Please wait a minute...
Frontiers of Chemical Science and Engineering

ISSN 2095-0179

ISSN 2095-0187(Online)

CN 11-5981/TQ

Postal Subscription Code 80-969

2018 Impact Factor: 2.809

Front. Chem. Sci. Eng.    2017, Vol. 11 Issue (1) : 58-65    https://doi.org/10.1007/s11705-017-1610-x
REVIEW ARTICLE
Cell-free systems in the new age of synthetic biology
Fernando Villarreal,Cheemeng Tan()
Department of Biomedical Engineering, University of California Davis, Davis, One Shields Avenue, CA 95616, USA
 Download: PDF(302 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

The advent of synthetic biology has ushered in new applications of cell-free transcription-translation systems. These cell-free systems are reconstituted using cellular proteins, and are amenable to modular control of their composition. Here, we discuss the historical advancement of cell-free systems, as well as their new applications in the rapid design of synthetic genetic circuits and components, directed evolution of biomolecules, diagnosis of infectious diseases, and synthesis of vaccines. Finally, we present our vision on the future direction of cell-free synthetic biology.

Keywords cell-free system      application     
Corresponding Author(s): Cheemeng Tan   
Online First Date: 12 January 2017    Issue Date: 17 March 2017
 Cite this article:   
Fernando Villarreal,Cheemeng Tan. Cell-free systems in the new age of synthetic biology[J]. Front. Chem. Sci. Eng., 2017, 11(1): 58-65.
 URL:  
https://academic.hep.com.cn/fcse/EN/10.1007/s11705-017-1610-x
https://academic.hep.com.cn/fcse/EN/Y2017/V11/I1/58
Fig.1  Sources of CFS for synthetic biology approaches. CFS can be generated through lysis of E. coli, wheat germ, L. tarentolaeor HeLa cells. The lysates contain all the components required for in vitro translation (ribosomes and translation machinery, TraM) and other protein impurities. The protein synthesis using recombinant elements (PURE) system is obtained by purifying each of the components of the TraM and the ribosomes
Fig.2  Optimization and expansion of CFS. Top: Orthogonal RNA polymerases (RNAP) from phage T7 and T3 can be used to transcribe proteins in CFS. In addition, σ transcription factors (TFs) can guide the transcription from particular promoters by RNAP II. Center: Stabilization of linear DNA can be achieved by adding DNA exonuclease (ExoDNAse)-inhibitor GamS. Furthermore, protein stability can be increased using chaperones or factors that introduce post-translational (Post-Trn) modifications. Turnover of mRNA and proteins can be achieved by incorporating specific RNA endonucleases (EndoRNAse) and/or proteases that recognize specific tags (**) in the target protein. Bottom: Target proteins with extended properties can also be produced by incorporating novel, non-natural amino acids (U1 in the figure) through orthogonal and mutant synthases
Fig.3  Applications for CFS in synthetic biology. Prototyping of genetic circuit. Genetic circuits can be rapidly assembled and tested in vitro. For example, oscillator circuits are selected based on their capacity to produce an oscillatory output. The selected circuits can be further characterized and eventually implemented in vivo. Adapted from [49]. Metabolic pathways. A sample pathway uses four enzymes (E1, E2, E3 and E4) to convert a substrate S into a product P. Two other enzymes (E5 and E6) result in unnecessary fluxes, which can be eliminated in CFS. Each enzyme can be purified from recombinant microbes or synthesized directly in CFS. The metabolic pathway can be optimized by testing different ratios and mutants of the enzymes (E4*). The reactions can also be optimized by modifying environmental conditions (B1 to B5) that contain different metals, ions, and cofactors. Modified from [50]. In vitro evolution. A DNA library coding for a pore forming protein is evolved using CFS. A fluorophore (green stars) in the extraliposomal milieu is transported into the liposome if the pore is properly formed. This way, liposomes with active pore-forming mutants are selected by flow cytometry sorting (FACS). The DNA in the selected liposomes is recovered and used in successive rounds of selection. Modified from [51]. Diagnostics. The CFS, together with a genetic circuit (sensor) responsive to specific molecules are printed on paper. Samples from patients can be added to the paper for diagnosis of disease biomarkers. Adapted from [52]
23 Kovtun O, Mureev S, Johnston W, Alexandrov K. Towards the construction of expressed proteomes using a Leishmania tarentolae based cell-free expression system. PLoS One, 2010, 5(12): e14388
https://doi.org/10.1371/journal.pone.0014388
24 Matsuda T, Watanabe S, Kigawa T. Cell-free synthesis system suitable for disulfide-containing proteins. Biochemical and Biophysical Research Communications, 2013, 431(2): 296–301
https://doi.org/10.1016/j.bbrc.2012.12.107
25 Chemla Y, Ozer E, Schlesinger O, Noireaux V, Alfonta L. Genetically expanded cell-free protein synthesis using endogenous pyrrolysyl orthogonal translation system. Biotechnology and Bioengineering, 2015, 112(8): 1663–1672
https://doi.org/10.1002/bit.25587
26 Albayrak C, Swartz J R. Using E. coli-based cell-free protein synthesis to evaluate the kinetic performance of an orthogonal tRNA and aminoacyl-tRNA synthetase pair. Biochemical and Biophysical Research Communications, 2013, 431(2): 291–295
https://doi.org/10.1016/j.bbrc.2012.12.108
27 Nevin D E, Pratt J M. A coupled in vitro transcription-translation system for the exclusive synthesis of polypeptides expressed from the T7 promoter. FEBS Letters, 1991, 291(2): 259–263
https://doi.org/10.1016/0014-5793(91)81297-L
28 Tabor S. Expression using the T7 RNA polymerase/promoter system. In: Current Protocols in Molecular Biology. New York: John Wiley & Sons, Inc, 2001
1 Lederman M, Zubay G. DNA-directed peptide synthesis I. A comparison of T2 and Escherichia coli DNA-directed peptide synthesis in two cell-free systems. Biochimica et Biophysica Acta, 1967, 149(1): 253–258
29 Shin J, Noireaux V. An E. coli cell-free expression toolbox: Application to synthetic gene circuits and artificial cells. ACS Synthetic Biology, 2012, 1(1): 29–41
https://doi.org/10.1021/sb200016s
30 Garamella J, Marshall R, Rustad M, Noireaux V. The all E. coli TX-TL toolbox 2.0: A platform for cell-free synthetic biology. ACS Synthetic Biology, 2016, 5(4): 344–355
https://doi.org/10.1021/acssynbio.5b00296
31 Chung B, Lee D Y. Computational codon optimization of synthetic gene for protein expression. BMC Systems Biology, 2012, 6(1): 134
https://doi.org/10.1186/1752-0509-6-134
32 Noren C J, Anthony-Cahill S J, Griffith M C, Schultz P G. A general method for site-specific incorporation of unnatural amino acids into proteins. Science, 1989, 244(4901): 182–188
https://doi.org/10.1126/science.2649980
33 Hong S H, Kwon Y C, Jewett M C. Non-standard amino acid incorporation into proteins using Escherichia coli cell-free protein synthesis. Frontiers in Chemistry, 2014, 2: 34
https://doi.org/10.3389/fchem.2014.00034
34 Oza J P, Aerni H R, Pirman N L, Barber K W, ter Haar C M, Rogulina S, Amrofell M B, Isaacs F J, Rinehart J, Jewett M C. Robust production of recombinant phosphoproteins using cell-free protein synthesis. Nature Communications, 2015, 6: 8168
https://doi.org/10.1038/ncomms9168
35 Singh-Blom A, Hughes R A, Ellington A D. An amino acid depleted cell-free protein synthesis system for the incorporation of non-canonical amino acid analogs into proteins. Journal of Biotechnology, 2014, 178: 12–22
https://doi.org/10.1016/j.jbiotec.2014.02.009
36 Shimizu Y, Inoue A, Tomari Y, Suzuki T, Yokogawa T, Nishikawa K, Ueda T. Cell-free translation reconstituted with purified components. Nature Biotechnology, 2001, 19(8): 751–755
https://doi.org/10.1038/90802
37 Wang H H, Huang P Y, Xu G, Haas W, Marblestone A, Li J, Gygi S P, Forster A C, Jewett M C, Church G M. Multiplexed in vivo his-tagging of enzyme pathways for in vitro single-pot multienzyme catalysis. ACS Synthetic Biology, 2012, 1(2): 43–52
https://doi.org/10.1021/sb3000029
38 Hillebrecht J R, Chong S. A comparative study of protein synthesis in in vitro systems: From the prokaryotic reconstituted to the eukaryotic extract-based. BMC Biotechnology, 2008, 8(1): 1–9
https://doi.org/10.1186/1472-6750-8-58
39 Kazuta Y, Matsuura T, Ichihashi N, Yomo T. Synthesis of milligram quantities of proteins using a reconstituted in vitro protein synthesis system. Journal of Bioscience and Bioengineering, 2014, 118(5): 554–557
https://doi.org/10.1016/j.jbiosc.2014.04.019
40 Li J, Gu L, Aach J, Church G M. Improved cell-free RNA and protein synthesis system. PLoS One, 2014, 9(9): e106232
https://doi.org/10.1371/journal.pone.0106232
41 Niwa T, Kanamori T, Ueda T, Taguchi H. Global analysis of chaperone effects using a reconstituted cell-free translation system. Proceedings of the National Academy of Sciences of the United States of America, 2012, 109(23): 8937–8942
https://doi.org/10.1073/pnas.1201380109
42 Gibson D G, Young L, Chuang R Y, Venter J C, Hutchison C A, Smith H O. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nature Methods, 2009, 6(5): 343–345
https://doi.org/10.1038/nmeth.1318
43 Pachuk C J, Samuel M, Zurawski J A, Snyder L, Phillips P, Satishchandran C. Chain reaction cloning: A one-step method for directional ligation of multiple DNA fragments. Gene, 2000, 243(1-2): 19–25
https://doi.org/10.1016/S0378-1119(99)00508-9
44 Chappell J, Jensen K, Freemont P S. Validation of an entirely in vitro approach for rapid prototyping of DNA regulatory elements for synthetic biology. Nucleic Acids Research, 2013, 41(5): 3471–3481
https://doi.org/10.1093/nar/gkt052
45 Sun Z Z, Yeung E, Hayes C A, Noireaux V, Murray R M. Linear DNA for rapid prototyping of synthetic biological circuits in an Escherichia coli based TX-TL cell-free system. ACS Synthetic Biology, 2014, 3(6): 387–397
https://doi.org/10.1021/sb400131a
46 Karzbrun E, Tayar A M, Noireaux V, Bar-Ziv R H. Programmable on-chip DNA compartments as artificial cells. Science, 2014, 345(6198): 829–832
https://doi.org/10.1126/science.1255550
47 Elowitz M B, Leibler S. A synthetic oscillatory network of transcriptional regulators. Nature, 2000, 403(6767): 335–338
https://doi.org/10.1038/35002125
48 Takahashi M K, Hayes C A, Chappell J, Sun Z Z, Murray R M, Noireaux V, Lucks J B. Characterizing and prototyping genetic networks with cell-free transcription-translation reactions. Methods (San Diego, CA), 2015, 86: 60–72
https://doi.org/10.1016/j.ymeth.2015.05.020
49 Niederholtmeyer H, Sun Z Z, Hori Y, Yeung E, Verpoorte A, Murray R M, Maerkl S J. Rapid cell-free forward engineering of novel genetic ring oscillators. eLife, 2015, 4: e09771
https://doi.org/10.7554/eLife.09771
50 Karim A S, Jewett M C. A cell-free framework for rapid biosynthetic pathway prototyping and enzyme discovery. Metabolic Engineering, 2016, 36: 116–126
https://doi.org/10.1016/j.ymben.2016.03.002
51 Fujii S, Matsuura T, Sunami T, Kazuta Y, Yomo T. In vitro evolution of a-hemolysin using a liposome display. Proceedings of the National Academy of Sciences of the United States of America, 2013, 110(42): 16796–16801
https://doi.org/10.1073/pnas.1314585110
52 Pardee K, Green A A, Ferrante T, Cameron D E, DaleyKeyser A, Yin P, Collins J J. Paper-based synthetic gene networks. Cell, 2014, 159(4): 940–954
https://doi.org/10.1016/j.cell.2014.10.004
53 Dudley Q M, Karim A S, Jewett M C. Cell-free metabolic engineering: Biomanufacturing beyond the cell. Biotechnology Journal, 2015, 10(1): 69–82
https://doi.org/10.1002/biot.201400330
54 Zhang Y H P. Production of biofuels and biochemicals by in vitro synthetic biosystems: Opportunities and challenges. Biotechnology Advances, 2015, 33(7): 1467–1483
https://doi.org/10.1016/j.biotechadv.2014.10.009
55 Zhang Y, Sun J, Zhong J J. Biofuel production by in vitro synthetic enzymatic pathway biotransformation. Current Opinion in Biotechnology, 2010, 21(5): 663–669
https://doi.org/10.1016/j.copbio.2010.05.005
56 Zawada J F, Yin G, Steiner A R, Yang J, Naresh A, Roy S M, Gold D S, Heinsohn H G, Murray C J. Microscale to manufacturing scale-up of cell-free cytokine production—a new approach for shortening protein production development timelines. Biotechnology and Bioengineering, 2011, 108(7): 1570–1578
https://doi.org/10.1002/bit.23103
57 Dudley Q M, Anderson K C, Jewett M C. Cell-free mixing of Escherichia coli crude extracts to prototype and rationally engineer high-titer mevalonate synthesis. ACS Synthetic Biology, 2016,
https://doi.org/10.1021/acssynbio.6b00154
58 Joyce G F. Forty years of in vitro evolution. Angewandte Chemie International Edition, 2007, 46(34): 6420–6436
https://doi.org/10.1002/anie.200701369
59 Roberts R W, Szostak J W. RNA-peptide fusions for the in vitro selection of peptides and proteins. Proceedings of the National Academy of Sciences of the United States of America, 1997, 94(23): 12297–12302
https://doi.org/10.1073/pnas.94.23.12297
60 Hanes J, Pluckthun A. In vitro selection and evolution of functional proteins by using ribosome display. Proceedings of the National Academy of Sciences of the United States of America, 1997, 94(10): 4937–4942
https://doi.org/10.1073/pnas.94.10.4937
61 Sumida T, Yanagawa H, Doi N.In vitro selection of fab fragments by mRNA display and gene-linking emulsion PCR. Journal of Nucleic Acids, 2012, 2012: 371–379
62 Stafford R L, Matsumoto M L, Yin G, Cai Q, Fung J J, Stephenson H, Gill A, You M, Lin S H, Wang W D, Masikat M R, Li X, Penta K, Steiner A R, Baliga R, Murray C J, Thanos C D, Hallam T J, Sato A K. In vitro fab display: A cell-free system for IgG discovery. Protein Engineering, Design & Selection, 2014, 27(4): 97–109
https://doi.org/10.1093/protein/gzu002
63 Zhou H, Yong J, Gao H, Li T, Xiao H, Wu Y. Mannanase man23 mutants library construction basing on a novel cell-free protein expression system. Journal of the Science of Food and Agriculture, 2016,
https://doi.org/10.1002/jsfa.8029
64 Dodevski I, Markou G C, Sarkar C A. Conceptual and methodological advances in cell-free directed evolution. Current Opinion in Structural Biology, 2015, 33: 1–7
https://doi.org/10.1016/j.sbi.2015.04.008
65 Guillen Schlippe Y V, Hartman M C T, Josephson K, Szostak J W. In vitro selection of highly modified cyclic peptides that act as tight binding inhibitors. Journal of the American Chemical Society, 2012, 134(25): 10469–10477
https://doi.org/10.1021/ja301017y
66 Miller O J, Bernath K, Agresti J J, Amitai G, Kelly B T, Mastrobattista E, Taly V, Magdassi S, Tawfik D S, Griffiths A D. Directed evolution by in vitro compartmentalization. Nature Methods, 2006, 3(7): 561–570
https://doi.org/10.1038/nmeth897
67 Soh S, Banaszak M, Kandere-Grzybowska K, Grzybowski B A. Why cells are microscopic: A transport-time perspective. Journal of Physical Chemistry Letters, 2013, 4(6): 861–865
https://doi.org/10.1021/jz3019379
68 Georgi V, Georgi L, Blechert M, Bergmeister M, Zwanzig M, Wustenhagen D A, Bier F F, Jung E, Kubick S. On-chip automation of cell-free protein synthesis: New opportunities due to a novel reaction mode. Lab on a Chip, 2016, 16(2): 269–281
https://doi.org/10.1039/C5LC00700C
69 Deans T L, Cantor C R, Collins J J. A tunable genetic switch based on RNAi and repressor proteins for regulating gene expression in mammalian cells. Cell, 2007, 130(2): 363–372
https://doi.org/10.1016/j.cell.2007.05.045
70 Ellis T, Wang X, Collins J J. Diversity-based, model-guided construction of synthetic gene networks with predicted functions. Nature Biotechnology, 2009, 27(5): 465–471
https://doi.org/10.1038/nbt.1536
71 Tawfik D S, Griffiths A D. Man-made cell-like compartments for molecular evolution. Nature Biotechnology, 1998, 16(7): 652–656
https://doi.org/10.1038/nbt0798-652
72 Griffiths A D, Tawfik D S. Miniaturising the laboratory in emulsion droplets. Trends in Biotechnology, 2006, 24(9): 395–402
https://doi.org/10.1016/j.tibtech.2006.06.009
73 Sharma B, Takamura Y, Shimoda T, Biyani M. A bulk sub-femtoliter in vitro compartmentalization system using super-fine electrosprays. Scientific Reports, 2016, 6: 26257
https://doi.org/10.1038/srep26257
74 Weitz M, Kim J, Kapsner K, Winfree E, Franco E, Simmel F C. Diversity in the dynamical behaviour of a compartmentalized programmable biochemical oscillator. Nature Chemistry, 2014, 6(4): 295–302
https://doi.org/10.1038/nchem.1869
75 Kapsner K, Simmel F C. Partitioning variability of a compartmentalized in vitro transcriptional thresholding circuit. ACS Synthetic Biology, 2015, 4(10): 1136–1143
https://doi.org/10.1021/acssynbio.5b00051
76 Sachse R, Dondapati S K, Fenz S F, Schmidt T, Kubick S. Membrane protein synthesis in cell-free systems: From bio-mimetic systems to bio-membranes. FEBS Letters, 2014, 588(17): 2774–2781
https://doi.org/10.1016/j.febslet.2014.06.007
77 Caschera F, Noireaux V. Compartmentalization of an all-E. coli cell-free expression system for the construction of a minimal cell. Artificial Life, 2016, 22(2): 185–195
https://doi.org/10.1162/ARTL_a_00198
78 Tan C, Saurabh S, Bruchez M P, Schwartz R, Leduc P. Molecular crowding shapes gene expression in synthetic cellular nanosystems. Nature Nanotechnology, 2013, 8(8): 602–608
https://doi.org/10.1038/nnano.2013.132
79 Hovijitra N T, Wuu J J, Peaker B, Swartz J R. Cell-free synthesis of functional aquaporin Z in synthetic liposomes. Biotechnology and Bioengineering, 2009, 104(1): 40–49
https://doi.org/10.1002/bit.22385
80 Maeda Y T, Nakadai T, Shin J, Uryu K, Noireaux V, Libchaber A. Assembly of MreB filaments on liposome membranes: A synthetic biology approach. ACS Synthetic Biology, 2012, 1(2): 53–59
https://doi.org/10.1021/sb200003v
81 Ishihara G, Goto M, Saeki M, Ito K, Hori T, Kigawa T, Shirouzu M, Yokoyama S. Expression of G protein coupled receptors in a cell-free translational system using detergents and thioredoxin-fusion vectors. Protein Expression and Purification, 2005, 41(1): 27–37
https://doi.org/10.1016/j.pep.2005.01.013
82 Pardee K, Green A A, Takahashi M K, Braff D, Lambert G, Lee J W, Ferrante T, Ma D, Donghia N, Fan M, Daringer N M, Bosch I, Dudley D M, O’Connor D H, Gehrke L, Collins J J. Rapid, low-cost detection of Zika virus using programmable biomolecular components. Cell, 2016, 165(5): 1255–1266
https://doi.org/10.1016/j.cell.2016.04.059
83 Davies D H, Liang X, Hernandez J E, Randall A, Hirst S, Mu Y, Romero K M, Nguyen T T, Kalantari-Dehaghi M, Crotty S, Baldi P, Villarreal L P, Felgner P L. Profiling the humoral immune response to infection by using proteome microarrays: High-throughput vaccine and diagnostic antigen discovery. Proceedings of the National Academy of Sciences of the United States of America, 2005, 102(3): 547–552
https://doi.org/10.1073/pnas.0408782102
84 Lu Y, Welsh J P, Chan W, Swartz J R. Escherichia coli-based cell free production of flagellin and ordered flagellin display on virus-like particles. Biotechnology and Bioengineering, 2013, 110(8): 2073–2085
https://doi.org/10.1002/bit.24903
85 Kanter G, Yang J, Voloshin A, Levy S, Swartz J R, Levy R. Cell-free production of scFv fusion proteins: An efficient approach for personalized lymphoma vaccines. Blood, 2007, 109(8): 3393–3399
https://doi.org/10.1182/blood-2006-07-030593
86 Lewis D D, Villarreal F D, Wu F, Tan C. Synthetic biology outside the cell: Linking computational tools to cell-free systems. Frontiers in Bioengineering and Biotechnology, 2014, 2: 66
https://doi.org/10.3389/fbioe.2014.00066
2 DeVries J K, Zubay G. DNA-directed peptide synthesis. II. The synthesis of the alpha-fragment of the enzyme beta-galactosidase. Proceedings of the National Academy of Sciences of the United States of America, 1967, 57(4): 1010–1012
https://doi.org/10.1073/pnas.57.4.1010
3 Zubay G, Lederman M, DeVries J K. DNA-directed peptide synthesis. 3. Repression of beta-galactosidase synthesis and inhibition of repressor by inducer in a cell-free system. Proceedings of the National Academy of Sciences of the United States of America, 1967, 58(4): 1669–1675
https://doi.org/10.1073/pnas.58.4.1669
4 Reichman M, Penman S. Stimulation of polypeptide initiation in vitro after protein synthesis inhibition in vivo in HeLa cells. Proceedings of the National Academy of Sciences of the United States of America, 1973, 70(9): 2678–2682
https://doi.org/10.1073/pnas.70.9.2678
5 Roberts B E, Gorecki M, Mulligan R C, Danna K J, Rozenblatt S, Rich A. Simian virus 40 DNA directs synthesis of authentic viral polypeptides in a linked transcription-translation cell-free system. Proceedings of the National Academy of Sciences of the United States of America, 1975, 72(5): 1922–1926
https://doi.org/10.1073/pnas.72.5.1922
6 Pelham H R. Use of coupled transcription and translation to study mRNA production by vaccinia cores. Nature, 1977, 269(5628): 532–534
https://doi.org/10.1038/269532a0
7 Bottomley W, Whitfeld P R. Cell-free transcription and translation of total spinach chloroplast DNA. European Journal of Biochemistry, 1979, 93(1): 31–39
https://doi.org/10.1111/j.1432-1033.1979.tb12791.x
8 Steggles A W, Wilson G N, Kantor J A, Picciano D J, Falvey A K, Anderson W F. Cell-free transcription of mammalian chromatin: transcription of globin messenger RNA sequences from bone-marrow chromatin with mammalian RNA polymerase. Proceedings of the National Academy of Sciences of the United States of America, 1974, 71(4): 1219–1223
https://doi.org/10.1073/pnas.71.4.1219
9 Yang H L, Ivashkiv L, Chen H Z, Zubay G, Cashel M. Cell-free coupled transcription-translation system for investigation of linear DNA segments. Proceedings of the National Academy of Sciences of the United States of America, 1980, 77(12): 7029–7033
https://doi.org/10.1073/pnas.77.12.7029
10 Kozak M. Context effects and inefficient initiation at non-AUG codons in eucaryotic cell-free translation systems. Molecular and Cellular Biology, 1989, 9(11): 5073–5080
https://doi.org/10.1128/MCB.9.11.5073
11 Gallie D R, Sleat D E, Watts J W, Turner P C, Wilson T M. The 5′-leader sequence of tobacco mosaic virus RNA enhances the expression of foreign gene transcripts in vitro and in vivo. Nucleic Acids Research, 1987, 15(8): 3257–3273
https://doi.org/10.1093/nar/15.8.3257
12 Jang S K, Krausslich H G, Nicklin M J, Duke G M, Palmenberg A C, Wimmer E. A segment of the 5′ nontranslated region of encephalomyocarditis virus RNA directs internal entry of ribosomes during in vitro translation. Journal of Virology, 1988, 62(8): 2636–2643
13 Yost C S, Hedgpeth J, Lingappa V R. A stop transfer sequence confers predictable transmembrane orientation to a previously secreted protein in cell-free systems. Cell, 1983, 34(3): 759–766
https://doi.org/10.1016/0092-8674(83)90532-9
14 Rothblatt J A, Meyer D I. Secretion in yeast: Reconstitution of the translocation and glycosylation of alpha-factor and invertase in a homologous cell-free system. Cell, 1986, 44(4): 619–628
https://doi.org/10.1016/0092-8674(86)90271-0
15 Kwon Y C, Jewett M C. High-throughput preparation methods of crude extract for robust cell-free protein synthesis. Scientific Reports, 2015, 5: 8663
https://doi.org/10.1038/srep08663
16 Fujiwara K, Doi N. Biochemical preparation of cell extract for cell-free protein synthesis without physical disruption. PLoS One, 2016, 11(4): e0154614
https://doi.org/10.1371/journal.pone.0154614
17 Wang X, Liu J, Zheng Y, Li J, Wang H, Zhou Y, Qi M, Yu H, Tang W, Zhao W M. An optimized yeast cell-free system: sufficient for translation of human papillomavirus 58 L1 mRNA and assembly of virus-like particles. Journal of Bioscience and Bioengineering, 2008, 106(1): 8–15
https://doi.org/10.1263/jbb.106.8
18 Niimi T. Leishmania tarentolae for the production of multi-subunit complexes. Advances in Experimental Medicine and Biology, 2016, 896: 155–165
https://doi.org/10.1007/978-3-319-27216-0_10
19 Guild K, Zhang Y, Stacy R, Mundt E, Benbow S, Green A, Myler P J. Wheat germ cell-free expression system as a pathway to improve protein yield and solubility for the SSGCID pipeline. Acta Crystallographica. Section F, Structural Biology and Crystallization Communications, 2011, 67(9): 1027–1031
https://doi.org/10.1107/S1744309111032143
20 Stech M, Quast R B, Sachse R, Schulze C, Wustenhagen D A, Kubick S. A continuous-exchange cell-free protein synthesis system based on extracts from cultured insect cells. PLoS One, 2014, 9(5): e96635
https://doi.org/10.1371/journal.pone.0096635
21 Yadavalli R, Sam-Yellowe T. HeLa based cell free expression systems for expression of plasmodium rhoptry proteins. Journal of Visualized Experiments, 2015, 100: e52772
22 Gagoski D, Polinkovsky M E, Mureev S, Kunert A, Johnston W, Gambin Y, Alexandrov K. Performance benchmarking of four cell-free protein expression systems. Biotechnology and Bioengineering, 2016, 113(2): 292–300
https://doi.org/10.1002/bit.25814
[1] You Han, Yulian Wang, Tengzhou Ma, Wei Li, Jinli Zhang, Minhua Zhang. Mechanistic understanding of Cu-based bimetallic catalysts[J]. Front. Chem. Sci. Eng., 2020, 14(5): 689-748.
[2] Mostafa R. Shirdar, Nasim Farajpour, Reza Shahbazian-Yassar, Tolou Shokuhfar. Nanocomposite materials in orthopedic applications[J]. Front. Chem. Sci. Eng., 2019, 13(1): 1-13.
[3] Kaojin Wang, Satu Strandman, X. X. Zhu. A mini review: Shape memory polymers for biomedical applications[J]. Front. Chem. Sci. Eng., 2017, 11(2): 143-153.
[4] Minghui Sun, Chen Chen, Lihua Chen, Baolian Su. Hierarchically porous materials: Synthesis strategies and emerging applications[J]. Front. Chem. Sci. Eng., 2016, 10(3): 301-347.
[5] Gang FAN,Le YANG,Zhijian CHEN. Water-soluble BODIPY and aza-BODIPY dyes: synthetic progress and applications[J]. Front. Chem. Sci. Eng., 2014, 8(4): 405-417.
[6] Yanni WU, Shijun LIAO. Review of SO42-/MxOy solid superacid catalysts[J]. Front Chem Eng Chin, 2009, 3(3): 330-343.
[7] ZHANG Bo, XING Jianmin, LIU Huizhou. Preparation and application of magnetic microsphere carriers[J]. Front. Chem. Sci. Eng., 2007, 1(1): 96-101.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed