Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Available online at www.sciencedirect.

com

Protein Expression and Purification 59 (2008) 55–63


www.elsevier.com/locate/yprep

A step-wise approach significantly enhances protein


yield of a rationally-designed agonist antibody fragment in E. coli
Bing Lin, Mark W. Renshaw, Kathleen Autote, Lynette M. Smith, Peter Calveley,
Katherine S. Bowdish, Shana Frederickson *
Alexion Antibody Technologies, Inc., 3985 Sorrento Valley Boulevard, Suite A, San Diego, CA 92121, USA

Received 7 August 2007, and in revised form 21 December 2007


Available online 17 January 2008

Abstract

Fab59 is a rationally-designed antibody fragment (Fab) that mimics the activity of the cytokine thrombopoietin (TPO). Fab59 activity
was initially detected directly from bacterial supernatants in a cell-based assay and was subsequently estimated to be equipotent to TPO
using purified material. However, the expression of Fab59 was insufficient to support in vivo characterization of the Fab due to extremely
low expression levels from its initial phage display expression vector. To boost expression, a new expression vector was designed and
constructed, and Fab59 light chain codons were optimized for bacterial expression. However, from this a new challenge arose, in that
the codon-optimized Fab59 was more toxic to Escherichia coli cells than parental Fab59. Co-expression of the bacterial chaperon protein
Skp alleviated this toxicity. A two-step purification method was used to isolate monomeric Fab59 from the periplasm. Although Fab59
was prone to form aggregates during the purification process, buffer modification efficiently eliminated this problem. Overall, optimiza-
tion of Fab59 expression and purification achieved a 100-fold increase in Fab59 production in E. coli relative to the starting yield. The
yield of purified monomeric Fab59 from a shake flask reached up to 3.5 mg/L, which was sufficient to support testing of the agonist
activity of purified monomeric Fab59 in vivo. Even higher yields may be achieved by purification of Fab present in the culture media,
as Skp most significantly increased accumulation of Fab59 in that location.
Ó 2008 Elsevier Inc. All rights reserved.

Keywords: Antibody; Agonist

Fab59 is a rationally-designed human antibody frag- supernatants or Fab purified in small scale demonstrated
ment that was created to mimic thrombopoietin (TPO)1 that Fab59 had activity equivalent to TPO in vitro. How-
and provide a possible mode of therapeutic intervention ever, early attempts to produce and purify Fab59 in larger
for thrombocytopenia, while avoiding the risk of generat- scale were hampered by poor expression levels in Esche-
ing neutralizing antibodies to endogenous TPO. Fab59 richia coli. Initial yields of purified Fab59 protein were
was engineered by grafting a peptide with cMpl receptor insufficient to support in vivo modeling of Fab59 activity,
(cMpl-R) binding capability [1] into the CDR regions of thereby hindering both further characterization and its
both the heavy and light chains of a well-characterized fully potential as a commercial drug candidate. In order to meet
human anti-tetanus toxoid Fab scaffold (Fab TT) [2]. Ini- the protein level requirements for in vivo modeling and
tial cell-based assays using unpurified Fab in bacterial increase viability as a potential commercial therapeutic
candidate, production levels of Fab59 needed to be dra-
matically increased. Efficient expression of well-folded,
*
Corresponding author. functional antibody fragments can be a challenge in com-
E-mail address: sfrederickson@alxnsd.com (S. Frederickson).
1
Abbreviations used: TPO, thrombopoietin; cMpl-R, cMpl receptor;
parison to other recombinant proteins expressed in
Fab TT, anti-tetanus toxoid Fab; RBS, ribosomal binding site; BCA, E. coli [3]. Disulfide bond formation and assembly of Fab
bicinchoninic acid; TBS, tris-buffered saline. light and heavy chains generally require the oxidized envi-

1046-5928/$ - see front matter Ó 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.pep.2008.01.002
56 B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63

ronment found in the periplasmic space of E. coli, although PCR amplification. The HA-epitope tag and the His puri-
some Fabs can be expressed in the cytoplasm of specialized fication tag were arranged so that they would be expressed
E. coli strains [4]. Furthermore, use of a rational design on the C-terminal end of the Fab’s heavy chain.
strategy, increasingly used for new therapeutic approaches, For sub-cloning of the Fab genes into the new vectors,
adds a new layer of complexity. While CDR regions nor- 1.5 kb cDNA encoding the Fab light chain and heavy chain
mally encompass a higher degree of diversity than the was released from the vector pRL4 by double digestion
remaining antibody sequence, and thus may be a more with EcoRI/NheI, and then ligated into the expression vec-
accepting region for peptide grafts, expression and proper tor pAEV1 pre-digested with EcoRI/NheI. For construc-
folding of the engineered protein may be compromised. tion of pAEV1-HCG, a 670 bp light chain fragment was
Nonetheless, expression in E. coli still provides a conve- recovered from pRL4-TT by double digestion with SacI/
nient, fast and cost-efficient way to generate material for XbaI, and then ligated into the pAEV1-59 double digested
research and therapeutic purposes. with the same restriction enzymes. For the construction
Like other recombinant proteins, which are difficult to pAEV1-LCG, an 870 bp heavy chain fragment was recov-
express, the poor expression of Fab59 could be due to ered from pRL4-TT by double digestion with XhoI/NheI,
improper folding, insolubility, intracellular degradation and then ligated into pAEV1-59 vector double digested
or toxicity to the E. coli cells [5,6]. We therefore initiated with the same restriction enzymes. These constructs are
a step-wise process to first identify which factors were listed in Fig. 1.
rate-limiting to Fab59 production in E. coli and then deter-
mine how to modify them accordingly to effect an increase Codon optimization
in protein accumulation and yield. We found that poor
expression of Fab59 was specific to the light chain, due The codon-optimized synthetic light chain gene was
to both extremely low LC expression levels and insolubility designed based on the codon preference of highly expressed
of the light chain. Therefore, our expression optimization genes in E. coli [7]. A synthetic light chain was assembled
of Fab59 was mainly focused on increasing the light chain from 52 oligonucleotides (35–45 bases each) with 15–20
expression level and improving solubility. This was system- overlapping complementarily bases (Operon Inc.). Oligos
atically accomplished by first codon optimization, second, were phosphorylated, annealed and ligated into a full-
using a tightly controlled strong promoter, third low tem- length fragment. The full-length synthetic light chain was
perature and low levels of IPTG induction, and fourth then PCR amplified. The PCR products were digested by
co-expression of a bacterial chaperon protein Skp. Further- XbaI/NotI and ligated back to pAEV1-59 pre-digested
more, the purification process was optimized to efficiently with XbaI/NotI.
avoid protein aggregation. After optimization of expres-
sion and purification, the yield of Fab59 was increased Bacterial chaperon protein Skp cloning
approximately 100-fold. This level was sufficient to allow
for the successful purification and isolation of enough The PCR primer pairs used for PCR amplifying the Skp
Fab59 to support preclinical development. Highly pure gene from bacterial genomic DNA were designed based on
monomeric and correctly folded Fab59 with intact disulfide
bonds was isolated from bacterial periplasmic extracts TPO TPO
pRL4-59 P lac Omp LC Pel HC
using a two-step purification method. The agonist activity
TPO TPO
of the purified monomeric Fab59 was then demonstrated Omp LC Pel HC
pAEV1-59 P T7
to be equipotent to rhTPO in vitro in cMpl-R signaling,
luciferase reporter-based assays, and furthermore stimu- P T7 Omp LC Pel HC
pAEV1-TT
TPO
lated increased platelet levels in vivo [2]. Thus, this broadly Omp LC Pel HC
pAEV1-LCG P T7
applicable systematic approach allowed us to overcome the TPO
existing bottlenecks in Fab59 protein production in E. coli pAEV1-HCG P T7
Omp LC Pel HC
TPO TPO
that occurred in our initial expression system, as well as to Omp LC Pel HC
pAEV1-O59 P T7
solve new problems that developed as a result of increased TPO TPO
accumulated protein levels. pAEV1-O59Skp P T7 Omp LC Pel HC Skp

Fig. 1. Schematic diagram of the expression cassettes and constructs


Materials and methods
evaluated in our studies. Omp: OmpA leader sequence. Pel: PelB leader
sequence. LC: light chain (h). HC: heavy chain ( ). TPO :TPO mimetic
Expression vector construction peptide with selected flanking residues (j). Skp: E. coli 17 KDa
periplasmic chaperon protein Skp ( ). Plac: lac promoter. PT7: T7/lac
T7-based expression vector pAEV1 was developed by promoter. pAEV1-O59: codon-optimized light chain ( ) Fab59 con-
structs. pAEV1-TT: fully human Fab TT. In all cases where TPO is shown
modifying the pET28a vector (Novagen) so that lacI was
the TPO mimetic peptide cassette was grafted into either the CDR3 region
replaced by lacIq. In addition a ribosomal binding site in the heavy chain (pAEV1-HCG) or into the CDR2 region in the light
(RBS) and poly-linker in pET28a were replaced by the chain (pAEV1-LCG) or both in the heavy chain and in the light chain
OmpA RBS and an EcoRI/NheI/HA-His/SacII linker by (Fab59 constructs).
B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63 57

the bacterial Skp DNA sequence in GenBank (GenBank SB medium in a 3-L shaker flask. When bacterial culture
Accession No. M21118). The 50 primer was flanked with OD600 reached 0.7, IPTG was added to a final concentra-
NdeI: gaaacatatgaaaaagtggttattagctgcag, and the 30 primer tion of 0.02 mM and temperature was changed to 22 °C
was flanked with XhoI: gaaactcgagttatttaacctgtttcagatcgtc. in a 120 rpm shaker. After 16–20 h of induction, the bacte-
To PCR amplify Skp, a freshly grown Top10F’ cell colony rial pellet was collected for Fab purification.
on a LB plate was inoculated into a 50 ll PCR mixture (1
TaqPlus buffer, 0.1 mM dNTP, 1 pmol primers, 2.5 U Taq- Fab59 purification
Plus, Stratagene). The PCR reaction was performed on a
Perkin Elmer PCR machine with 95 °C 5000 –62 °C 5000 – Bacterial pellets were suspended in 50 ml binding buffer
72 °C 4000 for 25 cycles. PCR amplified products were (1 PBS buffer, or HEPES buffer (100 mM HEPES, 0.5 M
digested with NdeI/XhoI and ligated into the pAEV1-59 NaCl, 10% glycerol, 0.02% Tween-80)). After gentle soni-
vector digested with the same enzymes. Skp DNA sequence cation, samples were cleared by centrifugation at
was confirmed by sequencing. 15,000 rpm at 4 °C for 15 min. The supernatant was col-
lected and filtered through 0.8 lm and then 0.2 lm filtra-
Small-scale Fab expression tion units (NALGENEÒ, Apogene Technologies). Fab59
contained in the supernatant was then bound to an anti-
Overnight starter cultures were made by inoculating sin- human Fab affinity column pre-equilibrated with binding
gle colonies from a LB plate into 2 ml cultures of SB med- buffer on a FPLC (ÃKTA, Amersham Biosciences).
ium with the appropriate antibiotics and 2% glucose in a Fab59 was then eluted with elution buffer (0.2 M glycine
37 °C shaker. One hundred microliters of the overnight buffer, pH 2.2, or a buffer with 0.2 M glycine–HCl, 10%
starter cultures was inoculated into 2 ml of SB medium glycerol, 0.02% Tween-80, pH 2.7), and immediately neu-
(antibiotics, 2% glucose). The bacterial cultures were incu- tralized to pH 7.0 with neutralization buffer (1 M Tris–
bated in a 37 °C shaker until OD600 reached 0.5–0.9. Then HCl, 4 M NaCl, pH 9.0). The neutralized elute was dia-
IPTG was added for induction and the shaker temperature lyzed against TBS buffer (20 mM Tris–HCl, pH 7.4,
was changed to 30 °C. Four hours after induction, the bac- 0.15 M NaCl) or HEPES buffer overnight. After dialysis,
terial culture medium supernatants and pellets were col- the sample was concentrated down to approximately 1 ml
lected for Western blot. Bacterial pellets were suspended and loaded onto a Superdex75 (Amersham Biosciences)
in 1 PBS buffer and sonicated gently on ice using a power SEC column (10 cm  110 cm) pre-equilibrated with the
setting of 35%, with pauses at 10 s intervals (for three inter- dialysis buffer. Size exclusion chromatography was per-
vals) in order to release most of the proteins from the bac- formed on the FPLC with a flow rate of 0.4 ml/min. Mono-
terial periplasm while avoiding breaking open the cells as meric Fab59 fractions were collected. Bacterial endotoxins
monitored by release of DNA into the buffer. The condi- were removed from Fab59 by passing through a Sarto-
tions were established in pilot experiments using side-by- bindÒ Membrane Adsorber Q15 (Sartorius). The Fab59
side comparisons with the conventional osmotic shock sample was then concentrated using a Centriprep YM-30
method of releasing periplasmic proteins [8]. To isolate (Millipore Corp.) and filter-sterilized by MinisartÒ-Plus
total soluble proteins (cytoplasmic and periplasmic), bacte- (Sartorius) syringe tip filter.
rial pellets in PBS buffer were sonicated heavily on ice using The Ni-NTA agarose resin used for His-tag purification
a power setting of 45% and pauses at 20 s intervals for six was purchased from Invitrogen, and the process for the
intervals, followed by recovery of the supernate after spin- His-tag purification was based on the protocol provided
ning down the sonicated mixture. To isolate insoluble pro- with the kit. Protein concentration was determined by
teins, the bacterial pellets from overnight growth were using the bicinchoninic acid (BCA) protein assay reagent
heavily sonicated as above. After spinning down the soni- (Bio-Rad).
cated mixture, the insoluble proteins were recovered from
the pellet following three PBS washes. SDS–PAGE and Western blot

Overnight induction at room temperature Protein samples were combined with SDS–PAGE load-
ing buffer (with or without DTT) and heated at 100 °C for
A single colony was inoculated into 2 ml of SB medium, 10 min. For analysis of protein in the culture media or the
and cultured in a 37 °C shaker for OD600 to reach 0.7. periplasmic space, the samples were normalized to 10 ll of
IPTG was added to a final concentration of 0.02 mM, the culture volume (for Figs. 2, 3A, 5 and 6). For analysis
and the culture temperature was changed to 22 °C in a of total cellular protein (either the soluble or insoluble frac-
120 rpm shaker. The induction was continued overnight tion from sonication of the bacterial pellet), the samples
(16–20 h). were normalized by loading 10 lg of protein (Figs. 3B, C
To make 1 L of bacterial culture for Fab purification, and 4). The samples were resolved on a precast 4%–15%
one colony was inoculated into 100 ml SB medium and Gradient Ready Gel (Bio-Rad) following manufacturer’s
then cultured overnight in a 30 °C shaker. The next day, protocol. Proteins were then transferred onto a nitrocellu-
100 ml of the overnight culture was inoculated into 1-L lose membrane (Bio-Rad) using a Trans-Blot SD Semi-Dry
58 B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63

9
9

5
-5

-59
-5

-O
1

L4
EV

1
C

EV

EV
pR
pA

pA

pA
72K
A 43K
43K 25K

M
M

M
m

1m
M

1m

0m
05
1m

0 .5

0 .0
0.

0.

Fig. 4. Immunoblotting analysis demonstrating insoluble light chain


72K accumulation in E. coli total cell lysis. Insoluble protein samples were
prepared by small-scale Fab expression as described in Materials and
B 43K
methods. pAEV1-O59 contains a peptide grafted synthetic codon
optimized light chain. pAEV1-59 contains a peptide grafted wild-type
light chain. SDS–PAGE was performed under reducing conditions. The
Western blot was probed with an anti-human kappa light chain
Fig. 2. Immunoblotting analysis of Fab59 expression in E. coli periplasm. monoclonal antibody.
Periplasmic samples were prepared by small-scale Fab expression as
described in Materials and methods. Both gels were run under non-
reducing SDS–PAGE conditions and blots were probed with anti-human
light chain monoclonal antibody. (A) Comparing expression levels of Transfer Cell (Bio-Rad) according to manufacture’s proto-
Fab59 from two different expression vectors in two different hosts. Lane 1: col. The membranes were blocked overnight at 4 °C in 5%
pAEV1-59 was expressed in BL21(DE)StarTM cells and induced with 1 mM
skim milk powder in Tris-buffered saline (TBS). Anti-HA
IPTG. Lane 2: pRL4-59 was expressed in Top10F0 cells and induced with
1 mM IPTG. Lane 3: is a control (C) of pAEV1-59 in BL21(DE)StarTM monoclonal antibody (Sigma, 1:2000 dilution) was used
with no IPTG induction. (B) Evaluating expression levels of the construct to detect the heavy chain and anti-human light chain poly-
pAEV1-59 in E. coli periplasm under different IPTG induction concen- clonal antibody (Pierce, 1:5000) was used for light chain
trations in BL21(DE)StarTM cells. detection. After washing in TBS/0.1% Tween-20 (TBST)
three times (10 min each), the blots were probed with the
alkaline-phosphate conjugated goat anti-mouse IgG
G
CG

HC

(Pierce, 1:30,000 dilution). The blots were visualized by


T
59

-L

-T
1-
1-

using NBT–BCIP reagents (Pierce) following the provided


1
EV
EV
EV

EV
pA
pA

pA
pA

protocol.
72K
Luciferase assays
A
43K
The method for detecting agonist activity of Fab59
using a luciferase assay was described previously [2].
Briefly, NIH3T3 cells were co-transfected with either a con-
43K trol vector (pcDNA3.1, Invitrogen, Carlsbad, CA) or the
B 25K cMpl-R expressed in pcDNA3.1 and the Fos promoter/
luciferase reporter construct [2]. Co-transfections of 3T3
cells were performed by plating NIH 3T3 cells at 3  105
cells per 6 cm dish and then transfecting the following
43K day. NIH 3T3 cells were transfected using the Effectene lip-
C 25K ofection reagent (Qiagen), transfecting each plate with
0.1 lg pEGFP (Clonetech), 0.2 lg of the Fos promoter/
luciferase construct, 0.2 lg of pAdVAntage, and 0.5 lg of
either the empty control vector pcDNA3.1 (Invitrogen)
or the plasmid expressing the cMpl-R. 3T3 cells were
Fig. 3. Comparison of soluble Fab, light chain and heavy chain placed in 0.5% serum 24 h post transfection and incubated
accumulation for Fab59 versus singly grafted Fabs pAEV1-LCG and
pAEV1-HCG, and parental TT in E. coli. Protein samples were prepared
for an additional 24 h in this low serum media to serum
by small-scale Fab expression as described in Materials and methods. (A) starve the cells in order to reduce the background activa-
Fab expression for different constructs in E. coli periplasm. A non- tion of the Fos promoter. Fab59 samples and rhTPO
reducing gel was probed with an anti-human light chain monoclonal (inhouse purified) [2] were then applied to these cells for
antibody. (B) Soluble light chain expression in different constructs in
6 h. Cells were harvested and luciferase assays performed
E. coli. A reducing gel was probed with an anti-human light chain
monoclonal antibody. (C) Soluble heavy chain expression in different
using 50 lg of cell lysate using a Turner Designs TD20/
constructs in E. coli. A reducing gel was probed with an anti-HA 20 luminometer and beetle luciferin (Promega) according
monoclonal antibody. to manufacturer’s protocol.
B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63 59

Results tern blot (Fig. 3) of the reducing SDS–PAGE gel was used
to detect the total soluble light chain (Fig. 3B) and heavy
Fab59 expression is increased with the T7 promoter and chain (Fig. 3C) expression levels in E. coli. As shown in
affected by IPTG induction conditions Fig. 3C, there were no significant differences in heavy chain
expression levels in all the constructs, while the expression
The original expression vector containing Fab59, pRL4- levels of grafted light chain were very low in both con-
59, is a modified phage display vector with a lac promoter structs pAEV1-LCG and pAEV1-59, demonstrating that
(Fig. 1). To enhance transcription of Fab mRNA, we trans- poor expression of Fab59 resulted from reduced produc-
ferred the Fab genes to expression vector pAEV1, that car- tion of the grafted light chain in E. coli.
ries the T7 promoter, designated pAEV1-59. In order to
minimize the basal level leakage from the T7 promoter, Codon optimization of the Fab59 light chain increased
we added glucose to a final concentration of 2% in the bac- insoluble light chain accumulation in E. coli
terial culture medium. Induction, where indicated, used
1 mM IPTG. As shown in Fig. 2A, Fab59 was expressed Codon optimization has been widely used to achieve
well in BL21(DE)StarTM cells in the construct pAEV1-59, higher expression levels in heterologous recombinant pro-
while Fab59 expression in the original construct pRL4-59 tein expression systems [11–13]. To determine if the optimi-
in Top10F’ cells was barely detected. zation of codon usage to remove codons that are rarely
As demonstrated in Fig. 2B, IPTG greatly affected the used in E. coli would further improve Fab59 light chain
bacterial growth and recombinant protein expression in production, we optimized the grafted light chain gene
E. coli. Low IPTG (0.1–0.5 mM) induction has been used based on the codon usage of highly expressed genes in
to improve Fab expression in several previously reported E. coli. The expression level of the construct with the light
cases [9,10]. Here, we evaluated the expression level of chain codon optimized, pAEV1-O59, was tested by Wes-
pAEV1-59 with differing IPTG induction concentrations, tern blot. Surprisingly, Western blot analysis demonstrated
using 0.01, 0.05, 0.1, 0.5, and 1 mM IPTG. We found that that the construct pAEV1-O59 did not have significantly
IPTG induction concentrations higher than 0.1 mM inhib- improved Fab expression levels (data not shown). Impor-
ited the bacterial growth and resulted in lower expression tantly, we noticed that the growth rate of bacteria contain-
levels while lower concentrations of IPTG had little affect ing pAEV1-O59 was retarded and slowed significantly after
on bacterial growth, and induction at 0.05 mM resulted addition of IPTG, suggesting that codon optimization of
in a higher expression level of Fab59 as detected by Wes- the light chain had a growth inhibitory or toxic effect on
tern blot (Fig. 2B). At a 0.05 mM IPTG induction, we the E. coli cells not observed with the parental pAEV1-
obtained the highest expression level both in shaking cul- Fab59. As shown in Fig. 4, pAEV1-O59 contains much
tures and in bioreactor fermentation (data not shown), more insoluble light chain accumulation than the wild-type
demonstrating that IPTG induction concentration greatly pAEV1-59. These results suggested that though we success-
affected Fab59 expression. fully increased expression of the light chain, this led to
increased accumulation of an insoluble form of grafted
Reduced expression of Fab59 is light chain-specific light chain which was toxic to E. coli, and created addi-
tional issues to address.
Fab59 is a rationally-designed human antibody frag-
ment that was engineered by grafting a peptide with cMpl Co-expression of the bacterial chaperon protein Skp reduced
receptor (cMpl-R) binding capability [1] into the CDR pAEV1-O59 toxicity and increased Fab59 production and
regions of both the heavy and light chains of a well-charac- secretion into the culture medium
terized fully human anti-tetanus toxoid Fab scaffold (Fab
TT). Since expression of the original Fab scaffold, Fab In order to attempt to improve solubility of Fab59 we
TT, was strong compared to Fab59, we investigated co-expressed the bacterial chaperon protein Skp. The Skp
whether the drop in expression was due to grafting the pep- gene was cloned into pAEV1-O59 as part of the same
tide into the CDR of the light chain, the heavy chain, or operon as Fab59 under control of the same T7 promoter
both. To accomplish this, we switched light chains between (Fig. 1). In comparison to pAEV1-O59 without Skp, the
Fab59 and parental Fab TT which resulted in two addi- bacteria transformed with Skp (pAEV1-O59Skp) grew fas-
tional constructs in which only one chain contained a ter both before and after IPTG addition. To determine if
grafted peptide, pAEV1-LCG (light chain grafted) and the growth recovery of the bacteria containing pAEV1-
pAEV1-HCG (heavy chain grafted). The expression levels O59Skp was associated with reduced accumulation of the
of the constructs, pAEV1-TT, pAEV1-HCG, pAEV1- insoluble form of the grafted light chain, a Western blot
LCG, and pAEV1-59, were compared side-by-side. As was performed using the insoluble portion of an induced
shown by Western blot in Fig. 3A, both human Fab TT bacterial culture. Western blots were also performed exam-
and heavy chain grafted Fab HCG expressed well in ining Fab59 levels in the periplasm isolated from bacterial
E. coli. However, light chain grafted Fab LCG and pellets, and Fab59 secreted into culture medium superna-
Fab59 expression levels were reduced substantially. A Wes- tant after a 4-h induction at 30 °C. The results showed that
60 B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63

p
p
sk

sk
sk
59

T
59
59

59

-T
59
1-O

-0
1-O

1-O

1
1-O

EV
B

EV
EV
EV

EV
EV

pA
pA
pA
pA

pA
pA
A 72K
72K
43K
25K
43K 43K

Fig. 6. Immunoblotting analysis of Fab expression level in E. coli


p
sk

59

periplasm. The bacteria were induced at 0.02 mM IPTG at room


59

1-O
1-O

temperature for 16 h. A Western blot from a non-reducing SDS–PAGE


EV
EV

was probed with anti-human light chain monoclonal antibody.


pA
pA

72K
lower in overnight room temperature induction, as detected
C 43K by Western blot analysis and shown in Fig. 6.

Fab purification
Fig. 5. Immunoblotting analysis of Fab expression enhanced by Skp.
Protein samples were prepared by small-scale Fab expression as described
in Materials and methods. (A) Soluble Fab59 expression in the periplasm
Despite successful efforts to increase yield through codon
of E. coli. A Western blot from a non-reducing SDS–PAGE was probed optimization, introduction of bacterial Skp, and modifica-
with anti-human light chain monoclonal antibody. (B) Insoluble light tion of induction conditions, we still noted that nearly 40%
chain accumulation in E. coli from total cell lysis. A Western blot from a of the purified protein precipitated out during dialysis, and
reducing SDS–PAGE gel was probed with anti-human light chain more than 20% of the soluble Fab59 formed soluble
monoclonal antibody. (C) Fab59 accumulation in culture medium. A
Western blot from a non-reducing SDS–PAGE gel was probed with anti-
dimers/aggregates as shown by SEC profile (Fig. 7A),
human light chain monoclonal antibody. decreasing the yield and purity of the final product.
In initial purification attempts, we used 1 PBS as a
binding buffer, 0.2 M glycine buffer, pH 2.7, as an elution
the soluble Fab59 expression level was further increased buffer, and TBS buffer as a dialysis and SEC running buffer
slightly in the periplasm by co-expression of Skp protein in a two-step purification. However, we noted that Fab59
(Fig. 5A). As shown in Fig. 5B, the bacteria containing was sensitive to the low pH elution buffer in our process.
pAEV1-O59Skp accumulated somewhat less insoluble To address this, a subsequent attempt using a His-tag Ni-
grafted light chains than the bacteria containing pAEV1- NTA-based purification protocol with neutral elution con-
O59, which correlated with a reduction in Fab59 toxicity ditions was evaluated. Unfortunately the neutral elution
to E. coli. Interestingly, a much more dramatic difference conditions did not prevent Fab59 from aggregating.
was observed by Western blot of culture medium superna- Since it has previously been demonstrated that glycerol,
tant. As shown in Fig. 5C, significantly more fully assem- detergents and high ion strength can increase protein solu-
bled and secreted Fab59 was detected from pAEV1- bility and prevent protein aggregation in some cases
O59Skp than from pAEV1-O59. The increase in Fab59 [14,15], we switched to a HEPES loading buffer that con-
accumulation in the culture medium of pAEV1-O59Skp tained 10% glycerol, 0.02% Tween-80 and 0.5 M NaCl.
may be the result of increased secretion of properly folded For elution, we again used 0.2 M glycine elution buffer,
Fab59 from the periplasm. pH 2.7, but importantly added 10% glycerol and 0.02%
Tween-80 in the elution buffer. The elute from the column
Modification of induction conditions significantly increased was immediately neutralized and the NaCl concentration
Fab production in the modified expression system brought to 0.5 M. This strategy worked extremely well in
the purification of Fab59. No aggregation was observed
Although poor expression of Fab59 was partially resolved in the protein preparation during purification and dialysis,
by all of the changes described thus far, including codon- and no detectable dimers/aggregates were observed in the
optimized LC expression and the presence of the Skp chap- SEC profile (Fig. 7A). Importantly, no aggregation was
eron protein, further enhancements to protein expression observed to occur during 1 month of storage at 4 °C based
were investigated by re-evaluating IPTG induction condi- on SEC analysis, and no loss in agonist activity was
tions as well as the effects of temperature in the modified observed based on our in vitro luciferase reporter assay
expression system. We found that 0.02 mM IPTG induction (data not shown).
at room temperature overnight (16–20 h) gave the highest After purification optimization, Fab59 was purified to a
yield of Fab59. In comparison to wild-type pAEV1-TT, monomeric form with extremely high purity. Non-reducing
the pAEV1-O59Skp expression level was only 1- to 2-fold SDS–PAGE gel showed that the purified monomeric Fab
B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63 61

since conditions that efficiently prevent Fab59 aggregation


A O.D.
have been found using an optimized buffer, this should not
present a substantial difficulty.
Monomer
500 The activity of purified Fab59 was equivalent to recombinant
human TPO in cell-based assay
400
Purified monomeric Fab59 was tested in comparison to
300
recombinant human TPO in vitro for its ability to stimulate
cMpl-R present on cells in a luciferase reporter assay [2].
200
Dose dependent receptor activation was observed with
Dimer/aggregate addition of increasing amounts of Fab59, with comparable
activities from Fab59 preparations purified in two different
100
buffers (Fig. 8). No activity was observed in the control
vector transfected cells treated in an equivalent manner
20 40 60 Vol (ml) (data not shown). The results indicate that Fab59, recov-
ered following a series of step-wise expression and purifica-
1 2 3 4 tion improvements that significantly increased its yield, was
B an effective agonist capable of stimulating the cMpl -recep-
tor in a specific manner.

Discussion

50K Fab59 The rationally-designed agonist antibody fragment


40K LC dimer
Fab59 may provide therapeutic benefit in patients suffering
25K from low platelet counts, or thrombocytopenia, and is
20K
therefore a potential candidate for clinical development
[2]. However, large-scale bacterial production of the origi-
nal Fab59 was not feasible since its expression level was
Fig. 7. (A) SEC profiles of Fab59 on Superdex75. Solid line: Fab59 extremely low and barely detected by Western blot.
purified in regular binding and elution buffers. Dash line: Fab59 purified Poor expression of Fabs in E. coli can often be related to
in modified binding and elution buffers. (B) SDS–PAGE analysis of improper protein folding, intracellular degradation, aggre-
monomeric Fab59 purified from bacterial pellets (periplasmic preps). (1) gation in the periplasm and toxicity of the antibody frag-
Monomeric Fab59 from induction at room temperature (reducing
condition). (2) Standard molecular weight marker. (3) Monomeric
ment to the E. coli host [4,5]. Several different strategies
Fab59 from induction at room temperature (non-reducing condition). have been shown to improve expression levels efficiently
(4) Monomeric Fab59 from induction at 30 oC (non-reducing condition).
5
LC dimer, light chain dimer.
rhTPO
50 Kd Fab 59 monomer prep F1
Relative Fold Luciferase Activity

was very clean and free of detectable contaminates 50 Kd Fab 59 monomer prep F2
4
(Fig. 7B). Equal amounts of light chains and heavy chains
could be seen on a reducing SDS–PAGE gel, demonstrat-
ing that Fab59 was properly folded and assembled and that
3
the disulfide bonds were well formed. No detectable light
chain dimer contamination was observed in the monomeric
Fab59 purified from bacterial pellets induced at room tem-
perature. In contrast, some light chain dimer contamina- 2
tion in monomeric Fab59 purified from bacterial pellets
induced at 30 °C (Fig. 7B) was noted, suggesting that
Fab59 had a better folding and light/heavy chain paring 1
at room temperature induction rather than at 30 °C.
1000
100
10

Because the molecular weight (MW) of light chain dimers Concentration (pM)
is similar to the MW of Fab59, making SEC separation dif-
Fig. 8. Fab59 agonist activity demonstrated in cMpl receptor expressing
ficult, expression at room temperature was best for optimal
BaF3 cells using a luciferase reporter assay. rhTPO: purified recombinant
purity. Although SEC separation is useful at this scale, it human TPO protein (R&D Systems). Fifty kiloDalton Fab59 monomer
may be less feasible for large-scale manufacturing proto- prep F1: Fab59 monomer purified in PBS buffers. Fifty kiloDalton Fab59
cols therefore requiring additional optimization. However, monomer prep F2: Fab59 monomer purified in HEPES buffers.
62 B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63

for various Fabs. Toxicity of some Fabs can be greatly alle- and aggregation during passage through the bacterial per-
viated by modulating transcriptional and translational iplasm. Skp does this by recognizing unfolded or misfolded
rates, using methods such as low induction temperature, structures and interacting with them [32]. Aggregating pro-
low level IPTG induction, and modification of culture med- teins can be toxic to bacterial cells by acting as a sink for
ium [8–10,16–18], or by using more tightly controlled pro- other folding host proteins causing a stress response and
moters (tet and ara) [9, 19–21]. Co-expression of bacterial draining cell resources [33]. Therefore it is believed that
chaperons or heterologous disulfide isomerases [4,22] has co-expression of Skp alleviates the toxicity of aggregation
successfully improved Fab folding and disulfide bond for- prone recombinant proteins in E. coli by promoting correct
mation. In addition optimizing secretion signal sequences polypeptide folding. Skp has been used successfully with
could efficiently promote protein translocation to the bac- antibody fragments, including single chain antibodies
terial periplasm where proteins can be properly folded (scFv) [34,35] and Fabs [4]. In this study, we showed that
and form the correct disulfide bonds [23]. Other strategies Skp protein co-expression significantly reduced the toxicity
have been reported to enhance Fab expression levels such of Fab59 and reversed the growth retardation of E. coli
as expression in different hosts other than in E. coli cells bearing pAEV-O59. Skp mediated reduction of
[4,24], vector engineering to balance light chain and heavy Fab59 toxicity may result from the observed decrease in
chain production [10,25], and even more sophisticated pro- the accumulation of insoluble light chain.
tein point mutations [26] to remove critical surface exposed Interestingly, our results also demonstrated that co-
hydrophobic amino acid residues thereby increasing Fab expression of the Skp protein significantly increased assem-
solubility. However, each particular strategy may not be bled Fab59 ‘‘secretion” into the culture medium. The
applicable to every difficult to express protein since the rea- increased Fab59 appearance in the medium is not from cell
sons in each case will be different. Furthermore, multiple lysis since we did not see significant free chain accumula-
factors are likely at play in most cases. As a result, optimi- tion in the medium, even though there was significant sol-
zation of expression can be laborious and time-consuming. uble free chain accumulation in the periplasmic fraction.
In an effort to boost the Fab59 expression in E. coli in Skp mediated increase in light chain solubility correlated
this study, we first determined what factors were contribut- with a reduction of Fab59 toxicity and with an increase
ing to the low expression level of Fab59. In comparison to of Fab59 secretion into the medium. Increased soluble
the parental scaffold Fab TT or Fab with a peptide grafted LC likely allows for more Fab assembly within the peri-
heavy chain (Fab HCG), expression of Fabs containing a plasm, leading to more efficient release into the culture
grafted light chain (Fab LCG and Fab59) was dramatically supernate, though direct effects of Skp protein in promot-
decreased as noted for both Fab expression (non-reducing ing Fab secretion cannot be ruled out at this time.
gels) and light chain expression (reducing gel). We system- In a final step, the purification process from periplasmic
atically accomplished improvements by first codon opti- extracts was optimized to efficiently avoid protein aggrega-
mizing the light chain, second, using a tightly controlled tion. The use of glycerol, Tween-80 and increased ion
strong promoter, and third inducing at low temperature strength in the purification buffers efficiently prevented
with low levels of IPTG. Fab59 from forming insoluble and soluble aggregates,
Additionally, a significant decrease in solubility of light and did not interfere with downstream purification or func-
chain grafted Fabs was noted, both in E. coli cells and in tional assays. Purification of monomeric Fab59 from bac-
buffers used for purification. The solubility features of terial periplasmic extracts was accomplished in a two-step
Fab59 may be related to an increase of surface exposed protocol using SEC to remove antibody related contami-
hydrophobic residues residing within the grafted peptide nates, such as degradation products, free light and heavy
which was selected for optimal presentation within the chains, and dimers/aggregates. This two-step purification
Fab CDRs in order to achieve binding with target recep- provided a convenient, fast and straightforward method
tors, and therefore required for function. Poor expression to obtain highly pure monomeric and correctly folded
of Fab59 protein also appeared to be linked to grafted light Fab59 with intact disulfide bonds sufficient to support fur-
chain aggregation as a result of limited solubility, and sub- ther preclinical development. After expression and purifica-
sequent toxicity to E. coli. Other cases of poor light chain tion optimization, the final yields of purified monomeric
expression, and related toxicity to the cell, have been Fab59 reached 3.5 mg/L bacterial culture in a shake flask
reported [5,27]. In our case, the parental LC expressed well and represented a 100-fold increase from our starting yield.
on its own, but insertion of the TPO mimetic peptide into Although this was a significant improvement, even higher
CDR2 resulted in decreased LC expression and concomi- yields may be obtained in a larger-scale bioreactor, thereby
tant low Fab expression. To resolve this limitation, addi- increasing Fab590 s potential for practical and economical
tional analysis was undertaken. production.
The bacterial periplasmic protein Skp is a chaperon pro- In summary, Fab59 expression and purification from
tein that has been shown to improve solubility and reduce E. coli has been successfully enhanced 100-fold by combining
toxicity of recombinant proteins in E. coli [4,28–31]. Skp’s an alternate vector, codon optimization, optimized induc-
native role is to act on newly synthesized bacterial outer- tion conditions, co-expression of a chaperon protein and
membrane proteins, and protect them from misfolding improved purification processes. Following these efforts,
B. Lin et al. / Protein Expression and Purification 59 (2008) 55–63 63

the agonist activity of the purified monomeric Fab59 was [18] D. Humphreys, A.P. Chapman, D.G. Reeks, V. Lang, P.E. Stephens,
demonstrated to be equipotent to rhTPO in in vitro cMpl- Formation of dimeric Fabs in Escherichia coli: effort of hinge size and
isotype, presence of interchain disulphide bond, Fab’ expression
R signaling, luciferase reporter-based assays, and further- levels, tail piece sequences and growth conditions, J. Immunol.
more stimulated increased platelet levels in vivo [2]. Methods 209 (1997) 193–202.
[19] K. Alfthan, T. Kristiina, S. Dorothea, S. Ilkka, I. Tiina, V. Liisa,
References K. Sirkka, K. Matti, K.C. Jonathan, T.T. Tuula, Efficient
secretion of murine Fab fragments by Escherichia coli is deter-
[1] S.E. Cwirla, P. Balasubramanian, D.J. Duffin, C.R. Wagstrom, C.M. mined by the first constant domain of the heavy chain, Gene 128
Gates, S.C. Singer, A.M. Davis, R.L. Tansik, L.C. Mattheakis, C.M. (1993) 203–209.
Boytos, P.J. Schatz, D.P. Baccanari, N.C. Wrighton, R.W. Barrett, [20] F. Maeda, Y. Nagatsuka, S. Ihara, S. Aotsuka, Y. Ono, H. Inoko, M.
W.J. Dower, Peptide agonist of the thrombopoietin receptor as the Takekoshi, Bacterial expression of a human recombinant monoclonal
natural cytokine, Science 276 (1997) 1696–1699. antibody Fab fragment against Hepatitis B surface antigen, J. Med.
[2] S. Frederickson, M.W. Renshaw, B. Lin, L.M. Smith, P. Calveley, Virol. 58 (1999) 338–345.
J.P. Springhorn, K. Johnson, Y. Wang, X. Su, Y. Shen, K.S. [21] D. Rau, K. Kramer, B. Hock, Cloning, functional expression and
Bowdish, A novel rationally designed agonist antibody fragment that kinetic characterization of pesticide-selective Fab fragment variants
functionally mimics thrombopoietin, Proc. Natl. Acad. Sci. USA 103 derived by molecular evolution of variable antibody genes, Anal.
(2006) 14307–14312. Bioanal. Chem. 372 (2002) 261–267.
[3] R. Verma, E. Boleti, A.J. George, Antibody engineering: comparison [22] A. Ideno, F. Masahiro, I. Yoshitaka, K. Yoshikazu, M. Tadashi,
of bacterial, yeast, insect and mammalian expression systems, J. FK506 binding protein from the hyperthermophilic archaeon Pyro-
Immunol. Methods 216 (1998) 165–181. coccus horikoshii suppresses the aggregation of protein in Escherichia
[4] R. Levy, R. Weiss, G. Chen, B.L. Iverson, G. Georgiou, Production coli, Appl. Environ. Microbiol. 68 (2002) 464–469.
of correctly folded Fab antibody fragment in the cytoplasm of [23] D. Humphreys, M. Sehdev, A.P. Chapman, R. Ganesh, B.J. Smith,
Escherichia coli trxB gor mutants via the coexpression of molecular L.M. King, D.J. Glover, D.G. Reeks, P.E. Stephens, High-level
chaperones, Protein Express. Purif. 23 (2001) 338–347. periplasmic expression in Escherichia coli using a eukaryotic signal
[5] S. Kumar, J. Kalsi, D.S. Latchman, L.H. Pearl, D.A. Isenberg, peptide: Importance of codon usage at the 50 end of the coding
Expression of the Fabs of human auto-antibodies in Escherichia coli: sequence, Protein Express. Purif. 20 (2000) 252–264.
Optimization and determination of their fine binding characteristics [24] M. Venturi, C. Seifert, C. Hunte, High level production of functional
and cross-reactivity, J. Mol. Biol. 308 (2001) 527–539. antibody Fab fragment in an oxidizing bacterial cytoplasm, J. Mol.
[6] B.D. Stollar, Bacterial expression of anti-DNA antibody domains, Biol. 315 (2002) 1–8.
Methods 11 (1997) 12–19. [25] D.P. Humphreys, B. Carrington, L.C. Bowering, R. Ganesh, M.
[7] P.M. Sharp, W.H. Li, The codon adaptation index—a measure of Sehdev, B.J. Smith, L.M. King, D.G. Reeks, A. Lawson, A.G.
directional synonymous codon usage bias, and its potential applica- Popplewell, A plasmid system for optimization of Fab0 production in
tions, Nucleic Acids Res. 15 (1987) 1281–1295. Escherichia coli: importance of balance of heavy chain and light chain
[8] P.M. O’Brien, G. Maxwell, M.S. Campo, Bacterial expression and synthesis, Protein Express. Purif. 26 (2002) 309–320.
purification of recombinant bovine Fab fragments, Protein Express. [26] J.G. Wall, A. Pluckthun, The hierarchy of mutations influencing the
Purif. 24 (2002) 43–50. folding of antibody domains in Escherichia coli, Protein Eng. 12
[9] B. Seliko, G. Cosio, C. Garcia, B. Becerril, L.D. Possani, E. Horjales, (1999) 605–611.
Bacterial expression, purification and functional characterization of a [27] G. Forsberg, M. Forsgren, M. Jaki, M. Norin, C. Sterky, A.
recombinant chimeric Fab derived from murine mAb BCF2 that Enhorning, K. Larsson, M. Ericsson, P. Bjork, Identification of
neutralizes the venom of the scorpion Centruroides noxius Hoffmann, framework residues in a secreted recombinant antibody fragment that
Toxicon 43 (2004) 43–51. control production level and localization in Escherichia coli, J. Bio.
[10] S. Corisdeo, B. Wang, Functional expression and display of an Chem. 272 (1997) 12430–12436.
antibody Fab fragment in Escherichia coli : study of vector designs [28] J. Maynard, E.J. Adams, M. Krogsgaard, K. Petersson, C.W. Liu,
and culture conditions, Protein Express. Purif. 34 (2004) 270–279. K.C. Garcia, High-level bacterial secretion of single-chain ab T-cell
[11] M. Nalezkova, A. de Groot, M. Graf, P. Gans, L. Blanchard, receptors, J. Immunol. Methods 306 (2005) 51–67.
Overexpression and purification of Pyrococcus abyssi phosphopant- [29] D.K. Chatterjee, D. Esposito, Enhanced soluble protein expres-
etheine adenylytransferase from an optimized synthetic gene for sion using two new fusion tags, Protein Expr. Purif. 46 (2005)
NMR studies, Protein Express. Purif. 39 (2005) 296–306. 122–129.
[12] M.W. Laird, G.C. Sampey, K. Johnson, D. Zukauskas, J. Pierre, J.S. [30] T.A. Walton, M.C. Sousa, Crystal structure of Skp, a profoldin-like
Hong, B.A. Cooksey, Y. Li, O. Galperina, J.D. Karwoski, R.N. chaperone that protects soluble and membrane proteins from
Burke, Optimization of BLyS production and purification from aggregation, Mol. Cell. 15 (2004) 367–374.
Escherichia coli, Protein Express. Purif. 39 (2005) 237–246. [31] H. Boothmann, A. Pluckthun, Selection for a periplasmic factor
[13] E.P. Rocha, Codon usage bias from tRNA’s point of view: redun- improving phage display and functional periplasmic expression,
dancy, specialization, and efficient decoding for translation optimi- Nature Biotechnol. 16 (1998) 376–380.
zation, Genome Res. 14 (2004) 2279–2286. [32] J. Qu, C. Mayer, S. Behrens, O. Holst, J.H. Kleischmidt, The trimeric
[14] A. Neagu, M. Neagu, A. Der, Fluctuations and the Hofmeister effect, periplasmic chaperone Skp of Escherichia coli forms 1:1 complexes
Biophys. J. 81 (2001) 1285–1294. with outer membrane proteins via hydrophobic and electrostatic
[15] B.A. Kerwin, M.C. Heller, S.H. Levin, T.W. Randolph, Effects of interactions, J. Mol. Biol. 374 (2007) 91–105.
Tween 80 and sucrose on acute short-term stability and long-time [33] D. Missiakas, S. Raina, Protein folding in the bacterial periplasm, J.
storage at 20 °C of a recombinant hemoglobin, J. Pharm. Sci. 87 Bacteriol 179 (1997) 2465–2471.
(1998) 1062–1068. [34] A. Hayhurst, W.J. Harris, Escherichia coli Skp chaperone coexpres-
[16] H. Wlad, A. Ballagi, L. Bouakaz, Z. Gu, J.-C. Janson, Rapid two- sion improves solubility and phage display of single-chain antibody
step purification of a recombinant mouse Fab fragment expressed in fragments, Protein Express. Purif. 15 (1999) 336–343.
Escherichia coli, Protein Express. Purif. 22 (2001) 325–329. [35] A. Hayhurst, S. Happe, R. Mabry, Z. Koch, B.L. Iverson, G.
[17] J.G. Wall, A. Pluckthum, Effectors of overexpression folding mod- Georgiou, Isolation and expression of recombinant antibody frag-
ulators on the in vivo folding of heterologous proteins in Escherichia ments to the biological warfare pathogen Brucella melitensis, J.
coli, Curr. Opin. Biotechnol. 6 (1995) 507–516. Immunol. Methods 276 (2003) 185–196.

You might also like