Exciting news – Nature has published a collaboration between Salesforce and Tierra!Learn More 
The Tierra Blog
Discover the world of protein-powered cell-free research.
July 03, 2024 Cell-Free Protein Synthesis

Selecting a Cell-Free Protein Synthesis System

Navigate the cell-free system landscape and key influencing factors to optimize protein synthesis for your target proteins.

Picture a future where high-quality therapeutic proteins are manufactured in hours instead of days. Cell-free protein synthesis (CFPS) platforms enable reliable protein synthesis at any scale, driving biotherapeutic discovery and production. This protocol uses cellular lysates to extract the transcription-translation machinery and energy components, redirecting them to produce a target protein.  

As CFPS becomes more affordable, various systems have emerged, necessitating careful selection to optimize protein yield and quality[1]. Here, we explore the current CFPS landscape, factors influencing system choice, and the role of strain engineering in the evolution of CFPS. 

Prokaryotic CFPS Systems

Prokaryotes couple transcription and translation through ribosomes associating with mRNA before transcription is completed[2]. This simultaneous process minimizes naked mRNA accumulation and resource depletion, enhancing efficiency during protein synthesis[3]. The potential was first realized over 60 years ago with an E. coli-based CFPS system, leading to the synthesis of various therapeutically relevant proteins[4].  

Since then, multiple prokaryotic-based CFPS platforms have generated an array of biopharmaceutical proteins, including: 

E. coli
  • The first bacterial species used for CFPS, E. coli, is cost-effective due to its high growth rates, aiding in the discovery of SARS-CoV-2 antibodies and Shiga toxin characterization[5,6].
Vibrio spp.
  • Fast-growing Vibrio natriegens synthesizes proteins efficiently, supported by 3-PGA as an energy source, making it ideal for producing nonribosomal peptides and natural products such as antibiotics[7-9].
Streptomyces
  • Streptomyces has been a major source of natural products in medical care, from antibiotics to crop-protecting agents. Cells in this genus harbor enzyme-coding genes that confer strong capabilities to be a CFPS system[10]. Most notably, species in this genus can express phosphopantetheinyl transferases that activate carrier proteins involved in the biosynthesis of complex natural products like antibiotics and polyketides[11]. 
Anaerobes
  • The emerging use of anaerobic bacteria in CFPS is opening new possibilities for sustainable production[12]. Recently, companies like LanzaTech have announced the development of commercial-scale bioreactors that use anaerobes to metabolize gas emissions to produce ethanol and other compounds. 

In addition to these species, multiple strains within each confer distinct advantages for CFPS. For example, E. coli DE3 strains eliminate the need to add polymerases by inducing sufficient T7 RNA polymerase expression[13]. 

Even so, most prokaryotes produce a limited number of post-translational modifications, making them less suitable for producing complex proteins with glycosylation or disulfide bonds, which are essential for their function and therapeutic efficacy[14].  

Eukaryotic CFPS Systems

The challenge of producing proteins with post-translational modifications in prokaryotic systems has led to the increased use of eukaryotic CFPS systems. Several eukaryotic systems have been developed and tailored for protein synthesis, including: 

Spodoptera frugiperda 21 (Sf21)
  • Despite being a pest, the fall armyworm's cell extracts are valuable for producing integral membrane proteins[15,16].
Wheat germ
  • Wheat germ is used to produce proteins at the milligram scale. Although raw extracts are challenging to process, refined protocols minimize RNase activity and reduce yield variation. These improvements have enabled wheat germ systems to produce purified recombinant proteins and vaccine candidates, such as those for malaria[17-21].
Tobacco BY-2
  • Tobacco Bright Yellow-2 (BY-2) lysates offer high translational activity, quick preparation, and scalability to milligram and gram levels of protein. BY-2 cell lysates were used to produce the first FDA-approved recombinant pharmaceutical protein, Taliglucerase alfa (Elelyso®)[22].  
Human cell cultures
  • Various human cell lines, such as HEK293, HeLa, and CHO cells, are used to express diverse proteins using an in vitro coupled transcription/translation system. Like Sf21, human-based systems have ER-derived microsomes that facilitate the production of integral membrane proteins[23].  

Factors to Consider When Selecting a CFPS System

Like prokaryotic systems, eukaryotic CFPS systems have unique advantages and drawbacks that impact protein synthesis (Table 1). Each system's characteristics must be evaluated to select the best one for specific proteins. Here are key factors to consider when choosing a CFPS system: 

  • Protein Yield:
    Different CFPS systems produce varying yields based on the lysates' biochemical composition. Systems that couple transcription and translation typically generate higher yields. Additionally, DNA structures, like a 5’-UTR with an enhancer sequence, can boost transcription and protein synthesis[24,25].
  • Protein Type:
    Different proteins require specific reagents and biomolecules. For example, integral membrane proteins need lipid bilayers or vesicles for purity and integrity[26]. Therapeutic proteins like onconases, which cleave tRNAs, require systems that pulse tRNA intake or maintain tRNA stability[27].
  • Post-Translational Modifications:
    Some CFPS systems, particularly prokaryotic-based and wheat-germ systems, cannot perform post-translational modifications. For producing monoclonal antibodies with disulfide bridges or glycosylated proteins, eukaryotic CFPS systems are more suitable[28]. 

Evolution of CFPS Systems

Currently available prokaryotic CFPS systems can produce a wide variety of biopharmaceutical proteins. However, each species has unique strains that can further enhance protein yields and quality. Prokaryotic systems, particularly E. coli, can be engineered to synthesize non-canonical amino acids using strains like C321.DA[29].  

Eukaryotic CFPS systems can also be cloned to produce specific proteins at an industrial scale. Additionally, genomes from both prokaryotic and eukaryotic systems can be engineered to study biochemical pathways and assess protein interactions, driving R&D efforts[30,31]. 

CFPS systems offer diverse and versatile formats for synthesizing specific proteins, each with unique advantages and considerations. Understanding the landscape of prokaryotic and eukaryotic systems, as well as the key influencing factors such as protein type, yield, and post-translational modifications, is essential for optimizing protein synthesis.  

The future of advanced CFPS systems looks bright, with genetic engineering addressing shortcomings of today’s systems by enhancing protein yields, quality, and functionality. Moreover, the integration of automation, artificial intelligence, and machine learning is further streamlining protein production, enabling precise control and high-throughput screening at unprecedented scales. 

Work With Us

Accelerate your scientific discovery with the Tierra Protein Platform. Our platform integrates automation, computation, and high-throughput screening allowing you to synthesize custom proteins from diverse sources at the click of a button. Visit our ordering portal or contact Tierra Biosciences today to learn how our platform and expert guidance can drive the success of your programs. 

References

  1. Khambhati K, Bhattacharjee G, Gohil N, Braddick D, Kulkarni V, Singh V. Exploring the Potential of Cell-Free Protein Synthesis for Extending the Abilities of Biological Systems. Front Bioeng Biotechnol. 2019;7. doi:10.3389/fbioe.2019.00248
  2. Irastortza-Olaziregi M, Amster-Choder O. Coupled Transcription-Translation in Prokaryotes: An Old Couple With New Surprises.Front Microbiol. 2020;11:624830.doi:10.3389/fmicb.2020.624830
  3. McGary K, Nudler E. RNA polymerase and the ribosome: the close relationship. Curr Opin Microbiol. 2013;16(2):112-117.doi:10.1016/j.mib.2013.01.010
  4. Nirenberg MW, Matthaei JH. The dependence of cell-free protein synthesis in E. coli upon naturally occurring or synthetic polyribonucleotides. Proc Natl Acad Sci U S A. 1961;47(10):1588-1602. doi:10.1073/pnas.47.10.1588
  5. Hunt AC, Vögeli B, Hassan AO, et al. A rapid cell-free expression and screening platform for antibody discovery. Nat Commun. 2023;14(1):3897. doi:10.1038/s41467-023-38965-w
  6. Ramm F, Kaser D, König I, et al. Synthesis of biologically active Shiga toxins in cell-free systems. Sci Rep. 2024;14(1):6043.doi:10.1038/s41598-024-56190-3
  7. Failmezger J, Scholz S, Blombach B, Siemann-Herzberg M. Cell-Free Protein Synthesis From Fast-Growing Vibrio natriegens. Front Microbiol. 2018;9. doi:10.3389/fmicb.2018.01146
  8. Wiegand DJ, Lee HH, Ostrov N, Church GM. Cell-free Protein Expression Using the Rapidly Growing Bacterium Vibrio natriegens. JoVE (Journal of Visualized Experiments). 2019;(145):e59495. doi:10.3791/59495
  9. Ji X, Liu WQ, Li J. Recent advances in applying cell-free systems for high-value and complex natural product biosynthesis. Current Opinion in Microbiology. 2022;67:102142. doi:10.1016/j.mib.2022.102142
  10. J. Moore S, Lai HE, Li J, S. Freemont P. Streptomyces cell-free systems for natural product discovery and engineering. 2023;40(2):228-236. doi:10.1039/D2NP00057A
  11. Li J, Zhang L, Liu W. Cell-free synthetic biology for in vitro biosynthesis of pharmaceutical natural products. Synth Syst Biotechnol. 2018;3(2):83-89. doi:10.1016/j.synbio.2018.02.002
  12. SUN ZZ, Robertson DE, TREGO KS, CHIAO AC, IV LEM. Anaerobic cell-free systems and environments and methods for making and using same. Published online August 26, 2020. Accessed May 24, 2024. https://patents.google.com/patent/EP3697803A2/en
  13. Cole SD, Miklos AE, Chiao AC, Sun ZZ, Lux MW. Methodologies for preparation of prokaryotic extracts for cell-free expression systems. Synthetic and Systems Biotechnology. 2020;5(4):252-267. doi:10.1016/j.synbio.2020.07.006
  14. Macek B, Forchhammer K, Hardouin J, Weber-Ban E, Grangeasse C, Mijakovic I. Protein post-translational modifications in bacteria.Nat Rev Microbiol. 2019;17(11):651-664. doi:10.1038/s41579-019-0243-0
  15. Ezure T, Nanatani K, Sato Y, et al. A Cell-Free Translocation System Using Extracts of Cultured Insect Cells to Yield Functional Membrane Proteins. PLOS ONE. 2014;9(12):e112874. doi:10.1371/journal.pone.0112874
  16. Quast RB, Kortt O, Henkel J, et al. Automated production of functional membrane proteins using eukaryotic cell-free translation systems. Journal of Biotechnology. 2015;203:45-53. doi:10.1016/j.jbiotec.2015.03.015
  17. Harbers M. Wheat germ systems for cell-free protein expression. FEBS Letters. 2014;588(17):2762-2773. doi:10.1016/j.febslet.2014.05.061
  18. Scheele G, Blackburn P. Role of mammalian RNase inhibitor in cell-free protein synthesis. Proc Natl Acad Sci U S A.1979;76(10):4898-4902. doi:10.1073/pnas.76.10.4898
  19. Marcu K, Dudock B. Characterization of a highly efficient protein synthesizing system derived from commercial wheat germ. Nucleic Acids Res. 1974;1(11):1385-1397. doi:10.1093/nar/1.11.1385
  20. Goshima N, Kawamura Y, Fukumoto A, et al. Human protein factory for converting the transcriptome into an in vitro-expressed proteome,. Nat Methods. 2008;5(12):1011-1017. doi:10.1038/nmeth.1273
  21. Arumugam TU, Ito D, Takashima E, et al. Application of wheat germ cell-free protein expression system for novel malaria vaccine candidate discovery. Expert Rev Vaccines. 2014;13(1):75-85. doi:10.1586/14760584.2014.861747
  22. Buntru M, Vogel S, Spiegel H, Schillberg S. Tobacco BY-2 cell-free lysate: an alternative and highly-productive plant-based in vitro translation system. BMC Biotechnology. 2014;14(1):37. doi:10.1186/1472-6750-14-37
  23. Mikami S, Kobayashi T, Masutani M, Yokoyama S, Imataka H. A human cell-derived in vitro coupled transcription/translation system optimized for production of recombinant proteins. Protein Expr Purif. 2008;62(2):190-198. doi:10.1016/j.pep.2008.09.002
  24. Rosenblum G, Cooperman BS. Engine out of the Chassis: Cell-Free Protein Synthesis and its Uses. FEBS Lett. 2014;588(2):261-268. doi:10.1016/j.febslet.2013.10.016
  25. Mureev S, Kovtun O, Nguyen UTT, Alexandrov K. Species-independent translational leaders facilitate cell-free expression. Nat Biotechnol. 2009;27(8):747-752. doi:10.1038/nbt.1556
  26. Shinoda T, Shinya N, Ito K, et al. Cell-free methods to produce structurally intact mammalian membrane proteins. Sci Rep. 2016;6(1):30442. doi:10.1038/srep30442
  27. Salehi ASM, Smith MT, Bennett AM, Williams JB, Pitt WG, Bundy BC. Cell-free protein synthesis of a cytotoxic cancer therapeutic: Onconase production and a just-add-water cell-free system. Biotechnol J. 2016;11(2):274-281. doi:10.1002/biot.201500237
  28. Porche KB, Lanclos CE, Kwon YC. Challenging Post-translational Modifications in the Cell-free Protein Synthesis System. Synthetic Biology and Engineering. 2023;1(2):10011. doi:10.35534/sbe.2023.10011
  29. Martin RW, Des Soye BJ, Kwon YC, et al. Cell-free protein synthesis from genomically recoded bacteria enables multisite incorporation of noncanonical amino acids. Nat Commun. 2018;9(1):1203. doi:10.1038/s41467-018-03469-5
  30. Gupta MD, Flaskamp Y, Roentgen R, et al. Scaling eukaryotic cell-free protein synthesis achieved with the versatile and highyielding tobacco BY-2 cell lysate. Biotechnology and Bioengineering. 2023;120(10):2890-2906. doi:10.1002/bit.28461
  31. Silverman AD, Karim AS, Jewett MC. Cell-free gene expression: an expanded repertoire of applications. Nat Rev Genet. 2020;21(3):151-170. doi:10.1038/s41576-019-0186-3
More from the Blog
Therapeutics

Antibody-drug conjugates: A new era in targeted cancer therapy

Accelerating the discovery and development of effective antibody-drug conjugates with cell-free protein synthesis
Cell-Free Protein Synthesis

Breaking the bounds of biology: innovations in cell-free protein synthesis

Challenges, applications, and scalable solutions.
Therapeutics

From targets to treatments: insights from the cancer proteome

Exploiting the cancer proteome for therapeutic innovation and creating effective, easy-to-manufacture protein-based cancer therapies.

Protein synthesis can be easy, fast, and reliable.

Order ProteinsQuestions? Get in touch