine 5'-diphosphate-l-fucose biosynthetic enzymes. Among the four genes, 2'-FL was produced only by the α1,2-FT from Thermosynechococcus elongatus (Te2FT). Bifidobacterium thermacidophilum α1,2-FT (Bt2FT) showed high expression but was not active in E. coli ΔL M15. The other two α1,2-FTs were not expressed to a detectable level. During batch flask fermentation of Te2FT-expressing E. coli ΔL M15 cells, 0.49 g/L 2'-FL was obtained after 72 h of induction. This is comparable to the values previously reported for α1,2-FTs from Helicobacter pylori and Bacteroides fragilis.


Subject(s)
Escherichia coli/genetics , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trisaccharides/biosynthesis , Bacterial Proteins/genetics , Bacteroides fragilis/enzymology , Bacteroides fragilis/metabolism , Bifidobacterium/genetics , Bifidobacterium/metabolism , Cyanobacteria/enzymology , Cyanobacteria/genetics , DNA, Bacterial , Escherichia coli/metabolism , Fermentation , Gene Expression Regulation, Bacterial , Helicobacter pylori/enzymology , Helicobacter pylori/metabolism , Milk, Human , Oligosaccharides
SELECTION OF CITATIONS
SEARCH DETAIL
...