7
Cell-free expression — making a mark Frank Bernhard 1 and Yuzuru Tozawa 2 Cell-free protein production opens new perspectives for the direct manipulation of expression compartments in combination with reduced complexity of physiological requirements. The technology is therefore in particular suitable for the general synthesis of difficult proteins including toxins and membrane proteins as well as for the analysis of their functional folding in artificial environments. A further key application of cell-free expression is the fast and economic labeling of proteins for structural and functional applications. Two extract sources, wheat embryos and Escherichia coli cells, are currently employed for the preparative scale cell-free production of proteins. Recent achievements in structural characterization include cell-free synthesized membrane proteins and even larger protein assemblies may become feasible. Addresses 1 Institute of Biophysical Chemistry, Centre for Biomolecular Magnetic Resonance, Goethe University, Frankfurt am Main, Germany 2 Cell-Free Science and Technology Research Center, Ehime University, Matsuyama, Japan Corresponding author: Bernhard, Frank ([email protected]) Current Opinion in Structural Biology 2013, 23:374380 This review comes from a themed issue on New constructs and expression of proteins Edited by Imre Berger and Lorenz M Mayr For a complete overview see the Issue and the Editorial Available online 27th April 2013 0959-440X/$ see front matter, # 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.sbi.2013.03.012 Introduction Cell-free (CF) expression systems are rapidly evolving as an alternative option for general protein production as well as a primary choice for the synthesis of difficult targets. Frequent examples are toxic proteins [1,2] or membrane integrated proteins but also appli- cations for the CF production of vaccines or small bioactive peptides come into focus [3,4]. Preparative scale samples of proteins for structural approaches can be isolated from only few milliliters of reaction volumes with crude cell extracts from various organ- isms [5,6 ]. The complete control over the amino acid pool facilitates the efficient and versatile incorpora- tion of labeled amino acids as a prerequisite for structural approaches. CF expression is therefore an established option for the economic labeling of proteins analyzed by nuclear magnetic resonance (NMR) spectroscopy [7]. Currently, either extracts from Escherichia coli cells or from wheat germ embryos are efficient enough for pre- parative scale protein production and both systems are implemented as core platforms in structural genomics projects [8,9]. However, protocols for extract preparations from alternative sources such as insect and mammalian cells, protozoa or reconstituted from purified E. coli trans- lation components are emerging and continuously being improved in their efficiencies [1015]. They provide valuable options for applications with analytical amounts of proteins such as characterization of function or folding pathways [16]. In particular insect lysates could have an increased potential of post-translational protein modifi- cations after enrichment with microsome fractions or other supplements. CF expression protocols must be considered as result of subsequent optimization levels addressing yield and sample quality. Complexity of CF protein expression is largely reduced to the central transcription/translation process. The basic yield optimization is therefore a rou- tine approach resulting into high success rates by con- sidering fundamental issues such as template design and reaction compound concentrations. The subsequent sys- tematic modulation of protein quality by taking advantage of the open accessibility of CF reactions is a unique option and can require extensive screening of multitudes of additives or combinations thereof. Supple- mented compounds can act already co-translationally at nascent polypeptide chains and promote folding path- ways or protein stability. This unique option is in particu- lar valuable for designing artificial hydrophobic environments upon membrane protein synthesis and makes CF expression to the most versatile protein pro- duction system available (Figure 1). We summarize cur- rent principles for the preparative scale production of high quality protein samples in CF systems based on E. coli and wheat germ extracts. A further emphasis will be on emerging structural approaches with membrane proteins and on recently established tools for efficient protein labeling. Cell-free expression in E. coli extracts: the versatile working horse Extract preparation out of a variety of standard E. coli lab strains has become a routine and reliable procedure that can be carried out in most biochemical labs. While CF expression is still usually directed by the T7 RNA poly- merase, common promoters recognized by the endogen- ous E. coli RNA polymerase might be considered in future [17]. Routine Mg 2+ ion optimization is highly recom- mended for best performance [18]. Specific additives Available online at www.sciencedirect.com Current Opinion in Structural Biology 2013, 23:374380 www.sciencedirect.com

Cell-free expression—making a mark

  • Upload
    yuzuru

  • View
    216

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Cell-free expression—making a mark

Available online at www.sciencedirect.com

Cell-free expression — making a

markFrank Bernhard1 and Yuzuru Tozawa2

Cell-free protein production opens new perspectives for

the direct manipulation of expression compartments in

combination with reduced complexity of physiological

requirements. The technology is therefore in particular

suitable for the general synthesis of difficult proteins

including toxins and membrane proteins as well as for the

analysis of their functional folding in artificial environments.

A further key application of cell-free expression is the fast

and economic labeling of proteins for structural and

functional applications. Two extract sources, wheat

embryos and Escherichia coli cells, are currently employed

for the preparative scale cell-free production of proteins.

Recent achievements in structural characterization include

cell-free synthesized membrane proteins and even larger

protein assemblies may become feasible.

Addresses1 Institute of Biophysical Chemistry, Centre for Biomolecular Magnetic

Resonance, Goethe University, Frankfurt am Main, Germany2 Cell-Free Science and Technology Research Center, Ehime University,

Matsuyama, Japan

Corresponding author: Bernhard, Frank ([email protected])

Current Opinion in Structural Biology 2013, 23:374–380

This review comes from a themed issue on New constructs andexpression of proteins

Edited by Imre Berger and Lorenz M Mayr

For a complete overview see the Issue and the Editorial

Available online 27th April 2013

0959-440X/$ – see front matter, # 2013 Elsevier Ltd. All rights reserved.

http://dx.doi.org/10.1016/j.sbi.2013.03.012

IntroductionCell-free (CF) expression systems are rapidly evolving

as an alternative option for general protein production

as well as a primary choice for the synthesis of

difficult targets. Frequent examples are toxic proteins

[1,2] or membrane integrated proteins but also appli-

cations for the CF production of vaccines or small

bioactive peptides come into focus [3,4]. Preparative

scale samples of proteins for structural approaches can

be isolated from only few milliliters of reaction

volumes with crude cell extracts from various organ-

isms [5,6�]. The complete control over the amino acid

pool facilitates the efficient and versatile incorpora-

tion of labeled amino acids as a prerequisite for

structural approaches. CF expression is therefore an

established option for the economic labeling of

proteins analyzed by nuclear magnetic resonance

(NMR) spectroscopy [7].

Current Opinion in Structural Biology 2013, 23:374–380

Currently, either extracts from Escherichia coli cells or

from wheat germ embryos are efficient enough for pre-

parative scale protein production and both systems are

implemented as core platforms in structural genomics

projects [8,9]. However, protocols for extract preparations

from alternative sources such as insect and mammalian

cells, protozoa or reconstituted from purified E. coli trans-

lation components are emerging and continuously being

improved in their efficiencies [10–15]. They provide

valuable options for applications with analytical amounts

of proteins such as characterization of function or folding

pathways [16]. In particular insect lysates could have an

increased potential of post-translational protein modifi-

cations after enrichment with microsome fractions or

other supplements.

CF expression protocols must be considered as result of

subsequent optimization levels addressing yield and

sample quality. Complexity of CF protein expression is

largely reduced to the central transcription/translation

process. The basic yield optimization is therefore a rou-

tine approach resulting into high success rates by con-

sidering fundamental issues such as template design and

reaction compound concentrations. The subsequent sys-

tematic modulation of protein quality by taking

advantage of the open accessibility of CF reactions is a

unique option and can require extensive screening of

multitudes of additives or combinations thereof. Supple-

mented compounds can act already co-translationally at

nascent polypeptide chains and promote folding path-

ways or protein stability. This unique option is in particu-

lar valuable for designing artificial hydrophobic

environments upon membrane protein synthesis and

makes CF expression to the most versatile protein pro-

duction system available (Figure 1). We summarize cur-

rent principles for the preparative scale production of

high quality protein samples in CF systems based on E.coli and wheat germ extracts. A further emphasis will be

on emerging structural approaches with membrane

proteins and on recently established tools for efficient

protein labeling.

Cell-free expression in E. coli extracts: theversatile working horseExtract preparation out of a variety of standard E. coli lab

strains has become a routine and reliable procedure that

can be carried out in most biochemical labs. While CF

expression is still usually directed by the T7 RNA poly-

merase, common promoters recognized by the endogen-

ous E. coli RNA polymerase might be considered in future

[17]. Routine Mg2+ ion optimization is highly recom-

mended for best performance [18]. Specific additives

www.sciencedirect.com

Page 2: Cell-free expression—making a mark

Cell-free expression Bernhard and Tozawa 375

Figure 1

Strategies for cell-free protein production

(I) Basic reaction configurations

(a) Batch

(a) (b)

(b) CECF

Dialysismembrane

Micelles

Hybrid Micelles

Membrane protein precipitate CF expression of soluble membrane proteins Reconstituted membrane protein samples

Artificial Membranes

Liposomes

Bicelles

Nanodiscs

Mixed micelles

Lipids

RM

RM

20 hr16ºC

Diffusion

FM

FMRM Waste

ValvePumpunit

Dialysismembraneunit

mRNA

FM

(c) Bilayer (d) Filter-and-Feed

(II) Variation of expression conditions; Reaction modes for membrane protein synthesis

Current Opinion in Structural Biology

Configurations of CF reaction and variations of expression mode for producing membrane proteins. (Ia) One-compartment batch configuration for

throughput optimization or preparative expression; (Ib) Two-compartment CECF configuration with reaction mixture (RM) separated from a feeding

mixture (FM) by a dialysis membrane; (Ic) Bilayer configuration providing improved protein synthesis by slow diffusion mixing of the high-density RM

with the low-density FM; (Id) Filter-and-Feed configuration giving optimized protein yield by programmed intermittent exchange of FM and mRNA

across a dialysis membrane unit. II: Reaction modes for membrane protein production as (a) initial precipitate or (b) solubilized with micelles, hybrid

micelles (combination of different detergents or surfactants), mixed micelles (combination of lipids and detergent), or with artificial membranes (by

adding liposomes, nanodiscs or bicelles).

can significantly improve reaction protocols [19,20] and

efficiencies of the basic one compartment batch configur-

ation (Figure 1) could become sufficient for producing

NMR samples [21]. At special conditions, batch reactions

could even be scaled up to industrial scales in bioreactors

[22]. Still generally more efficient for standard lab scale

applications are continuous exchange cell-free (CECF)

configurations providing extra pools of fresh precursors

during the reaction (Figure 1) and several milligrams of

protein can be obtained per milliliter of reaction on a

routine basis [5,18,23].

www.sciencedirect.com

For the expression of each new target, protein yield and/or

quality optimization is often mandatory. It has been ident-

ified that poor initial protein synthesis is often correlated to

inefficient initiation of translation and systematic DNA

template optimization can have dramatic effects

[18,21,24,25]. For improved sample quality, the systematic

co-translational screening of protein stabilizers such as deter-

gents or lipids in case of membrane proteins is highly

recommended [18,23]. The disulfide bond formation in

eukaryotic proteins can be modulated by adjusting redox

conditions directly in the CF reactions [20,22,26–28].

Current Opinion in Structural Biology 2013, 23:374–380

Page 3: Cell-free expression—making a mark

376 New constructs and expressions of proteins

Bacterial CF extracts are routinely used for structural

approaches of soluble proteins of both eukaryotic and

prokaryotic origin. A frequent application is structure deter-

mination by NMR which is based on the excellent labeling

options offered by CF expression (see paragraph below). As

an example, more than 1000 solution NMR structures of

eukaryotic proteins or protein domains have been deter-

mined by the RIKEN Genomic Sciences Center [8].

Wheat germ extracts: mimicking eukaryoticenvironmentsEukaryotic CF translation systems derived from wheat

germ (WG) extracts have been fundamentally reinvented

by improved stability of translation activity, utilization of

non-capped mRNAs in combination with translational

enhancer cis elements [6�]. New translation reactions

and implementation of robotic systems facilitate through-

put approaches [29,30�]. The special bilayer configuration

complements the more common CECF configuration in

WGCF reactions (Figure 1). The recently developed

‘Filter-and-Feed’ translation method (Figure 1) enables

robotic synthesis of up to 50 milligrams protein in reaction

volumes of 10 milliliter in 18 hours [30�]. The WG is kept

in a dehydrated state for hibernation and most organelles

such as plastids, mitochondria and vacuoles are not yet fully

developed. Consequently, activity of amino acid metabo-

lizing enzymes is low. The hexaploid genome of cultivated

wheat crops results into increased grain sizes, thus facil-

itating the preparation of purified embryos. Although

wheat grains as well as other plant seeds contain various

enzyme inhibitors that affect translation, most of them are

localized in the endosperm. After the physical detachment

of the embryo part (germ) from endosperm, further exten-

sive washing of the germ followed by extraction results in

inhibitor-free translation efficient extracts [6�].

In general, preparation of WG extracts is more compli-

cated and time consuming if compared with the prep-

aration of E. coli lysates. User of WGCF systems therefore

may depend more on commercial sources. As an eukar-

yotic system, WG extracts may contain components that

are necessary for promoting correct folds of translated

eukaryotic multi-domain proteins [31]. However, eukar-

yotic as well as prokaryotic proteins can generally be

produced in both systems if protocols have been opti-

mized according to the individual system requirements.

WG extracts are considered for protein structural studies

since the early 2000s and have been adopted as one main

tool for structural studies of eukaryotic proteins by pre-

dominantly NMR approaches [9]. More recently, also

crystallization and X-ray analysis of proteins expressed

in the WGCF system becomes popular [32].

Cell-free production of membrane proteins:particular requirements of challenging targetsCF expression of membrane proteins deserves a particu-

lar consideration as it provides a new and versatile

Current Opinion in Structural Biology 2013, 23:374–380

pipeline for the efficient production of this usually very

difficult to synthesize class of proteins [23,33,34]. CF

extracts from E. coli as well as from WG tolerate a wide

range of detergents as additives for the co-translational

solubilization of CF expressed membrane proteins

(Figure 1) [23,33,35]. The portfolio of potential hydro-

phobic additives for membrane protein solubilization is

continuously increased by new surfactants, amphipols or

other compounds [36–40]. The particular compound(s)

suitable to result into optimal quality of a given mem-

brane protein has to be identified by systematic screening

and compound mixtures might be considered as well

[18,35,41,42].

If detergents should be avoided, the co-translational

insertion into provided lipid bilayers is becoming an

interesting option [42,43]. The addition of vesicles or

liposomes of defined compositions into CF reactions

could result into functionally active membrane proteins

that could be analyzed in natural lipid environments of

desired composition [43,44�,45,46]. The underlying

mechanisms for efficient membrane protein transloca-

tion in the artificial in vitro reaction environment are

currently subject of research and might differ from

known in vivo pathways [44�]. In particular, the recent

combination of CF expression with the nanodisc tech-

nology appears to be promising (Figure 1). The co-

translational or post-translational insertion of CF

expressed membrane proteins into nanodiscs with differ-

ent membrane compositions is emerging as an excellent

tool for their functional and structural characterization

[43,47,48�,49�]. In the absence of hydrophobic additives,

the CF synthesized membrane proteins will precipitate

but could retain in many cases at least partially folded

conformations [27,42,50,51]. After non-denaturating

post-translational solubilization in detergent, such CF

produced membrane protein precipitates are frequently

already suitable for functional and structural studies

[27,41,42,50,51,52�].

Structural studies of CF-expressed membrane proteins

have been approached so far in particular by NMR

spectroscopy (Table 1). The NMR structure of the C-

terminal fragment of human presenilin-1, a subunit of the

g-secretase complex, has been solved with re-solubilized

CF produced precipitates [51]. Systematic NMR screen-

ing of re-solubilized CF produced membrane protein

precipitates can result into the routine identification of

samples suitable for structural evaluation [50,52�]. The

NMR structure of a functional membrane protein cotran-

slational solubilized in the presence of detergent/lipid

mixtures was first solved from a bacterial proteorhodopsin

[53�]. Even the structural evaluation of CF expressed

membrane complexes appears to become feasible [54�]. A

542 kDa bacterial ATP synthase complex composed out

of 25 protomers did fully assemble during CF production

in the presence of detergents.

www.sciencedirect.com

Page 4: Cell-free expression—making a mark

Cell-free expression Bernhard and Tozawa 377

Table 1

Structural approaches with CF expressed membrane proteins

Protein Origin Characteristics Method Reference

Proteorhodopsin Bacteria Proton pump NMR [53�]

Rhodopsin II Acetabularia sp. Proton pump X-ray [57�]

Presenilin-1 CTF Human Subunit of g-secretase NMR [51]

ATP synthase Caldalkalibacillus sp. 542 kDa complex EM [54�]

hVDAC1 Human Ion channel X-ray [55]

EmrE E. coli Multidrug transporter X-ray [56]

Transmembrane domains of ArcB, QseC, KdpD E. coli Histidine sensor kinases NMR [50]

Six inner membrane proteins Human Unknown NMR [52�]

Reports on the crystallization of CF expressed membrane

proteins are still emerging and comprise lower resolution

crystals of the human voltage-dependent anion channel-1

[55] and crystals of the selenomethionine labeled deriva-

tive of the bacterial small multidrug transporter EmrE

[56]. A recent highlight was the 3.2 A structure of an

eukaryotic rhodopsin by in meso crystallization after CF

expression in the presence of a mixture of lipids and

detergent [57�].

Protein labeling in CF systems: essentialprerequisites for structural approachesEfficient labeling of proteins with non-natural or chemi-

cally modified amino acids is one of the major benefits of

CF expression systems for NMR and X-ray structural

analysis [5,8,9]. Toxic side effects caused by the incorp-

oration of amino acids modified with isotopes, biotin or

with fluorescent or photoreactive groups into proteins as

well as many scrambling problems resulting from meta-

bolic conversions of the supplied label precursors are

generally eliminated or significantly reduced by using

CF expression. The labeling costs in particular for

NMR are lower than by in vivo expression and can be

even more reduced by regeneration of unstable amino

acids [58]. Moreover, there are no restrictions for selective

or combinatorial labeling schemes. Consequently, WG as

well as E. coli extracts have been established as core

platforms for the preparation of labeled protein samples

suitable for NMR structural analysis and also for crystal-

lization [5,8,9]. At least 1200 entries in the Protein Data

Bank (PDB) can currently be attributed to cell-free

expressed proteins. During the last five years, the annual

number of new entries is relatively constant while first

structures of cell-free expressed membrane proteins start

to appear.

A variety of supplements and extract modifications have

been developed in order to increase label stability and to

reduce residual scrambling problems for NMR appli-

cations in E. coli as well as in WG extracts. Extracts from

scrambling enzyme deficient strains or addition of chemi-

cal inhibitors address specific scrambling problems [59–61]. A more general approach is the broadband inhibition

www.sciencedirect.com

of cofactor pyridoxal phosphate containing enzymes by

treatment of the CF extract with NaBH4 [62�]. Besides

uniform and amino acid specific labeling, a variety of

combinatorial labeling schemes have become standard

tools for the structural determination of CF expressed

proteins by NMR. Such specific labeling schemes are in

particular required for membrane proteins due to the

generally increased number of ambiguous assignments

[50,51,52�,53�]. The stereo array isotope labeling (SAIL)

approach by using synthesized amino acids with specific

labeling patterns almost exclusively relies on efficient CF

protein production in smaller volumes [63]. The potential

of SAIL applications for spectral simplification and sen-

sitivity enhancement of in particular larger proteins is still

becoming extended [64]. The selective study of func-

tional sites becomes possible by advanced strategies of

site-specific labeling approaches implementing suppres-

sor tRNAs mischarged with unnatural amino acids either

by chemical acylation or by engineered aminoacyl-tRNA

synthetases [65]. The recently improved protocols for the

CF labeling of proteins for NMR applications comp-

lement the already established tools for efficient incorp-

oration of selenomethionine for X-ray analysis or of

modified amino acids for functional studies [66].

ConclusionsCF expression systems based on either E. coli or WG

extracts are established tools for the preparative scale

production of protein samples for structural applications.

The considerable success rate in protein production in

combination with the high versatility in reaction modi-

fication allows to focus on selected targets by systematic

protocol development. The manifold options to design

artificial expression environments of proteins open new

avenues for their synthesis and characterization. CF

expression appears to become one method of choice

for the analysis of difficult proteins such as membrane

proteins, toxins or complexes and it is already a prime

working platform for the general production of labeled

samples for NMR applications. Future developments

including a broader variety of efficient CF extract

sources and improved protocols for the preparation of

crystallization grade membrane proteins come into sight.

Current Opinion in Structural Biology 2013, 23:374–380

Page 5: Cell-free expression—making a mark

378 New constructs and expressions of proteins

As major challenge, there still remains the preparative

scale CF production of post-translationally modified

proteins.

Acknowledgements

We thank Dr Yaeta Endo for his critical comments on this manuscript. Thiswork was supported by MEXT KAKENHI Grant Number 24117516 toY.T.

References and recommended readingPapers of particular interest, published within the period of review,have been highlighted as:

� of special interest�� of outstanding interest

1. Orth JHC, Schorch B, Boundy S, Ffrench-Constant R, Kubick S,Aktories K: Cell-free synthesis and characterization of a novelcytotoxic piersin-like protein from the cabbage butterfly Pierisrapae. Toxicon 2010, 57:199-207.

2. Villate M, Merino N, Blanco FJ: Production of meganucleases bycell-free protein synthesis for functional and structuralstudies. Protein Expr Purif 2012, 85:246-249.

3. Lee KH, Kwon YC, Yoo SJ, Kim DM: Ribosomal synthesis and insitu isolation of peptide molecules in a cell-free translationsystem. Protein Expr Purif 2010, 71:16-20.

4. Tsuboi T, Takeo S, Sawasaki T, Torii M, Endo Y: An efficientapproach to the production of vaccines against the Malariaparasite. Methods Mol Biol 2010, 607:73-85.

5. Kigawa T, Yabuki T, Yoshida Y, Tsutsui M, Ito Y, Shibata T,Yokoyama S: Cell-free production and stable-isotope labelingof milligram quantities of proteins. FEBS Lett 1999, 442:15-19.

6.�

Takai K, Sawasaki T, Endo Y: Practical cell-free proteinsynthesis system using purified wheat embryos. Nat Protoc2010, 5:227-238.

A detailed description of WG extract preparation for small and large-scaleCF protein production by hands-down operations.

7. Sobhanifar S, Reckel S, Junge F, Schwarz D, Kai L, Karbyshev M,Lohr F, Bernhard F, Dotsch V: Cell-free expression and stableisotope labelling strategies for membrane proteins. J BiomolNMR 2010, 46:33-43.

8. Kigawa T: Cell-free protein production system with the E. colicrude extract for determination of protein folds. Methods MolBiol 2010, 607:101-111.

9. Makino SI, Goren MA, Fox BG, Markley JL: Cell-free proteinsynthesis technology in NMR high-throughput structuredetermination. Methods Mol Biol 2010, 607:127-149.

10. Mikami S, Kobayashi T, Imataka H: Cell-free protein synthesiswith extracts from cultured human cells. Methods Mol Biol2010, 607:43-53.

11. Pratt CA, Mowry KL: Preparation of a highly active cell-freetranslation system from immature Xenopus laevis oocytes.Methods 2010, 51:101-105.

12. Shimizu Y, Ueda T: Pure technology. Methods Mol Biol 2010,607:11-23.

13. Wakiyama M, Kaitsu Y, Matsumoto T, Yokoyama S: Coupledtranscription and translation from polymerase chain reaction-amplified DNA in Drosophila Schneider 2 cell-free system. AnalBiochem 2010, 400:142-144.

14. Kovtun O, Mureev S, Jung W, Kubala MH, Wayne J, Kirill A:Leishmania cell-free protein expression system. Methods2011, 55:58-64.

15. Sato Y, Aizawa K, Ezure T, Ando E, Uozumi N: A simple fed-batchmethod for transcription and insect cell-free translation. JBiosci Bioeng 2012, 114:677-679.

Current Opinion in Structural Biology 2013, 23:374–380

16. Niwa T, Kanamori T, Ueda T, Taguchi H: Global analysis ofchaperone effects using a reconstituted cell-free translationsystem. Proc Natl Acad Sci USA 2012, 109:8937-8942.

17. Shin J, Noireaux V: Efficient cell-free expression with theendogenous E. coli RNA polymerase and sigma factor 70. JBiol Eng 2010, 4:8.

18. Rath P, Demange P, Saurel O, Tropis M, Daffe M, Dotsch V,Ghazi A, Bernhard F, Milon A: Functional expression of thePorAH channel from Corynebacterium glutamicum in cell-freeexpression systems: implications for the role of the naturaloccurring mycolic acid modification. J Biol Chem 2011,286:32525-32532.

19. Kim HC, Kim TW, Kim DM: Prolonged production of proteins in acell-free protein synthesis system using polymericcarbohydrates as an energy source. Process Biochem 2011,46:1366-1369.

20. Shingaki T, Nimura N: Improvement of translation efficiency inan Escherichia coli cell-free protein system using cysteine.Protein Expr Purif 2011, 77:193-197.

21. Michel E, Wuthrich K: High-yield Escherichia coli-based cell-free expression of human proteins. J Biomol NMR 2012, 53:43-51.

22. Zawada JF, Yin G, Steiner AR, Yang J, Naresh A, Roy SM, Gold D,Heinsohn HG, Murray CJ: Microscale to manufacturing scale-up of cell-free cytokine production — a new approach forshortening protein production development timelines.Biotechnol Bioeng 2011, 108:1570-1578.

23. Reckel S, Sobhanifar S, Durst F, Lohr F, Shirokov VA, Dotsch V,Bernhard F: Strategies for the cell-free expression ofmembrane proteins. Methods Mol Biol 2010, 607:187-212.

24. Kralicek AV, Radjainia M, Mohamad A, Nazratul AB, Carraher C,Newcomb RD, Mitra AK: A PCR-directed cell-free approach tooptimize protein expression using diverse fusion tags. ProteinExpr Purif 2011, 80:117-124.

25. Haberstock S, Roos C, Hoevels Y, Dotsch V, Schnapp G,Pautsch A, Bernhard F: A systematic approach to increase theefficiency of membrane protein production in cell-freeexpression systems. Protein Expr Purif 2012, 82:308-316.

26. Bundy BC, Swartz JR: Efficient disulfide bond formation invirus-like particles. J Biotechnol 2011, 154:230-239.

27. Keller T, Eggenberger B, Gorboulev V, Bernhard F, Uzelac Z,Gorbunov D, Witte C, Koppatz S, Doetsch V, Hunte C et al.: Thelarge extracellular loop of organic cation transporter 1influences substrate affinity and is pivotal for oligomerization.J Biol Chem 2011, 286:37874-37886.

28. Michel E, Wuthrich K: Cell-free expression of disulfide-containing eukaryotic proteins for structural biology. FEBS J2012, 279:3176-3184.

29. Sawasaki T, Hasegawa Y, Tsuchimochi M, Kamura N,Ogasawara T, Kuroita T, Endo Y: A bilayer cell-free proteinsynthesis system for high-throughput screening of geneproducts. FEBS Lett 2002, 514:102-105.

30.�

Beebe ET, Makino S, Nozawa A, Matsubara Y, Frederick RO,Primm JG, Goren MA, Fox BG: Robotic large-scale applicationof wheat cell-free translation to structural studies includingmembrane proteins. Nat Biotechnol 2011, 28:239-249.

The authors describe a robotic system, which is based on ‘Filter-and-Feed’ method, for large-scale protein synthesis including membraneproteins.

31. Goshima N, Kawamura Y, Fukumoto A, Miura A, Honma R,Satoh R, Wakamatsu A, Yamamoto J, Kimura K, Nishikawa T et al.:Human protein factory for converting the transcriptome intoan in vitro-expressed proteome. Nat Methods 2008, 5:1011-1017.

32. Watanabe M, Miyazono KI, Tanokura M, Sawasaki T, Endo Y,Kobayashi I: Cell-free protein synthesis for structuredetermination by X-ray crystallography. Methods Mol Biol 2010,607:149-161.

www.sciencedirect.com

Page 6: Cell-free expression—making a mark

Cell-free expression Bernhard and Tozawa 379

33. Nozawa A, Hideaki N, Tozawa Y: Production of membraneproteins through the wheat-germ cell-free technology.Methods Mol Biol 2010, 607:213-218.

34. Isaksson L, Enberg J, Neutze R, Goran KB, Pedersen A:Expression screening of membrane proteins with cell-freesynthesis. Protein Expr Purif 2012, 82:218-225.

35. Genji T, Nozawa A, Tozawa Y: Efficient production andpurification of functional bacteriorhodopsin with a wheat-germ cell-free system and a combination of Fos-choline andCHAPS detergents. Biochem Biophys Res Commun 2010,400:638-642.

36. Guild K, Zhang Y, Stacy R, Mundt E, Benbow S, Green A, Myler P:Wheat germ cell-free expression system as a pathway toimprove yield and solubility for the SSGCID pipeline. ActaCrystallogr Sect F 2011, 67:1027-1031.

37. Klammt C, Perrin MH, Maslennikov I, Renault I, Krupa M,Kwiatkowski W, Stahlberg H, Vale W, Choe S: Polymer-basedcell-free expression of ligand-binding family B G-proteincoupled receptors without detergents. Protein Sci 2011,20:1030-1041.

38. Park KH, Billon-Denis E, Dahmane T, Lebaupain F, Pucci B,Breyton C, Zito F: In the cauldron of cell-free synthesis ofmembrane proteins: playing with new surfactants. NatBiotechnol 2011, 28:255-261.

39. Wang X, Corin K, Baaske P, Wienken CJ, Jerabek-Willemsen M,Duhr S, Braun D, Zhang S: Peptide surfactants for cell-freeproduction of functional G protein-coupled receptors. ProcNatl Acad Sci USA 2011, 108:9049-9054.

40. Blesneac I, Ravaud S, Juillan-Binard C, Barret LA, Zoonens M,Polidori A, Miroux B, Pucci B, Pebay-Peyroula E: Production ofUCP1 a membrane protein from the inner mitochondrialmembrane using the cell free expression system in thepresence of a fluorinated surfactant. Biochim Biophys Acta:Biomembr 2012, 1818:798-805.

41. Junge F, Luh LM, Proverbio D, Schafer B, Abele R, Beyermann M,Dotsch V, Bernhard F: Modulation of G-protein coupledreceptor sample quality by modified cell-free expressionprotocols: a case study of the human endothelin A receptor. JStruct Biol 2010, 172:94-106.

42. Ma Y, Muench D, Schneider T, Sahl HG, Bouhss A,Ghoshdastider U, Wang J, Doetsch V, Wang X, Bernhard F:Preparative scale cell-free production and qualityoptimization of MraY homologues in different expressionmodes. J Biol Chem 2011, 286:38844-38853.

43. Periasamy A, Shadiac N, Amalraj A, Garajova S, Nagarajan Y,Waters S, Mertens HDT, Hrmova M: Cell-free synthesis ofmembrane (1,3)-b-D-glucan (curdlan) synthase: co-translational insertion in liposomes and reconstitution innanodiscs. Biochim Biophys Acta: Biomembr 2012, 1828:743-757.

44.�

Berrier C, Guivout I, Bayan N, Park KH, Mesneau A, Chami M,Pugsley A, Ghazi A: Coupled cell-free synthesis and lipidvesicle insertion of a functional oligomeric channel MscL:MscL does not need the insertase YidC for insertion in vitro.Biochim Biophys Acta: Biomembr 2011, 1808:41-46.

A detailed study of membrane protein insertion mechanisms into definedlipid bilayers.

45. Nozawa A, Ogasawara T, Matsunaga S, Iwasaki T, Sawasaki T,Endo Y: Production and partial purification of membraneproteins using a liposome-supplemented wheat cell-freetranslation system. BMC Biotechnol 2011, 11:35.

46. Long AR, O’Brian CC, Alder NN: The cell-free integration of apolytopic mitochondrial membrane protein into liposomesoccurs cotranslationally and in a lipid-dependent manner.PLoS One 2012:7.

47. Gao T, Petriova J, He W, Huser T, Kudlicki W, Voss J, Coleman MA:Characterization of de novo synthesized GPCRs supported innanolipoprotein discs. PLoS One 2012, 7:e44911.

48.�

Lyukmanova EN, Shenkarev ZO, Khabibullina NF, Kopeina GS,Shulepko MA, Paramonov AS, Mineev KS, Tikhonov RV,Shingarova LN, Petrovskaya LE et al.: Lipid–protein nanodiscsfor cell-free production of integral membrane proteins in a

www.sciencedirect.com

soluble and folded state: comparison with detergent micelles,bicelles and liposomes. Biochim Biophys Acta: Biomembr 2012,1818:349-358.

Systematic analysis of sample quality of membrane proteins synthesizedin different CF reaction environments

49.�

Roos C, Zocher M, Muller D, Munch D, Schneider T, Sahl HG,Scholz F, Wachtveitl J, Ma Y, Proverbio D et al.: Characterizationof co-translationally formed nanodisc complexes with smallmultidrug transporters, proteorhodopsin and with the E. coliMraY translocase. Biochim Biophys Acta: Biomembr 2012,1818:3098-3106.

Detailed analysis of the co-translationally insertion of different membraneproteins into preformed nanodiscs and characterization of lipid depen-dent activities.

50. Maslennikov I, Klammt C, Hwang E, Kefala G, Okamura M,Esquivies L, Mors K, Glaubitz C, Kwiatkowski W, Jeon YH et al.:Membrane domain structures of three classes of histidinekinase receptors by cell-free expression and rapid NMRanalysis. Proc Natl Acad Sci USA 2010, 107:10902-10907.

51. Sobhanifar S, Schneider B, Lohr F, Gottstein D, Ikeya T,Mlynarcyk K, Pulawski W, Ghoshdastider U, Kolinski M et al.:Structural investigation of the C-terminal catalytic fragment ofpresenilin 1. Proc Natl Acad Sci USA 2010, 107:9644-9649.

52.�

Klammt C, Maslennikov I, Bayrhuber M, Eichmann C, Vajpai N,Chiu EJC, Blain KY, Esquivies L, Kwon JHJ, Balana B et al.: Facilebackbone structure determination of human membraneproteins by NMR spectroscopy. Nat Methods 2012,9:834-839.

Detailed protocol for the routine structure determination of CF expressedmembrane proteins in detergent micelles.

53.�

Reckel S, Gottstein D, Stehle J, Lohr F, Verhoefen MK, Takeda M,Silvers R, Kainosho M, Glaubitz C, Wachtveitl J et al.: Solutionstructure of proteorhodopsin. Angew Chem Int Ed 2011,50:11942-11946.

First solution structure of a CF expressed membrane protein in functionalconformation.

54.�

Matthies D, Haberstock S, Joos F, Dotsch V, Vonck J, Bernhard F,Meier T: Cell-free expression and assembly of ATP synthase. JMol Biol 2011, 413:593-603.

Demonstration of the complete assembly of a large complex composedout of integrated membrane proteins as well as of soluble proteins indifferent CF reaction conditions.

55. Deniaud A, Liguori L, Blesneac I, Lenormand JL, Pebay-Peyroula E: Crystallization of the membrane protein hVDAC1produced in cell-free system. Biochim Biophys Acta: Biomembr2010, 1798:1540-1546.

56. Chen YJ, Pornillos O, Lieu S, Ma C, Chen AP, Chang G: X-raystructure of EmrE supports dual topology model. Proc NatlAcad Sci USA 2007, 104:18999-19004.

57.�

Wada T, Shimono K, Kikukawa T, Hato M, Shinya N, Kim SY,Kimura-Someya T, Shirouzu M, Tamogami J, Miyauchi S et al.:Crystal structure of the eukaryotic light-driven proton-pumping rhodopsin, Acetabularia rhodopsin II, from marinealga. J Mol Biol 2011, 411:986-998.

First in meso crystal structure of a CF-expressed eukaryotic membraneprotein.

58. Yokoyama J, Matsuda T, Koshiba S, Kigawa T: An economicalmethod for producing stable-isotope labeled proteins by theE. coli cell-free system. J Biomol NMR 2010, 48:193-201.

59. Calhoun KA, Swartz JR: Total amino acid stabilization duringcell-free protein synthesis reactions. J Biotechnol 2006,123:193-203.

60. Yokoyama J, Matsuda T, Koshiba S, Tochio N, Kigawa T: Apractical method for cell-free protein synthesis to avoid stableisotope scrambling and dilution. Anal Biochem 2011, 411:223-229.

61. Tonelli M, Singarapu KK, Makino S, Sahu SC, Matsubara Y,Endo Y, Kainosho M, Markley JL: Hydrogen exchange duringcell-free incorporation of deuterated amino acids and anapproach to its inhibition. J Biomol NMR 2011, 51:467-476.

62.�

Su XC, Loh CT, Qi R, Otting G: Suppression of isotopescrambling in cell-free protein synthesis by broadband

Current Opinion in Structural Biology 2013, 23:374–380

Page 7: Cell-free expression—making a mark

380 New constructs and expressions of proteins

inhibition of PLP enzymes for selective 15N-labelling andproduction of perdeuterated proteins in H2O. J Biomol NMR2011, 50:35-42.

Practical method for the general inhibition of most label scramblingenzymes in CF extracts.

63. Kainosho M, Torizawa T, Iwashita Y, Terauchi T, Mei Ono A,Guntert P: Optimal isotope labelling for NMR protein structuredeterminations. Nature 2006, 440:52-57.

64. Takeda M, Ono AM, Terauchi T, Kainosho M: Application of SAILphenylalanine and tyrosine with alternative isotope-labelling

Current Opinion in Structural Biology 2013, 23:374–380

patterns for protein structure determination. J Biomol NMR2010, 46:45-49.

65. Ozawa K, Loscha KV, Kuppan KV, Loh CT, Dixon NE, Otting G:High-yield cell-free protein synthesis for site-specificincorporation of unnatural amino acids at two sites. BiochemBiophys Res Commun 2012, 418:652-656.

66. Kang SH, Jun SY, Kim DM: Fluorescent labeling of cell-freesynthesized proteins by incorporation of fluorophore-conjugated nonnatural amino acids. Anal Biochem 2007, 360:1-6.

www.sciencedirect.com