Modular pathway engineering of key carbon‐precursor supply‐pathways for improved N‐acetylneuraminic acid production in Bacillus subtilis

N‐acetylneuraminic acid (NeuAc) is widely used as a nutraceutical for facilitating infant brain development, maintaining brain health, and enhancing immunity. Currently, NeuAc is mainly produced by extraction from egg yolk and milk, or via chemical synthesis. However, its low concentration in natural resources and its non‐ecofriendly chemical synthesis result in insufficient NeuAc production and environmental pollution, respectively. In this study, improved NeuAc production was attained via modular pathway engineering of the supply pathways of two key precursors—N‐acetylglucosamine (GlcNAc) and phosphoenolpyruvate (PEP)—and by balancing NeuAc biosynthesis and cell growth in engineered Bacillus subtilis. Specifically, we used a previously constructed GlcNAc‐producing B. subtilis as the initial host for NeuAc biosynthesis. First, we constructed a de novo NeuAc biosynthetic pathway utilizing glucose by coexpressing glucosamine‐6‐phosphate acetyl‐transferase (GNA1), N‐acetylglucosamine 2‐epimerase (AGE), and N‐acetylneuraminic acid synthase (NeuB), resulting in 0.33 g/l NeuAc production. Next, to balance the supply of the two key precursors for NeuAc biosynthesis, modular pathway engineering was performed. The optimal strategy for balancing the GlcNAc module and PEP supply module involved the use of an engineered, unique glucose and malate coutilization pathway in B. subtilis, supplied with both glucose (for the GlcNAc moiety) and malate (for the PEP moiety) at high strength. This led to 1.65 g/L NeuAc production, representing a 5.0‐fold improvement over the existing methods. Furthermore, to enhance the NeuAc yield on cell, glucose and malate coutilization pathways were engineered to balance NeuAc biosynthesis and cell growth via the blocking of glycolysis, the introduction of the Entner–Doudoroff pathway, and the overexpression of the malic enzyme YtsJ. NeuAc titer reached 2.18 g/L, with 0.38 g/g dry cell weight NeuAc yield on cell, which represented a 1.32‐fold and 2.64‐fold improvement over the existing methods, respectively. The strategy of modular pathway engineering of key carbon precursor supply pathways via engineering of the unique glucose‐malate coutilization pathway in B. subtilis should be generically applicable for engineering of B. subtilis for the production of other important biomolecules. Our study also provides a good starting point for further metabolic engineering to achieve industrial production of NeuAc by a Generally Regarded As Safe bacterial strain.

[1]  Min-Kyu Oh,et al.  A synthetic suicide riboswitch for the high-throughput screening of metabolite production in Saccharomyces cerevisiae. , 2015, Metabolic engineering.

[2]  A. Yang,et al.  A practical synthesis of zanamivir phosphonate congeners with potent anti-influenza activity. , 2011, Journal of the American Chemical Society.

[3]  Uwe Sauer,et al.  The PEP-pyruvate-oxaloacetate node as the switch point for carbon flux distribution in bacteria. , 2005, FEMS microbiology reviews.

[4]  Xin Yan,et al.  Cre/lox System and PCR-Based Genome Engineering in Bacillus subtilis , 2008, Applied and Environmental Microbiology.

[5]  Guocheng Du,et al.  Metabolic engineering of Bacillus subtilis fueled by systems biology: Recent advances and future directions. , 2017, Biotechnology advances.

[6]  Kam Y. Lau,et al.  Molecular characterization of a new N-acetylneuraminate synthase ( NeuB 1 ) from Idiomarina loihiensis , 2014 .

[7]  Xiaolin Shen,et al.  Establishing a synergetic carbon utilization mechanism for non-catabolic use of glucose in microbial synthesis of trehalose. , 2017, Metabolic engineering.

[8]  Kristala L J Prather,et al.  Retro-biosynthetic screening of a modular pathway design achieves selective route for microbial synthesis of 4-methyl-pentanol , 2014, Nature Communications.

[9]  Shuchi H. Desai,et al.  Two-dimensional isobutyl acetate production pathways to improve carbon yield , 2015, Nature Communications.

[10]  Recruiting alternative glucose utilization pathways for improving succinate production , 2013, Applied Microbiology and Biotechnology.

[11]  Dian Donnai,et al.  NANS-mediated synthesis of sialic acid is required for brain and skeletal development , 2016, Nature Genetics.

[12]  M. Inui,et al.  Metabolic engineering of Corynebacterium glutamicum for shikimate overproduction by growth-arrested cell reaction. , 2016, Metabolic engineering.

[13]  J. Stelling,et al.  Transcriptional regulation is insufficient to explain substrate-induced flux changes in Bacillus subtilis , 2013, Molecular systems biology.

[14]  Qian Wang,et al.  Pathway optimization and key enzyme evolution of N-acetylneuraminate biosynthesis using an in vivo aptazyme-based biosensor. , 2017, Metabolic engineering.

[15]  Chiam Yu Ng,et al.  Rational design of a synthetic Entner-Doudoroff pathway for improved and controllable NADPH regeneration. , 2015, Metabolic engineering.

[16]  Hongwu Ma,et al.  Engineering of Serine-Deamination pathway, Entner-Doudoroff pathway and pyruvate dehydrogenase complex to improve poly(3-hydroxybutyrate) production in Escherichia coli , 2014, Microbial Cell Factories.

[17]  Joerg M. Buescher,et al.  Metabolic Fluxes during Strong Carbon Catabolite Repression by Malate in Bacillus subtilis* , 2009, The Journal of Biological Chemistry.

[18]  W. Hsu,et al.  Production of N-acetyl-D-neuraminic acid by recombinant whole cells expressing Anabaena sp. CH1 N-acetyl-D-glucosamine 2-epimerase and Escherichia coli N-acetyl-D-neuraminic acid lyase. , 2007, Journal of biotechnology.

[19]  A. Smalås,et al.  Characterization of the N-acetylneuraminic acid synthase (NeuB) from the psychrophilic fish pathogen Moritella viscosa. , 2015, Carbohydrate research.

[20]  Y. Tsukada,et al.  Simple and large-scale production of N-acetylneuraminic acid from N-acetyl-D-glucosamine and pyruvate using N-acyl-D-glucosamine 2-epimerase and N-acetylneuraminate lyase. , 1998, Carbohydrate research.

[21]  Irnov Irnov,et al.  Mechanism of mRNA destabilization by the glmS ribozyme. , 2007, Genes & development.

[22]  J. Magano Synthetic approaches to the neuraminidase inhibitors zanamivir (Relenza) and oseltamivir phosphate (Tamiflu) for the treatment of influenza. , 2009, Chemical reviews.

[23]  B. Wang Sialic acid is an essential nutrient for brain development and cognition. , 2009, Annual review of nutrition.

[24]  Yinjie J. Tang,et al.  Examining Escherichia coli glycolytic pathways, catabolite repression, and metabolite channeling using Δpfk mutants , 2016, Biotechnology for Biofuels.

[25]  Qian Wang,et al.  Engineering of an N-acetylneuraminic acid synthetic pathway in Escherichia coli. , 2012, Metabolic engineering.

[26]  Xiao‐Zhou Zhang,et al.  Simple, fast and high‐efficiency transformation system for directed evolution of cellulase in Bacillus subtilis , 2010, Microbial biotechnology.

[27]  H. van Loveren,et al.  Safety of synthetic N‐acetyl‐d‐neuraminic acid as a novel food pursuant to Regulation (EC) No 258/97 , 2017, EFSA journal. European Food Safety Authority.

[28]  Guocheng Du,et al.  Modular pathway engineering of Bacillus subtilis for improved N-acetylglucosamine production. , 2014, Metabolic engineering.

[29]  E. Vimr,et al.  Purification and characterization of the Escherichia coli K1 neuB gene product N-acetylneuraminic acid synthetase. , 1997, Glycobiology.

[30]  Jamie H. D. Cate,et al.  Enhanced biofuel production through coupled acetic acid and xylose consumption by engineered yeast , 2013, Nature Communications.

[31]  D. M. Ryan,et al.  Rational design of potent sialidase-based inhibitors of influenza virus replication , 1993, Nature.

[32]  N. Strynadka,et al.  Structural and Mechanistic Analysis of Sialic Acid Synthase NeuB from Neisseria meningitidis in Complex with Mn2+, Phosphoenolpyruvate, and N-Acetylmannosaminitol* , 2005, Journal of Biological Chemistry.

[33]  Guocheng Du,et al.  Spatial modulation of key pathway enzymes by DNA-guided scaffold system and respiration chain engineering for improved N-acetylglucosamine production by Bacillus subtilis. , 2014, Metabolic engineering.

[34]  C. Boddy,et al.  Total Biosynthesis of Legionaminic Acid, a Bacterial Sialic Acid Analogue. , 2016, Angewandte Chemie.

[35]  Z. Cui,et al.  High-Level Expression and Secretion of Methyl Parathion Hydrolase in Bacillus subtilis WB 800 , 2005 .

[36]  K. Ochi,et al.  Glucose Uptake Pathway-Specific Regulation of Synthesis of Neotrehalosadiamine, a Novel Autoinducer Produced in Bacillus subtilis , 2006, Journal of bacteriology.

[37]  Guocheng Du,et al.  Pathway engineering of Bacillus subtilis for microbial production of N-acetylglucosamine. , 2013, Metabolic engineering.

[38]  L. I. Hallay Treatment of influenza. , 1966, Delaware medical journal.

[39]  W. Zhuang,et al.  Efficient immobilization of AGE and NAL enzymes onto functional amino resin as recyclable and high-performance biocatalyst , 2017, Bioprocess and Biosystems Engineering.

[40]  Long Liu,et al.  Microbial production of glucosamine and N-acetylglucosamine: advances and perspectives , 2013, Applied Microbiology and Biotechnology.

[41]  W. Zhuang,et al.  In Vivo Multienzyme Complex Coconstruction of N-Acetylneuraminic Acid Lyase and N-Acetylglucosamine-2-epimerase for Biosynthesis of N-Acetylneuraminic Acid. , 2017, Journal of agricultural and food chemistry.

[42]  Fuzhong Zhang,et al.  Modular pathway engineering for the microbial production of branched-chain fatty alcohols , 2017, Biotechnology for Biofuels.

[43]  M. Domach,et al.  Characterization of Growth and Acid Formation in a Bacillus subtilis Pyruvate Kinase Mutant , 2000, Applied and Environmental Microbiology.

[44]  R. Milo,et al.  Glycolytic strategy as a tradeoff between energy yield and protein cost , 2013, Proceedings of the National Academy of Sciences.

[45]  C. Boddy,et al.  Sialic acid and N-acyl sialic acid analog production by fermentation of metabolically and genetically engineered Escherichia coli. , 2007, Organic & biomolecular chemistry.

[46]  Z. Cui,et al.  High-Level Expression and Secretion of Methyl Parathion Hydrolase in Bacillus subtilis WB800 , 2005, Applied and Environmental Microbiology.

[47]  M. Hecker,et al.  Bacillus subtilis: from soil bacterium to super-secreting cell factory , 2013, Microbial Cell Factories.

[48]  Kam Y. Lau,et al.  Molecular characterization of a new N-acetylneuraminate synthase (NeuB1) from Idiomarina loihiensis. , 2015, Glycobiology.