Elsevier

Metabolic Engineering

Volume 47, May 2018, Pages 374-382
Metabolic Engineering

Development of an Escherichia coli-based biocatalytic system for the efficient synthesis of N-acetyl-D-neuraminic acid

https://doi.org/10.1016/j.ymben.2018.04.012Get rights and content

Highlights

  • Altering the balance of transcription improved Neu5Ac synthesis.

  • Structure-guided protein engineering increased Neu5Ac productivity significantly.

  • System optimization yielded an efficient E. coli biocatalyst for Neu5Ac synthesis.

Abstract

N-acetyl-d-neuraminic acid (Neu5Ac) is a valuable resource that has seen increasing demand in both medicine and biotechnology. Although enzymatic systems and whole-cell biocatalysts have been developed for the synthesis of Neu5Ac, low yield and productivity still hamper the use of these methods on larger scales. We report the creation of an Escherichia coli biocatalyst for the efficient synthesis of Neu5Ac using a metabolic and protein engineering strategy. Expression of the two enzymes, N-acetyl-D-glucosamine 2-epimerase (AGE) and Neu5Ac lyase (NAL), was balanced using promoter engineering. Genes encoding competing pathways and GlcNAc catabolism were deleted, and then a structure-guided process was used to identify a more efficient NAL and an AGE mutant with a higher rate of Neu5Ac synthesis. The resulting biocatalyst produced 351.8 mM Neu5Ac with a yield of 58.6% from GlcNAc. This work exemplifies the use of rational design and protein engineering to construct a complex bacterial biocatalyst that can serve as a platform for the large-scale synthesis of a useful biological material.

Introduction

Sialic acids, derivatives of the nine-carbon monosaccharide neuraminic acid, are distributed widely in animal tissues and in other species ranging from plants and fungi to some microorganisms. N-acetyl-D-neuraminic acid (Neu5Ac), the main sialic acid in biological systems, plays important roles in biological, pathological, and immunological recognition processes, such as cell adhesion, cell signaling, glycoprotein stability, bacterial virulence, and tumor metastasis (Chen and Varki, 2010, Maru et al., 2002, Schauer, 2000). As a potential raw material for the synthesis of the antiviral medication Zanamivir, Neu5Ac can be used to prevent influenza type A and B infections (Tao et al., 2010). Neu5Ac is also an important additive in dairy products that can strengthen the immunity of infants (Bode, 2006). However, the high cost of Neu5Ac, which is caused by the conventional methods of its production, has impeded its industrial application.

Neu5Ac has been prepared by extraction from natural sources, such as egg yolk, and by hydrolysis of colominic acid (Maru et al., 2002). These methods are limited by low yield and unsatisfactory stereoselectivity. The chemical synthesis of Neu5Ac has been unsuitable for large-scale production because of its tedious protection and deprotection steps (Maru et al., 2002, Xu et al., 2007). In addition to the processes mentioned above, a two-enzyme, sequential system comprising N-acetylglucosamine 2-epimerase (AGE, EC 5.1.3.8) and N-acetylneuraminic acid lyase (NAL, EC 4.1.3.3) has been developed to produce Neu5Ac from N-acetylglucosamine (GlcNAc) (Gao et al., 2011, Lee et al., 2004, Schauer, 2000, Soundararajan et al., 2009, Traving and Schauer, 1998). The original process was hampered by a lengthy reaction time, low yield and low productivity. To improve synthesis performance, AGE and NAL were purified, immobilized, and coupled within one reactor to produce Neu5Ac from GlcNAc (Hu et al., 2010). The conversion rate from GlcNAc to Neu5Ac improved to 73% within 24 h. Moreover, a chemoenzymatic synthesis of Neu5Ac from GlcNAc using E. coli NAL (EcNAL) displayed on the surface of Bacillus subtilis WB600 spores was able to produce Neu5Ac with a concentration of 53.9 g L−1 (Gao et al., 2011). However, these enzymatic processes are complicated and costly because the enzymes must be partially or completely purified, and ATP is required to activate AGE.

Compared with the isolation and purification of enzymes, whole-cell biocatalysts can be more readily and inexpensively prepared and are particularly useful for multi-enzyme reactions or those that require cofactor regeneration (Both et al., 2016). Two different E. coli systems have been developed for Neu5Ac synthesis. The first system couples two separate engineered strains to produce Neu5Ac; one overexpressing AGE and the other EcNAL. In the system produced in our laboratory, which used an AGE from Synechocystis sp. and EcNAL, the overall process generated 260.0 mM Neu5Ac (80.4 g/L) with a conversion yield of 43.3% from GlcNAc (Zhou et al., 2016). Lee et al. (2007a, b) used recombinant E. coli BL21(DE3) strains expressing Anabaena sp. CH1 AGE, and EcNAL to produce 0.4 M Neu5Ac with a yield of 33.3% from GlcNAc (Lee et al., 2007a, b). Zhang et al. (2010) constructed two recombinant E. coli strains expressing the AGE from Synechocystis sp. PCC6803 and EcNAL using a temperature-responsive expression system. Their system produced 61.9 mM Neu5Ac from 200 mM GlcNAc. The second system uses a single strain expressing both AGE and EcNAL to synthesize Neu5Ac. Lin et al. developed a single-cell biocatalytic process that achieved a maximum titer of 240 mM Neu5Ac (74.2 g/L) with a productivity of 6.2 g/L·h−1 and a conversion yield of 40% from GlcNAc.(Lin et al., 2013) In similar processes reported by Tao et al. (2011) and Sun et al. (2013), the maximal Neu5Ac concentrations were 191.5 and 198.4 mM, respectively.

However, low conversion yields hamper biotechnological Neu5Ac production at larger scales. Because of the reversibility of AGE and NAL in the two-enzyme Neu5Ac synthesis cascade (Yu et al., 2004) (Fig. 1), systematic process improvement should receive more attention. These efforts should include screening for novel enzymes and achieving an elegant balance of enzyme expression. Here, we report the creation of a biocatalyst for the efficient synthesis of Neu5Ac using a strategy that combines rational metabolic engineering with protein engineering in E. coli BL21(DE3). Using the indicated strategy, which can facilitate the comprehensive evaluation of biocatalysts involving multiple genes, we developed a high-yield, high-productivity platform for Neu5Ac biosynthesis suitable for use in a 5-L biocatalytic reaction system (Fig. 1).

Section snippets

Strains, plasmids and chemicals

The E. coli strains and plasmids used in this study are listed in Supplementary materials Table S1. Molecular cloning and vector propagation were performed in E. coli JM109. All targeted genes were deleted from E. coli BL21(DE3). Vector pMD18-T (TaKaRa Biotech, China) was used for cloning of PCR products. Plasmids pET-28a and pKK223-3 were purchased from Invitrogen for gene expression. Restriction enzymes, T4 DNA ligase, DNA polymerase, DNA markers and protein molecular weight markers were

Optimization of AGE and EcNAL expression pattern

After the slr and nanA genes were inserted into pET28a and the resulting expression vectors were inserted into E. coli BL21(DE3), enzyme assay and SDS-PAGE analysis showed that both proteins could be successfully expressed (Figs. 2a and 2b; Supplementary materials Table S3). To generate Neu5Ac-producing strains for further analysis, two operons containing the slr and nanA genes, one in the order slr-nanA and the other in the order nanA-slr, under control of the T7 promoter were constructed.

Conclusion

A new whole-cell biocatalysis process that produced 351.8 mM Neu5Ac with a yield of 58.6% from GlcNAc has been developed. This biocatalyst, which is based on E. coli, contains an expression plasmid containing two cistrons: one containing a codon-optimized synthetic gene encoding S. hominis NAL under control of the T7 promoter and the other containing a codon-optimized synthetic gene encoding the C372A mutant of Synechocystis sp. PCC 6803 AGE. Further improvements included elimination of the

Acknowledgments

We thank the Fundamental Research Funds for the Central Universities (no. JUSRP51611A), the Natural Science Foundation of Jiangsu province (no. BK20171138), the 111 Project (No. 111-2-06), Top-notch Academic Programs Project of Jiangsu Higher Education Institutions and the 863 program (no. 2013AA102101-5) for financial support. The work was carried out at the National Supercomputer Center in LvLiang of China, and the calculations were performed on TianHe-2.

Supplementary materials

The Supplementary materials include experimental methods of homology modeling and molecular docking, five figures, four tables, and text showing strains and plasmids (Table S1), oligonucleotides (Table S2), enzyme activity of AGE and NAL (Table S3), activity for wild-type and mutant AGE enzymes (Table S4), physical maps of the recombinant plasmids (Figure S1), optimization of process for Neu5Ac synthesis (Figure S2), sequence alignment of EcNAL and ShNAL (Figure S3), the time dependence of RMSD

References (32)

  • W. Sun et al.

    Construction and expression of a polycistronic plasmid encoding N-acetylglucosamine 2-epimerase and N-acetylneuraminic acid lyase simultaneously for production of N-acetylneuraminic acid

    Bioresour. Technol.

    (2013)
  • H. Yu et al.

    Chemoenzymatic synthesis of CMP–sialic acid derivatives by a one-pot two-enzyme system: comparison of substrate flexibility of three microbial CMP–sialic acid synthetases

    Bioorg. Med. Chem.

    (2004)
  • J.B. Zhou et al.

    Enhanced production of N-acetyl-D-neuraminic acid by whole-cell bio-catalysis of Escherichia coli

    J. Mol. Catal. B-Enzym

    (2016)
  • S. Blayer et al.

    Characterization of the chemoenzymatic synthesis of N-Acetyl-D-neuraminic acid (Neu5Ac)

    Biotechnol. Prog.

    (1996)
  • P. Both et al.

    Whole-cell biocatalysts for stereoselective C-H amination reactions

    Angew. Chem.

    (2016)
  • X. Chen et al.

    Advances in the biology and chemistry of sialic acids

    ACS Chem. Biol.

    (2010)
  • Cited by (19)

    • Enzyme engineering for biosynthetic cascades

      2021, Current Opinion in Green and Sustainable Chemistry
      Citation Excerpt :

      Using a combination of structure- and homology-guided engineering, the authors identified an improved variant that was 12-fold improved in specific activity, allowing substantial improvements in titer [4]. One notable feature of several in vivo cascades is the use of protein engineering to improve the catalytic rate on the native substrate of an enzyme [4,5,8]. Although natural evolution has optimized the catalytic efficiency of some enzymes on their native substrates, improvements in the catalytic rate can nevertheless be obtained in many cases, which is consistent with previously advanced notions of ‘moderately’ efficient enzymes whose catalytic potential is not fully optimized by natural selection [18,19].

    • An overview and future prospects of sialic acids

      2021, Biotechnology Advances
      Citation Excerpt :

      Meanwhile, in our previous work, we used a metabolic and protein engineering strategy to construct an efficient E. coli biocatalyst of Neu5Ac production, including expressing GlcNAc 2-epimerase and Neu5Ac lyase via promoter engineering, deleting competing pathways and GlcNAc catabolism genes, and improving the catalytic activities of GlcNAc 2-epimerase and Neu5Ac lyase by a structure-guided process (Chen et al., 2018). The Neu5Ac yield achieved by this E. coli biocatalyst reached 351.8 mM with GlcNAc as substrate (Chen et al., 2018). Whole-cell biotransformation for Neu5Ac production also has some disadvantages, including expensive substrates (e.g., GlcNAc and ManNAc), low quantities of products, poor conversion rates (e.g., <60% from GlcNAc), complex processes (e.g., cell collection and resuspension), and mass transfer resistance from the cell membrane, all of which may inhibit the industrial application of whole-cell biotransformation for Neu5Ac production (Tao et al., 2011; Zhang et al., 2019b).

    • Microbial production of sialic acid and sialylated human milk oligosaccharides: Advances and perspectives

      2019, Biotechnology Advances
      Citation Excerpt :

      Compared with extraction and chemical synthesis, the biological production of NeuAc, which includes enzyme catalysis, whole-cell catalysis, and de novo biosynthesis, offers many advantages, such as mild conditions and low cost (Comb and Roseman, 1960; Gao et al., 2011; Kang et al., 2012). In recent years, applications in metabolic engineering, protein engineering, and synthetic biology techniques have facilitated methods for NeuAc biosynthesis (Chen et al., 2018; Peters et al., 2018; Yang et al., 2017). Although whole-cell catalysis for NeuAc production have been previously summarized, a systematic discussion on recent advances for the production of NeuAc and sialyllactose fueled by metabolic engineering strategies, enzymatic engineering, and synthetic biology techniques is lacking and therefore relevant (Tao et al., 2010).

    View all citing articles on Scopus
    View full text