Sie sind auf Seite 1von 7

International Journal of Food Science and Technology 2015, 50, 1625–1631 1625

Original article
Lactulose production from lactose by recombinant cellobiose
2-epimerase in permeabilised Escherichia coli cells

Mingming Wang,1,2 Ruijin Yang,1,2* Xiao Hua,2 Qiuyun Shen,2 Wenbin Zhang2 & Wei Zhao2
1 State Key Laboratory of Food Science and Technology, Jiangnan University, 214122 Wuxi, China
2 School of Food Science and Technology, Jiangnan University, 214122 Wuxi, China
(Received 8 October 2014; Accepted in revised form 20 January 2015)

Summary Caldicellulosiruptor saccharolyticus cellobiose 2-epimerase (CsCE) catalyses the single substrate lactose
into lactulose and is the most efficient enzyme ever found for the enzymatic synthesis of lactulose. Etha-
nol-permeabilised Escherichia coli cells containing CsCE were used as biocatalysts for lactulose produc-
tion. The reaction conditions for maximum lactulose production were optimised to be 600 g L 1 lactose,
pH 7.5, 80 °C and 12.5 U mL 1 of whole-cell biocatalyst. After incubated at Na2HPO4–NaH2PO4 buffer
for 2 h, approximately 390.59 g L 1 lactulose was obtained with a conversion yield of 65.1%. The lowest
production and conversion yield of epilactose were also found in Na2HPO4–NaH2PO4 buffer with a final
concentration of 11.7 g L 1 and a conversion yield <2%. The results represent a promising technology to
attain high production and conversion yield of lactulose with a high purity on industrial scale.
Keywords Cellobiose 2-epimerase, lactose, lactulose, optimum reaction conditions, permeabilised Escherichia coli cells.

and b-galactosidase (EC 3.2.1.23) in free and immobi-


Introduction
lised forms (Kim et al., 2006; Mayer et al., 2010; Kha-
Lactose is a cheap disaccharide and several million tami et al., 2014). Low conversion yield and high costs
tons of lactose in the form of lactose-enriched cheese of enzymes are the main hurdles for the enzymatic
whey is regarded as waste and discarded annually (Mu method. In consideration of these major obstacles,
et al., 2013). Hence, lactose as a raw material for the many attempts aimed at increasing the lactulose con-
production of lactose derivatives is probably a practi- version yield and lowering the production costs have
cal way for its efficient use (Seki & Saito, 2012). Lactu- been carried out (Bednarski & Kulikowska, 2007;
lose (4-O-b-D-galactopyranosyl-D-fructose), another Tang et al., 2010; Song et al., 2012a,b). However, the
bioactive lactose derivative produced from the isomeri- enzymatic synthesis of lactulose has not yet been stud-
sation of lactose, is a nondigestible disaccharide that is ied on an industrial scale.
widely used in pharmaceutical, nutraceuticals and food Recently, cellobiose 2-epimerase from Caldicellulosi-
industries (Schumann, 2002; Aider & de Halleux, 2007; ruptor saccharolyticus has been reported to be effective
Olano & Corzo, 2009). Lactulose has a large market for direct conversion lactose into lactulose. The highest
and its demand increased considerably in recent years conversion yield (88%) and lactulose concentration
(Playne & Crittenden, 2009; Panesar & Kumari, 2011). (614 g L 1) were achieved (Park et al., 2011; Kim &
Commercially, lactose is exclusively produced by Oh, 2012; Kim et al., 2013; Ojima et al., 2013). Due to
alkaline isomerisation of lactose (Hicks & Parrish, the high conversion yield of lactulose from lactose and
1980; Kozempel et al., 1995; Panesar & Kumari, its thermostable property, biological synthesis of lactu-
2011). As a promising alternative to this chemical lose with CsCE will contribute significantly to the
method, enzymatic production of lactulose regarding industry. However, the complex separation and purifi-
product safety can result in clean production, and easy cation steps and activity loss of the purified enzymes
purification of the product would be desirable (Panesar during processing will sharply increase the production
& Kumari, 2011). Enzymatic synthesis of lactulose is expense. The use of whole cells as biocatalysts for lac-
commonly performed by b-glycosidase (EC 3.2.1.21) tulose production has advantages over purified enzymes
in many bioconversion processes because it is an eco-
*Correspondent: Fax: +86-510-85919150; nomical and easy to handle process. However, the per-
e-mail: yrj@jiangnan.edu.cn meability barrier of the cell envelope for substrates and

doi:10.1111/ijfs.12776
© 2015 Institute of Food Science and Technology
1626 Enzymatic production of lactulose M. Wang et al.

products often causes low reaction rates in whole cells and then centrifugated, and after freeze-dried, the per-
(Aider & de Halleux, 2007). To reduce the permeability meabilised E. coli cells were stored at 4 °C before being
barrier, ethanol permeabilisation of cells has been dem- used as a biocatalyst for lactulose production.
onstrated to be an economical, easy, convenient and
safe process for production of lactulose (Lee et al., Enzyme assay
2004; Panesar et al., 2007). Unless otherwise stated, the reaction was carried out
This study is concerned with the preparation of at 80 °C in Tris–HCl buffer (50 mM, pH 7.5) contain-
freeze-dried ethanol-permeabilised E. coli cells contain- ing 50 g L 1 lactose and 0.1 g L 1 purified enzyme or
ing CsCE and their use as biocatalysts for lactulose freeze-dried biocatalyst for 20 min. The enzymatic
production. Reaction conditions including pH, temper- reaction was stopped by boiling for 5 min. Controls
ature, reaction time, substrate concentration, volumet- were prepared at the same conditions with inactivated
ric activity of the biocatalyst, and molar ratio of enzyme. One unit (U) of the CsCE activity was defined
borate–lactose and buffer systems were optimised to as the amount of free enzyme or permeabilised E. coli
obtain maximum lactulose production. cells required to produce 1 lmol of lactulose from lac-
tose per min at 80 °C and pH 7.5.
The quantification of lactulose was based on the
Materials and methods
method described by Zhang et al. (2010b).
Materials
Effect of pH and temperature on enzyme activity of perme-
Caldicellulosiruptor saccharolyticus DSM 8903 was abilised E. coli cells
generously gifted by Department of Applied Microbi- The optimal pH was investigated using two different
ology, University of Lund (Sweden). Lactulose and pH buffer systems [PIPES buffer (50 mM, pH 6.0–7.0)
epilactose (HPLC grade) were purchased from Sigma- and Tris–HCl buffer (50 mM, pH 7.0–8.5)]. The effect
Aldrich (Shanghai, China). Water was obtained from of temperature on enzyme activity was varied from 40
a MilliQ water purification system (Millipore, Mols- to 95 °C in Tris–HCl buffer (50 mM, pH 7.5).
heim, France). All the other chemicals were of analyti-
cal grade except acetonitrile (HPLC grade) and Optimisation of reaction conditions for lactulose production
purchased from Sinopharm Chemical Reagent Co. Ltd by permeabilised E. coli cells
(Shanghai, China). All solutions were prepared with The various reaction conditions were optimized by
distilled water and used without further purification. varying the respective parameters. The volumetric
activity of the biocatalyst (2.5–20 U mL-1), the sub-
strate concentration of lactose (50–700 g L-1), and the
Methods
enzymatic reaction time (0–5 h) and the borate concen-
Microorganisms, plasmid, culture conditions and enzyme tration (0–3 M) were optimized to obtain the maxi-
purification mum lactulose production. Unless otherwise stated,
Caldicellulosiruptor saccharolyticus DSM 8903, Escheri- the reactions were carried out at 80°C in Tris–HCl
chia coli BL21 (DE3) and plasmid pET-28(a+) were buffer (50 mM, pH 7.5). Finally, the influence of buf-
used as the source of genomic DNA of cellobiose fer systems (a. Tris–HCl buffer; b. PIPES buffer; c.
2-epimerase, host cells and the expression vector. Na2HPO4–NaH2PO4 buffer; d. Deionized water; e.
Recombinant E. coli cells for enzyme expression were Na2HPO4–Citrate buffer; f. Borate buffer solution; g.
cultivated in 50 mL Luria-Bertani (LB) medium in 250- Britton-Robinson buffer) on the production of lactu-
mL flasks containing 10 lg mL 1 of Kanamycin at lose was also investigated. All the buffers were 50 mM
37 °C with shaking at 200 rpm min 1. When the opti- and adjusted to pH 7.5.
cal density reached 0.6 at 600 nm, lactose was added to
a final concentration of 10 g L 1 to induce cellobiose Analytical methods
2-epimerase expression. The culture was incubated with Lactulose produced during the enzymatic reactions were
shaking at 160 rpm min 1 at 25 °C for 20 h. quantitatively monitored by HPLC analysis as described
The purification step of the enzyme was carried out by Hua et al. (2013).
using Q Sepharose Fast Flow and Sephadex G-75 col-
umn chromatography (Gu et al., 2014).
Statistical analysis
Permeabilisation of recombinant E. coli cells The results expressed in various studies were reported
The cultivated cells were harvested by centrifugation at as mean  SD (standard deviation). Each value repre-
6000 g for 15 min and then resuspended in 40% (v/v) sents the mean for three independent experiments per-
ethanol, stirred at 4 °C for 30 min. The obtained per- formed in duplicate with average standard deviation
meabilised cells were washed twice with distilled water <5%. SPSS statistical software 17.0 (SPSS Inc.,

International Journal of Food Science and Technology 2015 © 2015 Institute of Food Science and Technology
Enzymatic production of lactulose M. Wang et al. 1627

Chicago, IL, USA) was used to perform the statistical higher ethanol concentration and longer permeabilisa-
analysis. tion time. Thus, treated with 40% (v/v) ethanol for
30 min were chosen as the optimum conditions for
E. coli cells permeabilisation in this study.
Results and discussion
Effects of pH and temperature on enzyme activity of
permeabilised E. coli cells and free enzyme were inves-
Permeabilisation and biochemical properties of
tigated. Both free enzyme and permeabilised E. coli
permeabilised E. coli cells
cells exhibited an optimal pH of 7.5 in 50 mM Tris–
To evaluate the effectiveness of ethanol as a permeabil- HCl buffer, and it can be noted that under neutral pH
ising agent, the cultivated E. coli cells were treated conditions, the relative activity was at a high level
with different ethanol concentration (0–80%, v/v) and (Fig. 1c). The activity-temperature profiles of the free
different permeabilisation time (0–75 min). Figure 1a,b enzyme and permeabilised E. coli cells are shown in
showed that the maximum activity of E. coli cells was Fig. 1d. The optimum temperature for the biocatalyst
obtained when treated with 40% (v/v) ethanol for and free enzyme was 80 °C. The free enzyme activity
30 min. In permeabilisation, the cell envelope is altered decreased significantly below and above 80 °C, while
to allow small molecules, such as lactose and lactulose the biocatalyst exhibited some thermal stability at tem-
to cross freely (Panesar et al., 2007). However, the peratures ranging from 75 to 85 °C (Fig. 1d). This
enzyme activity decreased may be attributed to the could be attributed to the microenvironment of the
denaturing effect of the ethanol on the enzyme and the cellobiose 2-epimerase in permeabilised E. coli cells.
leakage of the enzyme from the cells or cell lysis at The environment did not change greatly and the cells

(a) (c)

(b) (d)

Figure 1 Ethanol permeabilisation and biochemical properties of permeabilised Escherichia coli cells and free enzyme. The activity of
untreated E. coli cells was defined as 100% in (a and b). In (c and d), the one with the highest activity was taken as 100%.

© 2015 Institute of Food Science and Technology International Journal of Food Science and Technology 2015
1628 Enzymatic production of lactulose M. Wang et al.

could protect the enzyme activity conformation from concentration of permeabilised yeast cells for lactulose
distortion or damage by heat exchange. It is more effi- production was 10.4 g L 1. Up to this value, the pro-
cient to produce lactulose at high temperatures ductivity of lactulose increased with the adding of b-
because increased temperature favours substrate solu- galactosidase in cell mass. And then the productivity
bility; hence, high concentration of lactose can be used of lactulose decreased with increasing cell mass due to
and high concentration of lactulose can be obtained. steric hindrance. Thus, in this study, 12.5 U mL 1 was
In addition, high temperature also resulted in a higher chosen as the optimal volumetric activity of biocatalyst
reaction velocity of the enzyme, and a higher produc- for maximum lactulose production.
tivity of lactulose was achieved (Lee et al., 2004; Kim
& Oh, 2012). On the other hand, nonenzymatic brown-
Effect of the substrate concentration of lactose on
ing (Mallard reactions) could be initiated at high tem-
lactulose production using permeabilised E. coli cells as
peratures (Mayer et al., 2004). Future studies are
biocatalyst
needed to find new strains that display high CsCE
activity at low temperatures or facilitate lactulose As shown in Fig. 3, the production and conversion
production by improving the properties of cellobiose yield of lactulose were recognised as the two indexes
2-epimerase by protein engineering. This could mini- to determining the optimal concentration of lactose. It
mise the unwanted coloured by-products generated by was found that lactulose production significantly
nonenzymatic browning during biotransformation and increased with addition of lactose. When lactose con-
thus improve the product quality of lactulose. centration was varied from 50 to 700 g L 1, the lactu-
lose almost increased linearly from 30.5 to
356.3 g L 1. When the concentration of lactose was
Effect of the volumetric activity of the biocatalyst on
below 400 g L 1, the conversion yield of lactulose was
lactulose production
almost constant at about 60%. Above 400 g L 1, the
The effect of biocatalyst volumetric activity on the conversion yield of lactulose dramatically decreased to
maximum lactulose production was examined under approximately 55% at 600 g L 1 and 50.9% at
the given conditions with biocatalyst volumetric activ- 700 g L 1. Although the maximum lactulose produc-
ity ranging from 2.5 to 20 U mL 1. As shown in tion was obtained at 700 g L 1 lactose, the conversion
Fig. 2, it was distinctly found that the lactulose pro- yield was at a lower level. Taking the two key indexes
duction increased with the addition of biocatalyst, and into consideration, 600 g L 1 lactose was selected as
the maximum values were obtained at 12.5 U mL 1. the right substrate concentration for this study.
Lactulose concentration increased with the increase of The concentration of lactulose increasing with
biocatalyst, due to the increased amount of CsCE increase in lactose has also been found in other enzy-
present. However, when above 12.5 U mL 1, lactulose matic syntheses of lactulose by b-galactosidase. The
production was constant at approximately 275 g L 1. transgalactosylation activity of b-galactosidase increase
This phenomenon has also been found by other with decreasing water activity, caused by the high con-
researchers. Lee et al. (2004) found that the optimal centrations of substrates (Hanssen et al., 2001; Lee

Figure 2 Effect of the volumetric activity of the biocatalyst on Figure 3 Effect of substrate concentration of lactose on lactulose
lactulose production. production.

International Journal of Food Science and Technology 2015 © 2015 Institute of Food Science and Technology
Enzymatic production of lactulose M. Wang et al. 1629

et al., 2004). The decrease in conversion yield may be


Effect of borate on lactulose production using
caused by the high viscosity of lactose.
permeabilised E. coli cells as biocatalyst
Boric acid has successfully been used for achieving high
The time course of lactulose production using
yields in enzymatic production of D-psicose, D-taga-
permeabilised E. coli cells as biocatalyst
tose, L-ribulose and lactulose (Kim et al., 2008, 2013;
Time course of the production of lactulose from the Zhang et al., 2010a; Salonen et al., 2013). As shown in
single substrate lactose was determined by varying the Fig. 5, the influence of borate on lactulose production
reaction time from 0 to 6 h. As shown in Fig. 4, it was determined by varying the concentration of borate
was noted that up to 2 h the lactulose concentration from 0 to 3 M. When the molar ratio of borate–lactose
significantly increased from 0 to 318 g L 1. After 2 h, ranged from 0 to 1, the conversion yield of lactulose
the growth curve gradually became flatted. This phe- increased significantly, and the highest yield was
nomenon was also observed by other researchers, and achieved at a 1:1 molar ratio. After this ratio, the lactu-
it may attributed to the half-life of biocatalyst (Park lose yield decreased. Compared with the biological syn-
et al., 2011; Kim & Oh, 2012; Kim et al., 2013). The thesis in the absence of borate, the yield of lactulose
reaction time of lactulose production is of key impor- showed an approximately 13% increase from 53% to
tance for industrial application. Long reaction times 66% at a 1:1 molar ratio of borate–lactose. The
lead to a low productivity of lactulose. In addition, increased lactulose conversion yield can be explained by
long reaction times at high temperatures and high lac- the fact that the binding affinity of borate for lactulose
tose concentrations may lead to the generation of col- is 59 times higher than that for lactose (Vandenberg
oured by-products through nonenzymatic browning et al., 1994). The addition of borate in enzymatic lactu-
(Mayer et al., 2004). Hence, in this study, 2 h was cho- lose production allows the formation of borate–lactu-
sen as the optimal reaction time for lactulose produc- lose complex which means the newly formed lactulose
tion. After 2 h of enzymatic reaction, approximately was essentially removed and allows the continuation of
318.3 g L 1 lactulose was obtained. lactulose formation (Kim et al., 2013).
The reaction time needed to achieve a constant lac- The concentration of borate used in the reaction
tulose concentration by free cellobiose 2-epimerase solution in this study was 120 g L 1. The high con-
was 2 h. It was almost the same with that achieved centration of borate may cause numerous problems
by permeabilised E. coli cells. This result may be due including complex desalination by ion-exchange or
to the reduction of the permeability barrier caused by nanofiltration, severe environmental pollution and
treating the cells with ethanol. After ethanol perme- high cost. In addition, the stability of the borate–lac-
abilisation, the cells show a faster mass transfer of tose complex is strongly dependent on pH. In the
substrate and products which may result in a more weak basic environment of the reaction solution, the
efficient lactulose conversion similar to that of free coordination between lactulose and borate is pro-
enzyme. moted and this results in their difficult separation

Figure 5 Effect of the molar ratio of borate–lactose on lactulose


Figure 4 Effect of reaction time on lactulose production. production.

© 2015 Institute of Food Science and Technology International Journal of Food Science and Technology 2015
1630 Enzymatic production of lactulose M. Wang et al.

(Geffen et al., 2006). With respect to those results, it production was also found in Na2HPO4–NaH2PO4
can be concluded that it is cost-effective for lactulose buffer (50 mM, pH 7.5) with a final concentration of
production using biological synthesis in the absent of 11.7 g L 1 after 2 h (Fig. 6b). Further studies are
borate. needed to understand why the epilactose production
changed in different buffer systems.
According to literature, the maximum lactulose
Effect of different buffer systems on the production of
isomerisation rate was reported to be 20% when
lactulose and epilactose
2.1–8.6% of phosphates were added to a saturated
CsCE not only has the ability to convert lactose into lactose solution at 104 °C for 20–240 min (patent of
lactulose, but can also epimerise lactose into epilactose Great Brittan No. 2031430, 1978). It has been dem-
(Park et al., 2011). The yield of lactulose and the pro- onstrated that sulphites and phosphates have the
duction of epilactose in different buffer systems were ability to prevent oxidation of disaccharides. For this
investigated. Figure 6a shows that the lactulose reason, their use as catalysts for lactose isomerisa-
conversion yield was markedly different in various buf- tion reactions into lactulose allows the use of high
fer systems. When the reaction was performed in temperatures and lactose concentrations (Aider & de
Na2HPO4–NaH2PO4 buffer (50 mM, pH 7.5), the high- Halleux, 2007). In this study, a high lactose concen-
est conversion yield of lactulose was achieved with a tration and high temperature reaction system was
value of 65.1%. Interestingly, the lowest epilactose used. A Na2HPO4–NaH2PO4 buffer system resulted
in the highest lactulose conversion yield and the low-
est epilactose production. These are probably the
right conditions for the production of clean and safe
(a) lactulose with higher purity.

Conclusions
This study developed a biocatalytic method for lactu-
lose production using recombinant CsCE in ethanol-
permeabilised E. coli cells. Biocatalysts in the form of
whole cells have advantages over purified enzymes
because they are simpler, easy to handle and more eco-
nomical in many industrial bioconversion processes.
The maximum lactulose yield was 390.59 g L 1 with a
conversion yield of 65.1%. Na2HPO4–NaH2PO4 buffer
can reduce epilactose yield thus increase lactulose pro-
duction and the final concentration of epilactose was
(b) just 11.7 g L 1 with a conversion yield <2%. This
shows a great potential in the production of high pur-
ity lactulose.

Acknowledgment
The authors gratefully acknowledge the financial sup-
port provided by the National Key Technology R&D
Program in the 12th Five Year Plan of China
(2011BAD23B03), the Key Project of National Natu-
ral Science Fund (31230057) and 111 Project (B07029).
This work was also supported by the Fundamental
Research Funds for the Central Universities (JUD-
CF13001) and the Innovative Research Program for
Graduate Students of Jiangsu Province
(CXZZ13_0762). We thank Dr. (Serve) Kengen from
Figure 6 Effect of different buffer systems on the production of (a) Wageningen UR (Holland) and Dr. Ed van Niel from
lactulose and (b) epilactose. Unless otherwise stated, all the buffers University of Lund (Sweden) for the generous dona-
were adjusted to pH 7.5 in 50 mM. a: Tris-HCl buffer; b: PIPES buf- tion of the genomic DNA from Caldicellulosiruptor
fer; c: Na2HPO4–NaH2PO4 buffer; d: Deionized water; e: Na2HPO4- saccharolyticus DSM 8903 and we also thank Dr. Wu
Citrate buffer; f: Borate buffer solution; g: Britton-Robinson buffer. Li for valuable suggestions.

International Journal of Food Science and Technology 2015 © 2015 Institute of Food Science and Technology
Enzymatic production of lactulose M. Wang et al. 1631

tion of epilactose. Applied Microbiology and Biotechnology, 97,


References 1821–1827.
Aider, M. & de Halleux, D. (2007). Isomerization of lactose and lac- Ojima, T., Saburi, W., Yamamoto, T., Mori, H. & Matsui, H. (2013).
tulose production: review. Trends in Food Science & Technology, Identification and characterization of cellobiose 2-epimerases from
18, 356–364. various aerobes. Bioscience, Biotechnology, and Biochemistry, 77,
Bednarski, W. & Kulikowska, A. (2007). Influence of two-phase sys- 189–193.
tem composition on biocatalytic properties of b-galactosidase prep- Olano, A. & Corzo, N. (2009). Lactulose as a food ingredient.
arations. Chemical Papers, 61, 364–369. Journal of the Science of Food and Agriculture, 89, 1987–1990.
Geffen, N., Semiat, R., Eisen, M.S., Balazs, Y., Katz, I. & Dosoretz, Panesar, P.S. & Kumari, S. (2011). Lactulose: production, purifica-
C.G. (2006). Boron removal from water by complexation to polyol tion and potential applications. Biotechnology Advances, 29, 940–
compounds. Journal of Membrane Science, 286, 45–51. 948.
Gu, J., Yang, R., Hua, X., Zhang, W. & Zhao, W. (2015). Adsorp- Panesar, R., Panesar, P.S., Singh, R.S., Kennedy, J.F. & Bera, M.B.
tion-based immobilization of Caldicellulosiruptor saccharolyticus (2007). Production of lactose-hydrolyzed milk using ethanol per-
cellobiose 2-epimerase on Bacillus subtilis spores. Biotechnology meabilized yeast cells. Food Chemistry, 101, 786–790.
and Applied Biochemistry, 62, 237–244. Park, C.S., Kim, J.E., Choi, J.G. & Oh, D.K. (2011). Characteriza-
Hanssen, T., Andersson, M., Wehtje, E. & Adlercreutz, P. (2001). tion of a recombinant cellobiose 2-epimerase from Caldicellulosi-
Influence of water activity on the competition between b-glycosi- ruptor saccharolyticus and its application in the production of
dase catalysed transglycosylation and hydrolysis in aqueous hexa- mannose from glucose. Applied Microbiology and Biotechnology,
nol. Enzyme and Microbial Technology, 29, 527–534. 92, 1187–1196.
Hicks, K. & Parrish, F. (1980). A new method for the preparation Patent of Great Britain No. 2031430. Process for lactulose produc-
of lactulose from lactose. Carbohydrate Research, 82, 393–397. tion. 29. 09. 1978.
Hua, X., Yang, R., Shen, Q., Ye, F., Zhang, W. & Zhao, W. (2013). Playne, M. J. & Crittenden, R. G. (2009). Galacto-oligosaccharides
Production of 1-lactulose and lactulose using commercial beta- and other products derived from lactose. In: Advanced Dairy Chem-
galactosidase from Kluyveromyces lactis in the presence of fruc- istry: Lactose, Water, Salts and Major Constituents, 3rd edn, vol. 3
tose. Food Chemistry, 137, 1–7. (edited by P.L.H. McSweeney & P.F. Fox). Pp. 155. New York:
Khatami, S., Zokaee Ashtiani, F., Bonakdarpour, B. & Mehrdad, Springer.
M. (2014). The enzymatic production of lactulose via transglycosy- Salonen, N., Salonen, K., Leisola, M. & Nyyssola, A. (2013).
lation in conventional and non-conventional media. International D-Tagatose production in the presence of borate by resting Lacto-
Dairy Journal, 34, 74–79. coccus lactis cells harboring Bifidobacterium longum L-arabinose
Kim, Y.S. & Oh, D.K. (2012). Lactulose production from lactose as isomerase. Bioprocess and Biosystems Engineering, 36, 489–497.
a single substrate by a thermostable cellobiose 2-epimerase from Schumann, C. (2002). Medical, nutritional and technological proper-
Caldicellulosiruptor saccharolyticus. Bioresource Technology, 104, ties of lactulose. An update. European Journal of Nutrition, 41
668–672. (Suppl 1), I17–I25.
Kim, Y.S., Park, C.S. & Oh, D.K. (2006). Lactulose production Seki, N. & Saito, H. (2012). Lactose as a source for lactulose and
from lactose and fructose by a thermostable b-galactosidase from other functional lactose derivatives. International Dairy Journal, 22,
Sulfolobus solfataricus. Enzyme and Microbial Technology, 39, 110–115.
903–908. Song, Y S., Lee, H. U., Park, C. & Kim, S. W. (2012a). Batch and
Kim, N.H., Kim, H.J., Kang, D.I. et al. (2008). Conversion shift of continuous synthesis of lactulose from whey lactose by immobilized
D-fructose to D-psicose for enzyme-catalyzed epimerization by b-galactosidase. Food Chemistry, 136, 689–694.
addition of borate. Applied and Environment Microbiology, 74, Song, Y.S., Shin, H.Y., Lee, J.Y., Park, C. & Kim, S.W. (2012b).
3008–3013. b-Galactosidase-immobilised microreactor fabricated using a
Kim, Y.S., Kim, J.E. & Oh, D.K. (2013). Borate enhances the pro- novel technique for enzyme immobilisation and its application
duction of lactulose from lactose by cellobiose 2-epimerase from for continuous synthesis of lactulose. Food Chemistry, 133, 611–
Caldicellulosiruptor saccharolyticus. Bioresource Technology, 128, 617.
809–812. Tang, L., Li, Z., Dong, X., Yang, R., Zhang, J. & Mao, Z. (2010).
Kozempel, M.F., Kurantz, M.J., Craig, J.C. Jr & Hicks, K.B. Lactulose biosynthesis byb-galactosidase from a newly isolate
(1995). Development of a continuous lactulose process: separation Arthrobacter sp. Journal of Industrial Microbiology and Biotechnol-
and purification. Biotechnology Progress, 11, 592–595. ogy, 38, 471–476.
Lee, Y.J., Kim, C.S. & Oh, D.K. (2004). Lactulose production by Vandenberg, R., Peters, J.A. & Vanbekkum, H. (1994). The struc-
beta-galactosidase in permeabilized cells of Kluyveromyces lactis. ture and (local) stability-constants of borate esters of monosaccha-
Applied Microbiology and Biotechnology, 64, 787–793. rides and disaccharides as studied by B-11 and C-13 nmr-
Mayer, J., Conrad, J., Klaiber, I., Lutz Wahl, S., Beifuss, U. & spectroscopy. Carbohydrate Research, 253, 1–12.
Fischer, L. (2004). Enzymatic production and complete nuclear Zhang, Y.W., Jeya, M. & Lee, J.K. (2010a). L-Ribulose production
magnetic resonance assignment of the sugar lactulose. Journal of by an Escherichia coli harboring L-arabinose isomerase from Bacil-
Agricultural and Food Chemistry, 52, 6983–6990. lus licheniformis. Applied Microbiology and Biotechnology, 87,
Mayer, J., Kranz, B. & Fischer, L. (2010). Continuous production of 1993–1999.
lactulose by immobilized thermostable beta-glycosidase from Pyro- Zhang, Z., Wang, H., Yang, R. & Jiang, X. (2010b). A novel spec-
coccus furiosus. Journal of Biotechnology, 145, 387–393. trophotometric method for quantitative determination of lactulose
Mu, W., Li, Q., Fan, C., Zhou, C. & Jiang, B. (2013). Recent in food industries. International Journal of Food Science & Technol-
advances on physiological functions and biotechnological produc- ogy, 45, 258–264.

© 2015 Institute of Food Science and Technology International Journal of Food Science and Technology 2015

Das könnte Ihnen auch gefallen