Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 247
Filtrar
1.
J Agric Food Chem ; 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38598361

RESUMO

Difucosyllactose (DFL) is a significant and plentiful oligosaccharide found in human breast milk. In this study, an artificial metabolic pathway of DFL was designed, focusing on the de novo biosynthesis of GDP-fucose from only glycerol. This was achieved by engineering Escherichia coli to endogenously overexpress genes manB, manC, gmd, and wcaG and heterologously overexpress a pair of fucosyltransferases to produce DFL from lactose. The introduction of α-1,2-fucosyltransferase from Helicobacter pylori (FucT2) along with α-1,3/4-fucosyltransferase (HP3/4FT) addressed rate-limiting challenges in enzymatic catalysis and allowed for highly efficient conversion of lactose into DFL. Based on these results, molecular modification of HP3/4FT was performed based on computer-assisted screening and structure-based rational design. The best-performing mutant, MH5, containing a combination of five mutated sites (F49K/Y131D/Y197N/E338D/R369A) of HP3/4FT was obtained. The best strain BLC09-58 harboring MH5 yielded 45.81 g/L of extracellular DFL in 5-L fed-batch cultures, which was the highest titer reported to date.

2.
J Agric Food Chem ; 72(14): 8052-8059, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38563420

RESUMO

d-Allulose, a functional bulk sweetener, has recently attracted increasing attention because of its low-caloric-ness properties and diverse health effects. d-Allulose is industrially produced by the enzymatic epimerization of d-fructose, which is catalyzed by ketose 3-epimerase (KEase). In this study, the food-grade expression of KEase was studied using Bacillus subtills as the host. Clostridium sp. d-allulose 3-epimerase (Clsp-DAEase) was screened from nine d-allulose-producing KEases, showing better potential for expression in B. subtills WB600. Promoter-based transcriptional regulation and N-terminal coding sequence (NCS)-based translational regulation were studied to enhance the DAEase expression level. In addition, the synergistic effect of promoter and NCS on the Clsp-DAEase expression was studied. Finally, the strain with the combination of a PHapII promoter and gln A-Up NCS was selected as the best Clsp-DAEase-producing strain. It efficiently produced Clsp-DAEase with a total activity of 333.2 and 1860.6 U/mL by shake-flask and fed-batch cultivations, respectively.


Assuntos
Bacillus subtilis , Racemases e Epimerases , Racemases e Epimerases/genética , Racemases e Epimerases/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Frutose/metabolismo , Cetoses
3.
J Agric Food Chem ; 72(14): 7607-7617, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38563422

RESUMO

Gastrointestinal (GI)-associated viruses, including rotavirus (RV), norovirus (NV), and enterovirus, usually invade host cells, transmit, and mutate their genetic information, resulting in influenza-like symptoms, acute gastroenteritis, encephalitis, or even death. The unique structures of human milk oligosaccharides (HMOs) enable them to shape the gut microbial diversity and endogenous immune system of human infants. Growing evidence suggests that HMOs can enhance host resistance to GI-associated viruses but without a systematic summary to review the mechanism. The present review examines the lactose- and neutral-core HMOs and their antiviral effects in the host. The potential negative impacts of enterovirus 71 (EV-A71) and other GI viruses on children are extensive and include neurological sequelae, neurodevelopmental retardation, and cognitive decline. However, the differences in the binding affinity of HMOs for GI viruses are vast. Hence, elucidating the mechanisms and positive effects of HMOs against different viruses may facilitate the development of novel HMO derived oligosaccharides.


Assuntos
Leite Humano , Rotavirus , Lactente , Criança , Humanos , Leite Humano/química , Rotavirus/genética , Rotavirus/metabolismo , Sistema Imunitário , Antivirais/farmacologia , Oligossacarídeos/metabolismo
4.
Biotechnol Adv ; 73: 108355, 2024 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-38588907

RESUMO

Membraneless organelles (MLOs) formed by liquid-liquid phase separation (LLPS) have been extensively studied due to their spatiotemporal control of biochemical and cellular processes in living cells. These findings have provided valuable insights into the physicochemical principles underlying the formation and functionalization of biomolecular condensates, which paves the way for the development of versatile phase-separating systems capable of addressing a variety of application scenarios. Here, we highlight the potential of constructing synthetic MLOs with programmable and functional properties. Notably, we organize how these synthetic membraneless compartments have been capitalized to manipulate enzymatic activities and metabolic reactions. The aim of this review is to inspire readerships to deeply comprehend the widespread roles of synthetic MLOs in the regulation enzymatic reactions and control of metabolic processes, and to encourage the rational design of controllable and functional membraneless compartments for a broad range of bioengineering applications.

5.
J Agric Food Chem ; 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38659344

RESUMO

Lacto-N-difucohexaose II (LNDFH II) is a typical fucosylated human milk oligosaccharide and can be enzymatically produced from lacto-N-tetraose (LNT) by a specific α1,3/4-fucosyltransferase from Helicobacter pylori DMS 6709, referred to as FucT14. Previously, we constructed an engineered Escherichia coli BL21(DE3) with a single plasmid for highly efficient biosynthesis of LNT. In this study, two additional plasmids harboring the de novo GDP-L-fucose pathway module and FucT14, respectively, were further introduced to construct the strain for successful biosynthesis of LNDFH II. FucT14 was actively expressed, and the engineered strain produced LNDFH II as the major product, lacto-N-fucopentaose (LNFP) V as the minor product, and a trace amount of LNFP II and 3-fucosyllactose as very minor products. Additional expression of the α1,3-fucosyltransferase FutM1 from a Bacteroidaceae bacterium from the gut metagenome could obviously enhance the LNDFH II biosynthesis. After optimization of induction conditions, the maximum titer reached 3.011 g/L by shake-flask cultivation. During the fed-batch cultivation, LNDFH II was highly efficiently produced with the highest titer of 18.062 g/L and the productivity yield of 0.301 g/L·h.

6.
Int J Biol Macromol ; 266(Pt 1): 130955, 2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38499120

RESUMO

Lacto-N-fucopentaose V (LNFP V) is a typical human milk pentasaccharide. Multi-enzymatic in vitro synthesis of LNFP V from lactose was reported, however, microbial cell factory approach to LNFP V production has not been reported yet. In this study, the biosynthetic pathway of LNFP V was examined in Escherichia coli. The previously constructed E. coli efficiently producing lacto-N-tetraose was used as the starting strain. GDP-fucose pathway module and a regio-specific glycosyltransferase with α1,3-fucosylation activity were introduced to realize the efficient synthesis of LNFP V. The α1,3/4-fucosyltransferase from Bacteroides fragilis was selected as the best enzyme for in vivo biosynthesis of LNFP V from nine candidates, with the highest titer and the lowest by-product accumulation. A beneficial variant K128D was obtained to further enhance LNFP V titer using computer-assisted site-directed mutagenesis. The final strain EW10 could produce 25.68 g/L LNFP V by fed-batch cultivation, with the productivity of 0.56 g/L·h.

7.
J Agric Food Chem ; 72(13): 7179-7186, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38520358

RESUMO

Lacto-N-neotetraose (LNnT), a prominent neutral human milk oligosaccharide (HMO), serves as a pivotal structural element in complex HMO biosynthesis. Given its promising health effects for infants, the biosynthesis of LNnT is garnering greater interest. Using a previously engineered strain as a chassis, a highly effective LNnT producer was constructed. First, LNnT synthesis in Escherichia coli MG1655 was achieved by introducing ß1,3-N-acetylglucosaminyltransferase LgtA and ß1,4-galactosyltransferase CpsIaJ, coupled with the optimization of enzyme expression levels using various promoters. Subsequently, ugd underwent disruption, and the galE gene was enhanced by replacing its promoter with PJ23119 or Ptac. Then, a lux-type quorum sensing (QS) system was applied to achieve varied metabolic regulation. Additionally, systematic optimization of the QS promoters was conducted to further improve the LNnT titer in the shake flask. Finally, the extracellular titer of LNnT was 20.33 g/L, accompanied by a productivity of 0.41 g/L/h.


Assuntos
Escherichia coli , Percepção de Quorum , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Oligossacarídeos/metabolismo , Leite Humano/química
8.
J Agric Food Chem ; 72(11): 5860-5866, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38452259

RESUMO

Lacto-N-biose I (LNB), termed a Type 1 disaccharide, is an important building block of human milk oligosaccharides. It shows promising prebiotic activity by stimulating the proliferation of many gut-associated bifidobacteria and thus displays good potential in infant foods or supplements. Enzymatic and microbial approaches to LNB synthesis have been studied, almost all of which involve glycosylation of LNB phosphorylase as the final step. Herein, we report a new and easier microbial LNB synthesis strategy through the route "lactose → lacto-N-triose II (LNTri II) → lacto-N-tetraose (LNT) → LNB". A previously constructed LNT-producing Escherichia coli BL21(DE3) strain was engineered for LNB biosynthesis by introducing Bifidobacterium bifidum LnbB. LNB was efficiently produced, accompanied by lactose regeneration. Genomic integration of key pathway genes related to LNTri II and LNT synthesis was performed to enhance LNB titers. The final engineered strain produced 3.54 and 26.88 g/L LNB by shake-flask and fed-batch cultivation, respectively.


Assuntos
Acetilglucosamina/análogos & derivados , Escherichia coli , Leite Humano , Lactente , Humanos , Leite Humano/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Lactose/metabolismo , Oligossacarídeos/metabolismo
9.
J Agric Food Chem ; 72(8): 4317-4324, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38364314

RESUMO

Lacto-N-fucopentaose I (LNFP I) has recently been approved as generally recognized as safe, demonstrating its great commercial potential in the food industry. Microbial synthesis through metabolic engineering strategies is an effective approach for large-scale production of LNFP I. Biosynthesis of LNFP I requires consideration of two key points: high titer with low byproduct 2'-fucosyllactose (2'-FL) generation and high purity with low lacto-N-triose II (LNTri II) and lacto-N-tetraose (LNT) residues. Herein, α1,2-fucosyltransferase from Thermoanaerobacterium sp. RBIITD was screened from 16 selected LNFP I-producing glycosyltransferase candidates, showing the highest in vivo LNFP I productivity. Chromosomal integration of wbgO enhanced the LNFP I production by improving the precursor conversion from LNTri II to LNT. The best engineered strain produced 4.42 and 35.1 g/L LNFP I in shake-flask and fed-batch cultivation, respectively. The residual LNTri II and LNT were eliminated by further cultivation with a recombinant strain coexpressing Bifidobacterium bifidum ß-N-acetylhexosaminidase and lacto-N-biosidase. A strategy for LNFP I biosynthesis with high yield and purity was finally realized, providing support for its practical application in large-scale production.


Assuntos
Glicosiltransferases , Leite Humano , Trissacarídeos , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Leite Humano/química , Oligossacarídeos/química
10.
Food Chem ; 440: 138250, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38154282

RESUMO

Carbohydrate-active enzymes are accountable for the synthesis and degradation of glycosidic bonds among diverse carbohydrates. Fructosyl-transferases represent a subclass of these enzymes, employing sucrose as a substrate to generate fructooligosaccharides (FOS) and fructan polymers. This category primarily includes levansucrase (LS, EC 2.4.1.10), inulosucrase (IS, EC 2.4.1.9), and ß-fructofuranosidase (Ffase, EC 3.2.1.26). These three enzymes possess a similar five-bladed ß-propeller fold and employ an anomer-retaining reaction mechanism mediated by nucleophiles, transition state stabilizers, and general acids/bases. However, they exhibit distinct product profiles, characterized by variations in linkage specificity and molecular mass distribution. Consequently, this article comprehensively explores recent advancements in the catalytic characteristics, structural features, reaction mechanisms, and product specificity of levansucrase, inulosucrase, and ß-fructofuranosidase (abbreviated as LS, IS, and Ffase, respectively). Furthermore, it discusses the potential for modifying catalytic properties and product specificity through structure-based design, which enables the rational production of custom fructan and FOS.


Assuntos
Hexosiltransferases , Transferases , Transferases/metabolismo , beta-Frutofuranosidase/metabolismo , Hexosiltransferases/metabolismo , Oligossacarídeos/metabolismo , Frutanos/metabolismo , Catálise , Sacarose/metabolismo , Especificidade por Substrato
11.
J Agric Food Chem ; 71(48): 18943-18952, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37990968

RESUMO

Zearalenone (ZEN) and its derivatives are prevalent contaminants in cereal crops. This study investigated a novel thermostable ZEN lactonase (ZENM) from Monosporascus sp. GIB2. ZENM demonstrated its highest activity at 60 °C, maintaining over 90% relative activity from 50 to 60 °C. Notably, efficient hydrolysis of ZEN and its two derivatives was achieved using ZENM, with specific activities of 333 U/mg for ZEN, 316 U/mg for α-zearalenol (α-ZOL), and 300 U/mg for α-zearalanol (α-ZAL). The activity of ZENM toward α-ZOL is noteworthy as most ZEN lactonases rarely achieve such a high degradation rate of α-ZOL. Based on the sequence-structure analysis, five residues (L123, G163, E171, S199, and S202) conserved in other ZEN lactonases were substituted in ZENM. Of interest was the G163S mutant in the cap domain that displayed enhanced activity toward α-ZOL compared to the wild-type enzyme. Notably, the mutant G163S exhibited higher catalytic activity toward α-ZOL (kcat/Km 0.223 min-1 µM-1) than ZEN (kcat/Km 0.191 min-1 µM-1), preferring α-ZOL as its optimum substrate. In conclusion, a thermostable ZEN lactonase has been reported, and the alteration of residue G163 in the cap domain has been shown to modify the substrate specificity of ZEN lactonase.


Assuntos
Zearalenona , Zeranol , Zearalenona/metabolismo , Especificidade por Substrato
12.
Crit Rev Food Sci Nutr ; : 1-14, 2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37819266

RESUMO

Glucobiose is a range of disaccharides consisting of two glucose molecules, generally including trehalose, kojibiose, sophorose, nigerose, laminaribiose, maltose, cellobiose, isomaltose, and gentiobiose. The difference glycosidic bonds of two glucose molecules result in the diverse molecular structures, physiochemical properties and physiological functions of these glucobioses. Some glucobioses are abundant in nature but have unconspicuous roles on health like maltose, whereas some rare glucobioses display remarkable biological effects. It is unpractical process to extract these rare glucobioses from natural resources, while biological synthesis is a feasible approach. Recently, the production and application of glucobiose have attracted considerable attention. This review provides a comprehensive overview of glucobioses, including their natural sources and physicochemical properties like structure, sweetness, digestive performance, toxicology, and cariogenicity. Specific enzymes used for the production of various glucobioses and fermentation production processes are summarized. Additionally, their versatile functions and broad applications are also introduced.

13.
Int J Biol Macromol ; 253(Pt 4): 126919, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37717863

RESUMO

Currently, alginate oligosaccharides (AOS) become attractive due to their excellent physiological effects. AOS has been widely used in food, pharmaceutical, and cosmetic industries. Generally, AOS can be produced from alginate using alginate lyase (ALyase) as the biocatalyst. However, most ALyase display poor thermostability. In this study, a thermostable ALyase from Paenibacillus sp. YN15 (Payn ALyase) was characterized. It belonged to the polysaccharide lyase (PL) 31 family and displayed poly ß-D-mannuronate (Poly M) preference. Under the optimum condition (pH 8.0, 55 °C, 50 mM NaCl), it exhibited maximum activity of 90.3 U/mg and efficiently degraded alginate into monosaccharides and AOS with polymerization (DP) of 2-4. Payn ALyase was relatively stable at 55 °C, but the thermostability dropped rapidly at higher temperatures. To further improve its thermostability, rational design mutagenesis was carried out based on a combination of FireProt, Consensus Finder, and PROSS analysis. Finally, a triple-point mutant K71P/Y129G/S213G was constructed. The optimum temperature was increased from 55 to 70 °C, and the Tm was increased from 62.7 to 64.1 °C. The residual activity after 30 min incubation at 65 °C was enhanced from 36.0 % to 83.3 %. This study provided a promising ALyase mutant for AOS industrial production.


Assuntos
Paenibacillus , Paenibacillus/genética , Paenibacillus/metabolismo , Proteínas de Bactérias/química , Alginatos/metabolismo , Especificidade por Substrato , Concentração de Íons de Hidrogênio , Temperatura , Polissacarídeo-Liases/química , Oligossacarídeos/metabolismo
14.
J Agric Food Chem ; 71(40): 14678-14686, 2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37773050

RESUMO

2'-Fucosyllactose (2'-FL), the most typical human milk oligosaccharide, is used as an additive in premium infant formula. Herein, we constructed two highly effective 2'-FL synthesis producers via a de novo GDP-fucose pathway from engineered Escherichia coli MG1655. First, lacZ and wcaJ, two competitive pathway genes, were disrupted to block the invalid consumption of lactose and GDP-fucose, respectively. Next, the lacY gene was strengthened by switching its native promoter to PJ23119. To enhance the supply of endogenous GDP-fucose, the promoters of gene clusters manC-manB and gmd-fcl were strengthened individually or in combination. Subsequently, chromosomal integration of a constitutive PJ23119 promoter-based BKHT expression cassette (PJ23119-BKHT) was performed in the arsB and recA loci. The most productive plasmid-based and plasmid-free strains produced 76.9 and 50.1 g/L 2'-FL by fed-batch cultivation, respectively. Neither of them generated difucosyl lactose nor 3-fucosyllactose as byproducts.

15.
J Agric Food Chem ; 71(33): 12511-12518, 2023 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-37555443

RESUMO

Lacto-N-tetraose (LNT) is an important neutral human milk oligosaccharide (HMO) and acts as a significant core structure for complex HMO biosynthesis. We previously achieved high-yield LNT biosynthesis (57.5 g/L) using fed-batch fermentation; however, residual lacto-N-triose II (LNTri II) was also found (21.58 g/L). Here, we re-engineered an efficient LNT-producing Escherichia coli with low LNTri II accumulation using genetically stable LNTri II-producing strains with a genomic insertion of lgtA (encoding ß1,3-N-acetylglucosaminyltransferase). Comparable and low titers of LNT (3.73-4.61 g/L) and LNTri II (0.33-0.63 g/L), respectively, were obtained by introducing ß1,3-galactosyltransferase. To reduce residual LNTri II, the E. coli transporter gene setA was disrupted, obviously reducing the accumulation of LNTri II and LNT. Next, the gene encoding ß-N-acetylhexosaminidase (BbhI) was introduced into LNT-producing strains or E. coli BL21(DE3) for single- or mixed-strain cultivation, respectively. Finally, LNT was obtained (30.13 g/L) in a cocultivation system of mixed engineered strains without undesired LNTri II.


Assuntos
Escherichia coli , Oligossacarídeos , Humanos , Escherichia coli/genética , Oligossacarídeos/química , Trissacarídeos/análise , Leite Humano/química
16.
J Agric Food Chem ; 71(33): 12390-12402, 2023 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-37552889

RESUMO

Human milk oligosaccharides (HMOs) have received increasing attention because of their special effects on infant health and commercial value as the new generation of core components in infant formula. Currently, large-scale production of HMOs is generally based on microbial synthesis using metabolically engineered cell factories. Introduction of the specific glycosyltransferases is essential for the construction of HMO-producing engineered strains in which the HMO-producing glycosyltransferases are generally sugar nucleotide-dependent. Four types of glycosyltransferases have been used for typical glycosylation reactions to synthesize HMOs. Soluble expression, substrate specificity, and regioselectivity are common concerns of these glycosyltransferases in practical applications. Screening of specific glycosyltransferases is an important research topic to solve these problems. Molecular modification has also been performed to enhance the catalytic activity of various HMO-producing glycosyltransferases and to improve the substrate specificity and regioselectivity. In this article, various sugar nucleotide-dependent glycosyltransferases for HMO synthesis were overviewed, common concerns of these glycosyltransferases were described, and the future perspectives of glycosyltransferase-related studies were provided.


Assuntos
Glicosiltransferases , Leite Humano , Lactente , Humanos , Leite Humano/química , Glicosiltransferases/genética , Glicosiltransferases/química , Oligossacarídeos/química , Glicosilação , Açúcares/metabolismo
17.
Crit Rev Food Sci Nutr ; : 1-13, 2023 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-37485919

RESUMO

Enzymes can produce high-quality food with low pollution, high function, high acceptability, and medical aid. However, most enzymes, in their native form, do not meet the industrial requirements. Sequence-based and structure-based methods are the two main strategies used for enzyme modification. Molecular Dynamics (MD) simulation is a sufficiently comprehensive technology, from a molecular perspective, which has been widely used for structure information analysis and enzyme modification. In this review, we summarize the progress and development of MD simulation, particularly for software, force fields, and a standard procedure. Subsequently, we review the application of MD simulation in various food enzymes for thermostability and catalytic improvement was reviewed in depth. Finally, the limitations and prospects of MD simulation in food enzyme modification research are discussed. This review highlights the significance of MD simulation and its prospects in food enzyme modification.

18.
Synth Syst Biotechnol ; 8(3): 509-519, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37502821

RESUMO

N-Acetylneuraminic acid (Neu5Ac), the most common type of Sia, generally acts as the terminal sugar in cell surface glycans, glycoconjugates, oligosaccharides, lipo-oligosaccharides, and polysaccharides, thus exerting numerous physiological functions. The extensive applications of Neu5Ac in the food, cosmetic, and pharmaceutical industries make large-scale production of this chemical desirable. Biosynthesis which is associated with important application potential and environmental friendliness has become an indispensable approach for large-scale synthesis of Neu5Ac. In this review, the physiological roles of Neu5Ac was first summarized in detail. Second, the safety evaluation, regulatory status, and applications of Neu5Ac were discussed. Third, enzyme-catalyzed preparation, whole-cell biocatalysis, and microbial de novo synthesis of Neu5Ac were comprehensively reviewed. In addition, we discussed the main challenges of Neu5Ac de novo biosynthesis, such as screening and engineering of key enzymes, identifying exporters of intermediates and Neu5Ac, and balancing cell growth and biosynthesis. The corresponding strategies and systematic strategies were proposed to overcome these challenges and facilitate Neu5Ac industrial-scale production.

19.
J Agric Food Chem ; 71(28): 10701-10709, 2023 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-37409796

RESUMO

N-Acetylneuraminic acid (NeuAc) is the predominant sialic acid found in human cells and a human-identical milk monosaccharide. Due to its numerous health benefits, it has great commercial potential in the pharmaceutical, cosmetic, and food industries. Microbial synthesis via metabolic engineering strategies is an important approach to its large-scale production. In this study, a NeuAc synthetic pathway was constructed in Escherichia coli BL21(DE3) by deleting the competitive pathway genes and introducing two genes encoding UDP-N-acetylglucosamine (GlcNAc) 2-epimerase (NeuC) and NeuAc synthase (NeuB). UDP-GlcNAc pathway genes, glmS, glmM, and glmU, were overexpressed to strengthen precursor supply for enhancement of NeuAc synthesis. The microbial source of neuC and neuB was optimized, and their expression was fine-tuned. In addition, glycerol as the carbon source showed a much better effect on NeuAc synthesis than glucose. The final engineered strain produced 7.02 g/L NeuAc by shake-flask cultivation. The titer was enhanced to 46.92 g/L by fed-batch cultivation, with the productivity of 0.82 g/L/h and 1.05 g/g DCW.


Assuntos
Acetilglucosamina , Ácido N-Acetilneuramínico , Humanos , Acetilglucosamina/metabolismo , Vias Biossintéticas , Escherichia coli/genética , Escherichia coli/metabolismo , Engenharia Metabólica , Difosfato de Uridina/metabolismo
20.
Crit Rev Food Sci Nutr ; : 1-13, 2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37341681

RESUMO

Human milk oligosaccharides (HMOs) have attracted considerable attention owing to their unique physiological functions. Two important tetrasaccharides, lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT), are core structures of HMOs. Their safety has been evaluated and they can be added to infant formula as functional ingredients. The fucosylated derivatives of LNT and LNnT, mainly lacto-N-fucopentaose (LNFP) I, LNFP II, LNFP III, and lacto-N-difucohexaose I, exhibit prominent physiological characteristics, including modificating the intestinal microbiota, immunomodulation, anti-bacterial activities, and antiviral infection. However, they have received lesser attention than 2'-fucosyllactose. As precursors, LNT and LNnT are connected to one or two fucosyl units through α1,2/3/4 glycosidic bonds, forming a series of compounds with complex structures. These complex fucosylated oligosaccharides can be biologically synthesized using enzymatic and cell factory approaches. This review summarizes the occurrence, physiological effects, and biosynthesis of fucosylated LNT and LNnT derivatives and their future development.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...