Elsevier

Biotechnology Advances

Volume 43, 1 November 2020, 107575
Biotechnology Advances

Research review paper
Coenzyme A thioester-mediated carbon chain elongation as a paintbrush to draw colorful chemical compounds

https://doi.org/10.1016/j.biotechadv.2020.107575Get rights and content

Abstract

The biosynthesis of various useful chemicals from simple substrates using industrial microorganisms is becoming increasingly crucial to address the challenge of dwindling non-renewable resources. As the most common intermediate substrates in organisms, Coenzyme A (CoA) thioesters play a central role in the carbon chain elongation process of their products. As a result, numerous of chemicals can be synthesized by the iterative addition of various CoA thioester extender units at a given CoA thioester primer backbone. However, these elongation reactions and the product yields are still restricted due to the low enzymatic performance and supply of CoA thioesters. This review highlights the current protein and metabolic engineering strategies used to enhance the diversity and product yield by coupling different primers, extender units, enzymes, and termination pathways, in an attempt to provide a road map for producing a more diverse range of industrial chemicals.

Introduction

Csingle bondC bond forming reactions are critical steps in synthetic organic chemistry to construct the carbon framework of organic molecules.(Fesko and Gruber-Khadjawi, 2013; Lee et al., 2019) As an alternative, biological reactions carry out carbon chain elongation through the addition of various extender units to a carbon backbone have been exploited for the production of phenylpropanoids (Kallscheuer et al., 2016; Zhou et al., 2019), antibiotics (Go et al., 2015), organic acids (Zhao et al., 2018a; Zhao et al., 2018b) and alcohols(Ohtake et al., 2017; Yu et al., 2018). Furthermore, improving the diversity of biosynthesis will contribute to solving the decreasing energy resources and increasingly severe environmental pollution problems. Cellular anabolism beginning from a small starter molecule (such as CO2, formyl-CoA, acetyl-CoA and malonyl-CoA) and the addition of various carbon backbones at different positions is a promising strategy that leads to numerous chemicals (Cheong et al., 2016). Consequently, bio-based carbon chain elongation strategies are becoming increasingly popular.

In general, coenzyme A (CoA) and its thioesters are possibly the most common intermediate metabolites observed in cellular processes. Approximately 4% of the enzymes in all living cells are CoA dependent enzymes (Harijan, 2017). Hence, CoA thioester-mediated carbon rearrangements are very popular in nature (Cheong et al., 2016; Haapalainen et al., 2006). The structure and high nucleophilicity of CoA allow it to act as a nucleophile and react with carboxyl-containing molecules to generate its thioester derivatives (Gulick, 2009). Subsequently, the α‑carbon of CoA thioesters can be activated into α-carbanion by thiolases (Meriläinen et al., 2009; Meriläinen et al., 2008) and polyketide synthases (PKSs) (Heath and Rock, 2002). The activated α-carbanion could react with another molecule of a CoA thioester to extend the carbon chain. In general, the two thioesters used in the carbon chain elongation are defined as the “primer” and “extender” unit, respectively (Fig. 1) (Cheong et al., 2016). The extender unit supplies the α-carbanion to perform the nucleophilic attack on the acyl group of the primer unit. Consequently, a vast array of chemicals can be synthesized by combining different primers and extender units with Claisen-condensation enzymes (Fig. 1).

As described above, the enzymes that catalyze the Claisen-condensation reaction between the primer and extender units usually act as “gatekeepers” to control the diversity of the products formed. Generally, these enzymes can be simply separated into four categories: Thiolases, PKSs, carboxylases, and the other enzymes. The compatible substrates of these enzymes span a broad range from CO2 and formyl-CoA to bulky acyl/aryl-CoA (Chou et al., 2019; Haapalainen et al., 2006; Musiol-Kroll and Wohlleben, 2018; Palmer and Alper, 2019; Peter et al., 2015). Hence, a diverse range of either natural or non-natural products can be synthesized based on these enzymes. Specifically, when acetyl-CoA, malonyl-CoA and their analogues are used as extender units, iterative Claisen-condensation (ICC) reactions can further extend the diversity of the products by continuously adding them onto the end of the primer units (Kallscheuer et al., 2017). To terminate the ICC reaction, various termination pathways are coupled and to further enhance the diversity of the products formed. Overall, combining different primer and extender units as well as Claisen-condensation enzymes through a variety of ICC pathways and termination pathways can produce a large number of natural or non-natural products (Fig. 1).

Although there are a number of reviews on Csingle bondC bond formation, few of them have focused on carbon chain elongation using CoA thioesters (Fesko and Gruber-Khadjawi, 2013; Lee et al., 2019). Herein, we thoroughly examine the current status of CoA thioester-mediated carbon chain elongation. We describe the enzymes, pathways and strategies used to produce a diverse range of products. Likewise, we also emphasize the strategies used to improve the titer of the extender units as they relate to cell growth and the final titer of the products (Krivoruchko et al., 2015; Matsumoto et al., 2017; Takamura and Nomura, 1988).

Section snippets

CoA and its thioesters

CoA is an essential and ubiquitous cofactor in all living cells, which plays a crucial role in cellular catabolism and anabolism by controlling the enzymes activity and intermediate metabolism (Gout, 2018; Theodoulou et al., 2014). The stability of CoA and high reactivity of CoA thioesters determine their important functions in metabolic processes. Herein, we describe the biochemical properties and synthetic pathways of CoA and its thioester derivatives.

Enzymes in CoA thioester-mediated carbon chain elongation

Acyl/aryl-CoAs can be separated into two categories, primer and extender units. Based on enzyme-specific Claisen-condensation reactions, the primer and extender units can be used in a tremendous number of combinations. Hence, the enzymes involved in CoA thioester-mediated carbon chain elongation are crucial toward generating a diverse range of products (Fig. 1). Herein, we highlight the catalytic mechanisms and engineering strategies of these enzymes.

Metabolic engineering to improve the diversity and productivity of the products

By protein engineering, novel enzyme mutants were created, which enable generate greater product diversity. Furthermore, the concentration of the extender units, especially acetyl-CoA, malonyl-CoA and some branched extender units, directly determined the titer of target compounds (Krink-Koutsoubelis et al., 2018; Takamura and Nomura, 1988). Herein, we describe the strategies used to enhance the diversity and productivity of the target compounds.

Conclusion and prospects

Overall, CoA thioesters are core compounds in carbon chain elongation reactions. From starting molecules, such as CO2, acetyl-CoA and malonyl-CoA, a vast array of functionalized products can be synthesized based on flexible carbon chain elongation and termination pathways. To further enhance the diversity and productivity, protein and metabolic engineering strategies have been thoroughly explored to modify the substrate scope, enhance the catalytic properties, maintain the CoA balance and

Declaration of Competing Interest

The authors declare no competing financial interests.

Acknowledgements

This work was supported by the National Key R&D Program of China (2019YFA0905502), the National Natural Science Foundation of China (31900066, 21877053), the National First-class Discipline Program of Light Industry Technology and Engineering (LITE2018-24), the Top-Notch Academic Programs Project of Jiangsu Higher Education Institutions (TAPP) and the Fundamental Research Funds for the Central Universities (JUSRP51705A, JUSRP12056).

References (139)

  • M.E. Horsman et al.

    Polyketide synthase and non-ribosomal peptide synthetase thioesterase selectivity: logic gate or a victim of fate?

    Nat. Prod. Rep.

    (2016)
  • M. Jeschek et al.

    Biotin-independent strains of Escherichia coli for enhanced streptavidin production

    Metab. Eng.

    (2017)
  • N. Kallscheuer et al.

    Construction of a Corynebacterium glutamicum platform strain for the production of stilbenes and (2S)-flavanones

    Metab. Eng.

    (2016)
  • N. Kallscheuer et al.

    Reversal of β-oxidative pathways for the microbial production of chemicals and polymer building blocks

    Metab. Eng.

    (2017)
  • B. Khanppnavar et al.

    Genome-wide survey and crystallographic analysis suggests a role for both horizontal gene transfer and duplication in pantothenate biosynthesis pathways

    Biochim. Biophys. Acta Gen. Subj.

    (2019)
  • M. Klaus et al.

    Engineering strategies for rational polyketide synthase design

    Nat. Prod. Rep.

    (2018)
  • A. Krivoruchko et al.

    Microbial acetyl-CoA metabolism and metabolic engineering

    Metab. Eng.

    (2015)
  • R. Leonardi et al.

    Coenzyme A: back in action

    Prog. Lipid Res.

    (2005)
  • Z. Li et al.

    Structural basis for specificity and flexibility in a plant 4-coumarate:CoA ligase

    Structure

    (2015)
  • M.J. Li et al.

    De novo production of resveratrol from glucose or ethanol by engineered Saccharomyces cerevisiae

    Metab. Eng.

    (2015)
  • W.S. Liu et al.

    Improving acetyl-CoA biosynthesis in Saccharomyces cerevisiae via the overexpression of pantothenate kinase and PDH bypass

    Biotechnol. Biofuels.

    (2017)
  • T. Matsumoto et al.

    Engineering metabolic pathways in Escherichia coli for constructing a “microbial chassis” for biochemical production

    Bioresour. Technol.

    (2017)
  • Y. Miao et al.

    Recent developments in enzyme promiscuity for carbon–carbon bond-forming reactions

    Curr. Opin. Chem. Biol.

    (2015)
  • D.C. Monteiro et al.

    The structure of the PanD/PanZ protein complex reveals negative feedback regulation of pantothenate biosynthesis by coenzyme A

    Chem. Biol.

    (2015)
  • T. Ohtake et al.

    Metabolomics-driven approach to solving a CoA imbalance for improved 1-butanol production in Escherichia coli

    Metab. Eng.

    (2017)
  • X. Qian et al.

    Alcohol acyltransferase gene and ester precursors differentiate composition of volatile esters in three interspecific hybrids of Vitis labrusca× V. vinifera during berry development period

    Food Chem.

    (2019)
  • L. Ray et al.

    Recent advances in the biosynthesis of unusual polyketide synthase substrates

    Nat. Prod. Rep.

    (2016)
  • K.S. Shin et al.

    Introduction of an acetyl-CoA carboxylation bypass into Escherichia coli for enhanced free fatty acid production

    Bioresour. Technol.

    (2017)
  • J.L. Smith et al.

    Architecture of the polyketide synthase module: surprises from electron cryo-microscopy

    Curr. Opin. Struc. Biol.

    (2015)
  • A. Suyama et al.

    Production of 3-hydroxypropionic acid via the malonyl-CoA pathway using recombinant fission yeast strains

    J. Biosci. Bioeng.

    (2017)
  • T. Annaval et al.

    Evaluating ketoreductase exchanges as a means of rationally altering polyketide stereochemistry

    ChemBioChem

    (2015)
  • R.N. Armstrong

    Structure, catalytic mechanism, and evolution of the glutathione transferases

    Chem. Res. Toxicol.

    (1997)
  • C. Bathellier et al.

    Rubisco is not really so bad

    Plant Cell Environ.

    (2018)
  • C.L. Bayly et al.

    Towards precision engineering of canonical polyketide synthase domains: recent advances and future prospects

    Molecules

    (2017)
  • A. Bernard et al.

    Reconstitution of plant alkane biosynthesis in yeast demonstrates that Arabidopsis ECERIFERUM1 and ECERIFERUM3 are core components of a very-long-chain alkane synthesis complex

    Plant Cell

    (2012)
  • M.R. Blaisse et al.

    Discovery and engineering of pathways for production of α-branched organic acids

    J. Am. Chem. Soc.

    (2017)
  • I.W. Bogorad et al.

    Synthetic non-oxidative glycolysis enables complete carbon conservation

    Nature

    (2013)
  • B.B. Bond-Watts et al.

    Enzyme mechanism as a kinetic control element for designing synthetic biofuel pathways

    Nat. Chem. Biol.

    (2011)
  • B.M. Bonk et al.

    Rational design of thiolase substrate specificity for metabolic engineering applications

    Biotechnol. Bioeng.

    (2018)
  • T. Bretschneider et al.

    A ketosynthase homolog uses malonyl units to form esters in cervimycin biosynthesis

    Nat. Chem. Biol.

    (2012)
  • Y.J. Cao et al.

    Metabolic engineering of Escherichia coli for the production of hydroxy fatty acids from glucose

    BMC Biotechnol.

    (2016)
  • W. Cao et al.

    Enhanced pinocembrin production in Escherichia coli by regulating cinnamic acid metabolism

    Sci. Rep.

    (2016)
  • M. Casteels et al.

    The role of 2-hydroxyacyl-CoA lyase, a thiamin pyrophosphate-dependent enzyme, in the peroxisomal metabolism of 3-methyl-branched fatty acids and 2-hydroxy straight-chain fatty acids

    Biochem. Soc. T.

    (2007)
  • J.A. Chemler et al.

    Evolution of efficient modular polyketide synthases by homologous recombination

    J. Am. Chem. Soc.

    (2015)
  • S. Cheong et al.

    Energy-and carbon-efficient synthesis of functionalized small molecules in bacteria using non-decarboxylative Claisen condensation reactions

    Nat. Biotechnol.

    (2016)
  • A. Chou et al.

    2-Hydroxyacyl-CoA lyase catalyzes acyloin condensation for one-carbon bioconversion

    Nat. Chem. Biol.

    (2019)
  • J.M. Clomburg et al.

    The isoprenoid alcohol pathway, a synthetic route for isoprenoid biosynthesis

    Proc. Natl. Acad. Sci. U. S. A.

    (2019)
  • L.J. Duan et al.

    Biosynthesis and engineering of kaempferol in Saccharomyces cerevisiae

    Microb. Cell Factories

    (2017)
  • T.J. Erb et al.

    Synthesis of C-5-dicarboxylic acids from C-2-units involving crotonyl-CoA carboxylase/reductase: the ethylmalonyl-CoA pathway

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • T. Fehér et al.

    A sense of balance: experimental investigation and modeling of a malonyl-CoA sensor in Escherichia coli

    Front. Bioeng. Biotechnol.

    (2015)
  • Cited by (15)

    • Development of a growth coupled and multi-layered dynamic regulation network balancing malonyl-CoA node to enhance (2S)-naringenin biosynthesis in Escherichia coli

      2021, Metabolic Engineering
      Citation Excerpt :

      In comparison, repression of the activity of ACP in the present study exhibited a comparable result, which yielded a 7.5-fold improvement in malonyl-CoA production (Fig. 4). However, repression of fatty acid synthetic pathway significantly impaired the cellular growth (Wu et al., 2015; Xu et al., 2014; Zhou et al., 2020a). Hence, a dynamically regulated genetic circuit is essential for balancing the cell growth and production.

    View all citing articles on Scopus
    View full text