Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

Overcoming genetic heterogeneity in industrial fermentations

Abstract

Engineering the synthesis of massive amounts of therapeutics, enzymes or commodity chemicals can select for subpopulations of nonproducer cells, owing to metabolic burden and product toxicity. Deep DNA sequencing can be used to detect undesirable genetic heterogeneity in producer populations and diagnose associated genetic error modes. Hotspots of genetic heterogeneity can pinpoint mechanisms that underlie load problems and product toxicity. Understanding genetic heterogeneity will inform metabolic engineering and synthetic biology strategies to minimize the emergence of nonproducer mutants in scaled-up fermentations and maximize product quality and yield.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Evolution of populations during industrial fermentation.
Fig. 2: Causal factors and counterstrategies for genetic heterogeneity in large-scale biomanufacturing.
Fig. 3: Synthetic control solutions.

Similar content being viewed by others

References

  1. Glick, B. R. Metabolic load and heterologous gene expression. Biotechnol. Adv. 13, 247–261 (1995).

    Article  CAS  PubMed  Google Scholar 

  2. Nielsen, J. & Keasling, J. Engineering cellular metabolism. Cell 164, 1185–1197 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Lee, S. Y. & Kim, H. U. Systems strategies for developing industrial microbial strains. Nat. Biotechnol. 33, 1061–1072 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Van Dien, S. From the first drop to the first truckload: Commercialization of microbial processes for renewable chemicals. Curr. Opin. Biotechnol. 24, 1061–1068 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Burgard, A., Burk, M. J., Osterhout, R., Van Dien, S. & Yim, H. Development of a commercial scale process for production of 1,4-butanediol from sugar. Curr. Opin. Biotechnol. 42, 118–125 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Leavell, M. D., McPhee, D. J. & Paddon, C. J. Developing fermentative terpenoid production for commercial usage. Curr. Opin. Biotechnol. 37, 114–119 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Newbert, R. W., Barton, B., Greaves, P., Harper, J. & Turner, G. Analysis of a commercially improved Penicillium chrysogenum strain series: involvement of recombinogenic regions in amplification and deletion of the penicillin biosynthesis gene cluster. J. Ind. Microbiol. Biotechnol. 19, 18–27 (1997).

    Article  CAS  PubMed  Google Scholar 

  8. Azuma, T., Nakanishi, T. & Sugimoto, M. Isolation and characterization of a stable L-arginine producer from continuous culture broth of Corynebacterium acetoacidophilum. J. Ferment. Technol. 66, 279–284 (1988).

    Article  CAS  Google Scholar 

  9. Harris, R. J. et al. Assessing genetic-heterogeneity in production cell lines: detection by peptide mapping of a low level Tyr to Gln sequence variant in a recombinant antibody. Nat. Biotechnol. 11, 1293–1297 (1993).

    Article  CAS  Google Scholar 

  10. Zelder, O. & Hauer, B. Environmentally directed mutations and their impact on industrial biotransformation and fermentation processes. Curr. Opin. Microbiol. 3, 248–251 (2000).

    Article  CAS  PubMed  Google Scholar 

  11. Rugbjerg, P., Myling-Petersen, N., Porse, A., Sarup-Lytzen, K. & Sommer, M. O. A. Diverse genetic error modes constrain large-scale bio-based production. Nat. Commun. 9, 787 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Xiao, Y., Bowen, C. H., Liu, D. & Zhang, F. Exploiting nongenetic cell-to-cell variation for enhanced biosynthesis. Nat. Chem. Biol. 12, 339–344 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Rugbjerg, P., Sarup-Lytzen, K., Nagy, M. & Sommer, M. O. A. Synthetic addiction extends the productive life time of engineered Escherichia coli populations. Proc. Natl Acad. Sci. 115, 2347–2352 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gupta, A., Reizman, I. M. B., Reisch, C. R. & Prather, K. L. J. Dynamic regulation of metabolic flux in engineered bacteria using a pathway-independent quorum-sensing circuit. Nat. Biotechnol. 35, 273–279 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ceroni, F. et al. Burden-driven feedback control of gene expression. Nat. Methods 15, 387–393 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Yano, H. et al. Evolved plasmid-host interactions reduce plasmid interference cost. Mol. Microbiol. 101, 743–756 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Karim, A. S., Curran, K. A. & Alper, H. S. Characterization of plasmid burden and copy number in Saccharomyces cerevisiae for optimization of metabolic engineering applications. FEMS Yeast Res. 13, 107–116 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Bentley, W. E., Mirjalili, N., Andersen, D. C., Davis, R. H. & Kompala, D. S. Plasmid-encoded protein: The principal factor in the ‘metabolic burden’ associated with recombinant bacteria. Biotechnol. Bioeng. 35, 668–681 (1990).

    Article  CAS  PubMed  Google Scholar 

  19. Kafri, M., Metzl-Raz, E., Jona, G. & Barkai, N. The cost of protein production. Cell Rep. 14, 22–31 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Klein, T. et al. Overcoming the metabolic burden of protein secretion in Schizosaccharomyces pombe - A quantitative approach using 13C-based metabolic flux analysis. Metab. Eng. 21, 34–45 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Villaverde, A. & Carrió, M. M. Protein aggregation in recombinant bacteria: biological role of inclusion bodies. Biotechnol. Lett. 25, 1385–95 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Geiler-Samerotte, K. A. et al. Misfolded proteins impose a dosage-dependent fitness cost and trigger a cytosolic unfolded protein response in yeast. Proc. Natl Acad. Sci. 108, 680–685 (2011).

    Article  PubMed  Google Scholar 

  23. Kwon, S. K., Kim, S. K., Lee, D. H. & Kim, J. F. Comparative genomics and experimental evolution of Escherichia coli BL21(DE3) strains reveal the landscape of toxicity escape from membrane protein overproduction. Sci. Rep. 5, 16076 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nielsen, A. A. K. et al. Genetic circuit design automation. Science 352, aac7341–aac7341 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Pitera, D. J., Paddon, C. J., Newman, J. D. & Keasling, J. D. Balancing a heterologous mevalonate pathway for improved isoprenoid production in Escherichia coli. Metab. Eng. 9, 193–207 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Kazemi Seresht, A. et al. Long-term adaptation of Saccharomyces cerevisiae to the burden of recombinant insulin production. Biotechnol. Bioeng. 110, 2749–2763 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Michener, J. K., Nielsen, J. & Smolke, C. D. Identification and treatment of heme depletion attributed to overexpression of a lineage of evolved P450 monooxygenases. Proc. Natl Acad. Sci. USA 109, 19504–19509 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Carneiro, S., Ferreira, E. C. & Rocha, I. Metabolic responses to recombinant bioprocesses in Escherichia coli. J. Biotechnol. 164, 396–408 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Wu, G. et al. Metabolic burden: cornerstones in synthetic biology and metabolic engineering applications. Trends Biotechnol. 34, 652–664 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Linster, C. L., Van Schaftingen, E. & Hanson, A. D. Metabolite damage and its repair or pre-emption. Nat. Chem. Biol. 9, 72–80 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Zhu, M. M., Skraly, Fa & Cameron, D. C. Accumulation of methylglyoxal in anaerobically grown Escherichia coli and its detoxification by expression of the Pseudomonas putida glyoxalase I gene. Metab. Eng. 3, 218–225 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Tuite, N. L., Fraser, K. R. & Byrne, C. P. O. Homocysteine toxicity in Escherichia coli is caused by a perturbation of branched-chain amino acid biosynthesis. J. Bacteriol. 187, 4362–4371 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kizer, L., Pitera, D. J., Pfleger, B. F. & Keasling, J. D. Application of functional genomics to pathway optimization for increased isoprenoid production. Appl. Environ. Microbiol. 74, 3229–41 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tindall, K. R. & Stankowski, L. F. Molecular analysis of spontaneous mutations at the gpt locus in Chinese hamster ovary (AS52) cells. Mutat. Res. Genet. Toxicol. 220, 241–253 (1989).

    Article  CAS  Google Scholar 

  35. Zhang, Z., Shah, B. & Bondarenko, P. V. G/U and certain wobble position mismatches as possible main causes of amino acid misincorporations. Biochemistry 52, 8165–8176 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Cartwright, J. F., Anderson, K., Longworth, J., Lobb, P. & James, D. C. Highly sensitive detection of mutations in CHO cell recombinant DNA using multi-parallel single molecule real-time DNA sequencing. Biotechnol. Bioeng. 115, 1485–1498 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Zeck, A. et al. Low level sequence variant analysis of recombinant proteins: An optimized approach. PLoS One 7, e40328 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Lee, H., Popodi, E., Tang, H. & Foster, P. L. PNAS Plus: Rate and molecular spectrum of spontaneous mutations in the bacterium Escherichia coli as determined by whole-genome sequencing. Proc. Natl Acad. Sci. 109, E2774–E2783 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Renda, B. A., Hammerling, M. J. & Barrick, J. E. Engineering reduced evolutionary potential for synthetic biology. Mol. BioSyst. 10, 1668–1678 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Moxon, R., Bayliss, C. & Hood, D. Bacterial contingency loci: the role of simple sequence dna repeats in bacterial adaptation. Annu. Rev. Genet. 40, 307–333 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Beekwilder, J. et al. Polycistronic expression of a β-carotene biosynthetic pathway in Saccharomyces cerevisiae coupled to β-ionone production. J. Biotechnol. 192, 383–392 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Yoshikawa, T. et al. Amplified gene location in chromosomal dna affected recombinant protein production and stability of amplified genes. Biotechnol. Prog. 16, 710–715 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Schlegel, S., Genevaux, P. & de Gier, J. W. De-convoluting the genetic adaptations of E. coli C41(DE3) in real time reveals how alleviating protein production stress improves yields. Cell Rep. 10, 1758–1766 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Lovett, S. T., Hurley, R. L., Sutera, V. A., Aubuchon, R. H. & Lebedeva, M. A. Crossing over between regions of limited homology in Escherichia coli. RecA-dependent and RecA-independent pathways. Genetics 160, 851–859 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Jinks-Robertson, S. & Bhagwat, A. S. Transcription-associated mutagenesis. Annu. Rev. Genet. 48, 341–359 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Bzymek, M. & Lovett, S. T. Instability of repetitive DNA sequences: The role of replication in multiple mechanisms. Proc. Natl Acad. Sci. USA 98, 8319–8325 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mahillon, J. & Chandler, M. Insertion sequences. Microbiol. Mol. Biol. Rev. 62, 725–774 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Richardson, S. M. et al. Design of a synthetic yeast genome. Science 355, 1040–1044 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Xu, X. et al. The genomic sequence of the Chinese hamster ovary (CHO)-K1 cell line. Nat. Biotechnol. 29, 735–41 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kim, J. Y., Kim, Y. & Lee, G. M. CHO cells in biotechnology for production of recombinant proteins: current state and further potential. Appl. Microbiol. Biotechnol. 93, 917–930 (2012).

    Article  CAS  PubMed  Google Scholar 

  51. Kroll, J., Klinter, S., Schneider, C., Voß, I. & Steinbüchel, A. Plasmid addiction systems: Perspectives and applications in biotechnology. Microb. Biotechnol. 3, 634–657 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Scholes, D. T., Banerjee, M., Bowen, B. & Curcio, M. J. Multiple regulators of Ty1 transposition in Saccharomyces cerevisiae have conserved roles in genome maintenance. Genetics 159, 1449–1465 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Fahnert, B., Lilie, H. & Neubauer, P. Inclusion bodies: formation and utilisation. in Physiological Stress Responses in Bioprocesses. Advances in Biochemical Engineering 89, 93–142 (Springer, 2004).

  54. Hoffmann, F. & Rinas, U. Stress induced by recombinant protein production in Escherichia coli. in Physiological Stress Responses in Bioprocesses 89, 73–92 (Springer, 2004).

  55. Tippin, B., Pham, P. & Goodman, M. F. Error-prone replication for better or worse. Trends Microbiol. 12, 288–295 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Foster, P.L. Stress-induced mutagenesis in bacteria. in Critical Reviews in Biochemistry and Molecular Biology 42, 373–397 (Springer, 2007).

  57. Bailey, L. A., Hatton, D., Field, R. & Dickson, A. J. Determination of Chinese hamster ovary cell line stability and recombinant antibody expression during long-term culture. Biotechnol. Bioeng. 109, 2093–2103 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. International Conference on Harmonisation & FDA. Guidance on quality of biotechnological/biological products: Derivation and characterization of cell substrates used for production of biotechnological/biological products. US Fed. Regist. 63, 50244–50249 (1998).

  59. Mercier, S. M., Diepenbroek, B., Wijffels, R. H. & Streefland, M. Multivariate PAT solutions for biopharmaceutical cultivation : current progress and limitations. Trends Biotechnol. 32, 329–336 (2014).

    Article  CAS  PubMed  Google Scholar 

  60. Yusufi, F. N. K. et al. Mammalian systems biotechnology reveals global cellular adaptations in a recombinant CHO cell line. Cell Syst. 4, 530–542.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  61. Nakamura, K. et al. Sequence-specific error profile of Illumina sequencers. Nucleic Acids Res. 39, e90 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Salk, J. J., Schmitt, M. W. & Loeb, L. A. Enhancing the accuracy of next-generation sequencing for detecting rare and subclonal mutations. Nat. Rev. Genet. 19, 269–285 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Deatherage, D. E., Traverse, C. C., Wolf, L. N. & Barrick, J. E. Detecting rare structural variation in evolving microbial populations from new sequence junctions using breseq. Front. Genet. 5, 1–16 (2015).

    Article  CAS  Google Scholar 

  64. Rehder, D. S. et al. Expression vector-derived heterogeneity in a therapeutic IgG4 monoclonal antibody. MAbs 11, 145–152 (2019).

    Article  CAS  PubMed  Google Scholar 

  65. Harris, R. P. & Kilby, P. M. Amino acid misincorporation in recombinant biopharmaceutical products. Curr. Opin. Biotechnol. 30, 45–50 (2014).

    Article  CAS  PubMed  Google Scholar 

  66. Dorai, H. et al. Early prediction of instability of chinese hamster ovary cell lines expressing recombinant antibodies and antibody-fusion proteins. Biotechnol. Bioeng. 109, 1016–1030 (2012).

    Article  CAS  PubMed  Google Scholar 

  67. Csorgo, B., Feher, T., Timar, E., Blattner, F. R. & Posfai, G. Low-mutation-rate, reduced-genome Escherichia coli: An improved host for faithful maintenance of engineered genetic constructs. Microb. Cell Fact. 11, 11 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Choi, J. W., Yim, S. S., Kim, M. J. & Jeong, K. J. Enhanced production of recombinant proteins with Corynebacterium glutamicum by deletion of insertion sequences (IS elements). Microb. Cell Fact. 14, 207 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Nyerges, Á. et al. CRISPR-interference based modulation of mobile genetic elements in bacteria. Synth. Biol. 4, ysz008 (2019).

    Article  Google Scholar 

  70. Deatherage, D. E., Leon, D., Rodriguez, Á. E., Omar, S. K. & Barrick, J. E. Directed evolution of Escherichia coli with lower-than-natural plasmid mutation rates. Nucleic Acids Res. 46, 9236–9250 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Mikkelsen, M. D. et al. Microbial production of indolylglucosinolate through engineering of a multi-gene pathway in a versatile yeast expression platform. Metab. Eng. 14, 104–111 (2012).

    Article  CAS  PubMed  Google Scholar 

  72. Peng, B., Wood, R. J., Nielsen, L. K. & Vickers, C. E. An expanded heterologous GAL promoter collection for diauxie-inducible expression in Saccharomyces cerevisiae. ACS Synth. Biol. 7, 748–751 (2018).

    Article  CAS  PubMed  Google Scholar 

  73. Rugbjerg, P., Knuf, C., Förster, J. & Sommer, M. O. A. Recombination-stable multimeric green fluorescent protein for characterization of weak promoter outputs in Saccharomyces cerevisiae. FEMS Yeast Res. 15, fov085 (2015).

    Article  CAS  PubMed  Google Scholar 

  74. Kaas, C. S., Kristensen, C., Betenbaugh, M. J. & Andersen, M. R. Sequencing the CHO DXB11 genome reveals regional variations in genomic stability and haploidy. BMC Genomics 16, 160 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhang, H. & Wang, X. Modular co-culture engineering, a new approach for metabolic engineering. Metab. Eng. 37, 114–121 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Frumkin, I. et al. Gene architectures that minimize cost of gene expression. Mol. Cell 65, 142–153 (2017).

    Article  CAS  PubMed  Google Scholar 

  77. Sørensen, H. P. & Mortensen, K. K. Soluble expression of recombinant proteins in the cytoplasm of Escherichia coli. Microb. Cell Fact. 4, 1 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Xia, X.-X. et al. Native-sized recombinant spider silk protein produced in metabolically engineered Escherichia coli results in a strong fiber. Proc. Natl Acad. Sci. USA 107, 14059–14063 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Yu, T. et al. Reprogramming yeast metabolism from alcoholic fermentation to lipogenesis. Cell 174, 1549–1558.e14 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. Erb, T. J., Jones, P. R. & Bar-Even, A. Synthetic metabolism: metabolic engineering meets enzyme design. Curr. Opin. Chem. Biol. 37, 56–62 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Genee, H. J. et al. Functional mining of transporters using synthetic selections. Nat. Chem. Biol. 12, 1015–1022 (2016).

    Article  CAS  PubMed  Google Scholar 

  82. Hansen, E. H. et al. De novo biosynthesis of vanillin in fission yeast (Schizosaccharomyces pombe) and baker’s yeast (Saccharomyces cerevisiae). Appl. Environ. Microbiol. 75, 2765–2774 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Venayak, N., Anesiadis, N., Cluett, W. R. & Mahadevan, R. Engineering metabolism through dynamic control. Curr. Opin. Biotechnol. 34, 142–152 (2015).

    Article  CAS  PubMed  Google Scholar 

  84. Studier, F. W. Protein production by auto-induction in high density shaking cultures. Protein Expr. Purif. 41, 207–234 (2005).

    Article  CAS  PubMed  Google Scholar 

  85. Figge, R. & Vasseur, P. Use of inducible promoters in the production of methionine. EP2513322A1 (2018).

  86. Xu, P. Production of chemicals using dynamic control of metabolic fluxes. Curr. Opin. Biotechnol. 53, 12–19 (2018).

    Article  CAS  PubMed  Google Scholar 

  87. Dahl, R. H. et al. Engineering dynamic pathway regulation using stress-response promoters. Nat. Biotechnol. 31, 1039–46 (2013).

    Article  CAS  PubMed  Google Scholar 

  88. Taylor, N. D. et al. Engineering an allosteric transcription factor to respond to new ligands. Nat. Methods 13, 177–183 (2016).

    Article  CAS  PubMed  Google Scholar 

  89. Paddon, C. J. & Keasling, J. D. Semi-synthetic artemisinin: A model for the use of synthetic biology in pharmaceutical development. Nat. Rev. Microbiol. 12, 355–367 (2014).

    Article  CAS  PubMed  Google Scholar 

  90. Pitera, D.J., Newman, J.D., Kizer, J.L., Keasling, J.D. & Pfleger, B.F. Methods for increasing isoprenoid and isoprenoid precursor production by modulating fatty acid levels. US 8114645 B2 (2012).

  91. Sandoval, C. M. et al. Use of pantothenate as a metabolic switch increases the genetic stability of farnesene producing Saccharomyces cerevisiae. Metab. Eng. 25, 215–226 (2014).

    Article  CAS  PubMed  Google Scholar 

  92. Halls, C. & Yu, O. Potential for metabolic engineering of resveratrol biosynthesis. Trends Biotechnol. 26, 77–81 (2008).

    Article  CAS  PubMed  Google Scholar 

  93. Reyes, L. H., Almario, M. P., Winkler, J., Orozco, M. M. & Kao, K. C. Visualizing evolution in real time to determine the molecular mechanisms of n-butanol tolerance in Escherichia coli. Metab. Eng. 14, 579–590 (2012).

    Article  CAS  PubMed  Google Scholar 

  94. Ro, D. K. et al. Induction of multiple pleiotropic drug resistance genes in yeast engineered to produce an increased level of anti-malarial drug precursor, artemisinic acid. BMC Biotechnol. 8, 1–14 (2008).

    Article  CAS  Google Scholar 

  95. Mundhada, H. et al. Increased production of L-serine in Escherichia coli through adaptive laboratory evolution. Metab. Eng. 39, 141–150 (2017).

    Article  CAS  PubMed  Google Scholar 

  96. Koffas, M. A. G., Jung, G. Y. & Stephanopoulos, G. Engineering metabolism and product formation in Corynebacterium glutamicum by coordinated gene overexpression. Metab. Eng. 5, 32–41 (2003).

    Article  CAS  PubMed  Google Scholar 

  97. Raab, A. M., Gebhardt, G., Bolotina, N., Weuster-Botz, D. & Lang, C. Metabolic engineering of Saccharomyces cerevisiae for the biotechnological production of succinic acid. Metab. Eng. 12, 518–525 (2010).

    Article  CAS  PubMed  Google Scholar 

  98. Durfee, T. et al. The Complete genome sequence of Escherichia coli DH10B: Insights into the biology of a laboratory workhorse. J. Bacteriol. 190, 2597–2606 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Serero, A., Jubin, C., Loeillet, S., Legoix-Né, P. & Nicolas, A. G. Mutational landscape of yeast mutator strains. Proc. Natl Acad. Sci. USA 111, 1897–1902 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Summers, D. K. The kinetics of plasmid loss. Trends Biotechnol. 9, 273–8 (1991).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Porse, F. Lino and C. Hjort for helpful comments. The research leading to these results has received funding from the Novo Nordisk Foundation, Denmark, grant number NNF10CC1016517, and from the European Union Seventh Framework Programme (FP7-KBBE-2013-7-single-stage) under grant agreement 613745, Promys.

Author information

Authors and Affiliations

Authors

Contributions

P.R. and M.O.A.S. outlined and wrote the manuscript.

Corresponding author

Correspondence to Morten O. A. Sommer.

Ethics declarations

Competing interests

P.R. and M.O.A.S. are inventors of a pending patent application (WO2017055360) within product addiction filed by the Technical University of Denmark.

Additional information

Peer review information: Andy Marshall was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rugbjerg, P., Sommer, M.O.A. Overcoming genetic heterogeneity in industrial fermentations. Nat Biotechnol 37, 869–876 (2019). https://doi.org/10.1038/s41587-019-0171-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0171-6

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research