Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Food Chemistry 121 (2010) 307318

Contents lists available at ScienceDirect

Food Chemistry
journal homepage: www.elsevier.com/locate/foodchem

Review

Recent advances rening galactooligosaccharide production from lactose


Aaron Gosling a,b, Geoff W. Stevens a, Andrew R. Barber c, Sandra E. Kentish a, Sally L. Gras a,b,*
a
The Department of Chemical and Biomolecular Engineering, The University of Melbourne, Victoria 3010, Australia
b
The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
c
Innovative Food and Plants Division, South Australian Research and Development Institute, Regency International Centre, Days Road, Regency Park, SA 5010, Australia

a r t i c l e i n f o a b s t r a c t

Article history: The production of prebiotic galactooligosaccharides (GOS) by the b-galactosidase catalysed conversion of
Received 26 June 2009 lactose has become commercially important. Yet it remains a challenge to sufciently understand the
Received in revised form 21 October 2009 structure and activity of b-galactosidase, to increase the efciency of transgalactosylation and GOS pro-
Accepted 14 December 2009
duction and to improve the quality of GOS products in a rational way. This review covers the broad but
related aspects of GOS synthesis including: the structure and reaction mechanism of b-galactosidase, fac-
tors effecting yield and productivity of GOS synthesis systems, the structure of GOS products, models for
Keywords:
the kinetics of GOS synthesis and reactor congurations for GOS synthesis. It aims to couple recent dis-
Galactooligosaccharide
Galactosyl oligosaccharide
coveries with established knowledge to enhance understanding of the complex biochemistry of GOS
Galacto-oligosaccharide synthesis.
GOS 2009 Elsevier Ltd. All rights reserved.
Prebiotic
Galactosyl transfer
b-galactosidase
Lactose hydrolysis
Review

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
2. Reaction mechanism and structure of b-galactosidase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
3. Factors effecting GOS yields . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 309
4. Structure of GOS products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311
5. Modelling of the kinetics of GOS synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313
6. Reactor configurations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314
6.1. Whole cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314
6.2. Membrane processing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314
7. Outlook . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315

1. Introduction as galactooligosaccharides (GOS) are an example of functional


foods, as they are prebiotic (Van Loo et al., 1999). A prebiotic has
Functional foods are generally dened as foods or food ingredi- been dened as being a selectively fermented ingredient that al-
ents which impart a health benet above and beyond the nutri- lows specic changes, both in the composition and/or activity in
tional value expected from food. The group of foods referred to the gastrointestinal microora that confers benets upon host well
being and health (Roberfroid, 2007). Simply put, GOS are not di-
gested by humans or other animals, and selectively increase the
* Corresponding author. Address: The Department of Chemical and Biomolecular
benecial microora of the intestine, leading to health benets
Engineering, The University of Melbourne, Victoria 3010, Australia. Tel.: +61 3 8344
6281; fax: +61 3 8344 4153.
that are extensively recognised (Macfarlane, Steed, & Macfarlane,
E-mail address: sgras@unimelb.edu.au (S.L. Gras). 2008).

0308-8146/$ - see front matter 2009 Elsevier Ltd. All rights reserved.
doi:10.1016/j.foodchem.2009.12.063
308 A. Gosling et al. / Food Chemistry 121 (2010) 307318

GOS molecules (for example, Gal (b 1 ? 4) Gal (b 1 ? 4) Glc) are glycosidic bond of beta stereochemistry, there is broad specicity
typically synthesised by the enzymatic activity of b-galactosidase for the aglycan moiety. Consequently, the R1 group (Fig. 1) could
on lactose in a reaction known as transgalactosylation. Other car- equally represent an oligosaccharide, when the galactoside sub-
bohydrate-modifying enzymes, such as b-glucosidases and b-gly- strate is a GOS molecule. Structural features of b-galactosidase that
cosidases, can also catalyse this chemistry. b-Galactosidase are important in substrate docking interactions within the active
belongs to a class of hydrolytic enzymes and has long been used site have been reported and include groups participating in hydro-
in the dairy industry to hydrolyse lactose, producing glucose and gen bonding, an electrostatically bound sodium ion and aromatic
galactose. This hydrolytic activity increases the sweetness of a amino acid side chains forming a platform essential for binding
dairy product, and also lowers lactose concentration, which can hexose substrates (Gloster et al., 2004; Hidaka et al., 2002; Juers
be benecial to lactose intolerant consumers (Lomer, Parkes, & et al., 2001; Nerinckx, Desmet, & Claeyssens, 2003).
Sanderson, 2008). The competing transgalactosylation reaction After the substrate has docked in the active site of the b-galac-
and formation of GOS during b-galactosidase-catalysed conversion tosidase, catalysis can occur. It is currently accepted that the reac-
of lactose was rst observed in the early 1950s (Wallenfels, 1951). tion occurs with a retaining mechanism. The details of the catalysis
Since this time, GOS have become a commercially important prod- are well described (Brs, Moura-Tamames, Fernandes, & Ramos,
uct manufactured in Asia and Europe (Crittenden & Playne, 1996), 2008; Juers et al., 2001; Vasella, Davies, & Bhm, 2002; Zechel &
with a manufacturing facility also opening recently in Australia. Six Withers, 2000). A covalent bond forms between the galactosyl
thousand tonnes of GOS were manufactured in 2005 in Japan alone moiety and the enzyme, followed by galactosyl transfer to an
(Taniguchi, 2005). acceptor nucleophile (Fig. 1).
With increasing public awareness of nutrition (Cowburn & The reaction pathway diverges at this point. The result of the
Stockley, 2005) there has been increased demand for foods with reaction depends on the identity of the galactosyl acceptor, for
demonstrable health benets (Borra & Bouchoux, 2009). An exam- which there is broad specicity. When the galactosyl acceptor is
ple of this can be found with yoghurts, where consumers put a high water (i.e., R2 = H in Fig. 1), free galactose is released. The result
preference on products perceived to be low in fat (Valli & Traill, of the reaction is hydrolysis of the galactoside, leading to degrada-
2005). Dairy products processed exploiting the GOS-forming activ- tion of both lactose and GOS.
ity of b-galactosidase could contain lower sugar content (e.g., lac- When the reaction pathway diverges towards galactosyl trans-
tose) and higher soluble bre (e.g., GOS) and therefore have a fer and GOS synthesis, the galactosyl acceptor is another saccha-
competitive edge. This makes further increases in GOS manufac- ride (i.e., R2 = glucose, galactose, lactose, or a GOS molecule). The
ture likely. mechanism of transgalactosylation by b-galactosidase has received
This review provides an overview of the recent advances in GOS less attention compared to that of hydrolysis. To our knowledge, all
production from lactose for food applications. The importance of currently published data on the mechanism of transgalactosylation
the eld is emphasised by the literature activity surrounding it, draws inferences from the hydrolytic reaction, rather than directly
with recent reviews published on the health benets and charac- observing transgalactosylation. It is not known if the interactions
teristics of prebiotic oligosaccharides (including GOS) (Barreteau, in the active site differ when the acceptor is a saccharide, com-
Delattre, & Michaud, 2006; Gnzle, Haase, & Jelen, 2008; Macfar- pared to water. However, it is well known that different enzymes
lane et al., 2008) and b-galactosidase enzymology (Mlichova & display different selectivities for water and saccharides, as differ-
Rosenberg, 2006; Panesar, Panesar, Singh, Kennedy, & Kumar, ent enzymes yield varying amounts of GOS at the same lactose
2006; Trincone & Giordano, 2006). Here, the focus is on recent ad- concentration (Prenosil, Stuker, & Bourne, 1987b; Zarate & Lopez-
vances concerning the reaction mechanism and structure of b- Leiva, 1990). It seems likely that this difference in the GOS yields
galactosidase, factors effecting GOS yields the structure of GOS is a result of structural and/or mechanistic differences between
products, models for the kinetics of GOS synthesis, and reactor con- b-galactosidases from different sources. The structures of GOS
gurations for GOS synthesis. Specically, this review aims to in- products may also vary due to differences in the structure and/or
crease understanding of scientic issues related to production of mechanism of different b-galactosidases.
GOS on a commercial scale for sale as a food product. Mutagenesis studies have suggested, if not demonstrated, some
of the structural features which are important in inuencing the
propensity of an enzyme for transferring galactosyl moieties to
2. Reaction mechanism and structure of b-galactosidase saccharides rather than water. Deleting the sequence coding for
580 amino acid residues at the C-terminal of the b-galactosidase
The biochemical mechanism for b-galactosidase activity is gene encoding BIF3 from Bidobacterium bidum yielded an en-
highly relevant to the synthesis of GOS products as it explains zyme that produced GOS but had little or no hydrolytic activity
how b-galactosidase, renowned for its hydrolytic activity, can olig- (Jrgensen, Hansen, & Stougaard, 2001). This C-terminal section
omerise galactosides under kinetic control. This mechanism also of the protein contained a domain with homology to known galact-
offers insight into some of the environmental factors which inu- ose binding domains. It was hypothesised that deletion of this do-
ence GOS synthesis and yield. main allowed greater steric access to the active site, favouring
A brief summary of the critical catalytic events and the struc- saccharide acceptors. A patent on this truncated enzyme has been
tural features of the enzyme important for these events will be pre- led for applications including production of dairy foods and die-
sented here, as previous reviews are available on the hydrolysis tary supplements (Jrgensen, Hansen, & Stougaard, 2007).
reaction mechanism of both b-galactosidases specically (Mahon- Mutagenesis of single amino acids has also proved informative.
ey, 1998), and glycoside hydrolases more generally (Vocadlo & Da- The GOS yield and pH optima of the b-glucosidase from Pyrococcus
vies, 2008). Recent studies on the structure of b-galactosidase with furiosus were both increased by single amino acid substitutions
substrates and/or inhibitors bound in the active site are particu- and a mutant enzyme carrying both changes gave a GOS yield of
larly relevant and will form the emphasis of this section. approximately double the native enzyme (Hansson, Kaper, van
The rst event during b-galactosidase activity is the docking of a der Oost, de Vos, & Adlercreutz, 2000). It seems likely that negative
substrate molecule. When the substrate is lactose the glycan moi- public opinion towards the inclusion of genetically modied ingre-
ety is galactosyl and the aglycan moiety is glucosyl (represented as dients in food may render this interesting approach unsuitable for
the R1 group in Fig. 1). While b-galactosidase shows high substrate dairy processing. Perhaps the true value lies in the deeper under-
specicity requiring substrates with galactosyl glycans linked by a standing of the structural features of the enzyme which impact
A. Gosling et al. / Food Chemistry 121 (2010) 307318 309

Fig. 1. The reaction mechanism for b-galactosidase activity on galactosides (adapted from Juers et al. (2001)). R1 represents the aglycan moiety of the galactoside substrate;
R2 represents the nucleophile galactosyl acceptor.

on the maximum possible GOS yield, and this could aid in vitro and vant GOS producing b-galactosidases becomes available there will
in silico screening to identify novel native b-galactosidases with be opportunities for informative comparison with this model
higher GOS-producing characteristics. enzyme.
To aid the development of bioinformatic tools, more structural Another data set that could prove useful for building bioinfor-
data on the enzymes currently employed in GOS production is re- matic tools to identify optimum b-galactosidases exists in the Car-
quired. The X-ray crystal structures of b-galactosidase from sev- bohydrate-Active Enzymes database (http://www.cazy.org
eral microbial sources have been deposited in the Protein Data (Cantarel et al., 2009)). This database classies enzymes which
Bank (http://www.rcsb.org (Berman et al., 2000)) (Table 1). How- act on carbohydrates according to their primary amino acid se-
ever, none of the enzymes with solved structures are known to be quence (Henrissat, 1991). b-Galactosidases are placed in a broad
used in food processing. The structure of the Escherichia coli classication of glycoside hydrolases, which are further organised
enzyme is well characterised (Matthews, 2005) and the transga- into glycoside hydrolase families (GHF). Enzymes designated as
lactosylation characteristics of this enzyme have also been thor- b-galactosidases (E.C. 3.2.1.23) fall into four GHFs: 1, 2, 35 and
oughly described (Chen, Ou-Yang, & Yeh, 2003; Huber, Kurz, & 42. GHF 1, 2 and 35 contain enzymes other than b-galactosidases,
Wallenfels, 1976; Juers, Hakda, Matthews, & Huber, 2003; while only b-galactosidase has currently been classied into GHF
Mladenoska, Winkelhausen, & Kuzmanova, 2008; Richard, Wester- 42. It has been stated that b-galactosidases from GHF 2 generally
feld, Lin, & Beard, 1995). As structural data from industrially rele- have higher activity for both hydrolysis and transfer (van den
Broek, Hinz, Beldman, Vincken, & Voragen, 2008) but a systematic
study investigating correlations between amino acid sequence
homology and enzyme characteristics has not yet been reported.
Table 1
b-Galactosidases with crystal structures deposited in the Protein Databank (Berman Such a study is hampered by a lack of both published amino acid
et al., 2000). sequences for the commonly-used, commercially-available GOS
producing b-galactosidases, and standard reaction conditions to
Microbial Source Reference
compare the GOS-producing characteristics of different enzymes
Escherichia coli Juers et al. (2001)
(e.g., maximum GOS yield at a standard lactose concentration with
Thermus thermophilus A4 Hidaka et al. (2002)
Sulfolobus solfataricus Gloster et al. (2004) other dened reaction parameters).
Arthrobacter sp. C2-2 Sklov et al. (2005)
Bacteroides fragilis Ramagopal et al. (2009) 3. Factors effecting GOS yields
Bacteroides Palani, Kumaran, Burley, and Swaminathan
thetaiotamicron (2009)
Penicillium sp. Rojas et al. (2004) The maximum concentration of GOS typically obtained in a
reaction varies widely, depending on reaction conditions, as does
310 A. Gosling et al. / Food Chemistry 121 (2010) 307318

Table 2
Maximum GOS concentrations achieved in different reaction conditions.

Enzyme sourcea Reaction conditions Lactose Maximum Reference


conversion GOS yieldb
Initial lactose Temperature pH Buffer composition
(%)
concentration (C)
Sulfolobus 600 g/l 80 6.0 50 mM potassium phosphate 70 52.5% (w/w) Park et al., (2008)
solfataricus
Aspergillus oryzae 500 g/l 40 4.5 20 mM citrate phosphate 56 52% (w/w) Neri et al. (2009)
Bullera singularis 180 g/l 50 6.0 50 mM sodium phosphate 72 50% (w/w) Cho, Shin, and Bucke (2003)
Aspergillus oryzae 1.67 M 40 4.5 pH adjusted with 1 M HCl 55 31% (mol/ Iwasaki et al. (1996)
mol)
Saccharopolyspora 600 g/l 70 7.0 pH maintained by addition of 1 M 75 41% (w/w) Nakao et al. (1994)
rectivirgula NaOH, 10 lM MnCl2
Sterigmatomyces 200 g/l 60 5.0 100 mM sodium acetate 63 39% (w/w) Onishi and Tanaka (1995)
elviae
Lactobacillus 205 g/l 37 6.5 50 mM phosphate containing 80 38% (w/w) Splechtna et al. (2006)
reuteri 1 mM MgCl2
Pyrococcus 270 g/l 70 5.5 20 mM sodium citrate 80 33% (w/w) Petzelbauer, Reiter, Splechtna,
furiosus Kosma, and Nidetzky (2000)
Bidobacterium 400 g/l 45 6.8 30 mM phosphate 60 32.5% (w/w) Hsu et al. (2007)
longum
Sulfolobus 270 g/l 70 5.5 20 mM sodium citrate 72 26% (w/w) Petzelbauer et al. (2000)
solfataricus
Kluyveromyces 400 g/l 40 7.0 200 mM potassium phosphate 92 24.8% (w/w) Chockchaisawasdee et al. (2005)
lactis containing 2 mM MgCl2
Thermotoga 500 g/l 80 6.0 50 mM potassium phosphate 50 19% (w/w) Ji et al. (2005)
maritima
Pyrococcus 745 mmol/kg 80 5.0 0.2 mol/kg citrate 73 18.1% (w/w) Bruins et al. (2003a)
furiosus
a
All enzymes are b-galactosidases, except the Sulfolobus solfataricus and Pyrococcus furiosus derived b-glycosidases.
b
GOS concentration as a percentage of initial lactose concentration.

the GOS yield (as a percentage of the initial lactose). This is true for lactose hydrolysis for a B. bidum b-galactosidase (Dumortier,
even when using b-galactosidases recognised for their propensity Brassart, & Bouquelet, 1994), demonstrating the need for high lac-
to form GOS, as shown in Table 2 (see Prenosil et al. (1987b) and tose concentrations to achieve the maximum transgalactosylation
Zarate et al. (1990), for past compilations). A yield of 315 g/l GOS velocity.
from 600 g/l lactose (52.5% (w/w) yield) has been reported (Park, By comparison, the contribution of water activity to transgalac-
Kim, Lee, Kim, & Oh, 2008); it appears that yields over 50% are tosylation is easier to probe. Yields of GOS have been found to be
not often exceeded. More typical optimized yields are between higher when using organic solvent systems that lower the water
30% and 40% (w/w). activity of the reaction media (Cruz-Guerrero, Gmez-Ruiz, Vinie-
The time at which a reaction is harvested has an inuence on gra-Gonzlez, Brzana, & Garca-Garibay, 2006). In some instances,
the maximum GOS concentration (and therefore yield on initial this increased yield was shown to be due to reduced hydrolysis
lactose) because GOS are simultaneously synthesised and degraded (Chen, Wei, & Hu, 2001; Maugard, Gaunt, Legoy, & Besson, 2003),
by b-galactosidase. The concentration of GOS at its peak is deter- consistent with the commonly accepted theory that decreasing
mined by the relative kinetics of synthesis and degradation (Buch- the water activity of reaction media increases the yields of enzy-
holz, Kasche, & Bornscheuer, 2005). As such, factors which increase matic transfer reactions by decreasing competing hydrolysis reac-
the maximum concentration of GOS can be thought of as factors tions (Trincone & Giordano, 2006). The toxicity of organic solvents
which increase the rate of GOS synthesis or decrease the rate of limits their use in food applications but these studies do demon-
degradation. strate that decreasing water activity contributes to increasing
With this in mind, it is not surprising that the initial lactose con- GOS yields. Therefore, the increase in GOS yield noted with in-
centration is well accepted as one of the key factors inuencing the creased initial lactose concentration is likely to result in part from
maximum GOS concentration achieved (Mahoney, 1998). This a decreased water activity within the reaction media due to high
dependence on initial lactose concentration has at least two con- saccharide concentrations.
tributing factors: increased availability of saccharide galactosyl Temperature can increase the solubility of lactose, which is rel-
acceptors, and decreased availability of water. The former would atively low at ambient temperatures (c.a. 220 g/l at 25 C (White,
be expected to increase the rate of GOS synthesis, and the latter 2000). Much recent research has therefore been directed towards
would be expected to decrease both the rate of GOS degradation the discovery and characterisation of thermostable enzymes (Bru-
and lactose hydrolysis. However, the relative importance of these ins, Strubel, van Lieshout, Janssen, & Boom, 2003a; Chen et al.,
two contributions is unclear and will be discussed here. 2008; Hatzinikolaou et al., 2005; Ji, Park, & Oh, 2005; Park et al.,
It is difcult to quantitatively assess the contribution of saccha- 2008). These enzymes allow processes to be operated at increased
ride acceptor concentration on the maximum GOS concentration. temperatures and therefore with higher initial lactose concentra-
Traditional MichaelisMenten enzyme kinetic constants, and espe- tions, which in turn can deliver increases in the maximum GOS
cially the rate constant at saturating substrate concentrations kcat, concentration achieved. For example, 315 g/l GOS was produced
are rarely reported for GOS formation. This is perhaps due to the from a solution of 600 g/l initial lactose compared to 150 g/l GOS
difculties in accurately quantifying GOS formation rates. When produced from a solution of 300 g/l initial lactose when both solu-
the Michaelis constant (Km) is quoted for transgalactosylation, it tions were at 80 C (Park et al., 2008). Interestingly, while the max-
is typically orders of magnitude higher than for lactose hydrolysis; imum GOS concentration increased in this instance, the yield of
e.g., 800 mM lactose for transgalactosylation compared to 13 mM GOS per gram of initial lactose did not vary across this wide range
A. Gosling et al. / Food Chemistry 121 (2010) 307318 311

of lactose concentrations. The possibility of operating at higher maximum amount of GOS formed (Neri et al., 2009). Enzymes from
temperature using thermostable enzymes also offers the advan- different sources again display different characteristics. For exam-
tage of limiting potential for microbial contamination within the ple, lactose utilisation by b-galactosidase from Kluyveromyces lactis
process. Increases in yields will, however, need to be balanced with was inhibited by both glucose and galactose but only galactose re-
the increased loss of enzyme activity at higher temperatures. Mail- duced the maximum GOS concentration achieved with this en-
lard reactions between amino side-chains on the enzyme and sug- zyme (Chockchaisawasdee, Athanasopoulos, Niranjan, & Rastall,
ars have been shown to become signicant in inactivating a 2005).
thermostable enzyme at and above 80 C (Bruins, Van Hellemond,
Janssen, & Boom, 2003b).
Temperature seems to further affect GOS synthesis indepen- 4. Structure of GOS products
dently of lactose concentration. There are several examples of
studies which conclude that GOS yields increase with increasing GOS structures can differ in saccharide composition, regiochem-
temperature (Boon, Janssen, & vant Riet, 2000; Hsu, Lee, & Chou, istry of the glycosidic linkages and the degree of polymerisation, as
2007). However, individual enzymes clearly display different char- illustrated in Fig. 2. There are a number of important consequences
acteristics in their response to temperature, as there are also exam- arising from these structural differences: GOS structures give dif-
ples of GOS yield remaining constant with varied temperature ferent chemical properties, in vitro evidence suggests that prebiotic
(Iwasaki, Nakajima, & Nakao, 1996; Prenosil, Stuker, & Bourne, microbes grow differently on oligosaccharides with different struc-
1987a; Splechtna et al., 2006). tures (Rycroft, Jones, Gibson, & Rastall, 2001; Sanz, Cote, Gibson, &
It has long been recognised that pH affects the kinetics of lac- Rastall, 2006a, 2006b; Sanz, Gibson, & Rastall, 2005) and GOS
tose hydrolysis and GOS synthesis by the E. coli b-galactosidase dif- structure also affects characteristics relevant to their use in food
ferently (Huber et al., 1976). This suggests that it may be possible applications. Understanding the factors which inuence the struc-
to selectively control the rates of GOS synthesis and degradation by ture of the GOS products may therefore help to design processes
varying the pH of the reaction medium, thereby increasing GOS that produce oligosaccharide motifs with better properties and
yields. However, this may be a characteristic which varies between higher yield. Some of the vast number of structurally different
individual enzymes, similar to the effect of temperature on yield. GOS species produced by a variety of enzymes acting on lactose
Many recent studies have also found similar pH optima for GOS have been compiled previously (Prenosil et al., 1987b; Zarate
synthesis and galactoside hydrolysis (Hsu, Yu, & Chou, 2006; Hsu et al., 1990; Mahoney, 1998). In this section, recent research and
et al., 2007; Ji et al., 2005; Park et al., 2008). This was the case with generalisations about the structures of GOS produced by industri-
b-galactosidase from Sulfolobus solfataricus, which showed an opti- ally relevant enzymes are highlighted.
mal pH for GOS synthesis at 6.0, close to the optimal for hydrolysis The composition of GOS produced from lactose by b-galactosi-
of the model colorimetric substrate ortho-nitro phenol galactoside dase usually has the structure GalnGlc, where n indicates the de-
at pH 6.5 (Park et al., 2008). gree of polymerisation (DP), and is typically 15 (Mussatto &
Galactose and/or glucose are commonly recognised as inhibi- Mancilha, 2007). However, Galn Gal structures are also known
tors of lactose hydrolysis for many b-galactosidases, with galactose (Sanz, Sanz, & Martinez-Castro, 2002), as well as branched struc-
typically exerting a greater effect (Hatzinikolaou et al., 2005; Jura- tures, which contain multiple galactose residues attached to a sin-
do, Camacho, Luzn, & Vicaria, 2004). Galactose inhibition of lac- gle glucose moiety at the reducing terminal of the oligosaccharide
tose hydrolysis is generally competitive, as galactose can form (Yanahira et al., 1995).
galactosylenzyme intermediates with b-galactosidase, precluding It has been well established that the regiochemistry of the GOS
lactose from the active site. Glucose inhibition of lactose hydrolysis product is primarily controlled by the identity of the enzyme used
appears more complex, as it inhibits some b-galactosidases com- (Wallenfels & Weil, 1972), and it seems likely this is related to the
petitively (Boon, Janssen, & van der Padt, 1999) and some uncom- structure and/or reaction mechanism of the enzyme. For example,
petitively (Demirhan, Apar, & zbek, 2008). Inhibition by these the galactosyl moiety moves deeper into the active site of the E. coli
monosaccharides has also been recently shown to affect the b-galactosidase when galactosyl-enzyme intermediates forms
amount of GOS produced. The presence of galactose reduced the (Juers et al., 2001). This makes the galactosyl-like moiety less ste-
rate at which the b-galactosidase from Aspergillus oryzae consumed rically accessible to a nucleophilic acceptor. This inaccessibility
lactose more than the presence of glucose, and both glucose and would be expected to favour galactosyl transfer, resulting in the
galactose, separately and in combination, appeared to lower the formation of a 1 ? 6 bond with the most sterically open alcohol

Fig. 2. Examples of how GOS structure can vary. (A) Composition, (B) regiochemistry and (C) degree of polymerisation. Abbreviations are Gal for galactose and Glc for glucose.
312 A. Gosling et al. / Food Chemistry 121 (2010) 307318

group (C6OH) of a saccharide acceptor. This is consistent with the lus circulans b-galactosidase, the abundance of Gal (b1 ? 4) Gal
b1 ? 6 linkage in allolactose (Gal b1 ? 6 Glc), the major transga- (b1 ? 4) Glc decreased from 95% to 35% of the total trisaccharides
lactosylation product of the activity of the E. coli enzyme on lactose between 1 and 24 h, respectively, while Gal (b1 ? 4) Gal (b1 ? 3)
(Jobe & Bourgeoi, 1972). This 1 ? 6 linkage also appears to be the Glc increased its abundance from 4% to 18% of the total trisaccha-
preferred regiochemistry produced between dimers of galactose by rides over the same time interval (Yanahira et al., 1995). This
the E. coli b-galactosidase, as Gal (b1 ? 6) Gal was the only regio- highlights the opportunities for more selective production of spe-
isomer of di-galactose detected after reaction of a synthetic Gal do- cic structural motifs in GOS products through reaction
nor (o-nitrophenyl galactoside) with a synthetic Gal acceptor (3-O- engineering.
methyl Glc) (Yoon & Mckenzie, 2005). Many of the industrially rel- The degree of polymerisation (DP) may also affect the prebi-
evant b-galactosidases also preferentially produce b1 ? 6 linkages otic efcacy of GOS. There is in vitro evidence that some Bidobac-
between saccharides (Table 3), suggesting similar interactions be- terium species preferentially catabolise trimeric and tetrameric
tween the enzyme and galactosyl moieties in these systems. To our GOS over disaccharides (Gopal, Sullivan, & Smart, 2001) or lactose
knowledge, no mechanistic explanation has been offered to date to (Amaretti et al., 2007). It also seems likely that higher DP GOS
explain the preferential production of other linkages, such as would be more resistant to fermentation in the gut, and would
b1 ? 4 or b1 ? 3. therefore deliver a prebiotic effect in more distal locations in
It seems that b-galactosidases from prebiotic microbes produce the intestine. It has been suggested that controlling the DP can re-
distinctly structured GOS mixtures and grow more rapidly on the duce possible unpleasant side-effects linked to GOS consumption,
GOS which were produced by their own b-galactosidases (Rabiu, such as atulence and a laxative effect, due to osmotic pressure in
Jay, Gibson, & Rastall, 2001). This can translate to greater prebiotic the intestinal lumen (Topping & Clifton, 2001). DP also affects the
efcacy. It has been demonstrated in a human clinical trail that characteristics of GOS relevant to its incorporation into food
GOS produced by b-galactosidase from the prebiotic microbe B. products, such as sweetness and gelling properties (Delzenne,
bidum had different structure and signicantly greater prebiotic 2003).
potency compared to a commercial GOS mixture (Vivinal GOS As with regiochemistry, the typical DP of a GOS product is char-
produced by b-galactosidases from A. oryzae) (Depeint, Tzortzis, acteristic of the b-galactosidase used (Prenosil et al., 1987a) and is
Vulevic, IAnson & Gibson, 2008). The GOS produced by the B. bi- also a function of the reaction time. For example, trimeric GOS
dum b-galactosidase for this trial contained a majority of b1 ? 3 need to be present to act as galactosyl acceptors, before tetrameric
linkages, with a lesser amount of b1 ? 4 and b1 ? 6 linkages, GOS can be formed. In reactions catalysed by the b-galactosidase
while the commercial GOS contained primarily b1 ? 4 and from A. oryzae, the maximal concentration peak of the tetrameric
b1 ? 6 linkages. GOS occurs later than that of trimeric GOS (Neri et al., 2009). There
While the b-galactosidase directs GOS regiochemistry, this reg- has also been reports showing that penta- and hexasaccharides
iospecicity is not absolute, and GOS are inevitably produced as reach a maximum concentration well after the maximum concen-
mixtures of regioisomers (Mahoney, 1998). The linkages between tration of GOS has been reached (Albayrak & Yang, 2002), indicat-
sugars in these mixtures are formed and hydrolysed at different ing sacrices in total yield may be required to produce elevated
rates, so the reaction time inuences the structures present in a levels of high-DP GOS within the product mixture. This approach
GOS mixture. For example, during lactose conversion by the Bacil- may also lead to a decrease in the average DP of the mixture, as di-
meric GOS species can become dominant after the maximum con-
centration of GOS is reached and the degradation of higher DP GOS
Table 3 becomes increasingly important (Smart, 1991).
GOS-producing characteristics of commercially important b-galactosidases. Examples where the DP of the GOS product has been controlled
Microbial enzyme Yields with 5% Typical GOS Predominant by process parameters such as temperature, pH and lactose con-
source (w/v) initial degree of linkages centration have also been recently reported. A commercial prepa-
lactose polymerisation formed ration of Aspergillus aculeatus b-galactosidase produced higher
concentration maximum concentrations of high DP and trimeric GOS and de-
(%)
creased maximum concentrations of dimeric GOS with an increase
Cryptococcus laurentii 43b Mostly b1 ? 6 in reaction temperature. The same enzyme also produced higher
Ohtsuka et al. trisaccharides
maximum concentrations of trimeric GOS at pH 6.5 than at pH
(1990)
Bacillus circulans 41b Di- to b1 ? 4 Usui, 4.5, 5.5 or 7.5 without producing a signicant difference in dimeric
Mozaffar, pentasaccharides Kubota, and or high-DP GOS (Cardelle-Cobas, Villamiel, Olano, & Corzo, 2008).
Nakanishi, Ohi (1993) The maximum amount of tri- and tetrameric GOS and the ratio
Matsuno, and
of trimers to tetramers, produced by A. oryzae b-galactosidase
Kamikubo (1984)
Streptococcus 25m Disaccharides b1 ? 6 Toba,
operating in milk, could also be varied by altering the reaction tem-
thermophilus Tomita, Itoh, perature and the initial lactose concentration in this system (Chen,
Greenberg and and Adachi Hsu, & Chiang, 2002).
Mahoney (1983) (1981) Much potential for optimising the structure of a GOS product
Kluyveromyces fragilis 22b Roberts and Di- and b1 ? 6
exists, particularly for improvements in prebiotic efcacy. How-
Pazur, Tipton, Pettinati (1957) trisaccharides
Budovich, and ever, identifying structural motifs which considerably enhance
Marsh (1958) prebiotic bioactivity remains a challenge. Commercially-produced
Aspergillus oryzae 15b Iwasaki Di- to b1 ? 6 GOS are typically complex mixtures containing glucose, galactose
Toba, Yokota, and et al. (1996) hexasaccharides
and lactose, as well as many GOS species (Crittenden & Playne,
Adachi (1985)
Kluyveromyces lactis 14m Mozaffar, Di- to b1 ? 6
1996). The difculties in attributing increased prebiotic potency
Asp, Burvall, Nakanishi, and tetrasaccharides to a single structural feature of a saccharide in such mixtures is
Dahlqvist, Matsuno (1985) perhaps best exemplied by lactose itself being dubbed a
Hallgren, and conditional prebiotic (Adamczak, Charubin, & Bednarski, 2009;
Lundblad (1980)
Schaafsma, 2008; Szilagyi, 2004). More research is therefore
b
Lactose and buffer based reaction medium. required to purify and characterise the prebiotic qualities of indi-
m
Milk-based reaction medium. vidual GOS species.
A. Gosling et al. / Food Chemistry 121 (2010) 307318 313

5. Modelling of the kinetics of GOS synthesis cations, such as assuming that trimers were the largest GOS species
formed and that all trimers had a Gal2Glc composition. The t of
GOS synthesis is complex. Complexity is generated through dif- the data to the model was reasonable but nine constants required
ferent reactions (transfer and hydrolysis) acting on different sub- estimation.
strates (differently structured GOS species and lactose) at Simpler mechanistic models do exist. A model has been re-
different rates. Even the stereochemistry of the anomeric carbon ported which uses only six constants (one set to unity) to describe
changes the kinetics of b-galactosidase-catalysed hydrolysis of lac- the concentrations of trisaccharide, lactose, glucose and galactose
tose (Huber, Hurlburt, & Turner, 1981) and the stereochemistry of during the course of the reaction catalysed by the b-galactosidase
GOS molecules could also be expected to similarly alter the kinetics preparation from B. circulans (Boon et al., 1999). Although the sim-
of b-galactosidase catalysis. Despite these complexities, there have plicity of this model is attractive, it was less accurate for the lowest
been many recent publications that model the formation of GOS and highest initial lactose concentration reaction courses reported
over time (Table 4). These models can be categorised as either and appeared to become less accurate the longer the reaction pro-
mechanistic or empirical. gressed. Perhaps these deciencies were due to the simplication
Mechanistic models are built by initially proposing a reaction that all oligosaccharides were considered to be trimeric species
pathway. Rate constants are allocated to each step during catalysis, containing two galactose and one glucose residue. It is known that
and then experimentally estimated. The detailed knowledge of the the GOS products of B. circulans b-galactosidase activity frequently
reaction mechanism of b-galactosidase makes this approach do not have this composition or DP (Yanahira et al., 1995).
attractive. However, to limit the number of rate constants which Thus it seems that available mechanistic models strike a bal-
need experimental estimation, simplications are inevitably made ance between a poor t of the model to experimental data due to
about the reaction system. For example, it has been common for oversimplication and inappropriate estimation of a large number
studies to lump GOS molecules into DP classes (e.g., trimers, tetra- of rate constants. Models including more terms are generally able
mers, etc.), rather than attempt to study the rates of formation and to t experimental data more closely but the greater number of de-
degradation for all regioisomers within a DP class. When more grees of freedom in such a model can produce very large error mar-
simplications are made, the model can be less accurate, but more gins around individual terms.
convenient to construct, so a balance between the accuracy and the Mechanistic models can offer insights into which process
feasibility of making the model needs to be found. parameters can be usefully manipulated to optimise the reaction.
An example of a mechanistic model describing the change in The MichaelisMenten equation is a classic model most commonly
lactose, glucose, galactose, GOS disaccharides and GOS trisaccha- used for this purpose. This equation describes the reaction velocity
rides formed over time was proposed for the activity of b-galacto- as a function of substrate concentration, with the initial reaction
sidase from K. lactis on lactose (Kim, Ji, & Oh, 2004). This model velocity asymptotically approaching a theoretical maximum as
included complex considerations, such as competitive galactose substrate concentration increases. For the b-glycosidase from P.
inhibition and a preference for this enzyme to use glucose as the furiosus, the initial rate of galactose formation obeys this Michae-
galactosyl acceptor over lactose. The model also included simpli- lisMenten saturation behaviour but initial rates for the formation

Table 4
Kinetic models of GOS formation by b-galactosidase proposed in the literature.

Model description Number of Enzyme source Simplicationsa/variables with no signicant effect Complexitiesa/variables Reference
constantsa/ on GOS yieldb with signicant effect on
variables modelledb GOS yieldb
Mechanistic, saccharide 6 Bacillus GOS dened as only GalGalGlc. Gal inhibition Glc inhibition Boon et al.
concentration over circulans noted, but not modelled. GOS dened as tri and (1999)
time tetrasaccharides.
14 Aspergillus Hydrolysis of tetrasaccharides not included. No Viscosity of medium Iwasaki
oryzae distinction made between Gal and Glc. included et al.
(1996)
9 Kluyveromyces GOS dened as GalGlc dimers and trisaccharides. Glc and Gal inhibition Kim et al.
lactis (2004)
5 Pyrococcus. GOS dened as trisaccharides. Competitive Glc inhibition Bruins
furiosus et al.
(2003a)
16 Immobilised GOS dened as only 2 species; trimers and GalGal. Inhibition and mutarotation Bakken
Bacillus of Glc and Hill
circulans (1992)
Empirical, separate [L]i, T, R.T., [E]: [L]i Aspergillus [L]i, T [E]: [L]i, R.T. Chen et al.
yield of different DP oryzae (2002)
saccharides
Empirical, GOS [L]i, T, R.T., cell Bidobacterium T, R.T., cell concentration [L]i Roy et al.
concentration and concentration infantis whole (2002)
yield cells
Aspergillus [E], [L]i pH, T
oryzae
[E], [L]i, pH, T Kluyveromyces T [E], [L]i, pH Rustom
lactis et al.
Kluyveromyces [E], [L]i pH, T (1998)
fragilis

[L]i-Initial lactose concentration; [E]-enzyme concentration; T-temperature; and RT-reaction time.


a
Mechanistic models.
b
Empirical models.
314 A. Gosling et al. / Food Chemistry 121 (2010) 307318

of trisaccharides, glucose and the initial rate of lactose consump- the organism directly can circumvent these problems (Fukuda,
tion continued to increase with increasing lactose concentration Hama, Tamalampudi, & Noda, 2008). GOS yields of 36% (w/v) to
(Bruins et al., 2003a). As the difference between galactosyl transfer 43% (w/w) have been achieved in a whole cell system, demonstrat-
and hydrolysis rates appears to become greater at increasing lac- ing comparable yields to those achieved with isolated enzymes
tose concentrations, the results suggest that the optimal initial lac- (Goulas, Tzortzis, & Gibson, 2007).
tose concentration for GOS synthesis in this system is saturation. There have been studies describing examples of GOS production
Further complexity is encountered at high saccharide concen- employing probiotic microbial cells as catalysts (Roy et al., 2002,
trations, as the high concentrations of lactose which favour GOS Tzortzis, Goulas, & Gibson, 2005). A further report describes adding
formation also increase the viscosity of the reaction solution, a pre-incubation step during yoghurt making, that allows the star-
changing mass transfer rates and enzyme dynamics. The rate con- ter culture sufcient time to act as GOS producing biocatalysts
stants in a mechanistic model have been found to correlate to the (Lamoureux, Roy, & Gauthier, 2002). Given the argument that b-
viscosity of the reaction solution (Iwasaki et al., 1996). It can be galactosidase from probiotic organisms produce GOS with an en-
extrapolated from this nding that the mixing efciency and vig- hanced prebiotic effect (Rastall, 2004) (also see Structure of GOS
our are important effectors of the reaction rate at conditions of products section), this approach may provide an opportunity to
high saccharide concentration. produce GOS with superior prebiotic qualities without the need
Empirical models can also be used to mathematically describe for the manufacturer to purchase b-galactosidase.
the production of GOS. This approach involves tting equations An advantage of whole cells over isolated enzymes during the
to experimentally measured reaction time courses, in order to development of an optimal biocatalyst is the ability to evolve.
accurately determine and optimise GOS yields. Empirical ap- Changing an isolated protein typically involves manipulation at
proaches have the advantage of being able to include multiple pro- the DNA level. Regulation of genetically-modied food ingredients
cess parameters (such as initial lactose concentration, temperature makes this approach inappropriate and removes the most powerful
and pH) in a single model without the need for detailed reaction tool for optimising an isolated enzyme. However, standard breed-
mechanisms. This approach is well suited to more complex reac- ing and selection techniques offer options for improving food-
tion systems, such as milk or other dairy products, which have var- grade whole cell biocatalysts. Chemical mutagenesis followed by
iable composition. screening, to select for strains with increased b-galactosidase activ-
Several recent studies have applied a popular form of empirical ity, has been applied to probiotic strains, leading to increases of
optimisation, known as response surface methodology (Bas & Boy- 70222% in b-galactosidase activity (Ibrahim & OSullivan, 2000).
aci, 2007) to optimise GOS production (Table 4). These examples Screening of isolates of healthy intestinal microora without muta-
include modelling GOS synthesis by whole live Bidobacterium genesis also offers an opportunity for discovering novel whole cell
infantis RW-8120 cells (Roy, Daoudi, & Azaola, 2002). In such a sys- biocatalysts for GOS production (OSullivan, 2001).
tem, detailed knowledge of important reaction parameters, such as It is generally believed that the added complexity of using
the rate of lactose uptake into the cell, as well as the intracellular whole cells for biocatalysis can be a disadvantage compared to iso-
concentration of lactose, GOS and b-galactosidase, are difcult to lated enzymes (Woodley, 2006). However, whole cells also provide
determine. The effect of cell concentration, initial lactose concen- the opportunity to include additional metabolic functions such as
tration, reaction time and temperature were more easily examined the consumption of the monosaccharides glucose and galactose
and the model found that the initial lactose concentration was the from a GOS product. These monosaccharides do not show a prebi-
only signicant variable affecting GOS yield. otic effect, raise the caloric value of food containing unpuried
Interestingly, other empirical models (Chen et al., 2002; Rus- GOS preparations and can be considered as undesirable. For exam-
tom, Foda, & Lpez-Leiva, 1998) found no signicant effect of initial ple, mixtures of bacterial and yeast strains (B. bidum and Saccha-
lactose concentration on GOS yield. Given the wide acceptance that romyces cerevisiae) have been combined to produce GOS and to
initial lactose concentration is one of the major effectors of GOS preferentially consume 92% of the glucose, respectively, thereby
yields, these ndings may point to deciencies in these models. purifying the prebiotic carbohydrates of unwanted monosaccha-
rides (Goulas et al., 2007). A similar approach employed immobi-
lised Zymomonas mobilis cells (Crittenden & Playne, 2002) and
6. Reactor congurations
the selective monosaccharide catabolism of these cells may be
benecial. A study comparing monosaccharide removal from Vivi-
As for most reactions catalysed by a biological entity, there are
nal GOS using two chromatographic techniques (size exclusion or
many modes in which GOS producing reactions can be performed.
activated charcoal) and the addition of S. cerevisiae cells found that
The b-galactosidase can be soluble or immobilised. It can be iso-
cellular treatment gave the highest GOS yields (Hernandez, Ruiz-
lated after intra- or extracellular expression, or provided within
Matute, Olano, Moreno, & Sanz, 2009). However, it should be noted
whole cells. The mode in which the process is undertaken may
that the metabolic end products, such as ethanol, lactic acid or
change the behaviour of the enzyme, the concentrations of product
other metabolites, will affect taste and other characteristics of
and substrate around the enzyme and will certainly inuence the
the nal product when this approach is employed.
performance and economics of the process. A recent publication
reviewed the immobilisation of b-galactosidase for GOS production
(Panesar et al., 2006). Here, we present a review of the literature
6.2. Membrane processing
using two alternative approaches: (1) whole cells as biocatalysts
and (2) membrane reactors, emphasising the differences in the
The cost associated with using isolated enzymes can also be
outcomes of the process due to these alternative reactor formats.
ameliorated by recycling the biocatalyst. One means for enzyme
recycling involves ultraltration (UF). In this approach, a semi-per-
6.1. Whole cells meable membrane is used to retain the larger enzyme within the
bioreactor while allowing the passage of smaller molecules, such
Using resting or living whole cells removes the need for the iso- as substrate and product. Continuous removal of product saccha-
lation of the b-galactosidase enzyme. Protein isolation can be rides and replacement with fresh substrate allows the reaction to
demanding and costly and requires additional processing steps be continued indenitely while the integrity of the bioreactor is
after the culture of the b-galactosidase producing organism. Using maintained.
A. Gosling et al. / Food Chemistry 121 (2010) 307318 315

For example, a commercial preparation of the K. lactis b-galacto- dases are also likely to include thermotolerant enzymes, as in-
sidase reached its maximum GOS yield in a 1 h batch reaction, creased lactose solubility at higher temperatures generally leads
yielding 7.6 mg GOS per unit enzyme activity. GOS yields de- to increased GOS.
creased after this time. When the reaction was performed in a UF Solid inroads are being made into the historically challenging
membrane reactor, 6.4 mg GOS were produced per unit enzyme task of mathematically modelling this complex system. Effective
activity per hour for 4 h (Chockchaisawasdee et al., 2005) repre- models for GOS synthesis will allow GOS production to be opti-
senting a total increase in yield of 240%. mised. A careful choice of reactor conguration can further mini-
UF membranes can also give control over the average amount of mise production costs and promote desired product
time that lactose is exposed to the b-galactosidase. This is an characteristics.
important parameter to be able to control, given that it inuences Increases in GOS yield delivered by enhancing the transgalac-
GOS yield and structure. It has been found that shorter average res- tosylation reaction, through advances in reaction and enzyme
idence times in UF reactors gave higher GOS yields for both the K. engineering, will result in higher concentrations of GOS, alleviating
lactis and A. oryzae enzymes (Czermak et al., 2004). some of the need for further separation of GOS from other carbohy-
A recognised caveat to performing enzymatic reactions in UF drates. This will have an impact on both the economics of process-
reactors is that the shear forces and/or higher pressures present ing and the potential utilisation of GOS.
can cause enzyme denaturation (Bowen, 1993). However, there is Together these advances will lower the cost of GOS production
evidence that the benets of UF can outweigh this limitation. An and increase prebiotic potency. This should decrease the price of
increased transmembrane pressure in a UF unit gave higher rate GOS ingredients which will also be effective at lower concentra-
constants than an unpressured batch reaction using b-galactosi- tions, leading to health benets across society.
dase from A. oryzae (Matella, Dolan, & Lee, 2006). This study con-
cluded that the positive effects on reaction rates from increased Acknowledgements
mixing at higher uid pressures was greater than the negative ef-
fects due to shear-induced enzyme denaturation. This research was funded by an Australia-India Strategic Re-
The structure of the GOS product can also be inuenced by search Fund Program BF01-0024 from The Department of Innova-
using a continuous UF reactor. This was illustrated in a study com- tion, Industry and Science Research. We acknowledge Daniel
paring the behaviour of the b-galactosidase from Lactobacillus reu- Otieno for preliminary work leading to this study and Professor
teri in both a continuous UF reactor and a batch reactor (Splechtna, Chiranjib Bhattacharjee from Jadavpur University Kolkata and Pro-
Nguyen, & Haltrich, 2007). It was reported that the GOS product fessor P.K. Bhattacharya from the Indian Institute of Technology
from the continuous system had a higher percentage of disaccha- Kanpur India for useful discussions. We would also like to
rides. This was explained by the higher concentration of glucose acknowledge the Particulate Fluids Processing Centre, Special Re-
in the continuous system at steady state compared to the batch search Centre of the ARC, for access to equipment.
system. Glucose was fourfold preferred as an acceptor for transga-
lactosylation than lactose for this enzyme, so the higher glucose References
concentration in the UF system led to more GalGlc dimers. It
was also found that the continuous system produced a higher ratio Adamczak, M., Charubin, D., & Bednarski, W. (2009). Inuence of reaction medium
composition on enzymatic synthesis of galactooligosaccharides and lactulose
of b1- > 6 to b1- > 3 glycosidic linkages as the b1- > 3 bonds were from lactose concentrates prepared from whey permeate. Chemical Papers,
hydrolysed more rapidly than b1- > 6 bonds. 63(2), 111116.
Membranes with smaller pore sizes can also be used for purify- Albayrak, N., & Yang, S.-T. (2002). Production of galacto-oligosaccharides from
lactose by Aspergillus oryzae beta-galactosidase immobilized on cotton cloth.
ing GOS products from the monosaccharide hydrolysis products, Biotechnology and Bioengineering, 77(1), 819.
glucose and galactose. In one example, up to 88% of the di- and oli- Amaretti, A., Bernardi, T., Tamburini, E., Zanoni, S., Lomma, M., Matteuzzi, D., et al.
gosaccharides were recovered from a commercial GOS mixture (2007). Kinetics and metabolism of Bidobacterium adolescentis MB 239 growing
on glucose, galactose, lactose, and galactooligosaccharides. Applied and
using a nanoltration membrane with only 19% of the monosac- Environmental Microbiology, 73(11), 36373644.
charides remaining in the retentate stream (Goulas, Grandison, & Asp, N. G., Burvall, A., Dahlqvist, A., Hallgren, P., & Lundblad, A. (1980).
Rastall, 2003). Other literature on the benets of cost-effective Oligosaccharide formation during hydrolysis of lactose with Saccharomyces
Lactis lactase (Maxilact). 2 Oligosaccharide structures. Food Chemistry, 5(2),
membrane separation of monosaccharides from oligosaccharides
147153.
(Catarino, Minhalma, Beal, Mateus, & de Pinho, 2008; Feng, Chang, Bakken, A. P., & Hill, C. G. (1992). Hydrolysis of lactose in skim milk by immobilised
Wang, & Ma, 2009; Goulas, Kapasakalidis, Sinclair, Rastall, & beta-galactosidase (Bacillus circulans). Biotechnology and Bioengineering, 39(4),
Grandison, 2002) is available as a comparison to industrial chro- 408417.
Barreteau, H., Delattre, C., & Michaud, P. (2006). Production of oligosaccharides as
matography processes (Playne & Crittenden, 1996). promising new food additive generation. Food Technology and Biotechnology,
44(3), 323333.
Bas, D., & Boyaci, I. H. (2007). Modeling and optimization I: Usability of response
surface methodology. Journal of Food Engineering, 78(3), 836845.
7. Outlook Berman, H. M., Westbrook, J., Feng, Z., Gilliland, G., Bhat, T. N., Weissig, H., et al.
(2000). The protein data bank. Nucleic Acids Research, 28(Database issue),
Few enzymes are as well described as b-galactosidase. Yet gaps 235242.
Boon, M. A., Janssen, A. E. M., & van der Padt, A. (1999). Modelling and parameter
still exist in our knowledge of b-galactosidase-catalysed GOS syn- estimation of the enzymatic synthesis of oligosaccharides by beta-galactosidase
thesis. This review has highlighted important recent research that from Bacillus circulans. Biotechnology and Bioengineering, 64(5), 558567.
contributes to our understanding of b-galactosidase structure and Boon, M. A., Janssen, A. E. M., & vant Riet, K. (2000). Effect of temperature and
enzyme origin on the enzymatic synthesis of oligosaccharides. Enzyme and
activity, and drawn attention to areas where greater insight would Microbial Technology, 26(24), 271281.
be useful for the future rational improvement of GOS production Borra, S. T., & Bouchoux, A. (2009). Effects of science and the media on consumer
processes at a commercial scale. perceptions about dietary sugars. Journal of Nutrition, 139(6), 1214S1218S.
Bowen, R. (1993). Understanding ux patterns in membrane processing of protein
Opportunities exist to optimise features of b-galactosidase. For
solutions and suspensions. Trends in Biotechnology, 11(11), 451460.
example, systematic studies of different enzymes and their resul- Brs, N. F., Moura-Tamames, S. A., Fernandes, P. A., & Ramos, M. J. (2008).
tant GOS structures could be used to tailor desired characteristics Mechanistic studies on the formation of glycosidase-substrate and glycosidase-
in commercially applicable enzymes. Further enzyme engineering inhibitor covalent intermediates. Journal of Computational Chemistry, 29(15),
25652574.
could be directed towards enzymes that produce GOS with higher Bruins, M. E., Strubel, M., van Lieshout, J. F. T., Janssen, A. E. M., & Boom, R. M.
prebiotic efcacy. The next generation of optimised b-galactosi- (2003a). Oligosaccharide synthesis by the hyperthermostable beta-glucosidase
316 A. Gosling et al. / Food Chemistry 121 (2010) 307318

from Pyrococcus furiosus: Kinetics and modelling. Enzyme and Microbial Goulas, A. K., Grandison, A. S., & Rastall, R. A. (2003). Fractionation of
Technology, 33(1), 311. oligosaccharides by nanoltration. Journal of the Science of Food and
Bruins, M. E., Van Hellemond, E. W., Janssen, A. E. M., & Boom, R. M. (2003b). Agriculture, 83(7), 675680.
Maillard reactions and increased enzyme inactivation during oligosaccharide Goulas, A. K., Kapasakalidis, P. G., Sinclair, H. R., Rastall, R. A., & Grandison, A. S.
synthesis by a hyperthermophilic glycosidase. Biotechnology and Bioengineering, (2002). Purication of oligosaccharides by nanoltration. Journal of Membrane
81(5), 546552. Science, 209(1), 321335.
Buchholz, K., Kasche, V., & Bornscheuer, U. T. (2005). Equilibrium and kinetically Greenberg, N. A., & Mahoney, R. R. (1983). Formation of oligosaccharides by beta-
controlled reactions catalysed by enzymes. In Biocatalysts and enzyme galactosidase from Streptococcus thermophilus. Food Chemistry, 10(3), 195204.
technology (pp. 4246). Weinheim: Wiley-VCH Verlag GmbH& Co. Hansson, T., Kaper, T., van der Oost, J., de Vos, W. M., & Adlercreutz, P. (2000).
Cantarel, B. L., Coutinho, P. M., Rancurel, C., Bernard, T., Lombard, V., & Henrissat, B. Improved oligosaccharide synthesis by protein engineering of beta-glucosidase
(2009). The carbohydrate-active enzymes database (CAZy): An expert resource CelB from hyperthermophilic Pyrococcus furiosus. Biotechnology and
for glycogenomics. Nucleic Acids Research, 37(Database issue), D233238. Bioengineering, 73(3), 203210.
Cardelle-Cobas, A., Villamiel, M., Olano, A., & Corzo, N. (2008). Study of galacto- Hatzinikolaou, D. G., Katsifas, E., Mamma, D., Karagouni, A. D., Christakopoulos, P., &
oligosaccharide formation from lactose using Pectinex Ultra SP-L. Journal of the Kekos, D. (2005). Modeling of the simultaneous hydrolysis-ultraltration of
Science of Food and Agriculture, 88(6), 954961. whey permeate by a thermostable beta-galactosidase from Aspergillus niger.
Catarino, I., Minhalma, M., Beal, L. L., Mateus, M., & de Pinho, M. N. (2008). Biochemical Engineering Journal, 24(2), 161172.
Assessment of saccharide fractionation by ultraltration and nanoltration. Henrissat, B. (1991). A classication of glycosyl hydrolases based on amino acid
Journal of Membrane Science, 312(12), 3440. sequence similarities. Biochemical Journal, 280(2), 309316.
Chen, C. S., Hsu, C. K., & Chiang, B. H. (2002). Optimization of the enzymic process Hernandez, O., Ruiz-Matute, A. I., Olano, A., Moreno, F. J., & Sanz, M. L. (2009).
for manufacturing low-lactose milk containing oligosaccharides. Process Comparison of fractionation techniques to obtain prebiotic
Biochemistry, 38(5), 801808. galactooligosaccharides. International Dairy Journal, 19(9), 531536.
Chen, C. W., Ou-Yang, C. C., & Yeh, C. W. (2003). Synthesis of galactooligosaccharides Hidaka, M., Fushinobu, S., Ohtsu, N., Motoshima, H., Matsuzawa, H., Shoun, H., et al.
and transgalactosylation modeling in reverse micelles. Enzyme and Microbial (2002). Trimeric crystal structure of the glycoside hydrolase family 42 beta-
Technology, 33(4), 497507. galactosidase from Thermus thermophilus A4 and the structure of its complex
Chen, S.-X., Wei, D.-Z., & Hu, Z.-H. (2001). Synthesis of galacto-oligosaccharides in with galactose. Journal of Molecular Biology, 322(1), 7991.
AOT/isooctane reverse micelles by beta-galactosidase. Journal of Molecular Hsu, C. A., Lee, S. L., & Chou, C. C. (2007). Enzymatic production of
Catalysis B: Enzymatic, 16(2), 109114. galactooligosaccharides by beta-galactosidase from bidobacterium longum
Chen, W., Chen, H., Xia, Y., Zhao, J., Tian, F., & Zhang, H. (2008). Production, BCRC 15708. Journal of Agricultural and Food Chemistry, 55(6), 22252230.
purication, and characterization of a potential thermostable galactosidase for Hsu, C. A., Yu, R. C., & Chou, C. C. (2006). Purication and characterization of a
milk lactose hydrolysis from Bacillus stearothermophilus. Journal of Dairy Science, sodium-stimulated beta-galactosidase from Bidobacterium longum CCRC
91(5), 17511758. 15708. World Journal of Microbiology and Biotechnology, 22(4), 355361.
Cho, Y.-J., Shin, H.-J., & Bucke, C. (2003). Purication and biochemical properties of a Huber, R. E., Hurlburt, K. L., & Turner, C. L. (1981). The anomeric specicity of beta-
galactooligosaccharide producing beta-galactosidase from Bullera singularis. galactosidase and lac permease from Escherichia coli. Canadian Journal of
Biotechnology Letters, 25(24), 21072111. Biochemistry, 59(2), 100105.
Chockchaisawasdee, S., Athanasopoulos, V. I., Niranjan, K., & Rastall, R. A. (2005). Huber, R. E., Kurz, G., & Wallenfels, K. (1976). A quantitation of the factors which
Synthesis of galacto-oligosaccharide from lactose using beta-galactosidase from affect the hydrolase and transgalactosylase activities of beta-galactosidase
Kluyveromyces lactis: Studies on batch and continuous UF membrane-tted (E. coli) on lactose. Biochemistry, 15(9), 19942001.
bioreactors. Biotechnology and Bioengineering, 89(4), 434443. Ibrahim, S. A., & OSullivan, D. J. (2000). Use of chemical mutagenesis for the
Cowburn, G., & Stockley, L. (2005). Consumer understanding and use of nutrition isolation of food grade beta-galactosidase overproducing mutants of
labelling: A systematic review. Public Health Nutrition, 8(1), 2128. Bidobacteria, Lactobacilli and Streptococcus thermophilus. Journal of Dairy
Crittenden, R. G., & Playne, M. J. (1996). Production, properties and applications of Science, 83(5), 923930.
food-grade oligosaccharides. Trends in Food Science and Technology, 7(11), Iwasaki, K.-i., Nakajima, M., & Nakao, S.-i. (1996). Galacto-oligosaccharide
353361. production from lactose by an enzymic batch reaction using beta-
Crittenden, R. G., & Playne, M. J. (2002). Purication of food-grade oligosaccharides galactosidase. Process Biochemistry, 31(1), 6976.
using immobilised cells of Zymomonas mobilis. Applied Microbiology and Ji, E.-S., Park, N.-H., & Oh, D.-K. (2005). Galacto-oligosaccharide production by a
Biotechnology, 58(3), 297302. thermostable recombinant beta-galactosidase from Thermotoga maritima. World
Cruz-Guerrero, A. E., Gmez-Ruiz, L., Viniegra-Gonzlez, G., Brzana, E., & Garca- Journal of Microbiology and Biotechnology, 21(5), 759764.
Garibay, M. (2006). Inuence of water activity in the synthesis of Jobe, A., & Bourgeoi, S. (1972). Lac repressoroperator interaction 6. Natural inducer
galactooligosaccharides produced by a hyperthermophilic beta-glycosidase in of Lac Operon. Journal of Molecular Biology, 69(3), 397404.
an organic medium. Biotechnology and Bioengineering, 93(6), 11231129. Jrgensen, F., Hansen, O. C., & Stougaard, P. (2001). High-efciency synthesis of
Czermak, P., Ebrahimi, M., Grau, K., Netz, S., Sawatzki, G., & Pfromm, P. H. (2004). oligosaccharides with a truncated beta-galactosidase from Bidobacterium
Membrane-assisted enzymatic production of galactosyl-oligosaccharides from bidum. Applied Microbiology and Biotechnology, 57(56), 647652.
lactose in a continuous process. Journal of Membrane Science, 232(12), Jrgensen, F., Hansen, O. C. & Stougaard, P. (2007). Beta-galactosidase isolated from
8591. bidobacterium. Patent no. EP1283876.
Delzenne, N. M. (2003). Oligosaccharides: State of the art. Proceedings of the Juers, D. H., Hakda, S., Matthews, B. W., & Huber, R. E. (2003). Structural basis for the
Nutrition Society, 62(1), 177182. altered activity of Gly794 variants of Escherichia coli beta-galactosidase.
Demirhan, E., Apar, D. K., & zbek, B. (2008). Product inhibition of whey lactose Biochemistry, 42(46), 1350513511.
hydrolysis. Chemical Engineering Communications, 195(3), 293304. Juers, D. H., Heightman, T. D., Vasella, A., McCarter, J. D., Mackenzie, L., Withers, S. G.,
Depeint, F., Tzortzis, G., Vulevic, J., IAnson, K., & Gibson, G. R. (2008). Prebiotic et al. (2001). A structural view of the action of Escherichia coli (lacZ) beta-
evaluation of a novel galactooligosaccharide mixture produced by the galactosidase. Biochemistry, 40(49), 1478114794.
enzymatic activity of Bidobacterium bidum NCIMB 41171, in healthy Jurado, E., Camacho, F., Luzn, G., & Vicaria, J. M. (2004). Kinetic models of activity
humans: A randomized, double-blind, crossover, placebo-controlled for beta-galactosidases: inuence of pH, ionic concentration and temperature.
intervention study. American Journal of Clinical Nutrition, 87(3), 785791. Enzyme and Microbial Technology, 34(1), 3340.
Dumortier, V., Brassart, C., & Bouquelet, S. (1994). Purication and properties of a Kim, C. S., Ji, E.-S., & Oh, D.-K. (2004). A new kinetic model of recombinant beta-
beta-D-galactosidase from Bidobacterium bidum exhibiting a galactosidase from Kluyveromyces lactis for both hydrolysis and
transgalactosylation reaction. Biotechnology and Applied Biochemistry, 19(3), transgalactosylation reactions. Biochemical and Biophysical Research
341354. Communications, 316(3), 738743.
Feng, Y. M., Chang, X. L., Wang, W. H., & Ma, R. Y. (2009). Separation of galacto- Lamoureux, L., Roy, D., & Gauthier, S. F. (2002). Production of oligosaccharides in
oligosaccharides mixture by nanoltration. Journal of the Taiwan Institute of Yogurt containing bidobacteria and Yogurt cultures. Journal of Dairy Science,
Chemical Engineers, 40(3), 326332. 85(5), 10581069.
Fukuda, H., Hama, S., Tamalampudi, S., & Noda, H. (2008). Whole-cell biocatalysts Lomer, M. C. E., Parkes, G. C., & Sanderson, J. D. (2008). Review article: Lactose
for biodiesel fuel production. Trends in Biotechnology, 26(12), 668673. intolerance in clinical practice Myths and realities. Alimentary Pharmacology
Gnzle, M. G., Haase, G., & Jelen, P. (2008). Lactose: Crystallization, hydrolysis and and Therapeutics, 27(2), 93103.
value-added derivatives. International Dairy Journal, 18(7), 685694. Macfarlane, G. T., Steed, H., & Macfarlane, S. (2008). Bacterial metabolism and
Gloster, T. M., Roberts, S., Ducros, V. M.-A., Perugino, G., Rossi, M., Hoos, R., et al. health-related effects of galacto-oligosaccharides and other prebiotics. Journal
(2004). Structural Studies of the beta-glycosidase from Sulfolobus solfataricus in of Applied Microbiology, 104(2), 305344.
complex with covalently and noncovalently bound inhibitors. Biochemistry, Mahoney, R. R. (1998). Galactosyl-oligosaccharide formation during lactose
43(20), 61016109. hydrolysis: A review. Food Chemistry, 63(2), 147154.
Gopal, P. K., Sullivan, P. A., & Smart, J. B. (2001). Utilisation of galacto- Matella, N. J., Dolan, K. D., & Lee, Y. S. (2006). Comparison of galactooligosaccharide
oligosaccharides as selective substrates for growth by lactic acid bacteria production in free-enzyme ultraltration and in immobilized-enzyme systems.
including Bidobacterium lactis DR10 and Lactobacillus rhamnosus DR20. Journal of Food Science, 71(7), C363C368.
International Dairy Journal, 11(12), 1925. Matthews, B. W. (2005). The structure of E Coli beta-galactosidase. Comptes Rendus
Goulas, A., Tzortzis, G., & Gibson, G. R. (2007). Development of a process for the Biologies, 328(6), 549556.
production and purication of alpha- and beta-galactooligosaccharides from Maugard, T., Gaunt, D., Legoy, M. D., & Besson, T. (2003). Microwave-assisted
Bidobacterium bidum NCIMB 41171. International Dairy Journal, 17(6), synthesis of galacto-oligosaccharides from lactose with immobilized beta-
648656. galactosidase from Kluyveromyces lactis. Biotechnology Letters, 25(8), 623629.
A. Gosling et al. / Food Chemistry 121 (2010) 307318 317

Mladenoska, I., Winkelhausen, E., & Kuzmanova, S. (2008). Transgalactosylation / Rustom, I. Y. S., Foda, M. I., & Lpez-Leiva, M. H. (1998). Formation of
hydrolysis ratios of various beta-galactosidases catalyzing alkyl-beta- oligosaccharides from whey UF-permeate by enzymatic hydrolysis Analysis
galactoside synthesis in single-phased alcohol media. Food Technology and of factors. Food Chemistry, 62(2), 141147.
Biotechnology, 46(3), 311316. Rycroft, C. E., Jones, M. R., Gibson, G. R., & Rastall, R. A. (2001). A comparative in vitro
Mlichova, Z., & Rosenberg, M. (2006). Current trends of beta-galactosidase evaluation of the fermentation properties of prebiotic oligosaccharides. Journal
application in food technology. Journal of Food and Nutrition Research, 45(2), of Applied Microbiology, 91(5), 878887.
4754. Sanz, M. L., Cote, G. L., Gibson, G. R., & Rastall, R. A. (2006a). Inuence of glycosidic
Mozaffar, Z., Nakanishi, K., & Matsuno, R. (1985). Formation of oligosaccharides linkages and molecular weight on the fermentation of maltose-based
during hydrolysis of lactose in milk using beta-galactosidase from Bacillus oligosaccharides by human gut bacteria. Journal of Agricultural and Food
circulans. Journal of Food Science, 50(6), 16021606. Chemistry, 54(26), 97799784.
Mozaffar, Z., Nakanishi, K., Matsuno, R., & Kamikubo, T. (1984). Purication and Sanz, M. L., Cote, G. L., Gibson, G. R., & Rastall, R. A. (2006b). Selective fermentation
properties of beta-galactosidases from Bacillus circulans. Agricultural and of gentiobiose-derived oligosaccharides by human gut bacteria and inuence of
Biological Chemistry, 48(12), 30533061. molecular weight. FEMS Microbiology Ecology, 56(3), 383388.
Mussatto, S. I., & Mancilha, I. M. (2007). Non-digestible oligosaccharides: A review. Sanz, M. L., Gibson, G. R., & Rastall, R. A. (2005). Inuence of disaccharide structure
Carbohydrate Polymers, 68(3), 587597. on prebiotic selectivity in vitro. Journal of Agricultural and Food Chemistry,
Nakao, M., Harada, M., Kodama, Y., Nakayama, T., Shibano, Y., & Amachi, T. (1994). 53(13), 51925199.
Purication and characterization of a thermostable beta-galactosidase with Sanz, M. L., Sanz, J., & Martinez-Castro, I. (2002). Characterization of O-trimethylsilyl
high transgalactosylation activity from Saccharopolyspora rectivirgula. Applied oximes of disaccharides by gas chromatographymass spectrometry.
Microbiology and Biotechnology, 40(5), 657663. Chromatographia, 56(910), 617622.
Neri, D. F. M., Balcao, V. M., Costa, R. S., Rocha, I., Ferreira, E., Torres, D. P. M., et al. Schaafsma, G. (2008). Lactose and lactose derivatives as bioactive ingredients in
(2009). Galacto-oligosaccharides production during lactose hydrolysis by free human nutrition. International Dairy Journal, 18(5), 458465.
Aspergillus oryzae beta-galactosidase and immobilized on magnetic Sklov, T., Dohnlek, J., Spiwok, V., Lipovov, P., Vondrckov, E., Petrokov, H.,
polysiloxane-polyvinyl alcohol. Food Chemistry, 115(1), 9299. et al. (2005). Cold-active beta-galactosidase from Arthrobacter sp. C2-2 forms
Nerinckx, W., Desmet, T., & Claeyssens, M. (2003). A hydrophobic platform as a compact 660 kDa hexamers: Crystal structure at 1.9 resolution. Journal of
mechanistically relevant transition state stabilising factor appears to be present Molecular Biology, 353(2), 282294.
in the active centre of all glycoside hydrolases. FEBS Letters, 538(13), 17. Smart, J. B. (1991). Transferase reactions of the beta-galactosidase from
Ohtsuka, K., Tanoh, A., Ozawa, O., Kanematsu, T., Uchida, T., & Shinke, R. (1990). Streptococcus Thermophilus. Applied Microbiology and Biotechnology, 34(4),
Purication and properties of a beta-galactosidase with high galactosyl transfer 495501.
activity from Cryptococcus laurentii Okn-4. Journal of Fermentation and Splechtna, B., Nguyen, T.-H., & Haltrich, D. (2007). Comparison between
Bioengineering, 70(5), 301307. discontinuous and continuous lactose conversion processes for the production
Onishi, N., & Tanaka, T. (1995). Purication and properties of a novel thermostable of prebiotic galacto-oligosaccharides using beta-galactosidase from
galacto-oligosaccharide-producing beta-galactosidase from Sterigmatomyces Lactobacillus reuteri. Journal of Agricultural and Food Chemistry, 55(16),
elviae CBS8119. Applied and Environmental Microbiology, 61(11), 40264030. 67726777.
OSullivan, D. J. (2001). Screening of intestinal microora for effective probiotic Splechtna, B., Nguyen, T.-h., Steinbock, M., Kulbe, K. D., Lorenz, W., & Haltrich, D.
bacteria. Journal of Agricultural and Food Chemistry, 49(4), 17511760. (2006). Production of prebiotic galacto-oligosaccharides from lactose using
Palani, K., Kumaran, D., Burley, S. K., & Swaminathan, S. (2009). Crystal structure of a beta-galactosidases from Lactobacillus reuteri. Journal of Agricultural and Food
beta-galactosidase from Bacteroides thetaiotaomicron. http://www.rcsb.org/pdb/ Chemistry, 54(14), 49995006.
explore.do?structureId=3D3A Accessed 23.01.09. Szilagyi, A. (2004). Redening lactose as a conditional prebiotic. Canadian Journal of
Panesar, P. S., Panesar, R., Singh, R. S., Kennedy, J. F., & Kumar, H. (2006). Microbial Gastroenterology, 18(3), 163167.
production, immobilization and applications of beta-D-galactosidase. Journal of Taniguchi, H. (2005). Carbohydrate active enzymes for the production of
Chemical Technology and Biotechnology, 81(4), 530543. oligosaccharides. In C.-T. Hou (Ed.), Handbook of industrial biocatalysis. Boca
Park, H. Y., Kim, H. J., Lee, J. K., Kim, D., & Oh, D. K. (2008). Galactooligosaccharide Raton: CRC Press. pp. 20-120-23.
production by a thermostable beta-galactosidase from Sulfolobus solfataricus. Toba, T., Tomita, Y., Itoh, T., & Adachi, S. (1981). Beta-galactosidases of lactic acid
World Journal of Microbiology and Biotechnology, 24(8), 15531558. bacteria: Characterization by oligosaccharides formed during hydrolysis of
Pazur, J. H., Tipton, C. L., Budovich, T., & Marsh, J. M. (1958). Structural lactose. Journal of Dairy Science, 64(2), 185192.
characterization of products of enzymatic disproportionation of lactose. Toba, T., Yokota, A., & Adachi, S. (1985). Oligosaccharide structures formed during
Journal of the American Chemical Society, 80(1), 119121. the hydrolysis of lactose by Aspergillus oryzae beta-galactosidase. Food
Petzelbauer, I., Reiter, A., Splechtna, B., Kosma, P., & Nidetzky, B. (2000). Chemistry, 16(2), 147162.
Transgalactosylation by thermostable beta-glycosidases from Pyrococcus Topping, D. L., & Clifton, P. M. (2001). Short-chain fatty acids and human colonic
furiosus and Sulfolobus solfataricus. European Journal of Biochemistry, 267(16), function: Roles of resistant starch and nonstarch polysaccharides. Physiology
50555066. Reviews, 81(3), 10311064.
Playne, M., & Crittenden, R. (1996). Commercially available oligosaccharides. Trincone, A., & Giordano, A. (2006). Glycosyl hydrolases and glycosyltransferases in
Bulletin of the International Dairy Federation, 313, 1022. the synthesis of oligosaccharides. Current Organic Chemistry, 10(10), 11631193.
Prenosil, J. E., Stuker, E., & Bourne, J. R. (1987a). Formation of oligosaccharides Tzortzis, G., Goulas, A. K., & Gibson, G. R. (2005). Synthesis of prebiotic
during enzymatic lactose hydrolysis and their importance in a whey hydrolysis galactooligosaccharides using whole cells of a novel strain, Bidobacterium
process: Part II: Experimental. Biotechnology and Bioengineering, 30(9), bidum NCIMB 41171. Applied Microbiology and Biotechnology, 68(3), 412416.
10261031. Usui, T., Kubota, S., & Ohi, H. (1993). A convenient synthesis of beta-galactosyl
Prenosil, J. E., Stuker, E., & Bourne, J. R. (1987b). Formation of oligosaccharides disaccharide derivatives using the beta-galactosidase from Bacillus circulans.
during enzymatic lactose: Part I: State of Art. Biotechnology and Bioengineering, Carbohydrate Research, 244(2), 315323.
30(9), 10191025. Valli, C., & Traill, W. B. (2005). Culture and food: A model of yoghurt consumption in
Rabiu, B. A., Jay, A. J., Gibson, G. R., & Rastall, R. A. (2001). Synthesis and the EU. Food Quality and Preference, 16(4), 291304.
fermentation properties of novel galacto-oligosaccharides by beta- van den Broek, L. A. M., Hinz, S. W. A., Beldman, G., Vincken, J. P., & Voragen, A. G.
galactosidases from Bidobacterium species. Applied and Environmental J. (2008). Bidobacterium carbohydrases Their role in breakdown and
Microbiology, 67(6), 25262530. synthesis of (potential) prebiotics. Molecular Nutrition and Food Research, 52(1),
Ramagopal, U. A., Rutter, M., Toro, R., Hu, S., Maletic, M., Gheyi, T., et al. (2009). 146163.
Crystal structure of putative beta-galactosidase from Bacteroides fragilis. http:// Van Loo, J., Cummings, J., Delzenne, N., Englyst, H., Franck, A., Hopkins, M., et al.
www.rcsb.org/pdb/explore.do?structureId=3CMG. Accessed 16.06.09. (1999). Functional food properties of non-digestible oligosaccharides: A
Rastall, R. A. (2004). Bacteria in the gut: Friends and foes and how to alter the consensus report from the ENDO project (DGXII AIRII-CT94-1095). British
balance. Journal of Nutrition, 134(8), 2022S2026. Journal of Nutrition, 81(2), 121132.
Richard, J. P., Westerfeld, J. G., Lin, S., & Beard, J. (1995). Structure-reactivity Vasella, A., Davies, G. J., & Bhm, M. (2002). Glycosidase mechanisms. Current
relationships for beta-galactosidase (Escherichia coli, lac Z). 2. Reactions of the Opinion in Chemical Biology, 6(5), 619629.
galactosyl-enzyme intermediate with alcohols and azide ion. Biochemistry, Vocadlo, D. J., & Davies, G. J. (2008). Mechanistic insights into glycosidase chemistry.
34(37), 1171311724. Current Opinion in Chemical Biology, 12(5), 539555.
Roberfroid, M. (2007). Prebiotics: The concept revisited. Journal of Nutrition, 137(3), Wallenfels, K. (1951). Enzymatische synthese von oligosacchariden aus
830s837s. disacchariden. Naturwissenschaften, 38(13), 306.
Roberts, H. R., & Pettinati, J. D. (1957). Oligosaccharide production, concentration Wallenfels, K., & Weil, R. (1972). In P. D. Boyer (Ed.). The enzymes (vol. 7,
effects in the enzymatic conversion of lactose to oligosaccharides. Journal of pp. 617663). New York: Academic Press.
Agricultural and Food Chemistry, 5(2), 130134. White, J. S. (2000). Sugar, special sugars. In Kirk-othmer encyclopedia of chemical
Rojas, A. L., Nagem, R. A. P., Neustroev, K. N., Arand, M., Adamska, M., Eneyskaya, E. technology. doi:10.1002/0471238961.1916050323080920.a01. Accessed
V., et al. (2004). Crystal structures of beta-galactosidase from Penicillium sp. and 16.06.09.
its complex with galactose. Journal of Molecular Biology, 343(5), 12811292. Woodley, J. M. (2006). Choice of biocatalyst form for scalable processes. Biochemical
Roy, D., Daoudi, L., & Azaola, A. (2002). Optimization of galacto-oligosaccharide Society Transactions, 34(2), 301303.
production by Bidobacterium infantis RW-8120 using response surface Yanahira, S., Kobayashi, T., Suguri, T., Nakajima, M., Miura, S., Ishikawa, H., et al.
methodology. Journal of Industrial Microbiology and Biotechnology, 29(5), (1995). Formation of oligosaccharides from lactose by Bacillus circulans beta-
281285. galactosidase. Bioscience Biotechnology and Biochemistry, 59(6), 10211026.
318 A. Gosling et al. / Food Chemistry 121 (2010) 307318

Yoon, J. H., & Mckenzie, D. (2005). A comparison of the activities of three beta- Zechel, D. L., & Withers, S. G. (2000). Glycosidase mechanisms: Anatomy of a nely
galactosidases in aqueous-organic solvent mixtures. Enzyme and Microbial tuned catalyst. Accounts of Chemical Research, 33(1), 1118.
Technology, 36(4), 439446.
Zarate, S., & Lopez-Leiva, M. H. (1990). Oligosaccharide formation during enzymatic
lactose hydrolysis: A literature review. Journal of Food Protection, 53(3),
262268.

You might also like