You are on page 1of 29

Strategies to Optimize Protein Expression UNIT 5.

24
in E. coli
Dana M. Francis1 and Rebecca Page1
1
Brown University, Providence, Rhode Island

ABSTRACT
Recombinant protein expression in Escherichia coli (E. coli) is simple, fast, inexpensive, and
robust, with the expressed protein comprising up to 50 percent of the total cellular protein.
However, it also has disadvantages. For example, the rapidity of bacterial protein expression
often results in unfolded/misfolded proteins, especially for heterologous proteins that require
longer times and/or molecular chaperones to fold correctly. In addition, the highly reductive
environment of the bacterial cytosol and the inability of E. coli to perform several eukaryotic
post-translational modifications results in the insoluble expression of proteins that require these
modifications for folding and activity. Fortunately, multiple, novel reagents and techniques have
been developed that allow for the efficient, soluble production of a diverse range of heterologous
proteins in E. coli. This overview describes variables at each stage of a protein expression
experiment that can influence solubility and offers a summary of strategies used to optimize
soluble expression in E. coli. Curr. Protoc. Protein Sci. 61:5.24.1-5.24.29. 
C 2010 by John Wiley
& Sons, Inc.
Keywords: protein expression r E. coli r fusion proteins r proteases r heterologous protein r
purification tags r expression tags r expression strains and vectors r folded protein r
active protein

INTRODUCTION In spite of the development of multi-


Recombinant protein expression has rev- ple nonbacterial recombinant expression sys-
olutionized all aspects of the biological sci- tems over the last three decades (yeast, bac-
ences. Most significantly, it has dramatically ulovirus, mammalian cell, cell free systems;
expanded the number of proteins that can be in- see Table 5.24.1), Escherichia coli is still the
vestigated both biochemically and structurally. preferred host for recombinant protein expres-
Previously, protein production was the domain sion (Yin et al., 2007). The rationale is clear:
of experts, as purification from a natural source E. coli is easy to genetically manipulate, it is
(i.e., plants, rabbits, bovine) was often difficult inexpensive to culture, and expression is fast,
and time consuming. However, the availability with proteins routinely produced in one day.
of new commercial systems for recombinant Moreover, protocols for isotope-labeling for
protein expression, combined with advanced NMR spectroscopy and selenomethionine in-
protein purification techniques, has made pro- corporation for X-ray crystallography are well
tein production prevalent throughout the bio- established, making it highly suitable for struc-
logical and biomedical sciences. This has en- tural studies. Thus, E. coli has multiple, sig-
abled the research community to study thou- nificant benefits over other expression systems
sands of low abundance and novel proteins including cost, ease-of-use, and scale.
from a large variety of organisms. Notably, Despite its many advantages and
31 recombinant proteins were approved for widespread use, there are also disadvan-
therapeutic use between 2003 and 2006, high- tages to using E. coli as an expression host. In
lighting the importance of heterologous pro- contrast to eukaryotic systems, transcription
tein expression in biopharmaceutical research and translation are fast and tightly coupled.
(Walsh, 2006). As the number of recombi- Since many eukaryotic proteins require
nantly produced proteins increases, so too does longer times and/or the assistance of folding
an appreciation for the difficulties and limita- chaperones to fold into their native state,
tions inherent to this process. this rate enhancement often leads to a pool
Production of
Recombinant
Proteins

Current Protocols in Protein Science 5.24.1-5.24.29, August 2010 5.24.1


Published online August 2010 in Wiley Interscience (www.interscience.wiley.com).
DOI: 10.1002/0471140864.ps0524s61 Supplement 61
Copyright C 2010 John Wiley & Sons, Inc.
Supplement 61
5.24.2
E. coli
Expression in
Protein
Optimizing
Table 5.24.1 Comparison of Recombinant Protein Expression Systems

E. coli Yeast Insect cells Mammalian cells Cell-freea (E. coli) Cell-freea (wheat germ)

Average time of 30 min 90 min 18 hr 24 hr N/A N/A


cell division
Cost of Low Low High High High High
expression
Expression level High Low-High Low-High Low-Moderate Low-High Low-High
Success rate (% 40-60 50-70 50-70 80-95 Variable Variable
soluble)
Advantages Simple, low cost, Simple, low cost Post-translational Natural protein High yield, fast, Fast, flexible,
rapid, robust, high modifications configuration, flexible, disulfide-bonded and
yield, easy labeling for post-translational disulfide-bonded and membrane proteins,
structural studies modifications membrane proteins, post-translational
easy labeling for modifications
structural studies
Disadvantages No post-translational Less Slow, higher cost, Slow, high cost, High cost, less High cost, lower yield
modifications, post-translational production of lower yield post-translational than E. coli cell-free
insoluble protein, modifications, membrane proteins modifications, efficient system, efficient
production of production of is difficult production requires production requires
disulfide-bonded and membrane proteins highly-specialized highly specialized setup
membrane proteins is is difficult setup
difficult
a Spirin, 2004; Langlais et al., 2007; for other references, see text.

Current Protocols in Protein Science


of partially folded, unfolded, or misfolded, in- protein. Must the protein be soluble? Does the
soluble proteins (Oberg et al., 1994). Thus, final product need to be active? Is the native
some targets, especially larger multidomain protein conformation important? In some in-
and membrane proteins, either fail to express stances, such as antibody production, the pro-
in E. coli or express insolubly as inclusion duction of soluble protein is not necessary, as
bodies. Moreover, insolubility is not just re- the protein sequence, rather than the correct
stricted to heterologous proteins, as many bac- 3-dimensional fold, is required for successful
terial proteins also cannot be produced in sol- antibody production (Yan et al., 2007). For
uble form when overexpressed in E. coli (Vin- these cases, expression that causes the pro-
centelli et al., 2003). In addition, the reducing tein to be incorporated into inclusion bodies is
environment of the bacterial cytoplasm makes suggested, as the recombinant protein, while
the efficient production of disulfide-containing misfolded or unfolded, is highly enriched and
proteins challenging (Stewart et al., 1998; Ritz protected from proteases (Valax and Georgiou,
and Beckwith, 2001). Finally, E. coli lacks the 1993). However, most often, the objective of
machinery required to perform certain eukary- recombinant protein expression in E. coli is
otic post-translational modifications, such as to produce a protein product that is soluble,
glycosylation, which can be critical for the for- folded, and active.
mation of folded, active protein (Zhang et al., Expression in E. coli requires four ele-
2004). ments: (1) the protein of interest, (2) a bac-
Considerable efforts have been made in re- terial expression vector, (3) an expression cell
cent years to maximize the efficient produc- line, and (4) the equipment/materials for bacte-
tion of soluble recombinant proteins in bac- rial cell culture (i.e., shaker/media). There are
teria. A remarkable number of novel reagents multiple parameters that can be varied when
(new vectors, new host strains) and strategies optimizing an expression protocol, from se-
(chaperone co-expression, low temperature in- lecting a vector with the appropriate promoter,
duction) have been developed that allow many to choosing an appropriate induction temper-
of these disadvantages to be readily and suc- ature. With each selection affecting the solu-
cessfully overcome. It is this topic, how to opti- bility and activity of the protein product, this
mize soluble protein expression in E. coli, that task can appear daunting. However, decades of
is the focus of this review. Section I outlines work by individual protein chemists, coupled
a typical expression protocol for the produc- with the recent experiences of high-throughput
tion of heterologous proteins in E. coli. While structural genomics efforts, have resulted in
this protocol has proven highly successful for the identification of a consensus protocol that
a broad range of targets, every protein has its allows a diverse set of proteins to be success-
own unique set of biophysical characteristics fully expressed in E. coli. A flowchart of the
that often requires protocol changes in order protocol typically used by the authors to ex-
to successfully express the target. Thus, in the press a diverse set of proteins is shown in
second part of this review, critical parameters Figure 5.24.2 (Mustelin et al., 2005; Brown
that are essential to consider when designing et al., 2008; Critton et al., 2008). More ex-
a protein expression strategy are highlighted tensive protocols are also available (Peti and
(Fig. 5.24.1). These parameters have the com- Page, 2007; Gråslund et al., 2008a).
mon goal of maximizing the yield of soluble, The protocol outlined in Figure 5.24.2 con-
active protein. Section II describes optimiza- sists of nine steps. First, the optimal residue
tion of the target DNA, section III discusses boundaries of the desired protein product, the
modifications for the optimization of expres- “target protein,” are determined. The target
sion vectors, section IV details bacterial host gene is then subcloned into a bacterial ex-
strains that aid heterologous protein expres- pression vector that utilizes the T7 lacO pro-
sion, section V outlines optimization of pro- moter system (i.e., that used in the pET system)
tein expression conditions, and section VI de- and contains both an N-terminal hexahista-
scribes how to enhance soluble expression by dine (his6 )-tag and a tobacco etch virus (TEV)
coexpression with other proteins. protease site (Peti and Page, 2007). The T7
promoter system provides strong, robust ex-
I. REPRESENTATIVE PROTOCOL pression, the his6 -tag facilitates purification,
FOR EXPRESSING PROTEINS IN while the protease site allows the (his6 )-tag
BACTERIA to be proteolytically removed from the puri-
Before initiating a protein expression fied target protein. In the third step, the ex- Production of
project, it is essential to determine the pression vector is transformed into a deriva- Recombinant
Proteins
definitive use of the recombinantly produced tive of the BL21 (DE3) strain, such as BL21
5.24.3
Current Protocols in Protein Science Supplement 61
Essential elements: Variables:

1. gene of interest rare codons


domain boundaries
properties of protein
clone sequence
fusion
tag
promoter
2. expression vector protease site fusion tag: type
target
position
protein
removal

transform

protease deficient
3. E.coli cells codon supplemented
facilitate disulfide-bonds
express toxic proteins
express

4. expression temperature
condition inducer concentration
media type
coexpression with partner
protein or chaperone

Figure 5.24.1 Schematic overview of the topics covered in this review, highlighting the multiple
parameters (listed on the right) that can greatly impact the success of soluble expression.

(DE3)-RIL cells. These cells, which are com- acteristics of the protein, vector, host strain,
patible with the T7 promoter system, contain and/or expression conditions may need to be
plasmids that encode arginine, isoleucine, and modified in order for folded, active protein to
leucine tRNAs that are rare in E. coli, and be produced, it is not uncommon to see more
are deficient in both lon and ompT proteases, complicated protocols that test these variables
which minimizes in vivo degradation of the in parallel (Peti and Page, 2007). Thus, the re-
target protein. After the overnight starter cul- mainder of this review describes which factors
ture is used to inoculate the large-scale culture have the largest effect on soluble protein ex-
(step 5), the cells are grown to mid-log phase pression and how to change them in order to
(OD600 of ∼0.6 to 0.9) in Luria broth (LB) express folded, active protein in E. coli.
in baffled shaker flasks (which increases aer-
ation and thus yield) at 37◦ C with constant II. PROPERTIES OF THE GENE
shaking. The cultures are then transferred to a AND PROTEIN THAT INFLUENCE
lower temperature (18◦ C), and, once cooled, EXPRESSION AND SOLUBILITY
protein expression is induced using isopropyl- Here, we discuss critical characteristics of
β-thio-galactoside (IPTG). Expression is con- the gene and/or protein sequence that influence
tinued overnight with vigorous shaking (200 to its soluble expression in E. coli.
250 rpm) at 18◦ C. The lower expression tem-
perature facilitates the production of folded, Rare codons
soluble protein. Finally, the cells are pelleted One of the most common reasons that het-
by centrifugation and stored at −80◦ C until erologous proteins fail to express in E. coli
needed. is the presence of “rare” codons in the tar-
Optimizing
Protein This protocol is meant to serve as a start- get mRNA. Many proteins, especially hu-
Expression in ing point for designing an expression strategy man proteins, have mRNA sequences that in-
E. coli in E. coli. However, because multiple char- clude codons that are infrequently used in
5.24.4
Supplement 61 Current Protocols in Protein Science
Construct
design/ 1-7 days Step 1: Determine construct
cloning/ boundaries
sequencing

Step 2: Clone into vector (contains


T7 promoter, His6 tag, TEV
protease cleavage site);
sequence verify cloned
constructs

Day 1: 2 hr
Transform
procedure, Step 3: Transform cloned construct
and test
overnight into BL21(DE3)RIL cells
expression
incubation

Day 2:
Optional: Test expression using
3 hr cell
microexpression protocol
growth, 3 hr
(Peti & Page, 2007)
induction

Day 2: Step 4: Inoculate 50-100 ml LB


Starter culture 10 min culture with colony from
procedure, transformation plate or 100 μl
overnight from best microexpression
incubation (uninduced). Grow overnight
at 37°C with vigorous shaking.

Step 5: Inoculate large-scale LB


Large-scale Day 3: culture (1 liter) with 5-10 ml
expression 10 min starter culture from step 4

Step 6: Grow culture to mid-log


Day 3:
phase (OD600 0.6-0.9) at
~2-4 hr
37°C with vigorous shaking

Day 3:
Step 7: Cool culture to 18°C
1 hr at 4°C

Day 3: Step 8: Induce expression with


10 min IPTG (0.5-1.0 mM,
final concentration)

Days 3-4: Step 9: Incubate overnight at 18°C


16-20 hr with vigorous shaking

Figure 5.24.2 Flowchart of a general expression protocol used by the authors to express a
broad range of targets, from phosphatases, to neuronal scaffolding proteins, to bacterial signaling
proteins. The approximate time required to complete each segment of the protocol is listed to the
left of the corresponding step.

Production of
Recombinant
Proteins

5.24.5
Current Protocols in Protein Science Supplement 61
E. coli (rare codons) (Sharp and Li, 1987). This comparative studies have shown that, for most
includes codons for arginine (AGA, AGG), targets, either approach is equally effective
isoleucine (AUA), leucine (CUA), and pro- (Burgess-Brown et al., 2008). Thus, many re-
line (CCC). Target genes that contain signif- search groups, including those of structural ge-
icant numbers of individual rare codons, or nomics consortia, protein production facilities,
smaller numbers of tandem rare codons, are and the authors (see the standard protocol in
more likely to experience translational stalling Fig. 5.24.2), use codon-supplemented cells for
in E. coli, and thus often either completely fail all initial expression trials.
to express, express at very low levels, or are
expressed as truncated proteins (Kane, 1995, Protein size and domain boundaries
Cruz-Vera et al., 2004). Moreover, when they The evolution of eukaryotes has been char-
do express, these rare codon–rich genes can acterized by a significant increase in the size
also be incorrectly translated, as high level and complexity of proteins; e.g., the aver-
misincorporation of lysine for arginine at AGA age protein length in E. coli is 317 residues
codons has been observed for protein targets while in humans it is 510 residues (Netzer
expressed in E. coli (Calderone et al., 1996). and Hartl, 1997, Sakharkar et al., 2006). This
Fortunately, the codon bias of E. coli is increase in size is due to an increase in the
straightforward to overcome. Multiple Web number of complex, multidomain proteins, in
sites are now available that quantify the which individual domains have distinct and
number and the location of rare codons independent functions. Comparative studies
in a gene (e.g., the rare codon calculator, have shown that the probability of soluble ex-
RaCC; http://nihserver.mbi.ucla.edu/RACC/). pression in E. coli decreases with increasing
These programs also often highlight the num- molecular weight, especially for proteins >60
ber of consecutive rare codons. If the target kD (Canaves et al., 2004, Goh et al., 2004,
protein contains a significant number of rare Gråslund et al., 2008a). For example, a large-
codons, especially tandem rare codons, two scale study examining the protein properties
approaches can be taken. In the first, changes of 95 recombinantly expressed mammalian
are made to the gene. Specifically, the gene proteins found that smaller proteins (aver-
is “codon optimized,” i.e., the rare codons are age molecular weight of 22.8 kD) were of-
replaced with those that are common to the ten expressed solubly alone, while larger pro-
host. This can be achieved using site-directed teins (average molecular weight of 40.4 kD)
mutagenesis. However, it is often faster and were only solubly expressed when fused to
cheaper to simply have the codon-optimized solubility-enhancing tags (Dyson et al., 2004).
gene synthesized (this service is offered by In a separate study, small proteins (100 amino
multiple companies). Gene synthesis has the acids or less) were expressed solubly in E. coli
added benefit that most gene optimization al- at levels suitable for purification 47% of the
gorithms optimize not only rare codons, but time, while large proteins (600 to 800 amino
also mRNA secondary structure, which has acids) were expressed solubly only 33% of the
also been shown to affect translation effi- time (Gråslund et al., 2008a). Thus, when us-
ciency (Hatfield and Roth, 2007). In the sec- ing E. coli as an expression host, it is typically
ond approach, changes are made to the expres- advantageous to express individual protein do-
sion host. Namely, genes encoding the rare mains, as opposed to the full-length protein,
tRNAs are co-expressed with the wild-type whenever possible.
(non-optimized) target gene (Wakagi et al., The starting and ending residues of the
1998). E. coli strains are now available that target domain can also greatly affect ex-
contain plasmids that encode rare tRNAs [i.e., pression yield and solubility. For example,
BL21 (DE3)-RIL/RP/RILP cells from Strata- Klock et al. (2008) showed that deletion of
gene or Rosetta cells from Novagen; Schenk just four residues at either the N- or C-
et al., 1995; Tegel et al., 2009]. terminus can convert a solubly expressing pro-
Both approaches effectively overcome tein into one that expresses insolubly. In a
codon bias. For example, codon optimization separate study, Gråslund et al. (2008b) gen-
of the human β-defensin 2 (hBD2) gene led erated 10 constructs of a single target do-
to a nine-fold enhancement in the expression main of interest: full-length and 9 deletion
level (Peng et al., 2004) while co-expression constructs that differed in length from one
Optimizing of inorganic phosphatase from Sulfolobus sp. another by 2 to 10 residues at either the N-
Protein with tRNAArg (AGA codon) more than dou- or C-terminus. Thus, all available functional
Expression in bled its already high expression level (Wakagi and structural data should be used to deter-
E. coli
et al., 1998). Critically, recent large-scale, mine optimal boundaries for a protein domain
5.24.6
Supplement 61 Current Protocols in Protein Science
construct. For a protein of unknown domain active) contain LCRs, yet have been robustly
structure, threading the target protein sequence and solubly expressed in E. coli (Dyson and
onto a homologous protein structure (i.e., Wright, 2005; Dancheck et al., 2008). Because
SWISS-MODEL; Arnold et al., 2006) or us- LCRs may play an active role in mediating
ing structure-based/fold recognition sequence protein function or protein-protein interactions
alignment programs (i.e., FFAS; Jaroszewski (Karlin et al., 2002), their inclusion in an ex-
et al., 2005) can aid in determining the op- pression protein construct must be determined
timal domain boundaries. When a homolo- on a protein-by-protein basis.
gous protein structure is not available, the Disulfide bonds. The presence of disulfide
prediction of secondary structural elements bonds in a protein also negatively correlates
(i.e., PSIPRED; Jones, 1999) should be uti- with soluble expression in E. coli. The reduc-
lized. The disruption of predicted secondary ing environment of the bacterial cytoplasm
structural elements must be avoided. Many of makes the efficient production of disulfide-
the bioinformatics tools needed to carry out containing proteins, such as growth factors and
these types of analyses are freely available antibody FAB fragments, challenging (Stewart
at http://www.expasy.org. In addition, a pro- et al., 1998). Thus, the expression of disulfide-
gram has been developed that integrates these bond containing proteins in E. coli commonly
bioinformatics tools to aid in protein bound- results in the production of insoluble pro-
ary identification (the SGC Domain Boundary tein (due to misfolding) sequestered into in-
Analyzer; described in more detail in Gråslund clusion bodies (Veldkamp et al., 2007; Chen
et al., 2008b). Finally, it is typically advan- and Leong, 2009); for a review of refolding
tageous to subclone four or more constructs strategies, see Tsumoto et al., (2003). When
with different N- and C-terminal boundaries refolding conditions cannot be successfully
and test, in parallel, which construct results in identified, the target protein must be produced
the highest level of soluble expression (Brown solubly in vivo. The three most common strate-
et al., 2008). gies to express disulfide-containing proteins,
all of which are discussed later in this re-
Protein sequence view, are to try the following: (1) target the ex-
Hydrophobic residues, low complexity re- pressed protein to the E. coli periplasm, which
gions. In addition to molecular weight, other is highly oxidative (Leichert and Jakob, 2004),
biophysical properties of the protein, such (2) fuse the protein to thioredoxin (Lefebvre
as hydrophobicity and sequence complexity, et al., 2009a), and/or (3) express the protein
can influence expression yields. In the study in bacterial strains containing thioredoxin re-
by Dyson et al. (2004), 95 mammalian pro- ductase and glutathione reductase mutants (Xu
teins were fused to a variety of N- and et al., 2008).
C-terminal expression and purification tags Transmembrane segments. Finally, the sol-
in order to elucidate the properties of the ubility of the recombinantly expressed pro-
proteins and fusion tags that facilitate solu- tein will typically be compromised if the
ble expression. They determined that contigu- construct includes transmembrane-spanning
ous hydrophobic residues (AILFWV) and low regions. Thus, the soluble expression of
complexity regions (LCRs) negatively corre- transmembrane-containing proteins, espe-
late with soluble expression. LCRs are regions cially integral membrane proteins, in E. coli is
of biased sequence composition, such as ho- exceptionally challenging, requiring special-
mopolymeric runs, short-period repeats, and ized materials and strategies (Mohanty and
overrepresentations of one or a few residues Wiener, 2004; Gordon et al., 2008; Dvir and
that typically adopt disordered coil conforma- Choe, 2009).
tions (DePristo et al., 2006). This has also
been reported by other groups (Canaves et al., III. PROPERTIES OF THE VECTOR
2004; Gråslund et al., 2008a). Thus, it is THAT INFLUENCE EXPRESSION
common to design protein expression con- AND SOLUBILITY
structs to avoid hydrophobic residues and low Once the protein target and correspond-
complexity segments in the extreme N- and ing construct(s) are determined, it must be
C-termini (Peti and Page, 2007). However, subcloned into a vector that contains all
LCRs do not always inhibit soluble expres- DNA sequence elements that direct the tran-
sion. Many intrinsically unstructured proteins scription and translation of the target gene
(IUPs; proteins that do not adopt a single (Studier and Moffatt, 1986a). These elements Production of
Recombinant
folded conformation yet are still biologically include promoters, regulatory sequences, the Proteins

5.24.7
Current Protocols in Protein Science Supplement 61
Shine-Dalgarno box, transcriptional termina- protein). Second, it exhibits low basal tran-
tors, and origins of replication, among others. scriptional activity to prevent unwanted tran-
In addition, expression vectors contain a selec- scription prior to induction. Third, induction
tion element, typically an antibiotic-resistance is simple and cost-effective.
gene, to aid in plasmid selection within the host When selecting a promoter system, the na-
cell. Another critical feature of E. coli expres- ture of the protein target and its desired down-
sion vectors is the presence of a fusion tag. stream use must be considered. If the protein
In contrast to the elements described above, target is a toxic protein (like a ribonuclease),
the fusion tags are transcribed in-frame with one should consider using promoter systems
the construct of interest. When translated, a that have extremely low basal expression, such
single fusion protein, which includes the pro- as the araBAD promoter (Lee et al., 1987).
tein of interest and the fusion tag, is obtained. Alternatively, for maximal protein yields, a
Today, nearly all proteins are expressed with strong promoter should be selected, such as T7
some kind of fusion tag, and the number and or tac. Finally, for aggregation-prone proteins,
diversity of tags is continually increasing. a cold-shock promoter, in which expression is
carried out at low temperatures, may be tested.
Origin of replication Multiple promoters have been developed for
The origin of replication of a vector is the expression in E. coli and are summarized in
site where replication is initiated. It also de- Table 5.24.2. The four most widely used pro-
termines copy number of the vector in the moters are the T7 RNA promoter, the araBAD
host. The copy number for common E. coli promoter, hybrid promoters, and the cspA pro-
expression plasmids ranges from low (2 to moter.
20) to high (20 to 40). Typically, high-copy- T7 promoter (T7 RNA polymerase system).
number plasmids are desired for protein ex- The T7 RNA polymerase system is the most
pression in E. coli, as they result in the max- commonly used promoter system in E. coli.
imum protein yield for a given culture vol- Gene expression is driven by the T7 RNA poly-
ume (Jing et al., 1993; Huang et al., 1994). merase (from the T7 bacteriophage), which
The origin of replication is also important transcribes DNA five times faster than the bac-
to consider when carrying out protein co- terial RNA polymerase (Studier et al., 1990).
expression experiments in which two differ- Because E. coli lacks this enzyme, the poly-
ent plasmids, each of which contains a differ- merase must be delivered to the cell, via an
ent protein/biomolecule, are simultaneously inducible plasmid or, more often, by using an
transformed into the same expression cell E. coli strain that contains a chromosomal copy
(Johnston and Marmorstein, 2003). For these of the T7 polymerase gene (Studier and Mof-
experiments, the origins of the two plasmids fatt, 1986b). In the absence of an inducer, the
should be different to allow the cell to support polymerase, which itself is under the control
both expression vectors. of the lacUV5 promoter, is not produced, and
correspondingly, the gene of interest is not
Promoter systems transcribed (Studier et al., 1990). Upon ad-
Promoters are another element of the vec- dition of the nonhydrolyzable lactose analog,
tor that can have a profound effect on the IPTG, the T7 RNA polymerase is transcribed
strength and duration of transcription and, in and synthesized. The polymerase then initiates
turn, protein yield. Synthesis of mRNA is ini- transcription of the target gene by binding to a
tiated when RNA polymerase binds to a spe- T7 polymerase–specific promoter.
cific DNA sequence, the promoter, adjacent Once induced, most of the cellular machin-
to the target gene. This sequence contains the ery is devoted to the production of the re-
transcription start site, as well as two hexanu- combinant protein, comprising up to 50% of
cleotide sequences approximately 10 and 35 the total cellular protein (Studier and Moffatt,
bases upstream of the initiation site that di- 1986b; Studier et al., 1990). However, such
rect the binding of essential elements of the robust transcription can have undesirable ef-
polymerase machinery (Rosenberg and Court, fects. First, even minimal basal production of
1979; Hawley and McClure, 1983; Harley and T7 RNA polymerase results in “leaky” expres-
Reynolds, 1987). An effective promoter for ex- sion (expression prior to induction) of the tar-
pressing heterologous proteins in E. coli has get protein (Moffatt and Studier, 1987). This
Optimizing three characteristic features. First, it is strong, can be detrimental if the protein is toxic to the
Protein resulting in robust expression of the target gene host, resulting in cell death or growth arrest. To
Expression in (typically 10% to 50% of the total cellular minimize leaky expression, several host strains
E. coli

5.24.8
Supplement 61 Current Protocols in Protein Science
Table 5.24.2 Promoter Systems used to Direct Recombinant Protein Expression in E. coli

E. coli
promoter Description Induction Advantages Disadvantages
system

Most common promoter systems


T7 RNA T7 RNA polymerase IPTG High level of expression: Leaky expression: use
polymerase gene under the control accumulate up to 50% total pLysS strains for
of L8-UV5 lac cell protein expression of proteins
promoter Well characterized, used toxic to host
most often
Titrate expression using
Tuner strains
araBAD Promoter is controlled L-arabinose Tight regulation Repressed expression
promoter by AraC regulator (repressed by Titrate expression levels state is not always zero,
(PBAD ) glucose) from low to high gene-dependent
Low basal expression:
suitable for production of
proteins toxic to host
trc and tac −35 sequence from trp IPTG High level of expression: Very leaky expression:
promoter promoter and −10 accumulate 15%-30% of not optimal for
sequence from lacUV5 total cell protein expression of proteins
promoter (trc-17 bp toxic to host
spacing, tac-16 bp Newer, more efficient
spacing between systems are available
sequences)
cspA Promoter from the Temperature Efficient expression at low Leaky expression: not
promoter major cold-shock downshift from temperatures optimal for expression
protein in E.coli 37◦ C (expression Can improve folding, lower of proteins toxic to host
optimal between inclusion body formation Translational efficiency
10◦ C-25◦ C) Advantageous for expression slows at lower
of aggregation-prone and temperatures
proteolytically-sensitive Not titratable
proteins
Induction is cost efficient
Less common promoter systems
Phage Phage promoter that is Temperature shift Moderately high expression High basal level of
promoter pLa regulated by the from 30◦ C to 42◦ C Induction is cost efficient expression at
temperature-sensitive temperatures below
cI repressor 30◦ C
Induction cannot be
performed at low
temperatures
PhoA Promoter for the gene Lower phosphate Promotes secretion to the Phosphate limitation
promoterb of the periplasmic concentration in the periplasm can have negative
alkaline phosphatase growth medium Inexpensive induction effects on metabolism
of host cell
Not titratable
Limited media options
continued

Production of
Recombinant
Proteins

5.24.9
Current Protocols in Protein Science Supplement 61
Table 5.24.2 Promoter Systems used to Direct Recombinant Protein Expression in E. coli, continued

E. coli
promoter Description Induction Advantages Disadvantages
system

RecA Promoter for recA gene Nalidixic acid Tight regulation Not titratable
promoterc Regulated by lexA No growth media or
temperature restrictions
TetA Regulated by the tetR Anhydrotetracycline Moderately high expression Not titratable
promoterd repressor Low basal expression
Independent of E. coli strain
a See Elvin et al., 1990.
b See Kikuchi et al., 1981; Miyake et al., 1985; Chang et al., 1986.
c See Shirakawa et al., 1984; Olins and Rangwala, 1990.
d See Delatorre et al., 1984, Skerra, 1994.

[i.e., BL21(DE3)pLysS, Rosetta(DE3)pLysS, two logarithms (Guzman et al., 1995). This al-
Rosetta-gami(DE3)pLysS] have been devel- lows the expression level to be titrated over a
oped that contain a plasmid encoding T7 wide range of inducer concentrations, which
lysozyme, an enzyme that binds and inhibits can be important when trying to either max-
T7 polymerase (Moffatt and Studier, 1987). imize expression yields (higher L-arabinose
The concentrations of T7 lysozyme produced concentrations) or to increase the yield of
in these strains are sufficient to inhibit basal soluble protein (lower L-arabinose concentra-
transcription of target genes. However, it con- tions). It should be noted that although this
tinues to inhibit T7 RNA polymerase after system can efficiently repress gene expres-
expression has been induced, and thus, it is sion, the repression level is not always zero
typical for the level of recombinant protein ex- and the efficiency of repression is gene de-
pression to be significantly reduced in pLysS pendent (Guzman et al., 1995). Finally, stud-
versus non-pLysS strains. For nontoxic pro- ies that have directly compared protein yields
teins, this results in lower yields (Studier, from the araBAD and the T7 promoters have
1991). However, for toxic proteins, this often found that T7 promoters generally result in
results in higher yields because the cell growth higher expression yields (Goulding and Perry,
is not arrested prior to induction by the prema- 2003). For example, in our hands, we have
ture expression of the toxic protein (Yeo et al., seen protein expression yields increase by
2009). In addition, because T7 is such a strong 2- to as much as 10-fold by switching from
promoter, some translated proteins aggregate the araBAD to the T7 promoter system.
and form inclusion bodies because they fail Hybrid promoters: trc and tac promoters.
to fold before encountering another unfolded The trc and tac promoters are hybrids of
protein. In these cases, expression parameters naturally occurring promoters, consisting of
can be changed (see section IV) to maximize the −35 region of the trp promoter and the
the yield of soluble, folded protein. Alterna- −10 region of the lacUV5 promoter (Amann
tively, a weaker promoter can be used. et al., 1983; Deboer et al., 1983). The only
araBAD promoter. The arabinose promoter difference between these two systems is the
system (araBAD promoter) is a strong, titrat- spacing between the −35 and −10 consen-
able promoter which, unlike the T7 promoter, sus sequences, with 16 bp and 17 bp separa-
has almost no basal transcriptional activity tions in the tac and trc promoters, respectively
(Lee et al., 1987). Thus, it is advantageous (Brosius et al., 1985). Expression is induced
for the expression of highly toxic proteins. with IPTG (Brosius et al., 1985). Trc and tac
The induction agent for this promoter is L- are both considered to be strong promoters,
arabinose (Lee et al., 1987). In the absence with the trc promoter ∼90% as active as the tac
of L-arabinose, transcription is exceptionally promoter, and both can result in the accumu-
low and, if needed, can be even further sup- lation of up to 15% to 30% of the total cellular
pressed by the addition of glucose (Miyada protein (Brosius et al., 1985). Because these
Optimizing et al., 1984). As reported by Guzman et al., promoters are leaky, they can be problematic
Protein when expressing proteins that are toxic to the
Expression in protein expression levels increase linearly with
E. coli increasing concentrations of L-arabinose over cell (Brosius et al., 1985, Otto et al., 1995).

5.24.10
Supplement 61 Current Protocols in Protein Science
cspA promoter. Following a downshift in Common tags: hexahistidine (his6 ). The
growth temperature from 37◦ C to 10◦ C, the his6 tag does not enhance soluble expression,
bacterial cellular machinery is largely devoted but it does facilitate purification and because
to the production of 13 “cold-shock” proteins of its widespread use, it is described here.
(Jones et al., 1987). After 90 min, the major His6 -tagged fusion proteins are purified us-
cold-shock protein, CspA, accounts for 13% of ing immobilized metal affinity chromatogra-
total cellular protein (Goldstein et al., 1990). phy (IMAC; Porath et al., 1975). In IMAC,
Accordingly, the cspA promoter has been ex- metal ions, typically nickel or cobalt, are im-
ploited to direct the expression of recombi- mobilized to resin via a metal chelator, such
nant proteins at low temperatures. For this sys- as nitrilotriacetic acid, with only three or four
tem, induction is achieved by simply changing of the six metal coordination sites occupied.
the growth temperature of the bacterial culture The unprotonated histidines of the his6 -tagged
from 37◦ C to between 10◦ C and 25◦ C (Vasina fusion protein coordinate the metal, allowing
and Baneyx, 1996); no chemical inductant is the expressed protein to be readily purified
required. Moreover, because expression is car- from E. coli lysate. The bound proteins are
ried out at low temperatures, this system has then eluted using either imidazole or by low-
the added benefit that it promotes the solu- ering the pH. Because this tag is small (<1
ble expression of aggregation-prone proteins kD), it is frequently used in conjunction with
(Vasina and Baneyx, 1996, 1997). One draw- solubility-enhancing fusion tags to provide a
back is that the cold shock promoter is not bi-functional tag that facilitates both solubil-
completely repressed at higher temperatures, ity and purification. Increasing the length of
which can result in basal expression of the the tag to eight or even ten histidine residues
target protein (Qing et al., 2004). Finally, ex- typically increases the purity of the purified
pression levels using the cspA promoter are fusion protein (Tanaka et al., 1999; Mobley
typically lower than that seen with the T7 et al., 2007).
promoter. Common tags: Thioredoxin (Trx). Trx is a
small protein that catalyzes dithiol-disulfide
Fusion tags exchange reactions, an activity that is impor-
Fusion tags are proteins or peptides that are tant for diverse cellular processes. When over-
genetically fused to the target protein. They expressed in E. coli, it can also accumulate to
are useful because they can improve protein up to 40% of the total cellular protein, yet still
expression, promote folding, increase protein remain soluble (LaVallie et al., 1993, Lauber
solubility, and facilitate downstream processes et al., 2001). Thus, trx facilitates both expres-
such as purification and detection. However, sion and solubility. Its utility for enhancing
the “perfect” tag, i.e., one that can perform soluble expression was recently demonstrated
all of these tasks for every protein, still does in two studies. In the first, 30 full-length mam-
not exist. Thus, it is often necessary to test malian proteins were expressed with a series
multiple fusion tags to determine which tag of fusion tags in order to identify those tags
results in the highest yields of soluble pro- that promote soluble expression for the most
tein (Peti and Page, 2007; Brown et al., 2008) diverse set of targets. It was found that two
and to also use a combination of tags in or- tags, Trx and maltose-binding protein (MBP,
der to facilitate both expression and purifica- discussed below), consistently promoted solu-
tion (Nilsson et al., 1996; Pryor and Leiting, ble expression when fused to the N-terminus
1997; Routzahn and Waugh, 2002). For a com- of the target protein (Dyson et al., 2004). In a
prehensive review of fusion tags, see Terpe second, similar study, in which the expression
(2003). Many of the most commonly used fu- of 27 eukaryotic proteins of suitable size for
sion tags, their biophysical characteristics, and NMR studies were examined, Trx was ranked
their uses in expression and/or purification are highest for its ability to promote soluble pro-
listed in Table 5.24.3. The placement of the tag, tein expression (Hammarstrom et al., 2002).
either N-terminal or C-terminal, is also impor- Trx is also the fusion tag of choice if
tant as it can have a profound effect on sol- expressing a protein that contains disulfide
uble protein expression levels. Additionally, bonds. To overcome the highly reductive en-
the presence of a fusion tag may interfere with vironment of the bacterial cytosol, new bacte-
the biological activity of the recombinantly ex- rial strains, which contain mutations in two
pressed protein, and thus, in these cases, it may proteins that play key roles in maintain-
be important to enzymatically remove the tag ing the reducing environment of the cytosol, Production of
after the fusion protein has been purified. thioredoxin reductase (trxB) and glutathione Recombinant
Proteins

5.24.11
Current Protocols in Protein Science Supplement 61
Table 5.24.3 Characteristics of Commonly used Fusion Tags

Amino Size Purification Affinity


Tag Protein Source organism Comments
acids (kDa) aid matrix

Tags with solubility-enhancing properties


GST Glutathione 243 28.1 Schistosoma Yes Glutathione Forms dimer
S-transferase japonicum agarose in solution
MBP Maltose binding 390 43.0 Escherichia coli Yes Amylose Strong
protein resin solubility-
enhancer
DsbA Disulfide 228 25.4 Escherichia coli No Aids
oxidoreductase periplasmic
disulfide-bond
formation
NusA N-utilizing 535 59.3 Escherichia coli No Strong
substance A solubility-
protein enhancer
Trx Thioredoxin 135 14.7 Escherichia coli No Aids cytosolic
disulfide-bond
formation
Z-domaina Protein A IgG 91 10.6 Staphylococcus Yes Protein
ZZ repeat aureus A-sepharose
domain
GB1b Protein G β1 85 9.7 Streptococcus Yes IgG-resins Used often
domain sp. with proteins
for NMR
SUMO Small 99 11.1 Homo sapiens No
ubiquitin-like
modifier
SET Solubility- <40 T7 phage gene No Small, highly
enhancing 10B; synthetic acidic peptide
tags tags that limit
protein
aggregation
HaloTag-7c Catalytically- 296 34.0 Rhodococcus sp. Yes HaloLink Strong
inactive resin solubility-
derivative of enhancer
DhaA
Tags without solubility-enhancing properties
His6 Hexahistadine 6 0.8 Synthetic Yes Immobilized Often
metal resin combined with
solubility-
enhancing
tags
Inteind Protein splicing 128-1650 Variable Yes Chitin resin Remove from
element resin by
induced
self-cleavage
a See Nilsson et al., 1987; Zhao et al., 2005.
b See Bao et al., 2006.
c See Ohana et al., 2009.
d See Chong et al., 1998; Singleton et al., 2002.

5.24.12
Supplement 61 Current Protocols in Protein Science
reductase (gor), have been developed often precipitate after enzymatic cleavage of
(Origami and Rosetta-gami cells; discussed MBP, resulting in wasted time and resources
further in section IV). Because the formation (Lee et al., 1993; Saavedraalanis et al., 1994;
of disulfide bonds appears to be dependent Kishore et al., 1998). In order to verify that
on the presence of thioredoxin in trxB mutant MBP facilitates the production of folded, ac-
strains (Stewart et al., 1998), increased yields tive protein, and does not simply solubilize
of soluble, active protein may be obtained by misfolded proteins, it is advantageous to co-
fusing the target protein to Trx and express- express the MBP fusion protein with its site-
ing the Trx-fusion protein in trxB/gor mutant specific protease. In this system, the MBP fu-
strains. sion protein is proteolytically cleaved in vivo
Common tags: Maltose-binding protein by the co-expressed protease (Nallamsetty and
(MBP). MBP is a soluble periplasmic pro- Waugh, 2006). Proteins/protein constructs for
tein that binds maltose and delivers it to the which folding and solubility is enhanced by
MalFGK2 transporter (Fox et al., 2001). As a MBP will remain soluble following cleavage
fusion tag, MBP facilitates expression, solubil- while those that do not will precipitate. Small-
ity, and purification. It can also be used to tar- scale expression tests can be used to rapidly
get proteins to either the cytosol or periplasm identify which constructs benefit from fusion
(fusion proteins targeted to the periplasm in- to MBP. Because the MBP tag is proteolyti-
clude the endogenous malE signal sequence in cally removed in vivo, it is advantageous to
the MBP gene), although it is most frequently add a purification tag, typically a his-tag, after
used for cytosolic expression. Like Trx, MBP the protease site (i.e., MBP–TEV site–his6 tag–
constantly ranks as one of the most effective protein target).
fusion tags for promoting protein solubility Common tags: Glutathione-S-transferase
(Dyson et al., 2004; Niiranen et al., 2007). (GST). GST, a protein that catalyzes the nucle-
In addition, MBP has been shown to pas- ophilic attack of glutathione on electrophilic
sively promote the folding of its fused part- substrates in order to reduce their reactivity
ner (Kapust and Waugh, 1999; Nallamsetty with other biomolecules (Armstrong, 1997),
and Waugh, 2006, 2007). The mechanism by is another commonly used fusion tag. GST
which MBP promotes folding is not well un- facilitates both expression and purification,
derstood, but it is has been suggested that MBP but is not generally considered a solubility-
possesses chaperone-like qualities and may enhancing tag as many studies have shown
function by either interacting directly with its that it is a poor solubility enhancer (Espos-
fusion protein, stabilizing folding intermedi- ito and Chatterjee, 2006; Brown et al., 2008).
ates, or inhibiting protein aggregation (Kapust While GST (∼28 kDa) is smaller than MBP,
and Waugh, 1999; Fox et al., 2001). it dimerizes. Thus, if the target protein also
MBP is most commonly used as an N- forms oligomers, the use of GST can lead to
terminal tag. However, unlike Trx, which is aggregation and precipitation of the expressed
only effective as an N-terminal fusion tag, fusion protein, even though the target protein is
MBP has been shown to effectively enhance folded and active (Kaplan et al., 1997). In spite
protein solubility as both an N- and a C- of these disadvantages, GST is still widely
terminal fusion tag (Dyson et al., 2004). Thus, used because it binds tightly and specifically
MBP can be used effectively in both positions. to glutathione agarose, allowing the target pro-
Unlike Trx, MBP also functions as an affin- tein to be purified in a single step (Smith and
ity tag as it binds tightly to sugars, such as Johnson, 1988).
amylose or dextrin, which, when coupled to Recently developed tags: NusA. Recently,
agarose or sepharose beads, can be used to novel solubility tags have been developed
purify MBP fusion proteins from the E. coli with the common aim to enhance the solu-
lysate (Diguan et al., 1988). bility of a diverse set of targets with mini-
While the effective increase in solubility mal drawbacks (Chong et al., 1998; Zhang
and affinity properties are highly favorable, et al., 2004; Butt et al., 2005; Ohana et al.,
MBP also has less desirable traits, which is 2009). N-utilization substance A (NusA), a
why it is not the “perfect” tag. The pro- transcription termination/antitermination fac-
tein is very large (42 kD) and its presence tor in E. coli, is one of these new tags. Its
can interfere with biological activity of the utility as a solubility-enhancing fusion pro-
recombinant protein if not removed. More- tein was identified through a systematic study
over, MBP enhances solubility so strongly of more than 4000 E. coli proteins. In this Production of
that it can even solubilize unfolded/misfolded study, NusA was predicted to have a high Recombinant
Proteins
proteins. In these cases, the target proteins solubility probability when expressed in
5.24.13
Current Protocols in Protein Science Supplement 61
E. coli (>90%), and subsequent experiments fusion of SUMO to the N-terminus of GFP and
demonstrated that it was more effective at pro- MMP13, a protein highly recalcitrant to sol-
moting solubility than both GrpE and baterio- uble expression, dramatically enhanced their
ferritin (BFR; Davis et al., 1999). Since then, expression and solubility (Malakhov et al.,
multiple studies have shown that NusA effec- 2004). Subsequent studies have shown SUMO
tively enhances protein solubility for a diverse also effectively promoted the soluble expres-
set of protein targets (De Marco et al., 2004; sion of SARS coronavirus proteins (Zuo et al.,
Douette et al., 2005; Niiranen et al., 2007); in 2005b) and membrane proteins (Zuo et al.,
fact, in some cases it has been shown to be even 2005a). One of the advantages of SUMO is
more effective than MBP (Kohl et al., 2008). that it is small (11.2 kD). It also has its own,
However, NusA is not an affinity tag. Thus, highly specific protease (Ulp), which functions
it is typically coupled to the his6 -tag to facil- by recognizing the tertiary structure of the
itate purification (de Marco, 2006). Finally, it SUMO protein, rather than a short, specific
is also large (55 kDa), which can sometimes amino acid sequence. It cleaves the peptide
make proteolytic removal of the NusA fusion chain immediately after the C-terminal residue
protein difficult. of the SUMO protein. This is an advantage be-
Recently developed tags: SET. It was also cause, unlike when using other proteases, no
recently shown that small highly acidic pep- non-native residues are left on the target pro-
tide tags, which are based on the C-terminal tein following cleavage (Butt et al., 2005). The
portion of the T7 phage gene 10B, promote the only restriction of the SUMO protease is that
soluble expression of folded, active protein for the N-terminal amino acid of the target protein
highly aggregation prone proteins. These tags cannot be a proline, as this residue restricts ac-
are known as solubility enhancing tags (SET cess to the SUMO protease active site.
tags). Namely, Zhang and colleagues showed Facilitating the formation of disulfide
that a series of T7 phage gene 10B-based pep- bonds: export signal sequences. An addi-
tide tags, with net negative charges of greater tional method to produce disulfide bond–
or equal than −6, fully solubilized the highly containing proteins in E. coli is to export
aggregation-prone Ig variable-type domain, them to the periplasm, the space between the
CAR D1 (Zhang et al., 2004). Moreover, it inner cytoplasmic membrane and the exter-
was folded. Significantly, in the same study, a nal outer membrane of Gram-negative bac-
peptide tag with a net charge of −12 partially teria like E. coli. The bacterial periplasm is
solubilized the myelin P-zero protein, P0 ex, a highly oxidative and thus promotes the for-
protein that is highly recalcitrant to solubiliza- mation of disulfide bonds (for a review of
tion (Zhang, Y.B. et al., 2004). An advantage methods used to produce disulfide containing
of the SET tags is that they are small, typi- proteins in E. coli see de Marco, 2009). The
cally <40 amino acids. They are also highly E. coli maltose-binding protein is targeted to
acidic, with net charges between −6 and the periplasm in vivo via an N-terminal ex-
−18. Although the mechanism by which SET port signal sequence. Thus, when a construct
tags improve solubility is still under investi- of MBP that includes the export sequence is
gation, it has been suggested that the nega- used as a fusion tag, the fusion protein is ex-
tively charged fusion peptide increases electro- ported to the periplasm and, in turn, the oxida-
static repulsion between folding-intermediates tive environment promotes disulfide bond for-
(Zhang, Y.B. et al., 2004). This effectively de- mation (Riggs, 2000). Because MBP is used
lays polypeptide aggregation, increasing the to target proteins both to the cytoplasm and
time available for the target protein to properly to the periplasm, it is important to verify that
fold. the expression vector used localizes the tar-
Recently developed tags: SUMO. Another get protein to the desired cellular compart-
novel and increasingly routinely used tag is ment. In addition to MBP, DsbA, a Trx ho-
the small ubiquitin-like modifier (SUMO) tag. molog, has also been used as a fusion partner
Its development as a prokaryotic protein ex- to target and promote the proper folding of
pression system was based on the observa- proteins in the periplasm (Collinsracie et al.,
tion that the addition of ubiquitin to recom- 1995; Couprie et al., 2000; Winter et al., 2000).
binant proteins increases their solubility (Butt Finally, it is important to point out that pro-
et al., 1989; Wittliff et al., 1990). The utility of teins targeted to the periplasm have specialized
Optimizing SUMO as both an expression and solubility- protocols for purification, which differ from
Protein enhancing tag has been demonstrated in mul- those used for proteins targeted to the bacterial
Expression in tiple studies. One of the first showed that cytosol (Malik et al., 2007).
E. coli

5.24.14
Supplement 61 Current Protocols in Protein Science
Position of fusion tag. All described solu- ferent residues on the C-terminal side of the
bility and affinity tags can be fused either to the sessile bond, eliminating nonnative residues
N- or C-terminus of the target protein. How- on the N-terminus of the target protein (Kapust
ever, N-terminal fusions are the most com- et al., 2002). TEV protease is also active over a
mon and have the added benefit that they often diverse set of experimental conditions, such as
enhance soluble protein expression more suc- pH, buffers, and temperatures (Kapust et al.,
cessfully than C-terminal fusions (Sati et al., 2001, 2002). A disadvantage is that TEV pro-
2002; Dyson et al., 2004; Busso et al., 2005). tease is inhibited by a large number of de-
One of the reasons N- versus C-terminally tergents, making the removal of fusion tags
fused tags effectively improve protein ex- from integral membrane proteins, which re-
pression and solubility is because many of quire protein-detergent complexes for solubi-
the fusion tags are native E. coli proteins. lization, challenging (Mohanty et al., 2003). If
Thus, they provide an efficient 5 sequence detergents are an essential component of the
for the initiation of transcription (Esposito protein buffer, thrombin protease, which re-
and Chatterjee, 2006). Correspondingly, the tains its activity in a wide range of detergents,
N-terminal mRNA sequence and structure is can be used to cleave the fusion tag.
efficiently recognized by the ribosome and Sometimes a protease will fail to cleave the
compatible with robust translation (Cebe and fusion protein. Most often, this is due to steric
Geiser, 2006). In contrast, the transcription and hindrance in which the protease site is not ac-
translation initiation sites of target genes and cessible to the enzyme (Kapust and Waugh,
proteins are variable and can impede these pro- 2000; Lee et al., 2008). Including a short linker
cesses when tags are fused to the target protein of glycine, asparagine, or alanine residues be-
C-terminus. Notably, there are exceptions, as tween the protease site and the fusion pro-
MBP and SET tags have been shown to be tein often alleviates this problem (Esposito and
equally effective in enhancing solubility re- Chatterjee, 2006). Inefficient cleavage may
gardless of which terminus they are fused to also be overcome by altering the cleavage con-
(Dyson et al., 2004; Zhang, Y.B. et al., 2004), ditions, such as increasing protease concentra-
although this is still protein dependent. One tag tions, extending the cleavage time, and altering
that is often fused to the protein C-terminus temperature, among other parameters.
is the his6 -tag. Typically, it is equally effec-
tive at either location, but has the advantage
that when fused to the C-terminus, only fully
IV. E. COLI HOST STRAINS THAT
translated proteins are purified as incompletely
AID EXPRESSION OF
translated target proteins do not include the
HETEROLOGOUS PROTEINS
Several elements of the E. coli strain used
purification tag.
for recombinant protein production have a
Removal of the fusion tag. All fusion tags
large impact on the success of soluble expres-
have the potential to influence the behavior
sion. As mentioned in previous sections of this
of the expressed protein, and thus it is typ-
review, bacterial host strains have been specif-
ically desirable to remove the tag following
ically developed to aid the expression of het-
purification. This is achieved by engineering
erologous proteins (e.g., codon-supplemented
a protease-specific cleavage site (a sequence
cells to aid the expression of proteins with rare
of ∼7 amino acids that is specifically recog-
codons). Brief descriptions of commercially
nized by the protease) between the tag and
available E. coli strains designed for the spe-
target protein. Following expression and pu-
cific expression of proteins that are susceptible
rification, the corresponding protease is used
to proteolysis, contain rare codons, or require
to cleave the tag from the protein in vitro.
disulfide-bonds are provided below.
Several proteases are widely used for fu-
sion tag removal (see Table 5.24.4). However,
many enzymes, such as thrombin, factor Xa, Protease-deficient strains: BL21, BL21
and enterokinase, can promiscuously cleave (DE3), and BL21Star (DE3)
the target protein at nonspecific sites (Chang, E. coli BL21 and its derivatives are most
1985; Choi et al., 2001). Today, one of the frequently used for routine protein expres-
most widely used proteases is tobacco etch sion. These strains are deficient of ompT and
virus (TEV) protease. It has a number of ad- lon proteases. OmpT is a bacterial endopro-
vantages. First, it is more stringent and thus tease that readily cleaves T7 RNA polymerase
efficiently cleaves fusion tags without nonspe- (Grodberg and Dunn, 1988), while the Lon Production of
cific secondary cleavage (Phan et al., 2002). (La) protease, encoded by the lon gene, is an Recombinant
Proteins
Second, it can accommodate a number of dif- ATP-dependent enzyme that rapidly degrades
5.24.15
Current Protocols in Protein Science Supplement 61
Supplement 61
5.24.16
Table 5.24.4 Commonly used Proteases to Remove Fusion Tags from Recombinant Proteins

Protease Description Cleavage sitea Protease inhibitors Activity Comments

TEV Catalytic domain of the EXXYXQ-(G/S) PMSF, AEBSF, TLCK, pepstatin A, pH: 4-9 Sequence specificity more
Nuclear Inclusion a (NIa) Most common: bestatin, E-64, zinc (>5 mM), EDTA Temperature: 4◦ C-37◦ C (max. at stringent than factor Xa,
protein, a cysteine protease, ENLYFQ-X (1 mM), reagents that react with 34◦ C)
found in the tobacco etch cysteine; various detergents Can accommodate most buffers thrombin, and enterokinase
virus (TEV) Can be produced in E. coli
3Cb Recombinant form of the 3C EVLFQ-GP PMSF, TLCK, leupeptin, zinc pH: 3-10 High sequence specificity
protease from human (100 mM), urea (1 M), guanidine Temperature: 4◦ C-37◦ C (max. at Can be produced in E. coli
rhinovirus type 14 (1 M) 4◦ C)
Test activity in buffer of choice
Xac Factor Xa is a serine protease I(E/N)GR- PMSF, AEBSF, DFP, aprotinin, pH: 6.5-9 Nonspecific proteolysis
that converts prothrombin to Will not cleave at site antithrombin III, antipain, Temperature: 4◦ C-37◦ C may occur at secondary
thrombin followed by P or R α1-antitypsin, chymostatin, hirudin, Activity is highly reduced in sites
leupeptin, urea (100 mM), guanidine phosphate buffers compared to Cleavage usually
(10 mM), NaCl (100 mM), imidazole Tris or HEPES, CaCl2 should be performed near
(100 mM) included in cleavage reaction physiological conditions
Thr Thrombin is a serine protease LVPR-GS PMSF, AEBSF, aprotinin, pH: 5-10 Nonspecific proteolysis
that converts fibrinogen into antithrombin III, antipain, Temperature: 4◦ C-37◦ C (max. at may occur at secondary
fibrin α1-antitypsin, chymostatin, hirudin, 37◦ C) sites
leupeptin, reducing agents Can accommodate most buffers Cleavage usually
performed near
physiological conditions
EntK Catalytic subunit of bovine DDDDK- Serine protease inhibitors, PMSF, pH: 6.0-8.5 Nonspecific proteolysis
enterokinase imidazole (250 mM), NaCl (250 mM), Temperature: 4◦ C-37◦ C may occur at secondary
urea (2 M), SDS Activity is highly reduced in sites
phosphate buffers compared to
Tris or MES
a The “–” indicates the site of cleavage within the single letter amino acid code.
b See Miyashita et al., 1992.
c See Nagai and Thogersen, 1984; Jenny et al., 2003.

Current Protocols in Protein Science


misfolded and recombinant proteins (Phillips mation (see next section) (Milisavljevic et al.,
et al., 1984). Correspondingly, deletion of lon 2009).
and ompT is correlated with increased ex-
pression and stability of recombinant proteins Strains to express disulfide-bonded
(Gottesman, 1990). Since, the T7 RNA poly-
proteins: BL21 trxB, Origami,
merase system is the most widely used pro- Rosetta-gami
As discussed in section III, some host
moter system for protein expression, most
strains (BL21 trxB, Origami, Rosetta-gami)
BL21 strains, designated (DE3) strains, con-
have mutations in thioredoxin reductase (trxB)
tain a chromosomal copy of the T7 RNA poly-
and/or glutathione reductase (gor) genes, two
merase gene, allowing for simple and efficient
proteins that maintain the reducing environ-
expression of genes from T7-based expression
ment of the bacterial cytosol (Prinz et al.,
vectors.
1997). Consequently, these strains aid the for-
BL21Star (DE3) (Invitrogen), a derivative
mation of cytosolic disulfide bonds, greatly en-
of the BL21 (DE3) strain, contains an addi-
hancing the solubility of folded, active disul-
tional mutation in the rne gene (Lopez et al.,
fide bond–containing proteins produced in the
1999). This gene encodes RNase E, an enzyme
cytosol. Moreover, these strains can also be
that functions as an essential part of the “de-
more effective than exporting the target protein
gradosome” to actively degrade mRNA within
to the periplasm. For example, in one study,
the cell (Grunberg-Manago, 1999; Lopez et al.,
a trxB/gor mutant strain (FA113; a suppres-
1999; Carpousis, 2007). Consequently, the use
sor strain that overcomes the normally slow
of this strain results in an increase in mRNA
growth of trxB/gor mutant strains in the ab-
stability, and, in turn, protein expression. The
sence of reductant) increased the active yield
strain developers have routinely noted a 2- to
of multiple proteins [E. coli alkaline phos-
10-fold increase in heterologous protein ex-
phatase, a truncated form of the human tissue
pression when compared to non-RNase E de-
plasminogen activator (vtPA) and full-length
fective BL21 strains (Invitrogen). However,
human tPA, which contained two, nine, and
the basal expression level of the target gene
seventeen disulfide bonds, respectively] when
is also increased when using these strains,
compared to yields obtained by secretion of
and therefore, a variant that exhibits repressed
the proteins to the periplasm (Bessette et al.,
basal expression, BL21Star (DE3)pLysS (see
1999). Because inter-protein disulfide bonds
description of pLysS strains below), should be
can also form, leading to high-molecular-
used when expressing toxic proteins.
weight oligomers, these strains should only be
used to express proteins that require disulfide
Codon-supplemented strains: BL21-RP, bonds for proper folding.
BL21-RIL, BL21-RPIL, Rosetta, and
Rosetta-gami Strains to express toxic proteins:
As described in section II, differences in BL21-AI and pLysS
codon frequency between the target gene The BL21-AI host strain, in which the T7
and the expression host can lead to trans- RNA polymerase gene is under the control of
lational stalling, premature translation ter- the araBAD promoter, was developed to allow
mination, and amino acid mis-incorporation the expression of toxic genes from any T7-
(Kane, 1995). Instead of generating a codon- based expression vector. As described earlier,
optimized gene, this disparity may be over- the low basal activity of the araBAD promoter
come by supplying the rare tRNAs during ex- system is optimal for the expression of pro-
pression (Burgess-Brown et al., 2008). Numer- teins that are toxic to the host. In BL21-AI
ous bacterial strains (BL21-RP, BL21-RIL, cells, basal expression of the T7 RNA poly-
BL21-RPIL, Rosetta, and Rosetta-gami) are merase, and the subsequent target gene, is
available that contain plasmids that encode highly repressed in the absence of arabinose
rare tRNAs to promote the efficient expres- and the presence of glucose. This cell line has
sion of genes that contain high frequencies been used for the robust expression of several
of rare codons. The tRNAs that are supple- heterologous enzymes, with soluble yields ex-
mented in each strain differ, and therefore, ceeding 30 mg/liter (Chen et al., 2009; Yao
the appropriate host strain should be deter- et al., 2009).
mined on a protein-to-protein basis. Addition- As an alternative to the BL21-AI strain,
ally, the Rosetta-gami cell line, which con- several BL21 derivatives, known as pLysS
Production of
tains trxB/gor mutants, has the added benefit strains (also described in section III), express Recombinant
of facilitating cytoplasmic disulfide bond for- T7 phage lysozyme, an enzyme that effectively Proteins

5.24.17
Current Protocols in Protein Science Supplement 61
inhibits T7 RNA polymerase activity. Inhibi- temperatures, and thus lowering the expres-
tion of T7 RNA polymerase, in turn, decreases sion temperature also results in a reduction
basal expression of the target protein. Like in the degradation of proteolytically sensitive
BL21-AI strains, pLysS strains have been used proteins (Spiess et al., 1999; Hunke and Bet-
to successfully express toxic proteins under the ton, 2003; Pinsach et al., 2008).
control of T7 RNA polymerase (Jensen et al., Because of the profound increase in the
1999; Cinquin et al., 2001). yield of soluble protein at low temperatures,
Typically, protein expression is accom- it is strongly suggested to use a low induction
plished using host strains that have a combi- temperature as the default (see typical protocol
nation of the elements described above. For in Fig. 5.24.2). The bacterial culture should be
example, Origami (DE3) pLysS cells have cultivated at 37◦ C until mid-to-late log phase
trxB/gor mutations, promoting the produc- (Chou, 2007). The culture is then induced and
tion of disulfide-bonds, contain a chromoso- the recombinant protein synthesized between
mal copy of T7 RNA polymerase for the ef- 15◦ C to 25◦ C (Peti and Page, 2007; Gråslund
ficient expression of genes under the control et al., 2008a). Due to the reduced protein syn-
of this enzyme, and express pLysS to decrease thesis rate, longer induction times are neces-
basal expression and aid production of toxic sary to obtain a sufficient protein yield (typical
target proteins. This strain was essential for induction time: 4 hr at 37◦ C, 16 to 20 hr at
the soluble production of BSPH1, a novel hu- 18◦ C).
man sperm-binding protein that contains four
disulfide-bonds and is completely insoluble Concentration of the inducer
when expressed in BL21 (DE3) cells (Lefeb- In addition to lowering the growth temper-
vre et al., 2009b). Therefore, all properties of ature, a reduction in transcription rate can also
the protein should be considered when choos- be achieved by lowering the concentration of
ing the optimal host strain to express the pro- the induction agent. For example, the araBAD
tein of interest. promoter system is titratable, and thus lower
concentrations of the inducer, L-arabinose, re-
V. THE SOLUBILITY OF PROTEINS sult in the production of less protein (Guzman
CAN BE IMPROVED BY et al., 1995). Additionally, decreasing the
CHANGING EXPRESSION concentration of IPTG can also enhance the
CONDITIONS production of soluble protein. For example, the
In E. coli, transcription and translation are solubility of recombinant cyclomaltodextri-
tightly coupled, with ∼60,000 polypeptide nase (CDase) was shown to be highly sensitive
chains being synthesized per minute (Lorimer, to the concentration of the inducer. When the
1996). The use of strong expression promot- protein was induced using 0.05 mM IPTG, the
ers and high inducer concentrations can result protein was soluble and active; however, when
in nascent protein concentrations that are so the inducer concentration was doubled to 0.1
high that the proteins aggregate before folding. mM the expressed protein was insoluble and
Thus, reducing the rates of transcription and/or inactive (Turner et al., 2005). Thus, although
translation facilitates folding by allowing the the most common IPTG concentrations for
newly synthesized protein to fold before it ag- protein induction range from 0.1 to 1.0 mM,
gregates and forms inclusion bodies. Below a decrease to even lower levels can effect solu-
we describe the common expression condition bility. Finally, a derivative of the BL21 (DE3)
parameters that can be manipulated to enhance expression strain, containing a lacY1 deletion
protein solubility. mutation, has been developed that allows the
T7 promoter system to be titratable over a
Temperature range of IPTG concentrations (Turner et al.,
Lowering the expression temperature rou- 2005). This cell line, called Tuner (DE3), per-
tinely improves the solubility of recom- mits accurate tuning of the induction level and
binantly expressed proteins (Shirano and can, in some cases, promote the production of
Shibata, 1990; Kataeva et al., 2005; Volonte soluble protein (Turner et al., 2005).
et al., 2008; Piserchio et al., 2009). At lower
temperatures, cell processes slow down, lead- Media
ing to reduced rates of transcription, transla- Although fermentation has its clear ad-
Optimizing tion, and cell division (Chou, 2007), while also vantages, batch culture is the most com-
Protein leading to reduced protein aggregation (Vasina mon method to cultivate cells for recombi-
Expression in and Baneyx, 1997; Sahdev et al., 2008). More- nant protein expression. Since there is lim-
E. coli ited control over the growth parameters using
over, most proteases are less active at lower
5.24.18
Supplement 61 Current Protocols in Protein Science
this approach, all nutrients that are required result in rapid growth arrest and/or cell death.
for growth must be supplied from the begin- For some proteins, this toxicity can be mit-
ning by inclusion in the growth medium. Luria igated when the toxin is coexpressed with a
broth (LB) is the standard for the expression second protein that either binds and inhibits
of proteins. This broth is composed of bacto- the toxin or does not bind yet counteracts its
tryptone, which provides peptides, peptones activity. Both strategies allow the toxic pro-
and essential amino acids, yeast extract, which tein to be successfully solubly expressed to
provides vitamins and trace elements, and high levels. Two families of proteins that are
sodium chloride, which provides sodium ions routinely associated with cell toxicity are en-
to maintain osmotic balance (Sahdev et al., dogenous E. coli toxins, such as RelE, which
2008). Terrific broth (TB), the second most inhibits translation through mRNA cleavage
widely used expression medium, is formu- (Pedersen et al., 2003), and protein tyrosine ki-
lated to increase protein solubility and yield nases (PTK), which can promiscuously phos-
(Sahdev et al., 2008). M9, or minimal medium, phorylate nonnative E. coli targets (Wang, Y.H.
contains the minimum nutrients essential for et al., 2006). However, multiple groups have
bacterial species to grow. Minimal medium successfully expressed these proteins in E. coli
usually lacks amino acids, and thus is most by coexpressing them with an appropriate pro-
widely used for selective labeling, i.e., isotope tein partner. Thus, if the target protein is toxic
labeling for proteins studied using NMR spec- to the host cell, coexpression with a protein
troscopy (Reilly and Fairbrother, 1994; Pryor that antagonizes the toxic effect should be con-
and Leiting, 1997; Chen et al., 2006). sidered.
Toxin-antitoxin pairs. Toxin:antitoxin pairs
VI. ENHANCING SOLUBILITY BY (Gerdes et al., 2005) are composed of an unsta-
COEXPRESSION WITH OTHER ble antitoxin and a stable toxin. Under normal
PROTEINS conditions, the toxin and antitoxin associate
Some proteins only express solubly to form a tight, nontoxic complex. However,
when they are coexpressed with additional under conditions of stress, the antitoxins are
biomolecules, typically other proteins, like a degraded, leading to cell growth arrest due
binding partner, or molecular chaperones. In to the cellular effects of the toxin (inhibiting
these cases, the target protein is coexpressed replication by blocking DNA gyrase or, more
with a second protein that is encoded on ei- commonly, inhibiting translation via mRNA
ther the same plasmid or a separate plasmid. cleavage) (Gerdes et al., 2005). One such
Multiple vectors have been designed for coex- toxin:antitoxin system is mqsRA, which en-
pression of two or more proteins, such as the codes the MqsR toxin, a ribonuclease, and its
Duet vectors (Novagen; coexpression of two cognate MqsA antitoxin (Brown et al., 2009).
or more target proteins from the same vector We showed that overexpression of MqsR alone
using the T7 promoter system) or separate vec- results in growth arrest, as translation is in-
tors (Expression Technologies; coexpression hibited via mRNA cleavage by MqsR. How-
of two proteins using two different vectors). ever, coexpression of MqsR with MqsA, which
Moreover, multiple Duet vectors can be used binds and mitigates MqsR-mediated toxicity,
in the same cell, allowing the expression of up resulted in the robust expression of both pro-
to eight proteins simultaneously. Finally, mul- teins. In this case, MqsR and MqsA were in-
tiple vectors have been developed that contain duced simultaneously using IPTG from sep-
E. coli chaperones (GroEL/ES, dnaJKE, trig- arate T7 polymerase-based vectors (MqsR:
ger factor; Takara Bio), allowing the protein of pet28A, Kan resistance, pBR322 promoter;
interest to be coexpressed with one or a combi- MqsA: pAC21a, Amp resistance, pACYC pro-
nation of chaperones to facilitate proper fold- moter) (Brown et al., 2009). There are also
ing (Kyratsous et al., 2009). Below are a few other examples in which other bacterial toxins
examples of proteins that require coexpression have been solubly produced in E. coli by coex-
with either a partner protein or molecular chap- pression with their cognate antitoxins (Gerdes
erone for successful expression. et al., 2005).
More recently, these toxin-antitoxin sys-
Coexpression with a partner protein tems have been used as plasmid-stabilization
Expressing proteins whose activities dis- systems that can effectively increase recombi-
rupt E. coli growth and/or signaling are of- nant protein production levels. While the anti-
ten toxic to the cell. While host cells can be toxin protein is constitutively expressed from Production of
cultivated to a high density under repressive a plasmid that encodes the protein of interest, Recombinant
Proteins
conditions, induction of the toxic protein may the toxin gene is chromosomally incorporated
5.24.19
Current Protocols in Protein Science Supplement 61
and highly repressed in the presence of the of these folding-partners using coexpression
antitoxin (StabyExpress, Delphi Genetics). If methods has proven to effectively aid solubi-
the plasmid is lost from the host cell, the highly lization of target proteins.
labile antitoxin is readily degraded and the DnaK-DnaJ-GrpE and GroEL-GroES. The
activity of the toxin is no longer inhibited, DnaK-DnaJ-GrpE and GroEL-GroES systems
leading to cell growth arrest. This plasmid- are the most extensively characterized molec-
stabilization strategy, using the CcdA/CcdB ular chaperones in E. coli. Both groups
toxin-antitoxin pair, often increases the fi- are folding chaperones that bind to solvent-
nal recombinant protein production levels by exposed hydrophobic domains, preventing
three- to five-fold (Stieber et al., 2008). nascent peptide aggregation, and through it-
Kinase-phosphatase partners. Several, but erative rounds of ATP-driven conformational
not all, tyrosine kinases have proven difficult changes, mediate the folding/unfolding of
to express in E. coli. The expression of an their substrates (for an extensive review of
active kinase is often accompanied by low cytosolic molecular chaperones see Hartl and
yield, extensive degradation, insolubility, and Hayer-Hartl, 2002). While the soluble expres-
a heterogeneously autophosphorylated sample sion of certain proteins is improved by coex-
(Y.H. Wang et al., 2006). In contrast, catalyti- pression with DnaK-DnaJ-GrpE (Chen et al.,
cally inactive kinases can be expressed to high 2003), other proteins result in higher yields
yields with little difficulty. Thus, the activity of soluble protein when coexpressed with
of the kinase is not well tolerated by the host GroEL-GroES (Ayling and Baneyx, 1996;
cell. To counteract the negative effects of the Park et al., 2004; Sahu et al., 2009). Unfor-
kinase, multiple groups have coexpressed tyro- tunately, it is not possible to predict which,
sine kinases with an opposing tyrosine phos- if any, molecular chaperone system will im-
phatase. For example, Src kinase, a tyrosine prove protein solubility. Due to the reduced
kinase, either expresses insolubly or sponta- metabolic burden, it is recommended to test
neously mutates to generate an inactive kinase protein expression with the two chaperone sys-
when expressed alone. However, when coex- tems separately (for a thorough coexpression
pressed with PTP1B, a tyrosine phosphatase, protocol see Baneyx and Palumbo, 2003).
milligram amounts of soluble, active kinase In our laboratory, we have found that co-
were produced (Y.H. Wang et al., 2006). In expression of protein phosphatase 1 (PP1α),
this case, rather than using two different plas- a metal-dependent serine/threonine phos-
mids, an MBP-PTP1B-thrombin site-Src fu- phatase, with GroEL and GroES results in
sion protein was expressed from a single plas- the maximum yield of soluble protein (Kelker
mid. Following purification, the protein fusion et al., 2009). To ensure ample chaperones are
tag (MBP-PTP1B) was removed by throm- available to aid protein folding at the start of
bin cleavage (Y.H. Wang et al., 2006). This recombinant protein production, GroEL and
kinase-phosphatase coexpression method has GroES expression, which is under the con-
also proven effective for the soluble expres- trol of the araBAD promoter (induced with
sion of Met kinase, which was solubly coex- L-arabinose), is induced at a culture optical
pressed with PTP1B in which both proteins density of λ600 = 0.5, while PP1α, which is
were expressed using two compatible vectors under the control of the T7 promoter (in-
(Seeliger et al., 2005), and Abl kinase, which duced with IPTG), is induced later, at a cul-
was solubly coexpressed with YopH using a ture optical density of λ600 = 1.0. After 20 hr
single bicistronic vector (W.R. Wang et al., (10◦ C), the cultivated cells are pelleted and
2006). then resuspended in fresh medium that con-
tains 200 μg/ml chloroamphenical to block all
Coexpression with molecular ribosome activity and the cultures are allowed
chaperones to shake for an additional 2 hr. This last step
Small bacterial proteins typically fold allows for in vivo refolding and results in a
rapidly, due to fast folding kinetics. How- substantial increase in the amount of folded,
ever, larger bacterial and heterologous pro- active PP1α (Kelker et al., 2009).
teins fold more slowly, and thus require protein cpn10-cpn60. Unfortunately, most molec-
chaperones and folding catalysts to prevent ular chaperone systems display reduced ac-
aggregation and facilitate folding in E. coli tivity at reduced temperatures. For example,
Optimizing (Baneyx and Mujacic, 2004). While molecu- while GroEL/GroES is most efficient at 30◦ C,
Protein lar chaperones are ubiquitous in the cell, they this system is only 30% active at 12◦ C. As
Expression in are rapidly titrated during overexpression of discussed throughout the review, there are ex-
E. coli
recombinant proteins. Thus, supplementation tensive advantages to expressing recombinant
5.24.20
Supplement 61 Current Protocols in Protein Science
Overview of protein expression strategy Obstacles Potential solutions

Design construct of single,


globular domain
Step 1: Design protein construct based on Protein has MW >60 kD
available structural and functional data Use solubility-enhancing
fusion tag
Consider: 1. rare codons
2. protein size and domain Many rare codons or Codon optimize target gene
boundaries tandem rare codons
3. protein sequence: number of low Supplement rare tRNAs
complexity regions, contiguous
hydrophobic residues, and Use 2° structure prediction
disulfide bonds Domain boundaries are
unknown Thread sequence onto a
homologous protein
structure

Step 2: Select/design/clone appropriate


vector
Use tightly controlled
Consider: 1. plasmid copy number Expressing toxic protein promoter system
2. promoter (strength, basal
expression, mode of induction) Coexpress with chaperones
3. fusion tag (placement, solubility Expressing aggregation-
enhancement, affinity properties) prone protein Use solubility-enhancing
4. removal of fusion tag fusion tag

Protein susceptible to
Use protease deficient strain
Step 3: Choose expression strain proteolytic degradation
Consider: 1. protease susceptibility
Express in trxB/gor mutant
2. rare codon usage strain with Trx tag
Protein contains disulfide
3. disulfide bond requirements
bonds
Export to periplasm

Step 4: Test expression Cell death due to Use tightly controlled


Consider: 1. induction temperature high basal level of promoter system
2. inducer concentration expression or Coexpress with partner
3. choice of media toxic protein protein

Export to periplasm
No Disulfide bonds not formed
Express in
Express trxB/gor mutant
in trxB/gor mutant
soluble (contains
strain with
strain with Trx
Trx tag
tag
expression? disulfide bonds)
Aggregation of protein into
No
(lacks inclusion bodies
Yes Coexpress with chaperones
disulfide bonds)
Lower induction temperature
Step 5: Large scale expression using
conditions determined in Step 4 Lower inducer concentration
Use solubility-enhacing
No Un/Misfolded protein fusion tag

No active protein?
No Interference of fusion tag Cleave affinity tag

Figure 5.24.3 Flowchart depicting the critical factors to consider, common obstacles, and potential solutions for each
stage of protein expression in E. coli. The left column lists the major steps of recombinant protein expression with key
variables to consider. The middle column includes common obstacles encountered at each step, while possible solutions
for each obstacle are presented in the right column.

Production of
Recombinant
Proteins

5.24.21
Current Protocols in Protein Science Supplement 61
proteins at low temperatures. A second sys- with molecular chaperones, supplementing the
tem is available that is maximally functional cultivation medium with metals, growing the
at low temperatures: the cold-adapted chaper- culture and inducing protein expression at low
onins Cpn10 and Cpn60 from psychrophilic temperatures, using low concentrations of in-
bacterium, Oleispira antarctica. These chap- ductants, and cleaving the fusion tag after the
eronins, which have 74% and 54% amino acid protein has been stabilized by a PP1α ligand
identity with GroEL and GroES, respectively, (either an inhibitor or a PP1α binding pro-
are effective folding-modulators at low tem- tein). While time-consuming, the high yields
peratures (4◦ C to 12◦ C) (Amada et al., 1995). of soluble, active PP1α have been essential for
Using this system, a temperature-sensitive es- the successful completion of multiple projects
terase expressed at 10◦ C with Cpn10 and (Dancheck et al., 2008, Kelker et al., 2009);
Cpn60 exhibited a 180-fold increase in activity thus, the effort devoted to determining the op-
over expression at 37◦ C (Ferrer et al., 2004). A timal protocol was invaluable.
derivative of the BL21 host strain, ArcticEx-
press (Stratagene), that coexpresses these two
chaperonins, has been developed and used to ACKNOWLEDGMENTS
We thank Dr. Wolfgang Peti for careful
successfully express several proteins, includ-
reading of the manuscript.
ing interleukin-2 tyrosine kinase (Joseph and
Andreotti, 2008).
LITERATURE CITED
Amada, K., Yohda, M., Odaka, M., Endo, I.,
ANTICIPATED RESULTS AND Ishii, N., Taguchi, H., and Yoshida, M. 1995.
TIME CONSIDERATIONS Molecular-cloning, expression, and character-
All of the parameters listed in this unit, ization of Chaperonin-60 and Chaperonin-
from construct length to inducer concentra- 10 from a thermophilic bacterium, Thermus-
tion, can affect the solubility of recombinant Thermophilus Hb8. J. Biochem. 118:347–354.
proteins produced in E. coli. Accordingly, it is Amann, E., Brosius, J., and Ptashne, M. 1983. Vec-
often necessary to vary one or many elements tors bearing a hybrid Trp-Lac promoter use-
ful for regulated expression of cloned genes in
in the expression protocol to successfully ex-
Escherichia coli. Gene 25:167-178.
press soluble protein (Fig. 5.24.3). This can
Armstrong, R.N. 1997. Structure, catalytic mech-
mean testing scores of expression protocols,
anism, and evolution of the glutathione trans-
which is time consuming and costly. Thus, it ferases. Chem. Res. Toxicol. 10:2-18.
is advantageous to use small-scale expression
Arnold, K., Bordoli, L., Kopp, J., and Schwede, T.
tests to select an optimal construct and ex- 2006. The SWISS-MODEL workspace: A web-
pression conditions prior to scaling up. The based environment for protein structure homol-
microexpression protocol used in our labora- ogy modelling. Bioinformatics 22:195-201.
tory to determine expression, as well as protein Ayling, A. and Baneyx, F. 1996. Influence of
solubility, has been explicitly described (Peti the GroE molecular chaperone machine on the
and Page, 2007). It is recommended to use in vitro refolding of Escherichia coli beta-
galactosidase. Protein Sci. 5:478-487.
this strategy to determine an optimal proto-
col for large-scale expression. Be aware that Baneyx, F. and Mujacic, M. 2004. Recombinant
protein folding and misfolding in Escherichia
results from small-scale growths do not al-
coli. Nat. Biotechnol. 22:1399-1408.
ways translate to large-scale systems. While
Baneyx, F. and Palumbo, J.L. 2003. Improving het-
positive small-scale results can usually be re-
erologous protein folding via molecular chap-
produced in large-scale studies, proteins that erone and foldase co-expression. Methods Mol.
appear to have low or insoluble expression on a Biol. 205:171-197.
small-scale may be expressed in soluble form Bao, W.J., Gao, Y.G., Chang, Y.G., Zhang, T.Y.,
when grown on a larger scale (Gråslund et al., Lin, X.J., Yan, X.Z., and Hu, H.Y. 2006. Highly
2008a). efficient expression and purification system of
The importance of folded, active recombi- small-size protein domains in Escherichia coli
for biochemical characterization. Protein Expr.
nant protein to the proposed research project
Purif. 47:599-606.
defines the amount of time and effort that
Bessette, P.H., Aslund, F., Beckwith, J., and
is devoted to creating the optimal expression
Georgiou, G. 1999. Efficient folding of proteins
protocol. For example, five years were de- with multiple disulfide bonds in the Escherichia
voted to identifying the optimal expression and coli cytoplasm. Proc. Natl. Acad. Sci. U.S.A.
Optimizing purification protocol for protein phosphatase 96:13703-13708.
Protein 1 (PP1α) (Kelker et al., 2009). This opti- Brosius, J., Erfle, M., and Storella, J. 1985. Spacing
Expression in
E. coli mized protocol includes coexpressing PP1α of the −10 and −35 regions in the tac promoter.

5.24.22
Supplement 61 Current Protocols in Protein Science
Effect on its in vivo activity. J. Biol. Chem. Chen, L.F., Maloney, K., Krol, E., Zhu, B., and
260:3539-3541. Yang, J. 2009. Cloning, overexpression, purifi-
Brown, B.L., Hadley, M., and Page, R. 2008. Het- cation, and characterization of the maleylacetate
erologous high-level E. coli expression, purifi- reductase from sphingobium chlorophenolicum
cation and biophysical characterization of the strain ATCC 53874. Curr. Microbiol. 58:599-
spine-associated RapGAP (SPAR) PDZ domain. 603.
Protein Expr. Purif. 62:9-14. Chen, X., Tong, X.T., Xie, Y.H., Wang, Y., Ma, J.B.,
Brown, B.L., Grigoriu, S., Kim, Y., Arruda, J.M., Gao, D.M., Wu, H.M., and Chen, H.B. 2006.
Davenport, A., Wood, T.K., Peti, W., and Page, Over-expression and purification of isotopically
R. 2009. Three dimensional structure of the labeled recombinant ligand-binding domain of
MqsR:MqsA complex: A novel TA pair com- orphan nuclear receptor human B1-binding fac-
prised of a toxin homologous to RelE and an tor/human liver receptor homologue 1 for NMR
antitoxin with unique properties. PLoS Pathog. studies. Protein Expr. Purif. 45:99-106.
5:e1000706. Chen, Y. and Leong, S.S.J. 2009. Adsorptive refold-
Burgess-Brown, N.A., Sharma, S., Sobott, F., ing of a highly disulfide-bonded inclusion body
Loenarz, C., Oppermann, U., and Gileadi, O. protein using anion-exchange chromatography.
2008. Codon optimization can improve expres- J. Chromatogr. A 1216:4877-4886.
sion of human genes in Escherichia coli: A Chen, Y., Song, J.M., Sui, S.F., and Wang, D.N.
multi-gene study. Protein Expr. Purif. 59:94- 2003. DnaK and DnaJ facilitated the folding
102. process and reduced inclusion body formation
Busso, D., Delagoutte-Busso, B., and Moras, D. of magnesium transporter CorA overexpressed
2005. Construction of a set Gateway-based des- in Escherichia coli. Protein Expr. Purif. 32:221-
tination vectors for high-throughput cloning and 231.
expression screening in Escherichia coli. Anal. Choi, S.I., Song, H.W., Moon, J.W., and Seong, B.L.
Biochem. 343:313-321. 2001. Recombinant enterokinase light chain
Butt, T.R., Jonnalagadda, S., Monia, B.P., with affinity tag: Expression from Saccha-
Sternberg, E.J., Marsh, J.A., Stadel, J.M., Ecker, romyces cerevisiae and its utilities in fusion
D.J., and Crooke, S.T. 1989. Ubiquitin fusion protein technology. Biotechnol. Bioeng. 75:718-
augments the yield of cloned gene-products in 724.
Escherichia coli. Proc. Natl. Acad. Sci. U.S.A. Chong, S.R., Montello, G.E., Zhang, A.H., Cantor,
86:2540-2544. E.J., Liao, W., Xu, M.Q., and Benner, J. 1998.
Butt, T.R., Edavettal, S.C., Hall, J.P., and Utilizing the C-terminal cleavage activity of
Mattern, M.R. 2005. SUMO fusion technology a protein splicing element to purify recombi-
for difficult-to-express proteins. Protein Expr. nant proteins in a single chromatographic step.
Purif. 43: 1-9. Nucleic Acids Res. 26:5109-5115.

Calderone, T.L., Stevens, R.D., and Oas, T.G. Chou, C.P. 2007. Engineering cell physiology
1996. High-level misincorporation of lysine for to enhance recombinant protein production in
arginine at AGA codons in a fusion protein Escherichia coli. Appl. Microbiol. Biotechnol.
expressed in Escherichia coli. J. Mol. Biol. 76:521-532.
262:407-412. Cinquin, O., Christopherson, R.I., and Menz, R.I.
Canaves, J.M., Page, R., Wilson, I.A., and Stevens, 2001. A hybrid plasmid for expression of
R.C. 2004. Protein biophysical properties that toxic malarial proteins in Escherichia coli. Mol.
correlate with crystallization success in Ther- Biochem. Parasitol. 117:245-247.
motoga maritima: Maximum clustering strategy Collinsracie, L.A., McColgan, J.M., Grant, K.L.,
for structural genomics. J. Mol. Biol. 344:977- Diblasio-Smith, E.A., McCoy, J.M., and Laval-
991. lie, E.R. 1995. Production of recombinant
Carpousis, A.J. 2007. The RNA degradosome of bovine enterokinase catalytic subunit in Es-
Escherichia coli: An mRNA-degrading machine cherichia coli using the novel secretory fusion
assembled on RNase E. Annu. Rev. Microbiol. partner Dsba. Biotechnology 13:982-987.
61:71-87. Couprie, J., Vinci, F., Dugave, C., Quemeneur,
Cebe, R. and Geiser, M. 2006. Rapid and easy ther- E., and Moutiez, M. 2000. Investigation of the
modynamic optimization of the 5 -end of mRNA DsbA mechanism through the synthesis and
dramatically increases the level of wild type pro- analysis of an irreversible enzyme-ligand com-
tein expression in Escherichia coli. Protein Expr. plex. Biochemistry 39:6732-6742.
Purif. 45:374-380. Critton, D.A., Tortajada, A., Stetson, G., Peti, W.,
Chang, C.N., Kuang, W.J., and Chen, E.Y. and Page, R. 2008. Structural basis of substrate
1986. Nucleotide-sequence of the alkaline- recognition by hematopoietic tyrosine phos-
phosphatase gene of Escherichia coli. Gene phatase. Biochemistry 47:13336-13345.
44:121-125. Cruz-Vera, L.R., Magos-Castro, M.A., Zamora-
Chang, J.Y. 1985. Thrombin specificity - require- Romo, E., and Guarneros, G. 2004. Ribo-
ment for apolar amino-acids adjacent to the some stalling and peptidyl-tRNA drop-off dur-
thrombin cleavage site of polypeptide substrate. ing translational delay at AGA codons. Nucleic
Acids Res. 32:4462-4468. Production of
Eur. J. Biochem. 151:217-224. Recombinant
Proteins

5.24.23
Current Protocols in Protein Science Supplement 61
Dancheck, B., Nairn, A.C., and Peti, W. 2008. Esposito, D. and Chatterjee, D.K. 2006. Enhance-
Detailed structural characterization of unbound ment of soluble protein expression through the
protein phosphatase 1 inhibitors. Biochemistry use of fusion tags. Curr. Opin. Biotechnol.
47:12346-12356. 17:353-358.
Davis, G.D., Elisee, C., Newham, D.M., and Ferrer, M., Chernikova, T.N., Timmis, K.N.,
Harrison, R.G. 1999. New fusion protein sys- and Golyshin, P.N. 2004. Expression of
tems designed to give soluble expression in a temperature-sensitive esterase in a novel
Escherichia coli. Biotechnol. Bioeng. 65:382- chaperone-based Escherichia coli strain. Appl.
388. Environ. Microbiol. 70:4499-4504.
de Marco, A. 2006. Two-step metal affinity purifi- Fox, J.D., Kapust, R.B., and Waugh, D.S. 2001.
cation of double-tagged (NusA-His(6)) fusion Single amino acid substitutions on the surface
proteins. Nat. Protoc. 1:1538-1543. of Escherichia coli maltose-binding protein can
de Marco, A. 2009. Strategies for successful recom- have a profound impact on the solubility of fu-
binant expression of disulfide bond-dependent sion proteins. Protein Sci. 10:622-630.
proteins in Escherichia coli. Microb. Cell Fact. Gerdes, K., Christensen, S.K., and Lobner-Olesen,
8:26. A. 2005. Prokaryotic toxin-antitoxin stress
De Marco, V., Stier, G., Blandin, S., and de Marco, response loci. Nat. Rev. Microbiol. 3:371-
A. 2004. The solubility and stability of re- 382.
combinant proteins are increased by their fu- Goh, C.S., Lan, N., Douglas, S.M., Wu, B., Echols,
sion to NusA. Biochem. Biophys. Res. Commun. N., Smith, A., Milburn, D., Montelione, G.T.,
322:766-771. Zhao, H., and Gerstein, M. 2004. Mining the
Deboer, H.A., Comstock, L.J., and Vasser, M. 1983. structural genomics pipeline: Identification of
The Tac promoter - a functional hybrid derived protein properties that affect high-throughput
from the Trp and Lac promoters. Proc. Natl. experimental analysis. J. Mol. Biol. 336:115-
Acad. Sci. U.S.A. 80:21-25. 130.
Delatorre, J.C., Ortin, J., Domingo, E., Delamarter, Goldstein, J., Pollitt, N.S., and Inouye, M. 1990.
J., Allet, B., Davies, J., Bertrand, K.P., Wray, Major cold shock protein of Escherichia coli.
L.V., and Reznikoff, W.S. 1984. Plasmid vectors Proc. Natl. Acad. Sci. U.S.A. 87:283-287.
based on Tn10 DNA - gene-expression regulated Gordon, E., Horsefield, R., Swarts, H.G., de Pont,
by tetracycline. Plasmid 12:103-110. J.J., Neutze, R., and Snijder, A. 2008. Effec-
DePristo, M.A., Zilversmit, M.M., and Hartl, D.L. tive high-throughput overproduction of mem-
2006. On the abundance, amino acid com- brane proteins in Escherichia coli. Protein Expr.
position, and evolutionary dynamics of low- Purif. 62:1-8.
complexity regions in proteins. Gene 378:19- Gottesman, S. 1990. Minimizing proteolysis in Es-
30. cherichia coli - Genetic solutions. Methods in
Diguan, C., Li, P., Riggs, P.D., and Inouye, H. 1988. Enzymol. 185:119-129.
Vectors that facilitate the expression and purifi- Goulding, C.W. and Perry, L.J. 2003. Protein pro-
cation of foreign peptides in Escherichia coli by duction in Escherichia coli for structural stud-
fusion to maltose-binding protein. Gene 67:21- ies by X-ray crystallography. J. Struct. Biol.
30. 142:133-143.
Douette, P., Navet, R., Gerkens, P., Galleni, M., Gråslund, S., Nordlund, P., Weigelt, J., Hallberg,
Levy, D., and Sluse, F.E. 2005. Escherichia B.M., Bray, J., Gileadi, O., Knapp, S.,
coli fusion carrier proteins act as solubiliz- Oppermann, U., Arrowsmith, C., Hui, R., Ming,
ing agents for recombinant uncoupling protein J., dhe-Paganon, S., Park, H.W., Savchenko, A.,
1 through interactions with GroEL. Biochem. Yee, A., Edwards, A., Vincentelli, R.,
Biophys. Res. Commun. 333:686-693. Cambillau, C., Kim, R., Kim, S.H., Rao, Z., Shi,
Dvir, H. and Choe, S. 2009. Bacterial expression Y., Terwilliger, T.C., Kim, C.Y., Hung, L.W.,
of a eukaryotic membrane protein in fusion to Waldo, G.S., Peleg, Y., Albeck, S., Unger, T.,
various Mistic orthologs. Protein Expr. Purif. Dym, O., Prilusky, J., Sussman, J.L., Stevens,
68:28-33. R.C., Lesley, S.A., Wilson, I.A., Joachimiak,
A., Collart, F., Dementieva, I., Donnelly, M.I.,
Dyson, H.J. and Wright, P.E. 2005. Intrinsically Eschenfeldt, W.H., Kim, Y., Stols, L., Wu, R.,
unstructured proteins and their functions. Nat. Zhou, M., Burley, S.K., Emtage, J.S., Sauder,
Rev. Mol. Cell Biol. 6:197-208. J.M., Thompson, D., Bain, K., Luz, J., Gheyi,
Dyson, M.R., Shadbolt, S.P., Vincent, K.J., Perera, T., Zhang, F., Atwell, S., Almo, S.C., Bonanno,
R.L., and McCafferty, J. 2004. Production of sol- J.B., Fiser, A., Swaminathan, S., Studier, F.W.,
uble mammalian proteins in Escherichia coli: Chance, M.R., Sali, A., Acton, T.B., Xiao, R.,
Identification of protein features that correlate Zhao, L., Ma, L.C., Hunt, J.F., Tong, L.,
with successful expression. BMC Biotechnol. Cunningham, K., Inouye, M., Anderson, S.,
4:32. Janjua, H., Shastry, R., Ho, C.K., Wang, D.,
Wang, H., Jiang, M., Montelione, G.T., Stuart,
Elvin, C.M., Thompson, P.R., Argall, M.E., Hendry, D.I., Owens, R.J., Daenke, S., Schutz, A., Heine-
Optimizing P., Stamford, N.P.J., Lilley, P.E., and Dixon, N.E. mann, U., Yokoyama, S., Bussow, K., and
Protein 1990. Modified bacteriophage-lambda promoter
Expression in Gunsalus, K.C. 2008a. Protein production and
vectors for overproduction of proteins in Es- purification. Nat. Methods 5:135-146.
E. coli cherichia coli. Gene 87:123-126.
5.24.24
Supplement 61 Current Protocols in Protein Science
Gråslund, S., Sagemark, J., Berglund, H., Jing, G.Z., Huang, Z., Liu, Z.G., and Zou, Q. 1993.
Dahlgren, L.G., Flores, A., Hammarstroem, Plasmid Pkkh - an improved vector with higher
M., Johansson, I., Kotenyova, T., Nilsson, M., copy number for expression of foreign genes
Nordlund, P., and Weigelt, J. 2008b. The use in Escherichia coli. Biotechnol. Lett. 15:439-
of systematic N- and C-terminal deletions to 442.
promote production and structural studies of re- Johnston, K. and Marmorstein, R. 2003. Co-
combinant proteins. Protein Expr. Purif 58:210- expression of proteins in E. coli using dual ex-
221. pression vectors. Methods Mol. Biol. 205:205-
Grodberg, J. and Dunn, J.J. 1988. Ompt encodes the 213.
Escherichia coli outer-membrane protease that Jones, D.T. 1999. Protein secondary structure
cleaves T7-Rna polymerase during purification. prediction based on position-specific scoring
J. Bacteriol. 170:1245-1253. matrices. J. Mol. Biol. 292:195-202.
Grunberg-Manago, M. 1999. Messenger RNA sta- Jones, P.G., Vanbogelen, R.A., and Neidhardt, F.C.
bility and its role in control of gene expression 1987. Induction of proteins in response to low-
in bacteria and phages. Ann. Rev. Genet. 33:193- temperature in Escherichia coli. J. Bacteriol.
227. 169:2092-2095.
Guzman, L.M., Belin, D., Carson, M.J., and Joseph, R.E. and Andreotti, A.H. 2008. Bacterial
Beckwith, J. 1995. Tight regulation, modulation, expression and purification of Interleukin-2 Ty-
and high-level expression by vectors contain- rosine kinase: Single step separation of the chap-
ing the arabinose PBAD promoter. J. Bacteriol. eronin impurity. Protein Expr. Purif. 60:194-
177:4121-4130. 197.
Hammarstrom, M., Hellgren, N., van Den Berg, S., Kane, J.F. 1995. Effects of rare codon clusters
Berglund, H., and Hard, T. 2002. Rapid screen- on high-level expression of heterologous pro-
ing for improved solubility of small human pro- teins in Escherichia coli. Curr. Opin. Biotech-
teins produced as fusion proteins in Escherichia nol. 6:494-500.
coli. Protein Sci. 11:313-321.
Kaplan, W., Husler, P., Klump, H., Erhardt, J.,
Harley, C.B. and Reynolds, R.P. 1987. Analysis of Sluis Cremer, N., and Dirr, H. 1997. Conforma-
Escherichia coli promoter sequences. Nucleic tional stability of pGEX-expressed Schistosoma
Acids Res. 15:2343-2361. japonicum glutathione S-transferase: A detoxi-
Hartl, F.U. and Hayer-Hartl, M. 2002. Protein fication enzyme and fusion-protein affinity tag.
folding - Molecular chaperones in the cytosol: Protein Sci. 6:399-406.
From nascent chain to folded protein. Science Kapust, R.B. and Waugh, D.S. 1999. Escherichia
295:1852-1858. coli maltose-binding protein is uncommonly ef-
Hatfield, G.W. and Roth, D.A. 2007. Optimizing fective at promoting the solubility of polypep-
scaleup yield for protein production: Compu- tides to which it is fused. Protein Sci. 8:1668-
tationally Optimized DNA Assembly (CODA) 1674.
and translation engineering. Biotechnol. Annu. Kapust, R.B. and Waugh, D.S. 2000. Controlled in-
Rev. 13:27-42. tracellular processing of fusion proteins by TEV
Hawley, D.K. and McClure, W.R. 1983. Compila- protease. Protein Expr. Purif. 19:312-318.
tion and analysis of Escherichia coli promoter Kapust, R.B., Tozser, J., Fox, J.D., Anderson,
DNA-sequences. Nucleic Acids Res. 11:2237- D.E., Cherry, S., Copeland, T.D., and Waugh,
2255. D.S. 2001. Tobacco etch virus protease: Mech-
Huang, B.H., Shi, Z.T., and Tsai, M.D. 1994. anism of autolysis and rational design of sta-
A small, high-copy-number vector suitable for ble mutants with wild-type catalytic proficiency.
both in-vitro and in-vivo gene-expression. Gene Protein Eng. 14:993-1000.
151:143-145. Kapust, R.B., Tozser, J., Copeland, T.D., and
Hunke, S. and Betton, J.M. 2003. Temperature ef- Waugh, D.S. 2002. The P1  specificity of to-
fect on inclusion body formation and stress re- bacco etch virus protease. Biochem. Biophys.
sponse in the periplasm of Escherichia coli. Mol. Res. Commun. 294:949-955.
Microbiol. 50:1579-1589. Karlin, S., Brocchieri, L., Bergman, A., Mrazek,
Jaroszewski, L., Rychlewski, L., Li, Z., Li, W., and J., and Gentles, A.J. 2002. Amino acid runs in
Godzik, A. 2005. FFAS03: A server for profile– eukaryotic proteomes and disease associations.
profile sequence alignments. Nucleic Acids Res. Proc. Natl. Acad. Sci. U.S.A. 99:333-338.
33:W284-W288. Kataeva, I., Chang, J., Xu, H., Luan, C.H., Zhou,
Jenny, R.J., Mann, K.G., and Lundblad, R.L. 2003. J., Uversky, V.N., Lin, D., Horanyi, P., Liu, Z.J.,
A critical review of the methods for cleavage Ljungdahl, L.G., Rose, J., Luo, M., and Wang,
of fusion proteins with thrombin and factor Xa. B.C. 2005. Improving solubility of Shewanella
Protein Expr. Purif. 31:1-11. oneidensis MR-1 and Clostridium thermocellum
Jensen, P.Y., Bonander, N., Horn, N., Tumer, Z., and JW-20 proteins expressed into Esherichia coli.
Farver, O. 1999. Expression, purification and J. Proteome Res. 4:1942-1951.
copper-binding studies of the first metal-binding Kelker, M.S., Page, R., and Peti, W. 2009.
domain of Menkes protein. Eur. J. Biochem. Crystal structures of protein phosphatase-1 Production of
264:890-896. bound to nodularin-R and tautomycin: A novel Recombinant
Proteins

5.24.25
Current Protocols in Protein Science Supplement 61
scaffold for structure-based drug design of ser- protein, BSPH1. Mol. Hum. Reprod. 15:105-
ine/threonine phosphatase inhibitors. J. Mol. 114.
Biol. 385:11-21. Lefebvre, J., Boileau, G., and Manjunath, P. 2009b.
Kikuchi, Y., Yoda, K., Yamasaki, M., and Tamura, Recombinant expression and affinity purifica-
G. 1981. The nucleotide-sequence of the pro- tion of a novel epididymal human sperm-binding
moter and the amino-terminal region of alkaline- protein, BSPH1. Mol. Hum. Reprod. 15:105-
phosphatase structural gene (Phoa) of Es- 114.
cherichia coli. Nucleic Acids Res. 9:5671-5678. Leichert, L.I. and Jakob, U. 2004. Protein thiol
modifications visualized in vivo. PLoS Biol.
Kishore, U., Leigh, L.E.A., Eggleton, P., Strong, 2:e333.
P., Perdikoulis, M.V., Willis, A.C., and Reid, Lopez, P.J., Marchand, I., Joyce, S.A., and Dreyfus,
K.B.M. 1998. Functional characterization of a M. 1999. The C-terminal half of RNase E, which
recombinant form of the C-terminal, globular organizes the Escherichia coli degradosome,
head region of the B-chain of human serum com- participates in mRNA degradation but not rRNA
plement protein, C1q. Biochem. J. 333:27-32. processing in vivo. Mol. Microbiol. 33:188-199.
Klock, H.E., Koesema, E.J., Knuth, M.W., and Lorimer, G.H. 1996. A quantitative assessment of
Lesley, S.A. 2008. Combining the polymerase the role of the chaperonin proteins in protein
incomplete primer extension method for cloning folding in vivo. FASEB J. 10:5-9.
and mutagenesis with microscreening to ac-
celerate structural genomics efforts. Proteins Malakhov, M.P., Mattern, M.R., Malakhova, O.A.,
71:982-994. Drinker, M., Weeks, S.D., and Butt, T.R. 2004.
SUMO fusions and SUMO-specific protease for
Kohl, T., Schmidt, C., Wiemann, S., Poustka, A., efficient expression and purification of proteins.
and Korf, U. 2008. Automated production of re- J. Struct. Funct. Genomics 5:75-86.
combinant human proteins as resource for pro-
teome research. Proteome Sci. 6:10. Malik, A., Jenzsch, M., Lubbert, A., Rudolph, R.,
and Sohling, B. 2007. Periplasmic production
Kyratsous, C.A., Silverstein, S.J., DeLong, C.R., of native human proinsulin as a fusion to E. coli
and Panagiotidis, C.A. 2009. Chaperone-fusion ecotin. Protein Expr. Purif. 55:100-111.
expression plasmid vectors for improved sol-
ubility of recombinant proteins in Escherichia Milisavljevic, M.D., Papic, D.R., Timotijevic, G.S.,
coli. Gene 440:9-15. and Maksimovic, V.R. 2009. Successful pro-
duction of recombinant buckwheat cysteine-rich
Langlais, C., Guilleaume, B., Wermke, N., aspartic protease in Escherichia coli. J. Serb.
Scheuermann, T., Ebert, L., LaBaer, J., and Chem. Soc. 74:607-618.
Korn, B. 2007. A systematic approach for test-
ing expression of human full-length proteins in Miyada, C.G., Stoltzfus, L., and Wilcox, G. 1984.
cell-free expression systems. BMC Biotechnol. Regulation of the arac gene of Escherichia coli
7:64. - Catabolite repression, auto-regulation, and ef-
fect on arabad expression. Proc. Natl. Acad. Sci.
Lauber, T., Marx, U.C., Schulz, A., Kreutzmann, U.S.A. 81:4120-4124.
P., Rosch, P., and Hoffmann, S. 2001. Accurate
disulfide formation in Escherichia coli: Overex- Miyake, T., Oka, T., Nishizawa, T., Misoka, F.,
pression and characterization of the first domain Fuwa, T., Yoda, K., Yamasaki, M., and Tamura,
(HF6478) of the multiple Kazal-type inhibitor G. 1985. Secretion of human interferon-
LEKTI. Protein Expr. Purif. 22:108-112. alpha induced by using secretion vectors con-
taining a promoter and signal sequence of
LaVallie, E.R., DiBlasio, E.A., Kovacic, S., Grant, alkaline-phosphatase gene of Escherichia coli.
K.L., Schendel, P.F., and McCoy, J.M. 1993. J. Biochem. 97:1429-1436.
A thioredoxin gene fusion expression system
that circumvents inclusion body formation in the Miyashita, K., Kusumi, M., Utsumi, R., Komano,
E. coli cytoplasm. Biotechnology 11:187-193. T., and Satoh, N. 1992. Expression and
purification of recombinant 3c-proteinase
Lee, C.D., Sun, H.C., Hu, S.M., Chiu, C.F., of coxsackievirus-B3. Biosci. Biotechnol.
Homhuan, A., Liang, S.M., Leng, C.H., and Biochem. 56:746-750.
Wang, T.F. 2008. An improved SUMO fusion
protein system for effective production of native Mobley, C.K., Myers, J.K., Hadziselimovic, A., El-
proteins. Protein Sci. 17:1241-1248. lis, C.D., and Sanders, C.R. 2007. Purification
and initiation of structural characterization of
Lee, H.S., Berger, D.K., and Kustu, S. 1993. Activ- human peripheral myelin protein 22, an inte-
ity of purified nifa, a transcriptional activator of gral membrane protein linked to peripheral neu-
nitrogen-fixation genes. Proc. Natl. Acad. Sci. ropathies. Biochemistry 46:11185-11195.
U.S.A. 90:2266-2270.
Moffatt, B.A. and Studier, F.W. 1987. T7 lysozyme
Lee, N., Francklyn, C., and Hamilton, E.P. 1987. inhibits transcription by T7 rna-polymerase.
Arabinose-induced binding of arac protein to Cell 49:221-227.
aral2 activates the arabad operon promoter.
Proc. Natl. Acad. Sci. U.S.A. 84:8814-8818. Mohanty, A.K. and Wiener, M.C. 2004. Membrane
protein expression and production: Effects of
Optimizing Lefebvre, J., Boileau, G., and Manjunath, P. 2009a. polyhistidine tag length and position. Protein
Protein Recombinant expression and affinity purifica- Expr. Purif. 33:311-325.
Expression in tion of a novel epididymal human sperm-binding
E. coli

5.24.26
Supplement 61 Current Protocols in Protein Science
Mohanty, A.K., Simmons, C.R., and Wiener, M.C. Park, S.L., Kwon, M.J., Kim, S.K., and Nam, S.W.
2003. Inhibition of tobacco etch virus pro- 2004. GroEL/ES chaperone and low culture
tease activity by detergents. Protein Expr. Purif. temperature synergistically enhanced the solu-
27:109-114. ble expression of CGTase in E-coli. J. Microbiol.
Mustelin, T., Tautz, L., and Page, R. 2005. Struc- Biotechnol. 14:216-219.
ture of the hematopoietic tyrosine phosphatase Pedersen, K., Zavialov, A.V., Pavlov, M.Y., Elf, J.,
(HePTP) catalytic domain: Structure of a KIM Gerdes, K., and Ehrenberg, M. 2003. The bacte-
phosphatase with phosphate bound at the active rial toxin RelE displays codon-specific cleavage
site. J. Mol. Biol. 354:150-163. of mRNAs in the ribosomal A site. Cell 112:131-
Nagai, K. and Thogersen, H.C. 1984. Generation of 140.
beta-globin by sequence-specific proteolysis of Peng, L., Xu, Z.N., Fang, X.M., Wang, F., Yang, S.,
a hybrid protein produced in Escherichia coli. and Cen, P.L. 2004. Preferential codons enhanc-
Nature 309:810-812. ing the expression level of human beta-defensin-
Nallamsetty, S. and Waugh, D.S. 2006. Solubility- 2 in recombinant Escherichia coli. Protein Pept.
enhancing proteins MBP and NusA play a pas- Lett. 11:339-344.
sive role in the folding of their fusion partners. Peti, W. and Page, R. 2007. Strategies to maximize
Protein Expr. Purif. 45:175-182. heterologous protein expression in Escherichia
Nallamsetty, S. and Waugh, D.S. 2007. Muta- coli with minimal cost. Protein Expr. Purif. 51:1-
tions that alter the equilibrium between open 10.
and closed conformations of Escherichia coli Phan, J., Zdanov, A., Evdokimov, A.G., Tropea,
maltose-binding protein impede its ability to J.E., Peters, H.K., Kapust, R.B., Li, M.,
enhance the solubility of passenger proteins. Wlodawer, A., and Waugh, D.S. 2002. Struc-
Biochem. Biophys. Res. Commun. 364:639-644. tural basis for the substrate specificity of tobacco
Netzer, W.J. and Hartl, F.U. 1997. Recombi- etch virus protease. J. Biol. Chem. 277:50564-
nation of protein domains facilitated by co- 50572.
translational folding in eukaryotes. Nature Phillips, T.A., Vanbogelen, R.A., and Neidhardt,
388:343-349. F.C. 1984. Ion gene-product of Escherichia coli
Niiranen, L., Espelid, S., Karlsen, C.R., Mustonen, is a heat-shock protein. J. Bacteriol. 159:283-
M., Paulsen, S.M., Heikinheimo, P., and 287.
Willassen, N.P. 2007. Comparative expression Pinsach, J., de Mas, C., Lopez-Santin, J., Striedner,
study to increase the solubility of cold adapted G., and Bayer, K. 2008. Influence of process
Vibrio proteins in Escherichia coli. Protein Expr. temperature on recombinant enzyme activity
Purif. 52:210-218. in Escherichia coli fed-batch Cultures. Enzyme
Nilsson, B., Moks, T., Jansson, B., Abrahmsen, Microb. Technol. 43:507-512.
L., Elmblad, A., Holmgren, E., Henrichson, C., Piserchio, A., Ghose, R., and Cowburn, D. 2009.
Jones, T.A., and Uhlen, M. 1987. A synthetic Optimized bacterial expression and purification
igg-binding domain based on staphylococcal of the c-Src catalytic domain for solution NMR
protein-A. Protein Eng. 1:107-113. studies. J. Biomol. NMR 44:87-93.
Nilsson, J., Larsson, M., Stahl, S., Nygren, P.A., and Porath, J., Carlsson, J., Olsson, I., and Belfrage, G.
Uhlen, M. 1996. Multiple affinity domains for 1975. Metal chelate affinity chromatography, a
the detection, purification and immobilization of new approach to protein fractionation. Nature
recombinant proteins. J. Mol. Recognit. 9:585- 258:598-599.
594. Prinz, W.A., Aslund, F., Holmgren, A., and
Oberg, K., Chrunyk, B.A., Wetzel, R., and Fink, Beckwith, J. 1997. The role of the thioredoxin
A.L. 1994. Native-like secondary structure and glutaredoxin pathways in reducing protein
in interleukin-1-beta inclusion-bodies by at- disulfide bonds in the Escherichia coli cyto-
tenuated total reflectance Ftir. Biochemistry plasm. J. Biol. Chem. 272:15661-15667.
33:2628-2634. Pryor, K.D. and Leiting, B. 1997. High-level ex-
Ohana, R.F., Enccell, L.P., Zhao, K., Simpson, D., pression of soluble protein in Escherichia coli
Slater, M.R., Urh, M., and Wood, K.V. 2009. using a His(6)-tag and maltose-binding-protein
HaloTag7: A genetically engineered tag that en- double-affinity fusion system. Protein Expr.
hances bacterial expression of soluble proteins Purif. 10:309-319.
and improves protein purification. Protein Expr. Qing, G., Ma, L.C., Khorchid, A., Swapna, G.V.,
Purif. 68:110-120. Mal, T.K., Takayama, M.M., Xia, B., Phadtare,
Olins, P.O. and Rangwala, S.H. 1990. Vector for S., Ke, H., Acton, T., Montelione, G.T., Ikura,
enhanced translation of foreign genes in Es- M., and Inouye, M. 2004. Cold-shock induced
cherichia coli. Methods Enzymol. 185:115-119. high-yield protein production in Escherichia
coli. Nat. Biotechnol. 22:877-882.
Otto, C.M., Niagro, F., Su, X.Z., and Rawlings, C.A. Reilly, D. and Fairbrother, W.J. 1994. A novel
1995. Expression of recombinant feline tumor- isotope labeling protocol for bacterially ex-
necrosis-factor is toxic to Escherichia coli. Clin. pressed Proteins. J. Biomol. NMR 4:459-
Diagn. Lab. Immunol. 2:740-746. 462. Production of
Recombinant
Proteins

5.24.27
Current Protocols in Protein Science Supplement 61
Riggs, P. 2000. Expression and purification of re- Singleton, S.F., Simonette, R.A., Sharma, N.C., and
combinant proteins by fusion to maltose-binding Roca, A.I. 2002. Intein-mediated affinity-fusion
protein. Mol. Biotechnol. 15:51-63. purification of the Escherichia coli RecA pro-
Ritz, D. and Beckwith, J. 2001. Roles of thiol- tein. Protein Expr. Purif. 26:476-488.
redox pathways in bacteria. Annu. Rev. Micro- Skerra, A. 1994. Use of the tetracycline promoter
biol. 55:21-48. for the tightly regulated production of a murine
Rosenberg, M. and Court, D. 1979. Regulatory se- antibody fragment in Escherichia coli. Gene
quences involved in the promotion and termi- 151:131-135.
nation of rna-transcription. Annu. Rev. Genet. Smith, D.B. and Johnson, K.S. 1988. Single-
13:319-353. step purification of polypeptides expressed in
Routzahn, K. and Waugh, D. 2002. Differential ef- Escherichia coli as fusions with glutathione
fects of supplementary affinity tags on the solu- s-transferase. Gene 67:31-40.
bility of MBP fusion proteins. J. Struct. Funct. Spiess, C., Beil, A., and Ehrmann, M. 1999. A
Genomics 2:83-92. temperature-dependent switch from chaperone
Saavedraalanis, V.M., Rysavy, P., Rosenberg, L.E., to protease in a widely conserved heat shock
and Kalousek, F. 1994. Rat-liver mitochondrial protein. Cell 97:339-347.
processing peptidase - both alpha-subunit and Spirin, A.S. 2004. High-throughput cell-free sys-
beta-subunit are required for activity. J. Biol. tems for synthesis of functionally active pro-
Chem. 269:9284-9288. teins. Trends Biotechnol. 22:538-545.
Sahdev, S., Khattar, S.K., and Saini, K.S. 2008. Pro- Stewart, E.J., Aslund, F., and Beckwith, J. 1998.
duction of active eukaryotic proteins through Disulfide bond formation in the Escherichia
bacterial expression systems: A review of the coli cytoplasm: An in vivo role reversal for the
existing biotechnology strategies. Mol. Cell thioredoxins. EMBO J. 17:5543-5550.
Biochem. 307:249-264. Stieber, D., Gabant, P., and Szpirer, C. 2008. The
Sahu, S.K., Rajasekharan, A., and Gummadi, S.N. art of selective killing: Plasmid toxin/antitoxin
2009. GroES and GroEL are essential chaper- systems and their technological applications.
ones for refolding of recombinant human phos- Biotechniques 45:344-346.
pholipid scramblase 1 in E. coli. Biotechnol Lett. Studier, F.W. 1991. Use of bacteriophage-T7
31:1745-1752. lysozyme to improve an inducible T7 expres-
Sakharkar, M.K., Kangueane, P., Sakharkar, K.R., sion system. J. Mol. Biol. 219:37-44.
and Zhong, Z. 2006. Huge proteins in the human Studier, F.W. and Moffatt, B.A. 1986a. Use of
proteome and their participation in hereditary bacteriophage T7 RNA polymerase to direct se-
diseases. In Silico Biol. 6:275-279. lective high-level expression of cloned genes.
Sati, S.P., Singh, S.K., Kumar, N., and Sharma, A. J. Mol. Biol. 189:113-130.
2002. Extra terminal residues have a profound Studier, F.W. and Moffatt, B.A. 1986b. Use of
effect on the folding and solubility of a Plas- bacteriophage-T7 rna-polymerase to direct se-
modium falciparum sexual stage-specific pro- lective high-level expression of cloned genes.
tein over-expressed in Escherichia coli. Eur. J. J. Mol. Biol. 189:113-130.
Biochem. 269:5259-5263.
Studier, F.W., Rosenberg, A.H., Dunn, J.J., and
Schenk, P.M., Baumann, S., Mattes, R., and Dubendorff, J.W. 1990. Use of T7 rna-
Steinbiss, H.H. 1995. Improved high-level polymerase to direct expression of cloned genes.
expression system for eukaryotic genes in Methods Enzymol. 185:60-89.
Escherichia coli using T7 RNA polymerase and
rare ArgtRNAs. Biotechniques 19:196-200. Tanaka, T., Kubota, M., Samizo, K., Nakajima, Y.,
Hoshino, M., Kohno, T., and Wakamatsu, E.
Seeliger, M.A., Young, M., Henderson, M.N., 1999. One-step affinity purification of the G pro-
Pellicena, P., King, D.S., Falick, A.M., and tein beta gamma subunits from bovine brain us-
Kuriyan, J. 2005. High yield bacterial expres- ing a histidine-tagged G protein alpha subunit.
sion of active c-Abl and c-Src tyrosine kinases. Protein Expr. Purif. 15:207-212.
Protein Sci. 14:3135-3139.
Tegel, H., Steen, J., Konrad, A., Nikdin, H.,
Sharp, P.M. and Li, W.H. 1987. The codon Adapta- Pettersson, K., Stenvall, M., Tourle, S.,
tion Index–a measure of directional synonymous Wrethagen, U., Xu, L., Yderland, L., Uhlen,
codon usage bias, and its potential applications. M., Hober, S., and Ottosson, J. 2009. High-
Nucleic Acids Res. 15:1281-1295. throughput protein production–lessons from
Shirakawa, M., Tsurimoto, T., and Matsubara, scaling up from 10 to 288 recombinant proteins
K. 1984. Plasmid vectors designed for high- per week. Biotechnol. J. 4:51-57.
efficiency expression controlled by the portable Terpe, K. 2003. Overview of tag protein fusions:
reca promoter-operator of Escherichia coli. From molecular and biochemical fundamen-
Gene 28:127-132. tals to commercial systems. Appl. Microbiol.
Shirano, Y. and Shibata, D. 1990. Low tempera- Biotechnol. 60:523-533.
ture cultivation of Escherichia coli carrying a Tsumoto, K., Ejima, D., Kumagai, I., and Arakawa,
Optimizing rice lipoxygenase L-2 cDNA produces a soluble T. 2003. Practical considerations in refolding
Protein and active enzyme at a high level. FEBS Lett.
Expression in proteins from inclusion bodies. Protein Expr.
271:128-130. Purif. 28:1-8.
E. coli

5.24.28
Supplement 61 Current Protocols in Protein Science
Turner, P., Holst, O., and Karlsson, E.N. 2005. Op- human proinsulin in the periplasmic space of
timized expression of soluble cyclomaltodextri- Escherichia coli by fusion to DsbA. J. Biotech-
nase of thermophilic origin in Escherichia coli nol. 84:175-185.
by using a soluble fusion-tag and by tuning of in- Wittliff, J.L., Wenz, L.L., Dong, J., Nawaz, Z., and
ducer concentration. Protein Expr. Purif. 39:54- Butt, T.R. 1990. Expression and characteriza-
60. tion of an active human estrogen-receptor as a
Valax, P. and Georgiou, G. 1993. Molecular char- ubiquitin fusion protein from Escherichia coli.
acterization of beta-lactamase inclusion-bodies J. Biol. Chem. 265:22016-22022.
produced in Escherichia coli. 1. Composition. Xu, Y., Yasin, A., Tang, R., Scharer, J.M., Moo-
Biotechnol. Prog. 9:539-547. Young, M., and Chou, C.P. 2008. Heterologous
Vasina, J.A. and Baneyx, F. 1996. Recombinant pro- expression of lipase in Escherichia coli is lim-
tein expression at low temperatures under the ited by folding and disulfide bond formation.
transcriptional control of the major Escherichia Appl. Microbiol. Biotechnol. 81:79-87.
coli cold shock promoter cspA. Appl. Environ. Yan, F., Qian, M.L., Yang, F., Cai, F., Yuan, Z.,
Microbiol. 62:1444-1447. Lai, S.T., Zhao, X.Y., Gou, L.T., Hu, Z.G., and
Vasina, J.A. and Baneyx, F. 1997. Expression of Deng, H.X. 2007. A novel pro-apoptosis protein
aggregation-prone recombinant proteins at low PNAS-4 from Xenopus laevis: Cloning, expres-
temperatures: A comparative study of the Es- sion, purification, and polyclonal antibody pro-
cherichia coli cspA and tac promoter systems. duction. Biochemistry (Moscow) 72:664-671.
Protein Expr. Purif. 9:211-218. Yao, J.W., Patrone, J.D., and Dotson, G.D. 2009.
Veldkamp, C.T., Peterson, F.C., Hayes, P.L., Characterization and kinetics of phosphopan-
Mattmiller, J.E., Haugner, J.C., de la Cruz, N., tothenoylcysteine synthetase from Enterococcus
and Volkman, B.F. 2007. On-column refold- faecalis. Biochemistry 48:2799-2806.
ing of recombinant chemokines for NMR stud- Yeo, Y.J., Shin, S., Lee, S.G., Park, S., and Jeong,
ies and biological assays. Protein Expr. Purif. Y.J. 2009. Production, purification, and char-
52:202-209. acterization of soluble NADH-flavin Oxidore-
Vincentelli, R., Bignon, C., Gruez, A., Canaan, S., ductase (StyB) from Pseudomonas putida SN1.
Sulzenbacher, G., Tegoni, M., Campanacci, V., J. Microbiol. Biotechnol. 19:362-367.
and Cambillau, C. 2003. Medium-scale struc- Yin, J.C., Li, G.X., Ren, X.F., and Herrler, G. 2007.
tural genomics: Strategies for protein expres- Select what you need: A comparative evaluation
sion and crystallization. Accounts Chem. Res. of the advantages and limitations of frequently
36:165-172. used expression systems for foreign genes.
Volonte, F., Marinelli, F., Gastaldo, L., Sacchi, S., J. Biotechnol. 127:335-347.
Pilone, M.S., Pollegioni, L., and Molla, Zhang, Y.B., Howitt, J., McCorkle, S., Lawrence,
G. 2008. Optimization of glutaryl-7- P., Springer, K., and Freimuth, P. 2004. Pro-
aminocephalosporanic acid acylase expression tein aggregation during overexpression limited
in E-coli. Protein Expr. Purif. 61:131-137. by peptide extensions with large net negative
Wakagi, T., Oshima, T., Imamura, H., and charge. Protein Expr. Purif. 36:207-216.
Matsuzawa, H. 1998. Cloning of the gene for Zhang, Z.W., Gildersleeve, J., Yang, Y.Y., Xu, R.,
inorganic pyrophosphatase from a thermoaci- Loo, J.A., Uryu, S., Wong, C.H., and Schultz,
dophilic archaeon, Sulfolobus sp. strain 7, and P.G. 2004. A new strategy for the synthesis of
overproduction of the enzyme by coexpres- glycoproteins. Science 303:371-373.
sion of tRNA for arginine rare codon. Biosci.
Biotechnol. Biochem. 62:2408-2414. Zhao, Y.X., Benita, Y., Lok, M., Kuipers, B., van der
Ley, P., Jiskoot, W., Hennink, W.E., Crommelin,
Walsh, G. 2006. Biopharmaceutical benchmarks D.J.A., and Oosting, R.S. 2005. Multi-antigen
2006. Nature Biotechnol. 24:769-765. immunization using IgG binding domain ZZ as
Wang, W.R., Marimuthu, A., Tsai, J., Kumar, A., carrier. Vaccine 23:5082-5090.
Krupka, H.I., Zhang, C., Powell, B., Suzuki, Y., Zuo, X., Li, S., Hall, J., Mattern, M.R., Tran, H.,
Nguyen, H., Tabrizizad, M., Luu, C., and West, Shoo, J., Tan, R., Weiss, S.R., and Butt, T.R.
B.L. 2006. Structural characterization of autoin- 2005a. Enhanced expression and purification
hibited c-Met kinase produced by coexpression of membrane proteins by SUMO fusion in Es-
in bacteria with phosphatase. Proc. Natl. Acad. cherichia coli. J. Struct. Funct. Genomics 6:103-
Sci. U.S.A. 103:3563-3568. 111.
Wang, Y.H., Ayrapetov, M.K., Lin, X.F., and Sun, Zuo, X., Mattern, M.R., Tan, R., Li, S., Hall,
G.Q. 2006. A new strategy to produce active hu- J., Sterner, D.E., Shoo, J., Tran, H., Lim, P.,
man Src from bacteria for biochemical study of Sarafianos, S.G., Kazi, L., Navas-Martin, S.,
its regulation. Biochem. Biophys. Res. Commun. Weiss, S.R., and Butt, T.R. 2005b. Expres-
346:606-611. sion and purification of SARS coronavirus pro-
Winter, J., Neubauer, P., Glockshuber, R., and teins using SUMO-fusions. Protein Expr. Purif.
Rudolph, R. 2000. Increased production of 42:100-110.

Production of
Recombinant
Proteins

5.24.29
Current Protocols in Protein Science Supplement 61

You might also like