Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

Enzyme and Microbial Technology 38 (2006) 855–859

Large scale production of d-allose from d-psicose using continuous


bioreactor and separation system
Kenji Morimoto a , Chang-Su Park b , Motofumi Ozaki b , Kei Takeshita c ,
Tsuyoshi Shimonishi b , Tom Birger Granström a , Goro Takata a ,
Masaaki Tokuda a , Ken Izumori a,∗
a Rare Sugar Research Center, Kagawa University, Miki-cho, Kagawa 761-0795, Japan
b Kagawa Industry Support Foundation, Hayashi-cho, Takamatsu, Kagawa 761-0301, Japan
c Fushimi Pharmaceutical Co., Ltd., Nakatsu-cho Marugame, Kagawa 763-8605, Japan

Received 11 April 2005; received in revised form 9 August 2005; accepted 10 August 2005

Abstract
l-Rhamnose isomerase (l-RhI) from Pseudomonas stutzeri LL172 can convert d-psicose to d-allose. Partially purified recombinant l-RhI from
Escherichia coli was immobilized on BCW-2510 Chitopearl beads and utilized to produce d-allose. Total 20,000 units of immobilized enzyme
converted d-psicose to d-allose without remarkable decrease in the enzyme activity over 17 days. When 50% d-psicose (w/w) was applied to a
column with a flow rate of 0.8 ml/min at 42 ◦ C, approximately 30% d-psicose was isomerized to d-allose for 17 days. However, by reducing the flow
rate to 0.4 ml/min after 17 days, d-allose was transformed at the same rate for 13 days. The total of 27 l reaction mixture was separated by Simulated-
Moving-Bed Chromatograph system. Approximately 2.2 l/d of 50% (w/w) reaction mixture was separated continuously. After separation, d-allose
and d-psicose fractions were 3 l of approximately 10% (w/w) with 95% purity and 10 l of approximately 8% (w/w) with 95% purity per day,
respectively. The separated d-allose solution was concentrated up to about 50% and crystallized gradually by being kept at room temperature.
Crystals of d-allose were separated from the syrup by filtration and 1.65 kg crystals of 100% purity were obtained. The d-allose crystal yield from
the d-psicose substrate was approximately 10%.
© 2005 Elsevier Inc. All rights reserved.

Keywords: Rare sugars; d-Allose; l-Rhamnose isomerase; Simulated-Moving-Bed Chromatograph

1. Introduction We established the bioproduction strategy route named as


“Izumoring” [4]. To produce d-allose in large quantities, one of
Rare sugars including monosaccharides are rare in nature the best route is a two step reaction as follows: first d-psicose
and knowledge of their biological and physiological functions is produced from inexpensive monosaccharide d-fructose, and
have consequently been known little so far. We are working in the second step, d-psicose is isomerized to d-allose. We
on production of various kinds of rare sugars using microbial reported that mass production of d-psicose from d-fructose was
enzymes and whole cells. It has been proven that some rare achieved by using d-tagatose 3-epimerase (d-TE) from Pseu-
hexose sugars have exhibited physiologically active functions domonas cichorii ST-24 [5], d-psicose was separated from sugar
[1–3]. Rare sugars have received increasing attention in recent mixture by Simulated-Moving-Bed Chromatograph (JPN Pat.
years for a variety of usages, such as low-calorie carbohydrate No. P2001-354690A, 2001). l-Rhamnose isomerase (l-RhI),
sweeteners, inhibitor of microbial growth and bulking agents l-rhamnose ketol-isomerase [EC 5.3.1.14], which isomerizes
[1,2]. For instance, d-allose, one of rare aldo-hexose, has been l-rhamnose to l-rhamnulose, has been found in Escherichia
reported to have a possibility to be applied in pharmaceutical coli [6,7], Salmonella and Pseudomonas [8–10]. l-Rhamnose
industry, such as inhibitor of ischemia/reperfusion injury [3]. isomerase, which was produced constitutively, from Pseu-
domonas stutzeri LL172was reported to catalyze isomerizations
not only between l-rhamnose–l-rhamnulose but also between
∗ Corresponding author. Tel.: +81 87 891 3290; fax: +81 87 891 3289. d-allose–d-psicose (Fig. 1; [10,11]). We had reported that small
E-mail address: izumori@ag.kagawa-u.ac.jp (K. Izumori). scale of d-allose production was successful using this enzyme

0141-0229/$ – see front matter © 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.enzmictec.2005.08.014
856 K. Morimoto et al. / Enzyme and Microbial Technology 38 (2006) 855–859

Fig. 1. Schematic diagram of d-allose and d-psicose production from d-fructose catalyzed by l-rhamnose isomerase.

[10]. Moreover, l-rhi gene from this strain was successfully Tris–HCl buffer (pH 9.0), and disrupted by ultrasonicator. The cell free extract
cloned into E. coli and over-expressed [12]. was heat-treated at 50 ◦ C for 10 min. The supernatant was collected by cen-
In this paper, we describe continuous d-allose production trifugation at 8000 × g for 30 min. The supernatant was slowly added with 1M
MnCl2 to a final concentration 10 mM. And polyethylene glycol #6000 was
from d-psicose by use of immobilized enzyme of recombi- slowly added to a final concentration 5% (w/v) and the mixture was stirred for
nant l-RhI under the optimized condition. The immobilized 1 h at 4 ◦ C. This mixture was centrifuged at 12,000 × g for 30 min and precip-
enzyme has advantages such as reusability, less by-product itation was discarded. More polyethylene glycol #6000 was added to a final
formation, and long operation time. Furthermore, we estab- concentration 20% (w/v). After stirring for 1 h, the precipitation was collected
lished the separation condition for reaction mixture contain- by centrifugation at 12,000 × g for 30 min and suspended in a small quantity of
10 mM Tris–HCl buffer (pH 9.0). This solution was used as partially purified
ing d-allose by using Simulated-Moving-Bed Chromatograph enzyme.
system.
2.4. Enzyme and protein assay
2. Materials and methods
It is difficult to detect an isomerase activity from ketose to aldose, because
the amount of reducing ketose is not detected easily. Isomerase activity is gen-
2.1. Chemicals erally detected from aldose to ketose, and ketose conversion from aldose is
easy to know enzyme activity using cysteine–carbazole method [13]. Appro-
All chemical agents were purchased from Wako Pure Chemicals (Tokyo, priately, diluted partially purified enzyme, 50 ␮l, was incubated with 50 ␮l of
Japan). Chitopearl BCW-2510 resin was purchased from Fuji Spinning Co., 50 mM d-allose as a substrate at 30 ◦ C for 10 min. Accumulation of product,
Ltd. (Tokyo, Japan). It has primary, secondary and quaternary amine groups d-psicose, was measured by cysteine-carbazole method. One unit of l-RhI is
and binds to proteins with electrovalent mode. d-Psicose was prepared by our defined as the amount of enzyme that converts 1 ␮mol of d-allose to d-psicose
laboratory as described previously [5]. in 1 min. The reaction mixture was assayed by the HITACHI High-performance-
liquid-chromatography (HPLC), which is constructed by L-7490 refractive index
2.2. Microorganism culture condition detector, D-2500 chromato-integrator and Hitachi HPLC column GL-C611.
Chromatography was carried out at 60 ◦ C using 10−4 M NaOH solution at a
The l-rhi gene of Pseudomonas stutzeri LL172, which was expressed contin- flow rate of 1.0 ml/min. Protein concentration was measured according to the
uously, was inserted in pQE 60, named as pOI-01. This plasmid was transformed method of Bradford [14] with bovine serum albumin as a standard.
in E. coli JM 109 [12]. The recombinant E. coli JM109 was cultured in a medium
composed of 3.5% polypepton, 2.0% yeast extract, 0.5% NaCl and 100 ␮g/ml
2.5. Optimization of l-RhI immobilization condition
ampicillin in the four 15 l jar-fermentor at 28 ◦ C for 18 h, and recombinant
enzyme was induced by addition 0.5 mM IPTG and 1 mM MnCl2 in the culture The partially purified enzyme (100 units) was immobilized on 1, 2, 3 and
medium. After 5 h period, the cells were collected by continuous centrifugation 5 g of BCW-2510 Chitopearl beads at 4 ◦ C for 1 day. Immobilization efficiency
(8000 × g, 4 ◦ C). was calculated as follows: residual activity in the supernatant was analyzed
with d-allose as a substrate and then d-psicose amount was measured by
cysteine–carbazole method as described above.
2.3. Partial purification of the enzyme
2.6. d-Allose mass production
When l-RhI was completely purified, it became to be unstable according to
formation of aggregation. Therefore, we used stable partially purified enzyme The immobilized enzyme, which was prepared under the optimized condi-
which was prepared as follows. The collected cells were suspended in 10 mM tion, was charged in a column (4 cm × 40 cm) tempered at 42 ◦ C. Substrate 50%
K. Morimoto et al. / Enzyme and Microbial Technology 38 (2006) 855–859 857

(w/w) d-psicose dissolved in 10 mM Tris–HCl buffer (pH 9.0) was applied by


flow rate of 0.8 ml/min. When the enzyme activity was decreased, the flow rate
was reduced to 0.4 ml/min.

2.7. Separation of d-psicose and d-allose

Before separation, reaction mixture was desalted by passing through Amber-


lite IRA-411 (CO3 2− form) and DIAION SK1B (H+ form) ion-exchanged resin.
After desalting, the reaction mixture was separated to obtain mainly d-allose
and d-psicose by TREZONE system (Organo Corp.) which can operate the
Simulated-Moving-Bed Chromatograph. It consisted of 2 pumps, 26 valves and
4 columns. Each column was filled with 750 ml of ion exchanged resin (Ca2+
form). This system can separate a reaction mixture which is composed of two
main components.

2.8. Crystallization of d-allose Fig. 2. Effect of the Chitopearl BCW-2510 beads volume on d-allose production
from d-psicose. d-Psicose in 50 mM Tris–HCl (pH 9.0) was used as a substrate at
After separation of reaction mixture, d-allose fraction was evaporated to the 42 ◦ C. After incubation, converted d-allose was measured by HPLC as described
concentration of 50% (w/w). The syrup was kept at room temperature to allow in Section 2. Symbols: open circles, 1 g beads; open squares, 2 g beads; close
d-allose crystals to grow without adding seed crystals. squares, 3 g beads; open triangles, 5 g beads.

3. Results inactivated in the column and enzyme was gradually detached it


from resin, it became impossible to keep an equilibrium point.
3.1. Immobilization of l-RhI We made preliminary tests whether bioreactor could be used
continuously after enzyme activity was decreased. As a result,
When 100 U of the enzyme was immobilized, not less than the flow rate of 0.4 ml/min was able to maintain the optimal con-
90% of l-RhI was bound to 2, 3, or 5 g of BCW-2510 Chito- version rate. When the flow rate was dropped to 0.4 ml/min after
pearl beads. By contrast, only approximately 50% of enzyme 17 days, d-allose was produced with the some ratio for 13 days
was bound to 1 g beads (Table 1). In this study, we found that more in this study (Fig. 4). Eventually, bioreactor was able to
the maximum equilibrium point (33% d-allose) was achieved maintain 1 month for continuous operation and to produce reac-
after 5 h in all conditions (Fig. 2). The conversion efficiency tion mixture containing 5.02 kg of d-allose from 27 l (16.6 kg)
was, however, the lowest among all in the case of using of 1 g of 50% (w/w) d-psicose syrup for 1 month (Table 2). When the
beads. The best condition for immobilization of l-RhI on Chi- same amount of l-RhI, which was used for bioreactor construc-
topearl beads was achieved when 2 or 3 g of beads were used for tion in this study, was let to react as soluble enzyme, the d-allose
100 U of enzyme. Based on this result and cost-effectiveness, yield was approximately 30%.
we determined that we utilized 2 g beads for 100 U of
l-RhI.

3.2. Continuous conversion from d-psicose to d-allose

The partially purified l-rhamnose isomerase which was pre-


pared from 40 l culture has 20,000 U of enzyme activity. Based
on the result of the optimized immobilization condition, we pre-
pared a bioreactor using 400 g beads. As shown in Figs. 2 and 3,
reaction equilibration was 30 (d-allose): 70 (d-psicose) toward
50% (w/w) d-psicose as a substrate, although a small amount of
some by-products detected. The immobilized l-rhamnose iso-
merase was able to convert d-psicose to d-allose in the flow rate
of 0.8 ml/min without significant decrease in the enzyme activity
over the first 17 days. Since immobilized enzyme was gradually

Table 1
Immobilization efficiency of l-Rhl on Chitopearl BCW-2510
Chitopearl resin Residual activity in Efficiency (%)
volume (g) supernatant (U)

1 42.7 51.3
2 5.98 94.0
3 3.19 96.8
5 3.78 96.2 Fig. 3. HPLC analysis of d-allose from d-psicose by immobilized l-rhamnose
isomerase.
858 K. Morimoto et al. / Enzyme and Microbial Technology 38 (2006) 855–859

3.3. Separation of d-psicose and d-allose

As a result of the optimized separation condition, contin-


uously the 50% (w/w) mixture of a maximum approximately
2.2 l in 1 day could be processed. The separated d-allose frac-
tion volume was 3 l/d containing 10% (w/w) d-allose with 95%
purity and a minor amount of by-products (Fig. 5). d-Psicose
fraction volume was 10 l/d containing 8% (w/w) d-psicose with
95% purity (Fig. 5).

3.4. Crystallization of d-allose

Fig. 4. Bioconversion of d-allose from d-psicose by immobilized l-rhamnose The separated d-allose solution was concentrated up to about
isomerase. Bold arrow and broken arrow indicate a period of flow rate of 0.8 50% (w/w) and crystallized gradually at room temperature. The
and 0.4 ml/min, respectively. crystals of d-allose were separated from the solution by filtration.
Finally, we could obtain approximately 1.65 kg d-allose crystal
with 100% purity (Fig. 5).

Table 2
The yield of d-allose mass production

Reacted mixture (kg)a Separated d-allose (kg)a Separated d-psicose (kg)a Loss of sugars (kg)a Crystallized d-allose (kg)b

Total sugarc 16.6 3.50 9.61 3.49


d-Allosec 5.02 3.33 0.64 1.05 1.65
d-Psicose 11.36 0.00 8.96 2.40
By-productsc 0.22 0.17 0.00 0.05
a Reaction mixture indicates total weight after enzyme reaction. “Separated d-allose” and “separated d-psicose” indicate total weights after separation, respectively.

“Loss of sugars” indicates total weight of Loss by desalting and separation processes.
b “Crystallized d-allose” indicates obtained weight of d-allose crystal from separated d-allose fraction.
c “Total sugar”, “d-allose”, “d-psicose” and “by-product” indicate sugar weights contained in “reacted mixture”, “separated d-allose”, “separated d-psicose” and

“loss of sugars”, respectively.

Fig. 5. HPLC analysis of reaction mixture before and after separation and d-allose crystal: (A) indicates a deionized solution mixture; (B and C) indicate fractions
separated d-allose and d-psicose by TREZONE system, respectively; (D) indicates finally obtained d-allose crystal.
K. Morimoto et al. / Enzyme and Microbial Technology 38 (2006) 855–859 859

4. Discussions and some by-product. However, crystallization of d-allose was


performed in advance of d-psicose in the syrup mixture because
We previously reported d-psicose mass production from d- d-allose has a property to crystallize easier than d-psicose. Con-
fructose by d-tagatose 3-epimerase from Pseudomonas cichorii sequently, d-allose is crystallized specifically in the mixture. The
ST-24 [5], and separation of d-psicose and d-fructose sugar crystallized d-allose was able to be harvested easily by filtration
mixture by Simulated-Moving-Bed Chromatograph (Organo using 5-␮m filter paper, and the purity was 100% as determined
Corp, JPN Pat. No. P2001-354690A 2001). In this paper, we by HPLC analysis (Fig. 5). The yield of d-allose in this study
have reported d-allose mass production from d-psicose by l- was summarized in Table 2. Finally the crystal of d-allose was
rhamnose isomerase (l-RhI) from Pseudomonas stutzeri LL172. obtained 10% efficiency of reacted d-psicose weight. In this
P. stutzeri LL172 expressed constitutively l-rhi gene. How- study, we established the mass production of d-allose by con-
ever, expression level was too low to construct an efficient tinuous bioreactor and Simulated-Moving-Bed Chromatograph.
bioreactor. In order to increase the l-RhI expression level and From now on, we will carry out improvements of bioreactor per-
construct the bioreactor, we carried out the over-expression of formance and Simulated-Moving-Bed Chromatograph in order
l-rhi gene in E. coli JM109 [12]. This recombinant could pro- to increase reaction efficiency and d-allose yield from the mix-
duce l-RhI of 20-fold toward the volumetric yield in comparison ture of d-allose and d-psicose.
with expression level of wild type P. stutzeri LL172 [12]. Par-
tially purified enzyme prepared from recombinant was able to References
produce mainly d-allose from d-psicose, although it produced
[1] Livesey G, Brown JC. d-Tagatose is a bulk sweetener with zero energy
some d-fructose and other by-product (Fig. 3).
determined in rats. J Nutr 1996;126:1601–9.
In the previous report [10], d-allose was produced by batch [2] Levin GV, Zehner LR, Sanders JP, Beadle JR. Sugar substitutes: their
reaction using l-RhI immobilized on Chitopearl beads. The energy values, bulk characteristics, and potential health benefits. Am J
batch reaction has two problems as follows: (1) we have to sep- Clin Nutr 1995;62:1161–8.
arate reaction mixture from immobilized enzyme after reaction; [3] Hossain MA, Izuishi K, Tokuda M, Izumori K, Maeta H. d-Allose has a
strong suppressive effect against ischemia/reperfusion injury: a compar-
(2) l-RhI was damaged by drop of pH due to high concentra-
ative study with allopurinol and superoxide dismutase. J Hepatobiliary
tion d-psicose. Consequently, it was not possible to use it for a Pancreat Surg 2004;11:181–9.
long period. Therefore, we constructed a continuous bioreactor [4] Granström TB, Takata G, Tokuda M, Izumori K. Izumoring: a novel
where substrate was injected by a pump as described in Section and complete strategy for bioproduction of rare sugars. J Biosci Bioeng
2. As a result, declining rate of immobilized enzyme activity was 2004;97:89–94.
[5] Takeshita K, Suga A, Takada G, Izumori K. Mass production of d-
decreased even under high substrate condition and immobilized
psicose from d-fructose by a continuous bioreactor system using immo-
enzyme was able to produce d-allose continuously. The solu- bilized d-tagatose 3-epimerase. J Biosci Bioeng 2000;90:453–5.
tion after the reaction was very clean, thereby eliminating the [6] Wilson DM, Ajl S. Metabolism of l-rhamnose by Escherichia coli. I.
separation step to recover the immobilized enzyme for further l-Rhamnose isomerase. J Bateriol 1956;73:410–4.
reactions. The reason why the substrate concentration was 50% [7] Power J. The l-rhamnose genetic system in Escherichia coli K-12.
Genetics 1967;55:557–68.
(w/w) of d-psicose was that it is the best condition to reduce the
[8] Zarban AL, Heffernan S, Nishitani L, Ransone J, Wilcox G. Positive
formation of by-products as much as possible. Although we tried control of the l-rhamnose genetic system in Selmonella typhimurium
to use 20% (w/w) d-psicose, much more by-products were gen- LT2. J Bacteriol 1984;158:603–8.
erated as compared with 50% (w/w) d-psicose (data not shown). [9] Bhuiyan HS, Itami Y, Izumori K. Isolation of an l-rhamnose isomerase-
The bioreactor constructed in this study was able to convert d- constitutive mutant of Pseudomonas sp. strain LL172: purification and
characterization of the enzyme. J Ferment Bioeng 1997;84:319–23.
psicose to d-allose to the equilibrium point at the flow rate of
[10] Bhuiyan HS, Itami Y, Rokui Y, Katayama T, Izumori K. d-Allose
0.8 ml/min at 42 ◦ C, however, conversion ability was remark- production from d-psicose using immobilized l-rhamnose isomerase.
ably decreased after 17 days (Fig. 4), suggesting that the half J Ferment Bioeng 1998;85:539–41.
life of this bioreactor was approximately a half month in these [11] Leang K, Takada G, Fukai Y, Morimoto K, Granström TB, Izu-
conditions. After 17 days, the bioreactor could convert d-psicose mori K. Novel reactions of l-rhamnose isomerase from Pseudomonas
stutzeri and its relation with d-xylose isomerase via substrate specificity.
into d-allose until equilibrium point for more 13 days, when the
Biochim Biophys Acta 2004;1674:68–77.
flow rate was decreased down to 0.4 ml/min. Although, it was [12] Leang K, Ishimura A, Okita M, Takada G, Izumori K. Cloning,
possible to use for 10 more days, we did not try to use more 30 nucleotide sequence and overexpression of l-rhamnose isomerase gene
days to consider productivity. from Pseudomonas stutzeri in Escherichia coli. Appl Environ Microbiol
We tried to separate the reaction mixture to obtain pure d- 2004;70:3298–304.
[13] Dische Z, Borenfreund E. A new spectrophotometric method for the
allose crystal. The TREZONE system could separate 2.2 l of
detection of keto sugars and trioses. J Biol Chem 1951;192:583–7.
this reaction mixture per day. By this system, we can separate [14] Bradford MM. A rapid and sensitive method for the quantitation of
reaction mixture continuously for 10 days and obtained 3.6 l of protein utilizing the principle of protein–dye binding. Anal Biochem
purity 95% d-allose solution, although it contained d-psicose 1976;72:248–54.

You might also like