Elsevier

Biochemical Engineering Journal

Volume 141, 15 January 2019, Pages 182-189
Biochemical Engineering Journal

Regular article
Cell-free protein synthesis: Recent advances in bacterial extract sources and expanded applications

https://doi.org/10.1016/j.bej.2018.10.023Get rights and content

Highlights

  • New CFPS systems have been developed from different bacterial extract sources.

  • Applications of CFPS for next-generation biomanufacturing.

  • Future challenges and opportunities for the CFPS technology.

Abstract

Cell-free protein synthesis (CFPS) is a technology for the production of proteins in vitro, which has emerged as a robust platform for biotechnology and synthetic biology applications. Recently, the renewed scientific interest in CFPS technology has driven the rapid development of cell-free biotechnology. In this review, we focus on the very recent advances and applications of CFPS with a time frame from 2016 to the middle of 2018. First, we introduce newly developed CFPS systems from different microorganisms including Streptomyces species, Bacillus subtilis, Pseudomonas putida, and Vibrio natriegens. Then, we summarize emerging applications of CFPS for next-generation biomanufacturing. With that, we discuss the future perspectives of CFPS and its potential for cost-effective, high-yielding production of therapeutics, chemicals, and materials.

Introduction

Cell-free protein synthesis (CFPS), also known as in vitro transcription-translation, has emerged as a powerful platform for the production of recombinant proteins without the use of living, intact cells. While the CFPS system was first used to elucidate the genetic code in the 1960s [1], it has shown substantial utility as a protein production technology during the past two decades [[2], [3], [4], [5], [6], [7]]. CFPS systems use crude cell extracts [8] or purified components [9] to synthesize a wide variety of proteins that include therapeutic proteins [[10], [11], [12]], membrane proteins [[13], [14], [15]], metalloproteins [[16], [17], [18]], and proteins modified with non-standard amino acids [[19], [20], [21]]. Recently, a renaissance in CFPS systems not only expands the protein synthesis toolkit, but also leads to wide and exciting applications in the field of synthetic biology, for example, the prototyping of genetic circuits and metabolic pathways [22,23], designing of medical diagnostics and biosensors [24,25], biosynthesis of natural products [26,27], and engineering of microfluidic biochip devices [28,29], among others [[30], [31], [32]]. Overall, pioneering efforts by scientists and engineers have created simple, robust, cost-effective, and efficient CFPS platforms for the rapid synthesis, study, and engineering of proteins, as well as for compelling applications in synthetic biology and biotechnology.

In recent years, with the fast development and great progress of CFPS technology, some outstanding reviews have been published, which summarize the advancement of CFPS systems and their broad applications [[33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44]]. For example, Jewett and colleagues comprehensively described the technological advances in CFPS and its emerging applications for the production of protein libraries, personalized medicines, evolved proteins, and protein-based biomaterials [34,35]. Different CFPS systems, which are generated from various prokaryotic (e.g., Escherichia coli) and eukaryotic (e.g., wheat germ and Chinese hamster ovary) organisms, have also been reviewed, showing their versatile biological and application-based goals [[36], [37], [38], [39]]. Although multiple CFPS systems are available, each one has its own advantages and disadvantages, which are intensively summarized and compared in details by Kubick and co-workers [40]. In addition, several reviews document the combination of CFPS systems with other high-throughput devices like microfluidics and lab-on-a-chip for the development of new application platforms [43,44]. More recently, the renewed scientific interest in CFPS technology has driven the rapid development of cell-free biotechnology, which stimulates the establishment of novel CFPS platforms, as well as other new research areas like cell-free metabolic engineering, natural product biosynthesis, and portable, on-demand biomolecular manufacturing. These new results are exciting and, therefore, it is necessary to summarize the recent achievements in an effort to give the reader an updated overview of the current state-of-the-art CFPS technology.

In this review, we aim to highlight the very recent advances and applications of CFPS (Fig. 1), covering from 2016 to the middle of 2018. First, we introduce newly developed CFPS systems from different microorganisms. Then, we summarize emerging applications of CFPS for next-generation biomanufacturing. Finally, we discuss the future prospects of CFPS for synthetic biology and biotechnology applications.

Section snippets

New CFPS systems

Crude extract based CFPS systems synthesize target proteins by harnessing the intracellular catalysts (e.g., aminoacyl-tRNA synthetases, ribosomes, RNA polymerases, chaperones, etc.) that are necessary for transcription, translation, and protein folding from crude cell extracts of microbial, plant, or mammalian cells. When combined with necessary substrates, which include nucleotides, amino acids, energy substrates, DNA or mRNA templates, cofactors, and salts, these biological catalysts work as

CFPS applications

CFPS systems offer distinct advantages over in vivo microbial systems due to the open reaction environment, which allows for easy manipulation, direct monitoring, and rapid sampling. Recent advances in the CFPS technology have driven new applications for the manufacturing of therapeutics, modified proteins, natural products, and diagnostics/biosensors. Examples of such applications will be described in the following section.

Conclusions and perspectives

CFPS systems without using intact cells hold tremendous potential for the next-generation biomanufacturing of therapeutics, chemicals, and materials. Therefore, highly productive CFPS platforms are needed for the rapid, inexpensive, and efficient production of recombinant proteins. In particular, new organism based CFPS systems except the commonly used E. coli, wheat germ, and CHO, etc. need to be developed to synthesize proteins with diverse complexity and species origin. More recently, a

Conflict of interest

The authors declare no commercial or financial conflict of interest.

Acknowledgements

This work was supported by Shanghai Pujiang Program (18PJ1408000). Jian Li also acknowledges the starting grant of ShanghaiTech University.

References (106)

  • Y. Endo et al.

    Cell-free expression systems for eukaryotic protein production

    Curr. Opin. Biotechnol.

    (2006)
  • R. Sachse et al.

    Membrane protein synthesis in cell-free systems: from bio-mimetic systems to bio-membranes

    FEBS Lett.

    (2014)
  • S.M. Schinn et al.

    Protein synthesis directly from PCR: progress and applications of cell-free protein synthesis with linear DNA

    N. Biotechnol.

    (2016)
  • R.H. Baltz

    Renaissance in antibacterial discovery from actinomycetes

    Curr. Opin. Pharmacol.

    (2008)
  • J. Li et al.

    Expanding the palette of Streptomyces-based cell-free protein synthesis systems with enhanced yields

    Biochem. Eng. J.

    (2018)
  • R. Kelwick et al.

    Development of a Bacillus subtilis cell-free transcription-translation system for prototyping regulatory elements

    Metab. Eng.

    (2016)
  • R.B. Quast et al.

    Cotranslational incorporation of non-standard amino acids using cell-free protein synthesis

    FEBS Lett.

    (2015)
  • J. Li et al.

    Escherichia coli as a cell factory for heterologous production of nonribosomal peptides and polyketides

    N. Biotechnol.

    (2014)
  • E.B. Bahadır et al.

    Applications of commercial biosensors in clinical, food, environmental, and biothreat/biowarfare analyses

    Anal. Biochem.

    (2015)
  • V. Perumal et al.

    Advances in biosensors: principle, architecture and applications

    J. Appl. Biomed.

    (2014)
  • D.K. Karig

    Cell-free synthetic biology for environmental sensing and remediation

    Curr. Opin. Biotechnol.

    (2017)
  • M. Soltani et al.

    Reengineering cell-free protein synthesis as a biosensor: biosensing with transcription, translation, and protein-folding

    Biochem. Eng. J.

    (2018)
  • K. Pardee et al.

    Rapid, low-cost detection of Zika virus using programmable biomolecular components

    Cell

    (2016)
  • A.S.M. Salehi et al.

    Biosensing estrogenic endocrine disruptors in human blood and urine: a RAPID cell-free protein synthesis approach

    Toxicol. Appl. Pharmacol.

    (2018)
  • M.W. Nirenberg et al.

    The dependence of cell-free protein synthesis in E. coli upon naturally occurring or synthetic polyribonucleotides

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

    (1961)
  • J.G. Perez et al.

    Cell-free synthetic biology: engineering beyond the cell, Cold Spring Harb

    Perspect. Biol.

    (2016)
  • J.R. Swartz

    Universal cell-free protein synthesis

    Nat. Biotechnol.

    (2009)
  • Y. Shimizu et al.

    Cell-free translation reconstituted with purified components

    Nat. Biotechnol.

    (2001)
  • B.C. Bundy et al.

    Escherichia coli-based cell-free synthesis of virus-like particles

    Biotechnol. Bioeng.

    (2008)
  • Y. Lu et al.

    Production and stabilization of the trimeric influenza hemagglutinin stem domain for potentially broadly protective influenza vaccines

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

    (2014)
  • S.E. Min et al.

    Cell-free production and streamlined assay of cytosol-penetrating antibodies

    Biotechnol. Bioeng.

    (2016)
  • D. Schwarz et al.

    Preparative scale expression of membrane proteins in Escherichia coli-based continuous exchange cell-free systems

    Nat. Protoc.

    (2007)
  • R. Sachse et al.

    Synthesis of membrane proteins in eukaryotic cell-free systems

    Eng. Life Sci.

    (2013)
  • J.A. Schoborg et al.

    A cell-free platform for rapid synthesis and testing of active oligosaccharyltransferases

    Biotechnol. Bioeng.

    (2018)
  • M.E. Boyer et al.

    Cell-free synthesis and maturation of [FeFe] hydrogenases

    Biotechnol. Bioeng.

    (2008)
  • Y.C. Kwon et al.

    Integrating cell-free biosyntheses of heme prosthetic group and apoenzyme for the synthesis of functional P450 monooxygenase

    Biotechnol. Bioeng.

    (2013)
  • J. Li et al.

    Cell-free protein synthesis enables high yielding synthesis of an active multicopper oxidase

    Biotechnol. J.

    (2016)
  • B.C. Bundy et al.

    Site-specific incorporation of p-propargyloxyphenylalanine in a cell-free environment for direct protein-protein click conjugation

    Bioconjug. Chem.

    (2010)
  • S.H. Hong et al.

    Cell-free protein synthesis from a release factor 1 deficient Escherichia coli activates efficient and multiple site-specific nonstandard amino acid incorporation

    ACS Synth. Biol.

    (2014)
  • C. Albayrak et al.

    Direct polymerization of proteins

    ACS Synth. Biol.

    (2014)
  • Z.Z. Sun et al.

    Linear DNA for rapid prototyping of synthetic biological circuits in an Escherichia coli based TX-TL cell-free system

    ACS Synth. Biol.

    (2014)
  • K.Y. Wen et al.

    Cell-free biosensor for detecting quorum sensing molecules in P. aeruginosa-infected respiratory samples

    ACS Synth. Biol.

    (2017)
  • A.W. Goering et al.

    In vitro reconstruction of nonribosomal peptide biosynthesis directly from DNA using cell-free protein synthesis

    ACS Synth. Biol.

    (2017)
  • Y. Hori et al.

    Cell-free extract based optimization of biomolecular circuits with droplet microfluidics

    Lab Chip

    (2017)
  • A.M. Tayar et al.

    Synchrony and pattern formation of coupled genetic oscillators on a chip of artificial cells

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

    (2017)
  • M.A. Klocke et al.

    Engineering DNA nanotubes for resilience in an E. coli TXTL system

    Synth. Biol.

    (2018)
  • M. Rustad et al.

    Cell-free TXTL synthesis of infectious bacteriophage T4 in a single test tube reaction

    Synth. Biol.

    (2018)
  • A.D. Halleran et al.

    Cell-free and in vivo characterization of Lux, Las, and Rpa quorum activation systems in E. coli

    ACS Synth. Biol.

    (2018)
  • S. Chong

    Overview of cell-free protein synthesis: historic landmarks, commercial systems, and expanding applications

    Curr. Protoc. Mol. Biol.

    (2014)
  • S.H. Hong et al.

    Non-standard amino acid incorporation into proteins using Escherichia coli cell-free protein synthesis

    Front. Chem.

    (2014)
  • Cited by (0)

    1

    These authors are contributed equally.

    View full text