You are on page 1of 18

Biotechnology Advances 34 (2016) 827–844

Contents lists available at ScienceDirect

Biotechnology Advances

journal homepage: www.elsevier.com/locate/biotechadv

Research review paper

Review of Levan polysaccharide: From a century of past experiences to


future prospects
Ebru Toksoy Öner a, Lázaro Hernández b, Joan Combie c,⁎
a
IBSB, Department of Bioengineering, Marmara University, Istanbul, Turkey
b
Enzyme Technology Group, Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 e/ 150 and 190, PO Box 6162, Habana 10600, Cuba
c
Montana Biopolymers Inc., 119 Cathcart Circle, Winnsboro, SC 29180, USA,

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

Article history: Levan is a fascinating β-(2,6)-linked fructose polymer with an unusual combination of properties characterized
Received 7 January 2016 in this review. In nature, levan is synthesized from sucrose by a wide range of microorganisms and a few plant
Received in revised form 1 May 2016 species. Bacterial levans often have molecular weights over 500,000 Da, are commonly branched, and form com-
Accepted 4 May 2016
pact nanospheres offering a broad spectrum of applications. The most relevant genetic, biochemical and structur-
Available online 10 May 2016
al aspects of the biosynthetic enzyme levansucrase are detailed. Optimization of parameters for levan production
Keywords:
by intact bacteria and by the isolated enzyme is surveyed. The diversity of current and potential applications of
Levan levan is illustrated by a discussion of uses ranging from personal care and aquaculture to the medical and food
Polysaccharide industries.
Fructan © 2016 Elsevier Inc. All rights reserved.
Levansucrase

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 828
2. Levan in nature and its function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 828
3. Biosynthesis of levan-type fructans . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 828
3.1. Classification of bacterial fructosyltransferases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 829
3.2. Levansucrase gene expression and protein secretion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 829
3.3. The levansucrase reaction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 829
3.4. The synthesis of hetero-oligosaccharides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 830
3.5. Structure-function relationship . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 831
4. Microbial levan production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 832
4.1. Levan production by gram-positive bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 832
4.1.1. Genera Bacillus, Paenibacillus, Geobacillus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 832
4.1.2. Other gram-positive species . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 833
4.2. Levan production by gram-negative bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 833
4.2.1. Acetic acid bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 833
4.2.2. The halophile Halomonas sp. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 834
4.2.3. Other gram-negative species . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 834
4.3. Recombinant levan production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 835
4.4. Microbial co-production of levan and non-fructan products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 835
4.5. Commercial production. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836
5. Applications of levan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836
5.1. Personal care . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836
5.1.1. Safety and activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836
5.1.2. Hair care . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836
5.1.3. Whitener . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836
5.2. Medical applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836

⁎ Corresponding author.
E-mail addresses: ebru.toksoy@marmara.edu.tr (E.T. Öner), lazaro.hernandez@cigb.edu.cu (L. Hernández), joan@polysaccharides.us (J. Combie).

http://dx.doi.org/10.1016/j.biotechadv.2016.05.002
0734-9750/© 2016 Elsevier Inc. All rights reserved.
828 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

5.2.1. Healing wounds and burned tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836


5.2.2. Anti-irritant, anti-oxidant and anti-inflammatory activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 837
5.2.3. Weight loss . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 837
5.2.4. Lower cholesterol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 837
5.3. Prebiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 837
5.4. Aquaculture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 837
5.5. Films for packaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 838
5.6. Levan in food. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 838
5.7. Additional applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 838
5.7.1. Future perspectives - demand, availability, inventions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 839
6. Declaration of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 839
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 839
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 839

1. Introduction 2. Levan in nature and its function

Levan is an unusual non-structural polysaccharide present in several “Fructans: beneficial for plants and humans”. This title of a publica-
microorganisms and a few plant species. Consisting almost solely of tion by Ritsema and Smeekens (2003) captures the spirit of the diverse
fructosyl residues linked via the β-2,6 carbons, this fructan molecule is levan roles in nature. This paper, along with an earlier publication that
packed into nano-sized, spherical forms, providing it with a remarkably included bacteria (Vijn and Smeekens, 1999), highlight the value of
low intrinsic viscosity (Arvidson et al., 2006) and somewhat greater sta- the best known fructans, levan and inulin. Vereyken et al. (2003) looked
bility than that of linear counterparts. Produced by either intact microor- at possible mechanisms for the protective effects of fructans in natural
ganisms in submerged cultures or by the isolated enzyme, levan is usually settings. They hypothesized the 5-membered ring fructans interacted
synthesized from sucrose, syrups or molasses. With some variability with cell membranes in such a way as to stabilize them and thus seem-
caused by production conditions, the microbial source of the levansucrase ingly protect them from damage. They found the small, flexible head
adds more differences to the molecular weight, degree of branching groups of levan and inulin penetrated membrane surrogates unlike a
(Jakob et al., 2013; Runyon et al., 2014), diameter, intrinsic viscosity, sta- 6-membered ring. It may be that this interaction provides improved
bility and functionalities such as immunogenic activity and adhesive membrane stability and, in turn, a higher cell survival. Researchers
strength. have identified further valuable functions described in the applications
The levan story begins with natto, a traditional Japanese food that section of this review running the gamut from anti-inflammatory and
was considered to promote long life and good health. As one component anti-viral activity to films for food packaging.
of natto, levan has long attracted the interest of researchers looking for Levan, often together with other exopolysaccharides, has been found
natural products with health benefits. to be a structural component of some biofilms in different habitats.
Lippmann is often credited with first identifying levan since he used When present in the biofilm of soil species such as Bacillus subtilis,
the term “lävulan” to describe the gum recovered from molasses in the levan shields the microorganisms from desiccation as the water level
sugar beet industry (v. Lippmann, 1881). But it was the Australian bacte- changes, helps glue cells in a favorable environment and protects the
riologist Greig-Smith who proposed the term “levan” for the levorotation community from predatory organisms (Dogsa et al., 2013). Sucrose uti-
of polarized light and properties analogous to dextran in 1901 (Greig- lization by secreted levansucrase appears to be a fitness factor for the in
Smith, 1901). planta life of several bacterial species. Levan as a biofilm component con-
For more than a hundred years numerous uses have been identified tributes to the virulence of the plant pathogens Erwinia amylovora and
for levan polysaccharide but only a few have been commercially realized Pseudomonas syringae (Koczan et al., 2009; Mehmood et al., 2015). In
simply because it has not been widely available in meaningful quanti- the endophyte Gluconacetobacter diazotrophicus, levan in the biofilm
ties. While levan production in the laboratory is straightforward, bottle- acts as an oxygen difussional barrier helping create the microaerobic
necks in the downstream process have hampered scale up. Technical conditions required for nitrogen fixation (Hernández et al., 2000;
issues associated with handling large volumes of alcohol, impracticality Velázquez-Hernández et al., 2011). As a secondary but no less important
of dialysis on a large scale and unavailability of ultrafiltration equip- role, levan in biofilms constitutes an extracellular nutrient reservoir that
ment for multi-ton operations have complicated purification. Moreover, can be used as an energy source by bacteria under starvation conditions.
without prompt enzyme inhibition at the end of the fermentation step,
depolymerization quickly decimates levan. Drying of the sticky product 3. Biosynthesis of levan-type fructans
is another hurdle. Lyophilization and vacuum drying commonly
employed in the lab (Han, 1990; Kazak Sarilmiser et al., 2015) are not In nature, fructans are synthesized from sucrose by a restricted
readily scaled. Spray drying is widely used in industry but the adhesive number of plant species and many microorganisms, which include Ar-
strength of levan requires use of a carrier resulting in decreased product chaea, fungi and a wide range of bacteria. Plants synthesize fructans in
purity. the vacuole by the concerted action of various fructosyltransferases,
This review opens with a sketch of the functional roles of levan in na- each with its own preferential donor and acceptor substrates.
ture. It then moves on to a comprehensive discussion of levan biosyn- Sucrose:fructan 6-fructosyltransferase (6-SFT; EC 2.4.1.10), also recog-
thesis, detailing the most relevant genetic, biochemical and structural nized as sucrose:sucrose 6-fructosyltransferase (6-SST) by its dual
aspects of levansucrase enzymes. Optimization of parameters for levan ability to use a fructan molecule or sucrose as the acceptor substrate,
production by intact bacteria and by the isolated enzyme is surveyed. is responsible for the formation of β(2–6) fructosyl-fructose linkages
The diversity of current and potential applications of levan is illustrated in plant fructans. Alone, 6-SFT synthesizes linear levan chains direct-
by consideration of uses ranging from personal care and aquaculture to ly from sucrose via the trisaccharide intermediate 6-kestose. Other
the medical and food industries. The review closes with suggestions of natural fructosyl acceptors of this enzyme are 1-kestose and 6G-
innovative approaches to increasing productivity by levan producers kestotriose (Lasseur et al., 2011; and references therein). The production
and suggests heretofore untested sectors for its use. of levan neoseries or mixed levan (graminan)-type fructans requires
E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 829

the combined action of 6-SFT with a second, third, or even fourth guarantees the prevalence of levan synthesis over degradation when the
fructosyltransferase, depending on the plant species (Vijn and substrate sucrose is available.
Smeekens, 1999). 6-SFT is a key enzyme in fructan monocot biosynthe- Levansucrases are released to the external medium or are attached
sis. The evergreen frost-hardy species Pachysandra terminalis (family to the cell surface. Variations in the enzyme secretion routes have
Buxaceae) is the only dicot plant reported so far to contain a functional been described for different hosts. Levansucrase secretion in gram-
6-SST/6-SFT enzyme (Van den Ende et al., 2011). positive bacteria involves cleavage of signal-peptide containing precur-
Plant fructans generally have a low degree of polymerization sors (Rathsam et al., 1993; Steinmetz et al., 1985; van Hijum et al., 2004;
(DP b 101 to 102). Bacterial inulin and levan reach much higher molec- Waldherr et al., 2008). The vast majority of gram-negatives secrete
ular masses (103 to N 104 fructosyl units), offering a broader spectrum of levansucrase by a signal-peptide-independent pathway (Geier and
biotechnological applications. As bacterial levan is the polysaccharide Geider, 1993; Jakob et al., 2012; Li and Ullrich, 2001; Song et al., 1993;
addressed in this review, the current section will be focused on the bio- Song et al., 1998), with the most remarkable exception being
synthetic enzyme levansucrase. G. diazotrophicus. This endophytic bacterium is equipped with type-II
secretory machinery for translocation of the mature and fully folded
levansucrase across the outer membrane. The precursor enzyme is syn-
3.1. Classification of bacterial fructosyltransferases thesized with a 30-amino acid N-terminal signal peptide which is
cleaved off during transport to the periplasm (Arrieta et al., 2004;
Bacterial fructosyltransferases are multifunctional enzymes ca- Hernández et al., 1999a).
pable of directly converting sucrose [α-D-glucopyranosyl-(1,2)-β-
D -fructofuranoside] into highly polymerized fructans. The enzyme is 3.3. The levansucrase reaction
named inulosucrase (EC 2.4.1.9) when it synthesizes inulin, and
levansucrase (EC 2.4.1.10) when the polymerization product is levan. All levansucrases catalyze the transfer of fructosyl residues from su-
Levansucrases can also form β-(2,1) fructosyl-fructose linkages either crose to a variety of acceptor molecules, such as water (release of glu-
to synthesize inulin-type fructooligosaccharides (FOS) or to create cose and fructose), sucrose (synthesis of 1-kestose or 6-kestose), the
branching points that connect the β-(2,6) linked chains of the polymer. synthesized β-(2,6) linked oligofructans (formation and elongation of
The presence of inulosucrase has been reported only in gram-positive levan), and the released monosaccharides glucose (synthesis of sucrose
bacteria, specifically in species from three genera (Streptococcus, or blastose) and fructose (synthesis of inulobiose or levanbiose). De-
Leuconostoc and Lactobacillus) which may additionally contain pending on the enzyme origin, there are remarkable differences in the
levansucrases (Olvera et al., 2007; Anwar et al., 2010). The occurrence affinity for the fructosyl acceptors emerging during the reaction. In gen-
of levansucrase is wide spread in gram-positive and gram-negative eral, levansucrases from gram-positive bacteria (for instance, Bacillus
bacteria. subtilis, Bacillus megaterium and Streptococcus salivarius) produce
the polymer without accumulating the intermediate oligofructans
(Chambert et al., 1974; Homann et al., 2007; Song and Jacques, 1999),
3.2. Levansucrase gene expression and protein secretion whereas the enzymes from gram-negative species (for instance,
Gluconacetobacter diazotrophicus, Zymomonas mobilis, Erwinia amylovora,
A simple search in databases reveals the occurrence of putative and Pseudomonas syringae) render high levels of oligofructans with a
levansucrase genes in bacteria with different habitats. Most of the spe- consequent lower yield of levan (Bekers et al., 2002; Caputi et al., 2013;
cies contain a unique levansucrase gene which often constitutes the Hernández et al., 1995; Visnapuu et al., 2015). The accumulated
first component of a chromosomal operon. Exceptionally, the plant oligofructans are successively released from the enzyme after each
pathogen Pseudomonas syringae contains three levansucrase genes fructosyl transfer in the so-called non-processive reaction. Little of the
(two in the chromosome and one in a plasmid) but only two of them G. diazotrophicus levansucrase products 1-kestose and nystose were con-
are functionally expressed (Li and Ullrich, 2001). In the soil bacterium sumed either as a donor or acceptor substrate (Hernández et al., 1995),
B. subtilis, the levansucrase gene initiates an operon which is tightly con- explaining their constant high concentration even after sucrose deple-
trolled by a sucrose-inducible antitermination mechanism (Crutz et al., tion. All levansucrases yield polysaccharides of high molecular weight,
1990). In general, sucrose is required to induce levansucrase transcription which may differ in their size distribution. The high polymerization de-
in the genera Bacillus, Paenibacillus and Geobacillus (Choi et al., 2004; gree of bacterial levans can be explained by the fact that the growing
Homann et al., 2007; Inthanavong et al., 2013). By contrast, constitutive fructan chain remains bound to the enzyme as it is progressively elongat-
expression of the enzyme is a common feature in bacteria that reside ed through a processive mechanism (Ozimek et al., 2006). When the
in sucrose-containing habitats, for instance, Streptococcus salivarius original substrate sucrose is depleted, the synthesized polymer acts as
(Rathsam and Jacques, 1998), Actinomyces naeslundii (Bergeron et al., a fructosyl donor. Then, levansucrase cleaves the β-(2,6) linkages of the
2000); Zymomonas mobilis (Senthilkumar et al., 2009); Erwinia amylovora levan chain in an exo-type manner causing the consecutive release of
(Geier and Geider, 1993) and Gluconacetobacter diazotrophicus the terminal fructose until the reaction stops at a branching point
(Hernández et al., 2000). In the latter species, the first gene of the (Chambert et al., 1974; Méndez-Lorenzo et al., 2015). In this sense, the
single-copy levansucrase-exolevanase operon is in fact expressed consti- β-(2,1) fructosyl-fructose linkages appear to prevent branched levans
tutively, but the exolevanase transcription is induced at low fructose con- from extensive hydrolysis by its own synthesizing enzyme. Fig. 1 illus-
centrations and repressed by glucose, which is the first released product trates the structure of both linear and branched levans.
of the levansucrase reaction (Menéndez et al., 2009). Several bacteria Levansucrase exhibits Michaelis-Menten kinetic properties for
contain both levansucrase and endo- or exo-type levanase genes forming both the hydrolase and transferase activities (Chambert et al., 1974;
either an operon or located in separate loci. Gene expression studies and Hernández et al., 1995; Song and Jacques, 1999). By contrast, inulosucrase
evolutionary analysis indicate that levan synthesis and degradation are does not follow substrate-saturation kinetics (van Hijum et al., 2003), be-
exclusive but also complementary habitat-related processes in a bacterial having in this sense analogously to plant fructosyltransferases (Ritsema
host. Fructose, the degradation product of levan and inulin, activates et al., 2006). The ratio of hydrolysis versus transfructosylation activities
fructanase expression under cell-starvation conditions. Fructan hydroly- of both levansucrases and inulosucrases is highly dependent on the initial
sis is otherwise transcriptionally repressed by glucose, the immediate en- reaction conditions, but it varies during the course of the reaction. Su-
ergy source generated during sucrose transfructosylation (Bergeron and crose hydrolysis occurs optimally at about 50–60 °C, but fructan (FOS
Burne, 2001; Martin-Verstraete et al., 1999; Menéndez et al., 2009; and/or levan) formation is favored at lower temperatures (10–40 °C)
Zeng et al., 2006). The strictly regulated control of levanase transcription and with the increase of initial sucrose concentrations to 300 mM or
830 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

Fig. 1. Schematic representation of linear (A) and branched (B) levan.

more (Chambert and Gonzy-Treboul, 1976; Támbara et al., 1999; Vigants levan, while an increase in the ionic force resulted in a shift from the bi-
et al., 2013; Visnapuu et al., 2015). As sucrose nears depletion, hydrolysis modal type to a single low-molecular-weight (LMW) levan (7.2 kDa)
tends to become the predominant reaction. The addition of NaCl and (Chambert and Petit-Glatron, 1989; Tanaka et al., 1980). At high tem-
organic solvents in the reaction mixture generates water-restricted envi- perature (50 °C), levansucrase from Bacillus licheniformis RN-01 synthe-
ronments promoting the reactions of transfructosylation over hydrolysis sized HMW levan (612 kDa) as a major product, but when the
(Castillo and López-Munguía, 2004; Chambert and Petit-Glatron, 1989; temperature was decreased to 30 °C or NaCl was added to the reaction,
Trujillo et al., 2004). Levansucrases perform best in the pH range 5–7 LMW levan (11 kDa) was mainly synthesized (Nakapong et al., 2013).
and in most cases pH variations do not affect the rate of hydrolase/
transferase activities (Caputi et al., 2013; Hernández et al., 1995; 3.4. The synthesis of hetero-oligosaccharides
Homann et al., 2007; Kim et al., 2003; Visnapuu et al., 2015). Interestingly,
levansucrase from Zymomonas mobilis catalyzes almost exclusively the Sucrose is the natural and preferred substrate of levansucrases. The
synthesis of high DP levan at pHs below 6 in its insoluble microfibril enzyme can also react on raffinose [α-D-galactopyranosyl-(1,6)-α-D-
form, but behaves mostly as a hydrolase at pHs above 7 in its soluble glucopyranosyl-(1,2)-β-D-fructofuranoside] and probably other oligo-
dimer form (Goldman et al., 2008). Levan polymerization by the enzyme saccharides containing a terminal sucrose with the free fructose unit.
from other bacteria has not been associated with the formation of The KM values for sucrose (10–50 mM) are lower than those for raffi-
microfibrils. nose in all kinetically characterized levansucrases (Alamäe et al., 2012;
The reaction conditions may influence not only the output of Caputi et al., 2013 and references therein; Olvera et al., 2007). Two
transfructosylation but also the average size and number of branches decades ago, levansucrase from Pseudomonas syringae pv. phaseolicola
of the product levan. The synthesis of a bimodal levan molecular weight was reported to have a relatively high KM for sucrose (160 mM) and
distribution is a particular property of levansucrase from Bacillus was unable to react on raffinose (Hettwer et al., 1995). More recently,
species. Relevant reaction factors that modulate the polymer molecular in recombinant gene expression experiments, Alamäe et al. (2012) de-
weight distribution include: enzyme concentration, temperature, ionic termined that all three levansucrases (Lsc1, Lsc2 and Lsc3) of P. syringae
strength, and the presence of water-miscible organic solvents. A low pv. tomato are capable of synthesizing levan from raffinose as the sole
concentration of B. subtilis levansucrase (0.1 U/mL) favored the synthe- substrate but the affinity for raffinose was much lower than for sucrose.
sis of high-molecular-weight (HMW) levan (average size 2300 kDa) by The same authors also demonstrated that at least one levansucrase
promoting the processive elongation of the fructan chain, with negligi- (LscA) of P. chlororaphis subsp. aurantiaca uses raffinose as a fructosyl
ble effect of the initial substrate concentration (Porras-Domínguez et al., donor.
2015; Raga-Carbajal et al., 2016). In the nineteen eighties two papers Levansucrase shows a wide spectrum of non-conventional fructosyl
also referring to the B. subtilis enzyme reported that the presence of eth- acceptors, including mono and disaccharides, aromatic and aliphatic
anol, polyethylene glycol or acetonitrile favored the synthesis of HMW alcohols (Beine et al., 2008; Han et al., 2009; Mena-Arizmendi et al.,
E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 831

2011; Seibel et al., 2006; Visnapuu et al., 2011). Their combination with apo form and complexed with its acceptor substrate 6-kestose
the appropriate fructosyl donor allows the production of different hetero- (Lammens et al., 2012). To date it is still the only 3D structure of a
oligosaccharides. As the main example of biotechnological interest, plant FT available in databases. As other GH32 enzymes, 6-SST/6-SFT
levansucrase can transfer the fructosyl residue of sucrose to the acceptor displays a bimodular fold consisting of a five-bladed β-propeller catalyt-
lactose [β-D-galactopyranosyl-(1,4)-D-glucopyranose] to produce ic domain at the N-terminal part of the protein connected to a shorter C-
lactosucrose [β-D-galactopyranosyl-(1,4)-α-D-glucopyranosyl-(1,2)-β-D- terminal β-sandwich domain which is important for maintaining struc-
fructofuranoside], a sweetener with a prebiotic effect as reviewed in Mu tural stability (Fig. 2).
et al. (2013). The GH68 enzymes structurally characterized so far are levansucrases
from B. subtilis (Meng and Fütterer, 2003), Gluconacetobacter
3.5. Structure-function relationship diazotrophicus (Martinez-Fleites et al., 2005), Bacillus megaterium
(Strube et al., 2011) and Erwinia amylovora (Wuerges et al., 2015),
Bacterial fructosyltransferases (levansucrase and inulosucrase) belong inulosucrase from Lactobacillus johnsonii (Pijning et al., 2011), and
to family 68 of glycoside hydrolases (GH), and together with the family lactosucrose-producing β-fructofuranosidase from Microbacterium
GH32 members (eukaryotic fructosyltransferases, invertases, sucrose-6P saccharophilum K-1 (formerly known as Arthrobacter sp. K-1)
hydrolases and fructanases) comprise the β-fructofuranosidase clan GH- (Tonozuka et al., 2012), all of which display a single domain with the
J as defined in the CAZy database (http://www.cazy.org/). All these en- conserved five-bladed β-propeller architecture (Fig. 2). The amino acid
zymes operate via a double-displacement mechanism (also known as residues involved in binding and cleavage of the fructosyl-donor sub-
ping-pong mechanism) that involves the formation and subsequent hy- strate are perfectly superimposable in a deep funnel-shaped central cav-
drolysis of a covalent fructosyl-enzyme intermediate. The reaction occurs ity. Co-crystallization of catalytically inactive mutants with natural
with an overall retention of the anomeric configuration of the fructosyl ligands has revealed that sucrose is accommodated similarly at the active
residue. site of all GH68 enzymes (Meng and Fütterer, 2003; Meng and Fütterer,
The crystal structure of the levan-producing enzyme 6-SST/6-SFT 2008; Pijning et al., 2011). Three fully conserved carboxylic residues
from the eudicot species Pachysandra terminalis was determined in the (the catalytic triad) located at the bottom of the active site pocket cleave

Fig. 2. Overall structures of levan-synthesizing enzymes from different sources. (A) 6-SST/6-SFT from the plant species Pachysandra terminalis (PDB: 3UGF); (B) Levansucrase from the
gram-positive bacterium Bacillus subtilis (PDB: 1OYG); (C) Levansucrase from the gram-negative bacterium Gluconacetobacter diazotrophicus (PDB: 1W18). The plant 6-SST/6-SFT
(family GH32) displays a bimodular fold consisting of a five-bladed β-propeller catalytic domain and a C-terminal β-sandwich domain. The bacterial levansucrases (family GH68) display a
single domain with the conserved five-bladed β-propeller architecture. The blades 1 to 5 are colored in orange, blue, green, yellow and red, respectively. The three catalytic residues
(catalytic triad) of each enzyme are shown in ball-and-stick representation. The figures were prepared with Molecular Operating Environment (MOE), available at http://www.chemcomp.
com/MOE-Molecular_Operating_Environment.htm.
832 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

the glycosidic bond by functioning as the nucleophile (Asp), the acid/base levan production by Bacillus lentus V8 cultures on sugarcane molasses
catalyst (Glu) and the transition-state stabilizer (Asp) (Meng and (Abou-taleb et al., 2015), by Halomonas smyrnensis AAD6T cultures
Fütterer, 2003; Pons et al., 2004). Extensive mutagenesis experiments on sugar beet and starch molasses (Küçükaşık et al., 2011), by
support the idea of an invariant mechanism for the hydrolase activity of Microbacterium laevaniformans PTCC 1406 cultures on date syrup
levansucrases and inulosucrases. On the contrary, the lack of conservation (Moosavi-Nasab et al., 2010), by Paenibacillus polymyxa NRRL B-18475
in regions lining the surface of the active site cavity and presumably in- cultures on sugar beet molasses and sugarcane syrup (Han and Watson,
volved in binding the fructosyl acceptors appears to determine the differ- 1992) and by Zymomonas mobilis ATCC 31821 cultures on sugarcane
ences in the linkage type and chain length of the transfructosylation molasses and syrup (de Oliveira et al., 2007) have been reported. Besides
products observed among GH68 enzymes (Anwar et al., 2012; Homann the crude form (Abou-taleb et al., 2015; Moosavi-Nasab et al., 2010)
et al., 2007; Mardo et al., 2014; Strube et al., 2011). pretreated forms of syrups and molasses were also used as substrate.
Fructosyltransferases from several gram-positive species require Levan yields comparable to sucrose containing media were recovered
calcium ions for activity (van Hijum et al., 2006 and references therein). from P. polymyxa cultures when filter clarified sugarcane syrup was sup-
B. subtilis levansucrase and L. johnsonii inulosucrase contain a calcium- plemented with peptone or when beet molasses was desalted by passing
binding site at a similar position (Meng and Fütterer, 2003; Pijning it through a gel filtration column (Han and Watson, 1992). Other
et al., 2011), where Ca2+ can stabilize a region of important connections pretreatment methods included various combinations of acid treatment,
between active site residues. In G. diazotrophicus levansucrase, which is pH adjustment, tricalcium phosphate and activated carbon treatments
fully active in the absence of Ca2 +, a similar fold-stabilizing role is (Küçükaşık et al., 2011) and centrifugation followed by filtration (de
accomplished by a disulfide bridge (Martinez-Fleites et al., 2005). Oliveira et al., 2007).
These structural variations are not directly responsible for the different
product specificity of the three enzymes.
4.1. Levan production by gram-positive bacteria
4. Microbial levan production
4.1.1. Genera Bacillus, Paenibacillus, Geobacillus
Levan is produced extracellularly from sucrose-based substrates by a The commercial natto starter Bacillus subtilis (natto) Takahashi has
variety of bacteria, including the genera Acetobacter, Bacillus, Erwinia, long been investigated for high-level levan production. When the type
Gluconobacter, Halomonas, Microbacterium, Pseudomonas, Streptococcus and concentration of sugars, initial pH, cultivation temperature and ag-
and Zymomonas (Table 1). Like other microbial extracellular polymeric itation conditions were optimized, shaking cultures produced 49.4 g/L
substances (EPS), levan production is significantly affected by fermenta- levan within 21 h and GPC analysis of levan samples produced two
tion conditions such as temperature, pH, oxygen concentration, bioreac- clear peaks of 1794 kDa and 11.9 kDa molecular weight (Shih et al.,
tor configuration and culture medium. Generally, the polymer 2005). Yields obtained by submerged cultures were further improved
production is associated with microorganism growth. Upon the enzy- by using alginate-immobilized B. subtilis (natto) Takahashi cells reaching
matic cleavage of sucrose, the fructose moiety is added to the growing 70.6 g/L levan in 72 h (Shih et al., 2010). In this study, sucrose concentra-
levan chain. Part of the released glucose is metabolized for growth tion, medium pH, metal ions and agitation speed were optimized to
and the rest is accumulated in the fermentation medium. Development maintain the integrity of the beads. N70% of the biocatalytic activity
of strategies for the effective utilization of this excess glucose by co- was retained even after nine successive 72 hours long reaction cycles.
production of levan with other high value products has been a part of The presence of the two peaks corresponding to high and low molecular
many microbial levan production processes. For the production, both weights of the produced levan was found to be independent of metal ion
free and immobilized cells may be used and the fermentations may be addition (Shih et al., 2010). With the same microbial system, a recent
operated in batch, fed-batch and continuous modes. study demonstrated high yield levan production under controlled
Syrups and molasses are low cost substitutes for sucrose and they bioreactor conditions (Wu et al., 2013). In this study, a 10-L stirred
have been widely used as substrates for fermentative production of tank bioreactor was used and levan yields of 61 g/L and 100 g/L were re-
microbial polysaccharides (Özcan and Toksoy Öner, 2015). As such, covered from the batch and fed-batch suspension cultures, respectively.

Table 1
Levan production by various native bacteria exposed to sucrose-containing substrates.

Bacterial species and strain Substrate Yield (Time) Reference

Acetobacter xylinum NCIM 2526 Suc (60 g/L) 13.25 g/L (60 h) Srikanth et al. (2015)
Bacillus amyloliquefaciens NK-ΔLP Suc (250 g/L) 22.6 g/L (48 h) Feng et al. (2015a)
B. amyloliquefaciens NK-Q-7 Suc (250 g/L) 31.1 g/L (48 h) Feng et al. (2015b)
Suc (250 g/L) 57.95 g/L (60 h)
Bacillus lentus V8 Abou-taleb et al. (2015)
Scane molasses (250 g/L) 49.86 g/L (60 h)
Suc (196.8 g/L) 47.8 g/L (96 h)
Bacillus licheniformis NS032 Kekez et al. (2015)
Suc (397.6 g/L) 99.2 g/L (96 h)
Bacillus methylotrophicus SK 21.002 Suc (300 g/L) 100 g/L (16 h) Zhang et al. (2014)
Suc (250 g/L) 61.46 g/L (24 h)
Bacillus subtilis (natto) Takahashi Wu et al. (2013)
Suc (250 g/L and pulse feeding at 30 mg/mL) 100 g/L (120 h)
B. subtilis (natto) CCT7712 Suc (400 g/L) 111.6 g/L (16 h) dos Santos et al. (2013)
Sbeet molasses (30 g/L) 12.4 g/L (210 h) Küçükaşık et al. (2011)
Halomonas smyrnensis AAD6 Suc (50 g/L) + mannitol (30 g/L) 6.60 g/L (160 h) Ateş et al. (2013)
Suc (50 g/L) + 50 mM boric acid 8.84 g/L (160 h) Kazak Sarilmiser et al. (2015)
Suc (200 g/L) 48.9 g/L (48 h)
Microbacterium laevaniformans PTCC 1406 Moosavi-Nasab et al. (2010)
Date syrup (250 g/L) 10.48 g/L (48 h)
Paenibacillus polymyxa EJS-3 Suc (188 g/L) 35.26 g/L (60 h) Liu et al. (2010)
Pseudomonas fluorescens NCIM 2059 Suc (60 g/L) 15.42 g/L (6 days) Jathore et al. (2012)
Serratia levanicum NN Suc (200 g/L) 50 g/L (178 h) Kikuchi et al. (2010)
Zymomonas mobilis CCT 4494 (Immobilized cells) Suc (300 g/L) 112.53 g/L (18 h) Lorenzetti et al. (2015)
Z. mobilis NRRL B-14023 Suc (299 g/L) 40.2 g/L (42.3 h)
Silbir et al. (2014)
Z. mobilis NRRL B-14023 (Immobilized cells) Suc (299 g/L) 31.8 g/L (D = 0.14 h−1)

Suc: sucrose, Scane: sugarcane, Sbeet: sugarbeet.


E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 833

Another important aspect of this study is that it showed the possibil- in addition to the fructose branches observed in both structures. Fur-
ity of controlling the molecular weight of levan by means of fermenta- thermore, statistical optimization of medium composition pointed to
tion conditions. The total levan produced was the same regardless of the importance of sucrose, yeast extract and CaCl2 content of the medium
conditions but the ratios of high to low molecular weight versions var- and resulted in 35.26 g/L of levan produced by P. polymyxa EJS-3 cultures
ied. At low agitation speeds and high sucrose concentrations, more grown in sucrose containing complex medium (Liu et al., 2010).
low molecular weight levan was produced than high molecular weight
but the ratio was reversed at a higher speed and low sucrose concentra- 4.1.2. Other gram-positive species
tion (Wu et al., 2013). A lower ratio of enzyme to sucrose in the Microbacterium laevaniformans is another levan producer (Fuchs
submerged culture favors the initial production of more 6-kestose pre- et al., 1985). Detailed chemical characterization of the levan as well as
cursors which bind the enzyme and are elongated to form levan chains. its antitumor activity (Oh et al., 2004) and rheological properties (Bae
Thus, in the presence of excess sucrose, more levan chains are formed et al., 2008) were reported for the M. laevaniformans KCTC 9732. Chemical
but they are shorter. Less is known about the effects of the agitation characterization revealed that levan was made of 70.2% β(2–6) linkages,
speed but it is possible that high agitation rates may simply increase 17% terminal groups and 12.9% branching at the C1, C2 or C6 positions
the number of effective contacts between the enzyme and sucrose, with a β(1–2) linkage (Oh et al., 2004). The first systematic study on
thus increasing the polymerization rate. the levan production by this bacterium was reported by Moosavi-Nasab
In another study, B. subtilis CCT 7712 isolated from natto produced et al. (2010). In this study, when M. laevaniformans PTCC 1406 cultures
111.6 g/L levan from 400 g/L of sucrose in 16 h. When the produced poly- were grown under optimized fermentation conditions, more than four
mers were analyzed by molecular exclusion chromatography, two frac- times higher levan yields could be reached from sucrose as compared to
tions were obtained with molecular weights of 568 kDa and 50 kDa date syrup.
with abundances of 13.39% and 86.61%, respectively (dos Santos et al., Lactic acid bacteria of the genera Streptococcus, Leuconostoc and
2013). Hence the dominance of low molecular weight levan at high Lactobacillus are also known to produce levan (Olvera et al., 2007;
sucrose concentrations was also observed with this strain. Rathsam and Jacques, 1998; Van Geel-Schutten et al., 1999). However,
Bacillus methylotrophicus SK 21.002 isolated from soil samples collect- most of the studies in the literature are mainly focused on the enzyme
ed from beet and sugarcane gardens in China generated such a high yield production and characterization rather than levan production by the in-
that it was considered economically viable for industrial levan produc- tact bacterium in culture conditions.
tion. After optimization of production conditions, up to 100 g/L, low mo-
lecular weight levan could be recovered from cultures grown on 300 g/L 4.2. Levan production by gram-negative bacteria
sucrose (Zhang et al., 2014).
Optimization of initial pH, incubation temperature, shaking speed 4.2.1. Acetic acid bacteria
and inoculum size of Bacillus lentus V8 fermentations achieved yields Though the Acetobacter genus was known for producing levan
of 57.95 g/L or 49.86 g/L when cultures were grown on medium supple- (Han, 1990), the first comprehensive study on levan biosynthesis and
mented with sucrose or sugar cane molasses, respectively (Abou-taleb levansucrase enzyme was reported on the endophytic nitrogen-fixing
et al., 2015). The levansucrase reaction is optimal under slightly acidic bacterium Gluconacetobacter diazotrophicus (formerly known as
conditions (pH range 5–7) and this is supported by numerous studies. Acetobacter diazotrophicus) (Hernández et al., 1995). The authors found
For example, a study investigating the effects of sucrose and ammonium that G. diazotrophicus strain SRT4 converted sucrose into a branched
chloride concentrations and initial pH of the medium on levan produc- levan with the molecular weight above 2 × 106 Da (Hernández et al.,
tion in low (60–200 g/L) and high (300–400 g/L) sucrose systems by a 1995). In another study, the G. diazotrophicus type strain PAl 5 was re-
Bacillus licheniformis NS032 strain isolated from a petroleum sludge ported to produce 24.7 g/L levan in batch cultures growing on 100 g/L su-
sample pointed to the decisive role of the culture pH at the start of the crose and under biological N2-fixation (BNF) conditions which in turn
production (Kekez et al., 2015). The central and crucial role of the pH suggested the high potential of this strain as a microbial producer since
optima 6.97 and 7.37 for low and high sucrose systems was attributed it does not require N-source supplementation of the production medium
to the need to increase the initial pH at high sucrose concentrations (Molinari and Boiardi, 2013). For the same strain, enhanced levan forma-
that in turn suggested a correlation between the levan production and tion was reported when G. diazotrophicus PAl 5 cells were cultivated on a
the amounts of organic acids produced as metabolic end products. solid medium containing a high concentration of phosphate. However
The genus Paenibacillus is another microbial system that has this finding could not be verified with submerged cultures (Idogawa
attracted great interest due to the high biotechnological potential et al., 2014). The gram-negative aerobe Gluconacetobacter xylinus
of the EPS that include levan and curdlan (Liang and Wang, 2015). (formerly Acetobacter xylinum), well known for its ability to accumulate
Levan production by Paenibacillus polymyxa (formerly known as Bacillus high levels of cellulose from glucose, has also been shown to produce a
polymyxa) was first reported in 1943 (Hestrin et al., 1943). Then in 1990, water soluble polymer when grown on sucrose. The presence of a single
production conditions and characterization of levan by P. polymyxa fructose peak in the HPLC pattern of the acidic hydrolysate of the polymer
NRRL B-18475 strain were reported for cultures grown on sucrose by A. xylinum NCI 1005 as well as comparison of the NMR spectra with
(Han and Clarke, 1990). Levan produced by strain NRRL B-18475 had a that of commercial levan of Erwinia herbicola confirmed the levan-type
molecular weight of 2 × 106 Da and 71% β(2–6) linkages, 13% terminal structure of the polymer (Tajima et al., 1997). Another Gluconacetobacter
groups and 12% branching at the C1 position with a β(1–2) linkage. xylinus I-2281 strain isolated from vinegar fermentations was known to
When cultures were grown on low-cost fermentation substrates like produce a gluconacetan EPS composed of rhamnose, glucose, mannose,
sugar cane syrup and sugar beet molasses, the low levan yields were im- and glucuronic acid monomers. To investigate the effect of nutritional fac-
proved by supplementing the cane syrup with peptone and desalting tors on the nature, yield, and composition of EPS, cultures were grown on
the beet molasses (Han and Watson, 1992). The observed strong antiox- glucose, fructose and sucrose. Based on enzymatic assays and the molar
idant activity of the EPS produced by another strain, P. polymyxa EJS-3 ratio of monosaccharides of EPS hydrolysates, 75% gluconacetan and
(Liu et al., 2009), encouraged further investigation. Purification of the 25% levan (0.07 C-mol levan/C-mol sucrose yield) were found to be
crude EPS produced two fractions, EPS-1 and EPS-2 with respective mo- produced in the presence of sucrose (Kornmann et al., 2003). Recently,
lecular weights of 1.22 × 106 Da and 0.869 × 106 Da. Detailed structural microbial levan production by Acetobacter xylinum NCIM 2526
characterization revealed that EPS-1 and EPS-2 were composed of 79% (reclassified as Gluconacetobacter swingsii) cultures was investigated
and 72% β(2–6) linkages, 11% and 15% terminal groups and 10% and in a sucrose rich medium and by using both one-factor-at-a-time and
13% branching, respectively. Whereas both fractions were identified to factorial design. More than a 20-fold increase in levan yields could be
be levan type, EPS-2 had (2–1) linked mannose residues as branches achieved (Srikanth et al., 2015). In this study, the branched levan
834 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

structure was confirmed by sugar analysis, TGA, FTIR and NMR. Besides et al., 2013). Similar results have been reported for other microbial sys-
the levan producing acetic acid bacteria species like Gluconobacter tems. The levansucrase activity in G. diazotrophicus cultures was in-
oxydans, Gluconacetobacter xylinus and Gluconacetobacter creased when using mannitol or sorbitol as the carbon source for cells
diazotrophicus, Jakob et al. (2013) reported Gluconobacter frateurii growth. This increase of levansucrase activity could be associated with
TMW 2.767, Gluconobacter cerinus DSM 9533T, Neoasaia chiangmaiensis an enhancement of gene expression which positively influenced the en-
NBRC 101099 and Kozakia baliensis DSM 14400 as high level levan pro- zyme secretion rate (Hernández et al., 1999b). On the other hand, the
ducing strains of acetic acid bacteria. Levan from K. baliensis stood out use of high doses of the isolated B. subtilis levansucrase resulted in a sig-
with an exceptionally high MW of 2466 MDa. nificant reduction of the levan size (Raga-Carbajal et al., 2016). Ongoing
studies on the Halomonas levansucrase enzyme will shed more light
4.2.2. The halophile Halomonas sp. onto the effect of reaction conditions on the structural features of the pro-
The gram-negative, aerobic bacterium Halomonas sp. was reported as duced levan (Toksoy Öner, personal communication).
the first halophilic levan producer microorganism by Poli et al. (2009). For such systems-based approaches, Next Generation Sequencing
The recovered levan was an unbranched linear molecule and had a mo- (NGS) technologies play a central role by enabling high-throughput
lecular weight of N1 × 106 Da when analyzed by density gradient centri- genomic data at very high speed with a relatively low cost. Hence to
fugation. The producer strain was identified as a novel species of the gain more insight into the genomic organization of this halophilic
genus Halomonas and named Halomonas smyrnensis AAD6T (Poli et al., levan producer and to elucidate its biotechnological and industrial po-
2013). Since the strain combines the advantages of osmoadaptation tential, the whole genome of H. smyrnensis AAD6T was obtained by NGS.
and halophilicity that enable unsterile production under high salinity, op- Moreover, this microbial strain was found to have many poten-
timization studies were initiated to improve the yields in semi-chemical tial applications in biotechnology. In addition to being a levan pro-
media. For this, effects of initial carbon and nitrogen concentrations, pro- ducer, H. smyrnensis AAD6T also has the capacity to produce
duction pH, NaCl and nitrogen pulses, nitrogen and phosphorous limita- cationic exopolysaccharides consisting of partially acetylated N-
tions, trace elements and thiamine content of the basal production acetylgalactosamine and N-acetylglucosamine residues known to be in-
medium were systematically investigated. Supplementing the medium volved in the formation of a pellicle at the air-liquid interface of static
with boric acid stood out as the most effective stimulator by improving cultures (Diken et al., 2015). Recent studies point to the important
the sucrose utilization of the cultures and increasing the levan yields up role of Pel in cellular adhesion by maintaining the structural stability
to five-fold (Kazak Sarilmiser et al., 2015). When the molar-mass and of biofilms as well as to the advantages provided by the ability to pro-
molar-mass distributions of levan samples were analyzed by GPC-LS, duce multiple exopolysaccharides with different charges in protecting
the linear relationship between RMS radius and molar-mass confirmed the cells from diverse antimicrobials (Jennings et al., 2015). Besides
the linear structure. The molecular weights of 9.861 × 106 Da and levan and Pel, the H. smyrnensis AAD6T strain also has the genetic capac-
1.483 × 106 Da were determined for levan produced in the absence and ity to accumulate polyhydroxyalkanoates (PHAs) and osmoprotectants
presence of boric acid, respectively. and to exhibit arsenic resistance (Diken et al., 2015).
In another study, levan production by H. smyrnensis AAD6T cultures
was investigated in the presence of starch and sugar beet molasses 4.2.3. Other gram-negative species
which had been subjected to different pretreatment methods like clar- Levan production by Zymomonas mobilis was first reported in 1966
ification, pH adjustment, sulfuric acid, tricalcium phosphate, and acti- when levan biosynthesis was found to be the reason behind the low bio-
vated carbon treatments as well as different combinations (Küçükaşık mass yields observed in the presence of sucrose (Dawes and Ribbons,
et al., 2011). In this study, the highest levan yields were reached in 1966). In this study, a branched levan structure was confirmed by
pretreated sugar beet molasses. Removal of heavy metals and increases periodate oxidation, partial hydrolysis and methylation analysis. Prom-
in iron concentration were found to result in diminished maintenance ising levels of antitumor and immunomodulatory activities of levans
of the cellular integrity and lower yields. produced by Z. mobilis cultures (Calazans et al., 1997) were shown to
To transform microbial hosts into highly efficient cellular factories, be associated with the chain length with maximum inhibition observed
metabolic systems engineering is being used to redirect microbial me- with samples of 0.457 × 106 Da (Calazans et al., 2000). These findings
tabolism for the overproduction of chemicals of interest. In fact, an in- led to more systematic studies on optimization of production conditions
creasing number of bio-based chemicals are being produced profitably to achieve high titers (Bekers et al., 2003). By statistical optimization in
at industrial scale by use of engineered strains (Dai and Nielsen, 2015). the presence of sucrose, levan yields of 14.67 g/L and 21.69 g/L were re-
In the field of metabolic systems engineering, in silico genome-scale ported for the ZAG-12 (Melo et al., 2007) and ATCC 31821 (de Oliveira
models are valuable tools for deciphering the molecular mechanisms et al., 2007) strains, respectively. In another study, disruption of the ex-
and identification of targets for strain improvement. Such a genome- tracellular Z. mobilis sucrase gene (sacC) resulted in high levan yields,
scale model was constructed for H. smyrnensis AAD6T with the aim to most probably due to the three-fold increase in levansucrase (SacB) ac-
improve the productivity levels by elucidation of the interrelations be- tivities in Z. mobilis CT2 mutant cultures (Senthilkumar et al., 2004).
tween metabolic pathways and levan biosynthesis mechanisms (Ateş When Z. mobilis CCT 4494 wild type cells were chemically mutated by
et al., 2013). In this study, the in silico metabolic model was systematical- N-methyl N-nitro N-nitrosoguanidine (NTG) treatment, the ZW mutant
ly analyzed to identify critical network elements (i.e., enzymes, biochem- increased the 30.3 g/L levan yields of the wild type cultures up to
ical transformations, and metabolites) related to the levan biosynthesis 42.76 g/L in 24 h. Moreover, this study also showed the necessity of
mechanism. Results pointed out the significant relationship between maintaining anaerobic conditions and the influences of carbon/inorganic
levan biosynthesis and three metabolic pathways, namely the glycolysis, nitrogen and organic/inorganic nitrogen ratios on the levan yields (Alegre
pentose-phosphate pathway, and fructose-mannose metabolism where et al., 2005). Whereas the use of low-cost substrates like sugar beet juice
the critical reactions were concentrated around three branch points, and syrup (Bekers et al., 2001) and sugar cane molasses (de Oliveira et al.,
ribulose 5-phosphate, glucose 6-phosphate, and mannitol. Supplementa- 2007) resulted in low levan yields, Z. mobilis ATCC 31821 cultures grow-
tion with mannitol of the sucrose-based fermentation medium resulted ing on sugar cane syrup were found to reach 15.5 g/L levan concentra-
in a twofold increase in levan production in parallel with increased tions comparable to the 21.69 g/L levan yields in commercial sucrose
sucrose depletion rate, accumulated extracellular glucose, and decreased (de Oliveira et al., 2007). In another systematic study, initial substrate
fructose uptake rate. When the levan fractions obtained in the presence concentration, incubation time and initial pH of Z. mobilis B-14023
of mannitol were analyzed by GPC-LS, they were found to be batch cultures were optimized by statistical design and 40.2 g/L was re-
6.633 × 105 Da, which was shorter than previous studies with this mi- ported as the maximum levan concentration under optimum conditions
crobial system and suggested a link to the levansucrase activity (Ateş (Silbir et al., 2014). Moreover, continuous levan production in a packed
E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 835

bed bioreactor was also demonstrated in this study where Ca-alginate expressed under its own signal peptide or fused to the S. cerevisiae
immobilized Z. mobilis B-14023 cells were used. When Z. mobilis CCT acid phosphatase signal sequence. The first successful study on levan
4494 cells were immobilized by entrapment in alginate and PVA producing strains of the yeast S. cerevisiae was reported only recently
beads, levan concentrations in the range of 18.84 to 112.53 g/L were re- where either the sucrose synthase (SuSy) gene from potato or the
ported for different pH, sucrose concentration and fermentation periods spinach sucrose transporter (SUT) gene were integrated into the ge-
(Lorenzetti et al., 2015). nome of an invertase deficient S. cerevisiae strain. When these integrand
The gram-negative, nitrate-reducing bacterium Pseudomonas strains were transformed with the levansucrase gene (M1FT) from
fluorescens is known to produce levan under anaerobic conditions Leuconostoc mesenteroides, both sucrose accumulating strains were
(Fuchs, 1956). When the levan production profile of P. fluorescens found to produce levan. When grown in rich medium containing
NCIM 2059 was investigated under agitating and static culture condi- sucrose, the strain co-expressing the levansucrase and SUT accumulated
tions, higher levan titers were achieved under static conditions. Medium 7.75 g/L branched and short chain (polymerization degree of 280 equiv-
optimization studies resulted in the production of 15.42 g/L levan from alent to an approximate molecular weight of 45 kDa) levan both intra
60 g/L sucrose (Jathore et al., 2012). Comparison of the FTIR spectra and extracellularly (Franken et al., 2013). The methylotrophic yeast
as well as 13C and 1H NMR spectra of levan from P. fluorescens with Hansenula polymorpha was used to express Z. mobilis levansucrase
those from Z. mobilis and P. polymyxa suggested a branched structure enzyme (Park et al., 2004). Similarly, P. pastoris was also used to excrete
due to the high similarity of the peaks. G. diazotrophicus (Trujillo et al., 2001) and L. mesenteroides (Kang et al.,
Serratia sp. bacteria isolated from soil samples were identified as 2011) levansucrase. However these yeast systems are rather useful for
levan producers for the first time by Kojima et al. (1993). The high high-level enzyme production for in vitro levan production purposes.
molecular weight polymer (4.4 × 106 Da) was characterized by a low Transgenic plants have been approached as alternative hosts for the
degree of branching. Large scale levan production by the same Serratia production of levan with different degrees of polymerization. The idea
levanicum NN bacteria in a 200 L jar fermenter resulted in 39 g/L levan of in vivo conversion of the plant sucrose into a high DP levan was
production within the first 10 h. Then by controlling the pH of the me- first explored in tobacco and potato plants expressing the levansucrase
dium and lowering the production temperature down to 4 °C, the levan gene from B. subtilis (Ebskamp et al., 1994; Van der Meer et al., 1994).
level reached 50 g/L after seven days of cultivation (Kikuchi et al., 2010). Since then, other crops that do not produce fructans naturally such as
Levan production by Erwinia herbicola SRI403 isolated from a maize and sugar beets have been genetically modified to produce
crushing mill at a sugar factory was investigated by Blake et al. (1982). bacterial type levan using different gene sources (see Banguela and
The levan concentration in batch cultures growing on 156 g/L sucrose Hernández, 2006 for review). When levansucrase from G. diazotrophicus
reached 13.2 g/L after two days of incubation. One interesting feature was targeted to the vacuoles of transgenic tobacco, levan accumulated
of levan from Erwinia herbicola SRI403 was its very low degree of to levels ranging from 30 to 70% (w/w) of the total dry weight of mature
branching occurring at every 20 residues on average (Blake et al., leaves, the highest yield reported so far for a plant grown in soil (Banguela
1982). In another study, concentrations of medium components includ- et al., 2011). In general, plants expressing bacterial fructosyltransferase
ing the nitrogen, carbon and phosphorous sources as well as fermenta- genes exhibit aberrant phenotypes such as stunting and leaf bleaching,
tion parameters like pH and incubation time were optimized for so they are not good candidates for biotechnological applications. The
levan production by Erwinia herbicola ATCC 15552 batch cultures. production of low DP levans by foreign plant 6-SFT appears to a more fea-
Under optimized conditions, cultures converted 19% of sucrose to sible approach to achieve healthy transgenic plants. Transgenic sugar beet
levan when grown in a continuous production mode (Keith et al., roots expressing a timothy 6-SFT gene accumulated linear levan of short
1991). This study also pointed to the importance of analysis methods chains (DP 3 to N 40) to amounts that reached 20 to 75 mg per gram
for measuring the molecular weight of levan. An order of magnitude (fresh weight), without causing visible alterations of the plant phenotype
lower values (1.1–1.6 × 106 Da) were obtained with Gel Permeation (Matsuhira et al., 2014).
Chromatography (GPC) as compared to Low-angle Laser Light Scattering
(LALLS) method. 4.4. Microbial co-production of levan and non-fructan products

4.3. Recombinant levan production Microbial exopolysaccharide producers usually have the genetic ca-
pacity to produce a mixture of different polymers rather than just one
The bacterium Escherichia coli has been the predominant host used type of EPS as the product (Özcan and Toksoy Öner, 2015). Simulta-
for the production of heterologous levansucrases. Strong induced pro- neous production of multiple microbial products is a desired biorefinery
moters (for instance, T7 RNA polymerase promoter) allows high expres- approach but the processes are highly limited by the diversity of the mi-
sion levels of the transgene but the recombinant enzyme often forms crobial systems' metabolic requirements for each product (Sukan et al.,
insoluble inclusion bodies. The use of constitutive promoters appears 2015). For example, the commercial natto starter Bacillus subtilis (natto)
to be a better choice to achieve the production of soluble and properly Takahashi cultures were found to produce poly(gamma-glutamic acid)
folded levansucrases although at lower yields (Alamäe et al., 2012; (PGA) in medium containing L-glutamic acid without sucrose whereas
Hernández et al., unpublished results). E. coli is unable to secrete pro- levan was the sole product when bacteria were cultivated in medium
teins beyond its periplasm into the medium, which is a major inconve- containing sucrose without L-glutamate (Shih et al., 2005). By optimiz-
nience for the downstream purification process in scaled fermentations. ing the sucrose and glutamic acid concentrations of the medium, copro-
On the other hand, intact cells containing active levansucrase in the duction of levan and PGA was achieved in a single-batch fermentation
periplasm have been permeabilized using toluene and successfully (Shih and Yu, 2005). The same group investigated the sequential
reacted with sucrose to directly produce levan (Jang et al., 2001; Kang production of levan with poly-ε-lysine (ε-PL) and ethanol where the
et al., 2004b). Toluene-treated cells would require several water levan and B. subtilis (natto) cells were selectively removed from the
washings before usage in the food and feed industry, an approach not first fermentation cultures by ultrafiltration and the remnants contain-
recommended to establish a large-scale industrial production process. ing the accumulated glucose were fed to the second stage for the ethanol
Contrary to E. coli, the yeasts Saccharomyces cerevisiae and Pichia production by Z. mobilis cultures (Shih et al., 2010) or ε-PL production by
pastoris have both the GRAS (generally recognized as safe) status and Streptomyces albulus cultures (Shih et al., 2011).
the ability to efficiently secrete recombinant proteins to the culture me- Co-production of levan and PGA was also reported for Bacillus
dium. There are only a few reports in the literature about using yeast for amyloliquefaciens NK-1. In a recent study, to obtain high purity levan,
microbial levan production. Scotti et al. (1996) reported the cytoplasmic the pgs cluster encoding for PGA biosynthesis genes was deleted. After
accumulation of B. subtilis levansucrase in S. cerevisiae cells when optimization of culture conditions with the newly constructed NK-ΔLP
836 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

strain, up to a ten-fold increase in levan yields was reported (Feng et al., forward because the low intrinsic viscosity of aqueous levan solutions
2015a). This strain was further improved by disrupting six extracellular does not greatly impact the physical properties of finished products.
protease genes and the tasA gene encoding the major biofilm matrix Since levan is a nonionic polymer, it has broad chemical compatibility
protein. Mutant NK-Q-7 shaking cultures reached 31.1 g/L levan con- with other ingredients and polymers. One use for levan is as a hair
centrations with a 14.4% increase in yields (Feng et al., 2015b). spray resin. Because of its low viscosity, water and water/ethanol solu-
Similarly, coproduction of levan together with sorbitol (Viikari, tions can be sprayed using existing pump spray and aerosol technology
1984) and ethanol (Bekers et al., 1990; Bekers et al., 1999) were report- (Carson, 2015).
ed for Z. mobilis cultures. Lactic acid bacteria are also known to produce Rhodia Inc. developed a levan-based surfactant with cationic
a mixture of dextran and levan. In order to produce pure dextran, levan substituents for a conditioning shampoo (Gunn et al., 2009). A
production has been suppressed by changing the pH in Lactobacillus carboxymethylated levan was found to increase the tensile strength of
reuteri cultures (Van Geel-Schutten et al., 1999) or by UV radiation of chemically damaged hair. Scanning electron micrographs of hair dam-
Leuconostoc mesenteroides cultures (Siddiqui et al., 2014). aged by bleaching showed a decline in the lifting of cuticle layers caused
by bleaching after the hair had been treated with derivatized levan (Kim
4.5. Commercial production et al., 2012).

Perhaps the earliest commercial scale production of levan was at 5.1.3. Whitener
Economics Laboratory Inc., St. Paul, MN, USA in the early 1970s. The Hyperpigmentation is a darkening of the skin caused by an excess of
company did not sell the levan directly but rather used it in-house to dark brown melanin. The production pathway starts with the oxidation
produce levan hydrolase sold as a slime control agent for paper mills of tyrosine, first to L-DOPA and then to L-dopaquinone, followed by po-
(Combie, personal communication). IGI Biotechnology Inc. produced lymerization. Tyrosinase catalyzes the rate-limiting, first two steps in
levan for a short time and patented a use for levan in food applications melanogenesis so researchers have been seeking tyrosinase inhibitors
(Mays and Dally, 1989). as a means of damping overproduction of melanin. A number of com-
Today levan is produced commercially by a number of companies, pounds do lighten hyper-pigmented areas on the skin but issues with
including Natural Polymers Inc., Bainbridge, GA, USA using Bacillus poor stability in water, oxidation by light and skin irritation have led
subtilis (Combie, personal communication), Real Biotech Co., Ltd., to searches for alternatives.
Chungnam, Korea, which has produced levan with Zymomonas mobilis A 2006 patent application claims the use of levan to inhibit melanin
(Kim et al., 2005a) and Advance Co., Ltd., Tokyo, Japan, a long time production by significantly slowing tyrosinase activity, resulting in a
user of Streptococcus salivarius (Takahashi et al., 1994). At the “beautifully whitening effect” (Masayo and Takayuki, 2006). Use of an
moment, Rahn AG sells the most widely distributed, finished products interesting combination of levan and ascorbic acid is the subject of an-
containing levan in the person care sector, Proteolea® to rejuvenate other patent application. It is claimed that linking the fructosyl moiety
the appearance of skin and Slimexir® which is touted as a silhouette to ascorbic acid makes a compound with improved stability to oxidation
refiner (Rahn, 2012; Rahn, 2013). (Kim et al., 2005b).

5. Applications of levan 5.2. Medical applications

The unique combination of properties that distinguish levan from 5.2.1. Healing wounds and burned tissue
most other polysaccharides has long attracted attention. While com- Some of the most exciting research with medical applications has
mercial use has been limited, the increasing availability with the addi- been on levan-based thin films for healing damaged tissue. Compared
tion of more companies to the producer list is anticipated to bring with other polymers, what advantage does levan bring to these films?
more levan-based items into the market. A few applications are de- One intriguing idea comes from the work of Sturzoiu et al. (2011).
scribed here with a few more listed in a table at the end of this section. They reported that levan has a role in metalloproteinase activation, a
key step in the healing of tissues that have been burned or mechanically
5.1. Personal care damaged.
Costa et al. (2013) assembled levan-phosphonate and chitosan layer-
The focus on safety and green products by the personal care and by-layer to form films on glass slides. The fabrication was driven by elec-
cosmetics arena is a particularly attractive venue for levan. trostatic self-assembly. Phosphated levan-chitosan films on glass slides
had remarkable lap shear strength of 2500 ± 300 kPa. They also investi-
5.1.1. Safety and activity gated the density of cells adhering to chitosan-derivatized levan films
Levan excels at meeting all safety criteria. The Human Repeated compared with chitosan-alginate films prepared in a similar manner.
Insult Patch Test (HRIPT) and Chorioallantoic Membrane Vascular Cells adhered in far greater numbers to the levan-based films as com-
Assay (CAMVA) have shown no skin or eye irritation nor allergic contact pared to those where the levan was replaced with chitosan (110 cells
sensitization and no cytotoxicity in an Agar Diffusion Test (Montana per mm2 for levan versus 20 cells per mm2 for chitosan).
Polysaccharides, 2015). The bonus is the range of functionalities for In another study, comprehensive characterization of cast films of
levan. This biopolymer is an effective moisturizer, with a Transepidermal levan blended with chitosan and poly(ethylene oxide) (PEO) revealed
Water Loss (TEWL) nearly identical to hyaluronic acid (Kim et al., 2005a). that stability, flexibility, and transparency of the ternary blends could
It functions as an anti-irritant, counteracts inflammation, lightens hyper- be enhanced by varying the ratios of the components. Levan enhanced
pigmented patches on the skin (Kang et al., 2009; Kim et al., 2004; Kim biocompatibility was shown by the increased cell attachment and
et al., 2005b; Masayo and Takayuki, 2006), stimulates the breakdown of proliferation (Bostan et al., 2014).
fat (Rahn, 2013) and forms a tightening film on the skin (Rahn, 2012, Bone regeneration is hindered by poor adhesion or development of a
2013). fibrous capsule. Methacrylated levan beads with glass nanoparticles
showed superior adhesive strength to similar beads prepared with
5.1.2. Hair care alginate (Leite et al., 2014).
The main use for levan in hair care products is as a film-forming, hair Moreover, levan was also found to be a suitable polymer for laser-
holding ingredient. Levan finds many uses in hair fixative products. based technologies. Sima et al. (2012) utilized matrix-assisted pulsed
Simply adding it to existing hair holding gels, mousses, muds and anti- laser evaporation (MAPLE) to make films for drug delivery, microsensors
frizz products to increase their hair holding properties is fairly straight and tissue regeneration. They prepared films with a gradient of levan and
E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 837

oxidized levan. Cellular proliferation was highest on oxidized levan with or as an arteriosclerosis reducing agent (Haber et al., 2006). Belghith
a substantial amount of growth observed around the boundary area be- et al. (2012) tested the effect of levan on rats fed a cholesterol-rich
tween the oxidized levan and the native levan (Axente et al., 2014; diet. Test animals were treated with 5% (w/w) levan dissolved in their
Sima et al., 2011). drinking water for 2 months. The levan treated rats had increased levels
of HDL cholesterol and lower levels of LDL cholesterol, demonstrating
5.2.2. Anti-irritant, anti-oxidant and anti-inflammatory activities the beneficial effect.
“All chronic diseases have two common components - oxidative
stress and inflammation” (Kaimal, 2009). At its most basic level, inflam- 5.3. Prebiotics
mation is a good thing; it is the manifestation of the immune system ini-
tiating the healing process in response to an irritant or the toxicity of Prebiotics promote proliferation of beneficial bacteria in the colon
reactive oxidant species. Too much inflammation, however, can be pain- and inhibit harmful microorganisms. Several studies have looked at var-
ful and injurious. For formation of an inflammatory lesion, circulating ious aspects of the role levan or levan-type fructooligosaccharides have
cells must adhere to the lumen of the blood vessels. Sedgwick et al. on probiotic bacteria and complex intestinal microbiota (Adamberg
(1984) found that levan significantly reduced leucocyte adhesion and et al., 2014; Adamberg et al., 2015; Arrizon et al., 2014; Marx et al.,
the effect was seen whether they used vascular endothelial cells or 2000; Porras-Domínguez et al., 2014; Sonnenburg et al., 2010; Visnapuu
glass leading them to deduce that levan affects the leucocyte itself and et al., 2015 and references therein). For example, Adamberg et al.
not the blood vessel cell. While allowing some effect due to counter (2015) studied how levan reshapes microbiota in human fecal samples
irritant properties, they concluded that levan may act by directly reduc- using metagenomic and metabolomic methods. They showed levan
ing processes involved in stimulating leucocytes to adhere to blood enriched Bacteroides, Escherichia, Streptococcus and Faecalibacterium
vessel walls. and provided a metabolic scheme to describe conversion of levan in
Working with levan and sulfated levan produced by B. subtilis, it was cooperative metabolism of gut bacteria.
found that two fractions were particularly potent free radical scavengers Beneficial effects of levan on the intestinal microbial community in
when tested against 1,1-diphenyl-2-pycryl-hydrazyl (DPPH) radicals. the gut of farmed animals have also been studied. One series of experi-
The fraction of DPPH bleaching required to reach the half maximal scav- ments on swine looked at levan as a potential alternative to prophylactic
enging concentration (SC50) was as low as 1.5 μg per mL (Abdel-Fattah use of antibiotics. Growing pigs were given 700 kDa levan at a rate of 0,
et al., 2012). This compared favorably with the SC50 for ascorbic acid of 0.05, 0.1 and 0.2% of the diet over a 42 day period. Average daily gain,
8.0 μg per mL (Ghareeb et al., 2013). The antioxidant activity of levan the gain to feed ratio and the total tract digestibility optimally increased
from Acetobacter xylinum was 81% of ascorbic acid (Srikanth et al., 2015). with the 0.1% rate of levan addition (Li and Kim, 2013). A trial on older,
finishing pigs gave similar results. Levan had a positive impact on
5.2.3. Weight loss weight gain over the 42 day period, Lactobacillus counts were increased
Kang et al. (2004a) found adding levan to the diets of laboratory rats and E. coli levels decreased (Zhao et al., 2013a). The value of levan in the
suppressed obesity and hyperlipidemia. Even on a high fat diet, the daily diet of chickens was also investigated. Levan added to the feed im-
weight gain was reduced from 2.58 ± 0.29 g/day for the controls to proved the gain to feed ratio, increased the weight of the breast meat,
1.61 ± 0.21 g/day for rats on a diet supplemented with 10% levan. decreased the amount of ammonia excreted, increased the levels of
There was also a significant reduction in white fat development, adipo- beneficial bacteria (Lactobacillus and Bifidobacteria) in the cecum and
cyte hypertrophy, and the development of hyperinsulinemia and hyper- decreased the harmful bacteria (E. coli and Clostridium perfringens) in
lipidemia in a dose-dependent manner. Serum triglycerides, free fatty the cecum (Zhao et al., 2013b).
acids and serum cholesterol were also reduced in rats receiving levan.
It was suggested that upregulated uncoupling protein (UCP) mRNA ex- 5.4. Aquaculture
pression may have helped suppress the development of obesity through
increased energy expenditure. As for other farmed animals, there is interest in replacing prophylac-
Building on the known ability of some polysaccharides to have a tic antibiotics with safe alternatives to build up non-specific immunity
beneficial effect in weight control and the widespread use of ginseng of aquacultured fish. Early studies using intraperitoneally administered
as an herbal medicine, Oh et al. (2014) looked at levan, fermented levan had limited success (Wang and Wang, 1997), but in light of the
ginseng and a combination of the two. Levan powder (100 mg/kg/day), beneficial immunological effect by levan on non-aquatic species, the
dried fermented ginseng (150 mg/kg/day) or a combination of levan role of the administration route was considered. In a 75 day trial on
and ginseng were added to the high fat diet of mice for 11 weeks. Cyprinus carpio (common carp), levan was added to the feed. Survival
Mice on a high fat diet supplemented with the mix of levan and rates were determined 10 days after the fish were challenged with an
fermented ginseng had a final total body weight of 11.7 ± 0.8 g, signif- intraperitoneal dose of Aeromonas hydrophila. 100% of the fish on the
icantly less than the 16.1 ± 1.1 g for the control group. The ginseng plus 0.5% levan diet survived (Rairakhwada et al., 2007), suggesting the pos-
levan diet also resulted in a reduction of white adipose tissue weight, sibility that the benefits of levan were conferred by the interaction with
fasting blood glucose, insulin resistance index and leptin compared the microbial population of the gastrointestinal tract. The Central Insti-
with controls. tute of Fisheries Education, Mumbai, followed up with a series of studies
on a different species of carp, Labeo rohita, finding only a 60% survival
5.2.4. Lower cholesterol rate but still an improvement over controls (Gupta et al., 2008).
An extensive study on the effects of high molecular weight levan on Elevated temperatures are stressful for some fish species. Levan was
cholesterol levels showed benefits of the approach. Levan was added to added to the feed of Labeo rohita acclimated at 26 °C. Selected fish were
the cholesterol-free diet of rats at a rate of 1 or 5% (w/w) over the course transferred to thermostatic aquaria where the water temperature was
of one month. Serum cholesterol in the animals was significantly raised at a constant rate of 0.3 °C/min. When the fish exhibited a loss
decreased by both doses of levan. It was noted that fecal excretions of of equilibrium, the temperature was designated the critical thermal
sterols and lipids were higher in rats receiving levan than in the control maximum. The temperature increase was continued until fish death
animals, so one theory of the mechanism was that levan prevented was recorded as the lethal thermal maximum. Fish on the diet including
sterol absorption (Yamamoto et al., 1999). Ishihara (1996) found 1.25% levan had significantly higher critical thermal maxima and lethal
levan-based agents suppressed blood lipids and body fat even when thermal maxima. High levels of heat shock protein (HSP) were found
high-calorie foods were eaten. A later patent application claimed in fish raised on a diet with levan while no HSP was found in controls.
levan as part of a treatment for hypercholesterolemia, hyperlipidemia The authors suggested that the enhanced tolerance to temperature
838 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

stress was due to the stimulation of a nonspecific defense mechanism properties was useful. When levan was included in a wheat bread
(Gupta et al., 2010). recipe, as little as 1% levan produced bread that was 18–26% softer
Huang et al. (2014) studied the value of levan in orange-spotted than the control.
grouper (Epinephelus coioides H.). Levan was added in pellet feed for Even our favorite treats have been subjected to the addition of levan.
84 days. The grouper were then challenged with a common pathogen A patent has been obtained for a confectionery based on fructans, in-
for salt water fish, Vibrio harveyi. The group of fish receiving the 2.5% cluding levan, to make a healthy candy with heart and digestive
dose of levan had the largest increase in growth and a 90% survival benefits (Cordero, 2010).
rate (compared with 50% survival of the controls).

5.5. Films for packaging 5.7. Additional applications

Bio-based food packaging materials are needed for safer and healthier In addition to the abovementioned applications, many additional
packaging and to reduce accumulation in the environment (Vijayendra uses have been reported in the literature. Some of those are listed in
and Shamala, 2014). Without long, flexible moieties in levan, films the following table. (See Table 2.)
made from this polymer are too brittle for practical use but addition of
clay or other plasticizers overcomes this issue.
A meticulous analysis of the interaction between exfoliated mont-
morillonite clay and levan provided insight into the interacting forces Table 2
Additional applications.
that result in levan-based films that are clear, flexible and tough.
Sodium montmorillonite was exfoliated and blended at 1, 5 or 10 wt% Application Description References
with a solution of B. subtilis levan. Following ultrasonication, water Anti-cancer Mice tumor free after 5–8 month Sinai et al. (1976)
was partially evaporated to produce viscous suspensions. The thickened activity treatment.
liquids were coated onto Mylar sheets and dried. Adding only 1% mont- Molecular weight/branching critical. Calazans et al.
morillonite to the formulation increased thermal stability but did not (2000), Yoo et al.
(2004)
significantly enhance mechanical properties of the cast films. In fact,
Sulfated levan raised caspase activity, Abdel-Fattah et al.
the slight negative effect suggested that the clay disrupted the levan effective radical scavenger. (2012), Bayoumi
structure but the clay platelets were too far apart to be bridged by et al.
levan. Ten percent montmorillonite made films with a tensile modulus (2012), Bayoumi
424% greater than that of pure levan and with 485% greater toughness. et al. (2013)
Quarternization enhanced anti-cancer Esawy et al. (2013)
It was theorized that at 10% montmorillonite, the clay platelets were activity.
aligned and close enough to be bridged by uncoiled levan molecules. Anti-bacterial Decreased foodborne pathogens. OD600 Byun et al. (2014)
Working in parallel with the molecular network of levan, the montmo- activity reduced from 1.9 to as low as 1.0 at 48 h.
rillonite added significant mechanical reinforcement. FTIR spectra sup- Potential to prevent and treat Donald et al. (2014)
gram-positive infections.
plied evidence that the interaction was via hydrogen bonding between
Anti-viral EC50 0.35 (μg/mL) against Herpes simplex. Koichi and Masahiro
the hydroxyl groups on the levan and oxygen molecules on the surface activity (2002)
of the montmorillonite (Chen et al., 2014). Bacillus subtilis levan activity against Ahmed et al. (2010),
One property of levan-based films that makes them particularly avian influenza H5N1 virus and enteric Esawy et al.
attractive for food packaging is the fact that they form a good oxygen adenovirus type 40. (2011), Esawy et al.
(2012)
barrier. The oxygen permeability of levan films formulated with 10% Diabetes Decreased plasma glucose in diabetic Dahech et al. (2011)
montmorillonite and 5% polyethylene glycol was b 0.05 cm3/m2 − day rats; no effect on normal rats.
(Montana Polysaccharides, 2015). Enhanced Difructose anhydride IV produced from Saito and Tomita
calcium levan stimulated calcium absorption (2000)
absorption from
5.6. Levan in food
rat small intestine.
Pharmaceuticals Conjugate allergenic drugs with levan. Moreno (1979)
Levan phosphate as a fat substitute was suggested by Roberts and Timed release drugs. Sezer et al. (2015)
Garegg (1998). Another patent application covers the use of the Open matrix network for orally Gupta et al. (2015)
smooth, fat-like properties of fructans to improve the mouthfeel, disintegrating tablets.
Decrease Sulfated levan protective against lethal Higginbotham
taste and spreadability of dairy products (Booten et al., 1998). toxicity doses of the drugs 48/80, polymyxin B (1959)
Other advantages of levan as a fat replacement include the fact that and stilbamidine.
the high molecular weight levan is too large to be detected by taste LD50 for shrimp to antiviral Poli et al. (2009)
sensors and the volatility is too low to emit detectable odors. A re- sesquiterpene hydroquinone (avarol)
decreased over 50-fold.
cent patent application describes how to make yogurt containing
Decreased acute toxicity of copper for Kekez et al. (2015)
levan to be consumed as a functional food (Xiao et al., 2014). Daphnia from 0.14 to 0.44 mg per liter.
Exploiting the texture levan can bring to a food, Vincent et al. Increased survival of carp fry to Gupta et al. (2013)
(2005) suggested inclusion of levan in fermented cereal, milk, and insecticide, fipronil.
juices. Mays and Dally (1989) developed a colloid system stabilized Adhesive Tensile strength on bare aluminum 500 Montana
to 1500 psi. Polysaccharides
by levan in the form of an emulsion, aerosol or foam. (2015)
Effective delivery of essential dietary micronutrients was demon- Electrospinning Levan electrospun from distilled water. Manandhar et al.
strated by coating selenium, cobalt and iron nanoparticles with levan. (2009)
The approach rendered the elements safer for intestinal and microbial Inhibition of Among most effective polysaccharides Finkenstadt et al.
metal tested as corrosion inhibitors. (2011)
cells and enhanced the stability of the preparation. The 2 products in 1
corrosion
approach offered all the benefits of nontoxic versions of vital elements Renewable Chemical conversion of fructose to Román-Leshkov
while simultaneously transporting a functional prebiotic to the intestinal transportation 2,5-dimethylfuran (DMF), potential et al. (2006)
microbes (Bondarenko et al., 2016). fuel liquid
Studying methods to increase the shelf life of bread, Jakob et al. biofuel with greater energy content than
bioethanol.
(2012) found the ability of levan to form a microgel with hydrocolloid
E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 839

5.7.1. Future perspectives - demand, availability, inventions Abou-taleb, K.A., Abdel-Monem, M.O., Yassin, M.H., Draz, A.A., 2015. Production, purifica-
tion and characterization of levan polymer from Bacillus lentus V8 strain. Br.
Various economic, political and social drivers call for more environ- Microbiol. Res. J. 5, 22–32. http://dx.doi.org/10.9734/BMRJ/2015/12448.
mentally responsible products. In turn, this has prompted manufac- Adamberg, S., Tomson, K., Vija, H., Puurand, M., Kabanova, N., Visnapuu, T., Jõgi, E.,
turers to seek renewable raw materials with a good safety profile. An Alamäe, T., Adamberg, K., 2014. Degradation of fructans and production of propionic
acid by Bacteroides thetaiotaomicron are enhanced by the shortage of amino acids.
unusual combination of properties allows levan to stand apart from Front. Nutr. 1 (21), 1–10. http://dx.doi.org/10.3389/fnut.2014.00021.
competitors. The low intrinsic viscosity, high adhesive strength and Adamberg, K., Tomson, K., Talve, T., Pudova, K., Puurand, M., Visnapuu, T., Alamäe, T.,
the ability to gel alcohol is not found in any other water soluble, film Adamberg, S., 2015. Levan enhances associated growth of Bacteroides, Escherichia,
Streptococcus and Faecalibacterium in fecal microbiota. PLoS One 10 (12), e0144042.
forming, biodegradable polymer. The well-established safety and numer-
http://dx.doi.org/10.1371/journal.pone.0144042.
ous studies on health benefits are bonuses. Ahmed, E.F., Mansour, N.M., Helmy, W.A., El-Senousy, W.M., El-Safty, M.M., Esawy, M.A.,
The issue has been producing levan at an economically viable price. 2010. Production of an antiviral levan from novel honey Bacillus subtilis isolates. JGEB
Research to improve levan yields has been mainly limited to conven- 8, 71–79 (Available from URL: http://www.readbag.com/jgeb-eg-pdf-vol8-n1-9).
Alamäe, T., Visnapuu, T., Mardo, K., Mäe, A., Zamfir, A.D., 2012. Levansucrases of Pseudomonas
tional approaches like optimization of production conditions. On the bacteria: novel approaches for protein expression, assay of enzymes, fructooligosaccha-
other hand, systems based approaches that rely heavily on high- rides and heterooligofructans. In: Rauter, A.P., Lindhorst, T.K. (Eds.), Carbohydrate
throughput omics technologies have gained increasing attention in ChemistrySpecialist Periodical Reports Vol. 38. Royal Society of Chemistry, Cambridge,
pp. 176–191. http://dx.doi.org/10.1039/9781849734769-00176.
various areas of industrial biotechnology with many success stories Alegre, R.M., Wendt, R., Rigo, M., 2005. Levan production by isolated mutants of
(Dai and Nielsen, 2015). However, there are very few omics data avail- Zymomonas mobilis. Rev. Ciênc. Exatas Nat. 7, 103–112.
able from levan producers. This data is confined to the genome level and Anwar, M.A., Kralj, S., Villar Piqué, A., Leemhuis, H., van der Maarel, M.J.E.C., Dijkhuizen, L.,
2010. Inulin and levan synthesis by probiotic Lactobacillus gasseri strains: characteriza-
from a diverse family of bacterial levan producers. Whole genome tion of three novel fructansucrase enzymes and their fructan products. Microbiology
data of bacteria proven to produce levan has been reported for Bacillus 156, 1264–1274. http://dx.doi.org/10.1099/mic.0.036616-0.
subtilis (Kunst et al., 1997) Gluconacetobacter diazotrophicus PAl 5 Anwar, M.A., Leemhuis, H., Pijning, T., Kralj, S., Dijkstra, B.W., Dijkhuizen, L., 2012. The role of
conserved inulosucrase residues in the reaction and product specificity of Lactobacillus
(Bertalan et al., 2009), Halomonas smyrnensis AAD6T (Sogutcu et al.,
reuteri inulosucrase. FEBS J. 279, 3612–3621. http://dx.doi.org/10.1111/j.1742-4658.
2012) and Zymomonas mobilis strains ATCC 31821 (Seo et al., 2005) 2012.08721.x.
and NRRL B-14023 (Kouvelis et al., 2014). But, genome data need to Arrieta, J.G., Sotolongo, M., Menéndez, C., Alfonso, D., Trujillo, L.E., Soto, M., Ramírez, R.,
be enriched by integrating with transcriptome, proteome or metabolome Hernández, L., 2004. A type II protein secretory pathway required for levansucrase
secretion by Gluconacetobacter diazotrophicus. J. Bacteriol. 186, 5031–5039.
data in order to gain a holistic view of the microbial system rather than http://dx.doi.org/10.1128/JB.186.15.5031-5039.2004.
focusing on individual factors. Hence integrating systems biology to cur- Arrizon, J., Hernández-Moedano, A., Toksoy Öner, E., González-Avila, M., 2014. In vitro
rent levan research will provide a deeper understanding of levan biosyn- prebiotic activity of fructans with different fructosyl linkage for symbiotics elaboration.
Int. J. Probiotics Prebiotics 9, 69–76 (Available from URL: https://www.researchgate.
thesis and associated mechanisms. In the microbial system, the holistic net/publication/269223654_IN_VITRO_PREBIOTIC_ACTIVITY_OF_FRUCTANS_W_ITH_
approach to improvement of the amount of levan produced from a DIFFERENT_FRUCTOSYL_LINKAGE_FOR_SYMBIOTICS_ELABORATION).
given amount of substrate must incorporate consideration of the total Arvidson, S.A., Rinehart, B.T., Gadala-Maria, F., 2006. Concentration regimes of solutions of
levan polysaccharide from Bacillus sp. Carbohyd. Polym. 65, 144–149. http://dx.doi.
levansucrase activity present and the catalytic features of this enzyme.
org/10.1016/j.carbpol.2005.12.039.
Structural diversity and use of different in vivo and in vitro bioactiv- Ateş, Ö., Arga, K.Y., Toksoy Öner, E., 2013. The stimulatory effect of mannitol on levan bio-
ity assays in structure-function analysis studies prevents making gener- synthesis: lessons from metabolic systems analysis of Halomonas smyrnensis AAD6T.
alizations in relation to the chemical structure and conformation to the Biotechnol. Prog. 29, 1386–1397. http://dx.doi.org/10.1002/btpr.1823.
Axente, E., Sima, F., Sima, L.E., Erginer, M., Eroglu, M.S., Serban, N., Ristoscu, C., Petrescu,
observed biological activity of levan. Unfortunately, there are very few S.M., Toksoy Öner, E., Mihailescu, I.N., 2014. Combinatorial MAPLE gradient thin
in silico studies on levan. Hence there is a clear need for analyzing the film assemblies signaling to human osteoblasts. Biofabrication 6, 1–9. http://dx.doi.
3D structure of levan by using molecular dynamics simulations. Such org/10.1088/1758-5082/6/3/035010.
Bae, I.Y., Oh, I.-K., Lee, S., Yoo, S.-H., Lee, H.G., 2008. Rheological characterization of levan
studies could provide valuable information on the conformational polysaccharides from Microbacterium laevaniformans. Int. J. Biol. Macromol. 42,
preferences, intramolecular hydrogen-bonding patterns, torsional dy- 10–13. http://dx.doi.org/10.1016/j.ijbiomac.2007.08.006.
namics, and configurational entropies of levan-based polymers under Banguela, A., Hernández, L., 2006. Fructans: from natural sources to transgenic plants.
Biotecnol. Apl. 23, 202–210 (Available from URL: https://www.researchgate.net/
different conditions.
publication/228488099_Fructans_From_natural_sources_to_transgenic_plants).
This review covers applications supported by research. The upcom- Banguela, A., Arrieta, J.G., Rodríguez, R., Trujillo, L.E., Menéndez, C., Hernández, L., 2011. High
ing availability of reasonably priced levan in commercial quantities will levan accumulation in transgenic tobacco plants expressing the Gluconacetobacter
allow consideration of heretofore untested arenas such as 3D printing diazotrophicus levansucrase gene. J. Biotechnol. 154, 93–98. http://dx.doi.org/10.1016/
j.jbiotec.2011.04.007.
and ecofriendly pest control. Bayoumi, F.S., El Amir, A.M., El Deeb, S.O., Abd el-Zaher, H., Haiam, S.A., 2012. Immuno-
modulatory and chemo preventive activity of Bacillus subtilis sulphated levan. Life
6. Declaration of interest statement Sci. J. 9, 2846–2856 (Available from URL: http://www.lifesciencesite.com).
Bayoumi, F.S., El Amir, A.M., El Deeb, S.O., el-Zaher, H.A., Haiam, S.A., 2013. In vitro study of
a bacterial polysaccharide as anti-cancer. J. Appl. Sci. Res. 9, 2007–2018.
The authors of this article declare that they have no conflicts of Beine, R., Moraru, R., Nimtz, M., Na'amnieh, S., Pawlowski, A., Buchholz, K., Seibel, J., 2008.
interest of any kind. Synthesis of novel fructooligosaccharides by substrate and enzyme engineering.
J. Biotechnol. 138, 33–41. http://dx.doi.org/10.1016/j.jbiotec.2008.07.1998.
Bekers, M.J., Shvinka, J.E., Pankova, L.M., Laivenieks, M.G., Mezbarde, I.N., 1990. A
Acknowledgements simultaneous sucrose bioconversion into ethanol and levan by Zymomonas
mobilis. Appl. Biochem. Biotechnol. 24/25, 265–274. http://dx.doi.org/10.1007/
BF02920251.
The authors wish to thank Emma Carpenter and Alexis Musacchio Bekers, M., Linde, R., Danilevich, A., Kaminska, E., Upite, D., Vigants, A., Scherbaka, R., 1999.
for their help in the preparation of Fig. 2. The authors also thank the Sugar beet diffusion juice and syrup as media for ethanol and levan production
reviewers whose comments contributed to the improvement of the by Zymomonas mobilis. Food Biotechnol. 13, 107–119. http://dx.doi.org/10.1080/
08905439609549964.
manuscript quality. E. Toksoy Öner gratefully acknowledges the financial
Bekers, M., Linde, R., Upite, D., Kaminska, E., Viesturs, U., 2001. Sugar beet juice fermenta-
support provided by the Scientific and Technological Research Council of tion by Zymomonas mobilis attached to stainless steel wire spheres. Acta Biotechnol.
Turkey (TUBITAK projects 114M239 and 115O495) for levan research by 21, 89–96. http://dx.doi.org/10.1002/1521-3846(200102)21:1b89::AID-ABIO89N3.0.
IBSB. CO;2-8.
Bekers, M., Laukevics, J., Upite, D., Kaminska, E., Vigants, A., Viesturs, U., Pankova, L.,
Danilevics, A., 2002. Fructooligosaccharide and levan producing activity of Zymomonas
References mobilis extracellular levansucrase. Process Biochem. 38, 701–706. http://dx.doi.org/10.
1016/S0032-9592(02)00189-9.
Abdel-Fattah, A.M., Gamal-Eldeen, A.M., Helmy, W.A., Esawy, M.A., 2012. Antitumor and Bekers, M., Upite, D., Kaminska, E., Grube, M., Laukevics, J., Vina, I., Vigants, A., Zikmanis, P.,
antioxidant activities of levan and its derivative from the isolate Bacillus subtilis 2003. Fructan biosynthesis by intra- and extracellular Zymomonas mobilis levansucrase
NRC1aza. Carbohyd. Polym. 89, 314–322. http://dx.doi.org/10.1016/j.carbpol.2012. after simultaneous production of ethanol and levan. Acta Biotechnol. 23, 85–93.
02.041. http://dx.doi.org/10.1002/abio.200390012.
840 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

Belghith, K.S., Dahech, I., Hamden, K., Feki, A., Mejdoub, H., Belghith, H., 2012. Hypolipid- dos Santos, L.F., Bazani Cabral De Melo, F.C., Martins Paiva, W.J., Borsato, D., Corradi
emic effect of diet supplementation with bacterial levan in cholesterol-fed rats. Int. Custodio Da Silva, M.L., Pedrine Colabone Celligoi, M.A., 2013. Characterization and
J. Biol. Macromol. 50, 1070–1074. http://dx.doi.org/10.1016/j.ijbiomac.2012.02.024. optimization of levan production by Bacillus subtilis natto. Rom. Biotechnol. Lett. 18,
Bergeron, L.J., Burne, R.A., 2001. Roles of fructosyltransferase and levanase-sucrase of 8413–8422 (Available from URL: http://hdl.handle.net/11449/113391).
Actinomyces naeslundii in fructan and sucrose metabolism. Infect. Immun. 69, Ebskamp, M.J.M., van der Meer, I.M., Spronk, B.A., Weisbeek, P.J., Smeekens, S.C.M., 1994.
5395–5402. http://dx.doi.org/10.1128/IAI.69.9.5395-5402.2001. Accumulation of fructose polymers in transgenic tobacco. Nat. Biotechnol. 12,
Bergeron, L.J., Morou-Bermudez, E., Burne, R.A., 2000. Characterization of the 272–275. http://dx.doi.org/10.1038/nbt0394-272.
fructosyltransferase gene of Actinomyces naeslundii WVU45. J. Bacteriol. 182, Esawy, M.A., Ahmed, E.F., Helmy, W.A., Mansour, N.M., El-Senousy, W.M., El-Safty, M.M.,
3649–3654 (Available from URL: http://www.ncbi.nlm.nih.gov/pmc/articles/ 2011. Production of levansucrase from novel honey Bacillus subtilis isolates capable
PMC94534/). of producing antiviral levans. Carbohydr. Polym. 86, 823–830. http://dx.doi.org/10.
Bertalan, M., Albano, R., de Pádua, V., Rouws, L., Rojas, C., et al., 2009. Complete genome se- 1016/j.carbpol.2011.05.035.
quence of the sugarcane nitrogen-fixing endophyte Gluconacetobacter diazotrophicus Esawy, M.A., Ahmed, E.F., Helmy, W.A., Mansour, N.M., El-Senousy, W.M., El-Safty, M.M.,
Pal5. BMC Genom. 10, 450. http://dx.doi.org/10.1186/1471-2164-10-450. 2012. Antiviral levans from Bacillus spp. isolated from honey. In: Karunaratne,
Blake, J.D., Clarke, M.L., Jansson, P.E., McNeil, K.E., 1982. Fructan from Erwinia herbicola. D.N. (Ed.), The Complex World of Polysaccharides. InTech publisher, pp. 195–214
J. Bacteriol. 151, 1595–1597 (Available from URL: http://www.ncbi.nlm.nih.gov/ http://dx.doi.org/10.5772/52483.
pmc/articles/PMC220442/). Esawy, M.A., Abdel-Fattah, A.M., Ali, M.M., Helmy, W.A., Salama, B.M., Taie, H.A.A.,
Bondarenko, O.M., Ivask, A., Kahru, A., Vija, H., Titma, T., Visnapuu, M., Joost, U., Pudova, K., Hashem, A.M., Awad, G.E.A., 2013. Levansucrase optimization using solid state fer-
Adamberg, S., Visnapuu, T., Alamäe, T., 2016. Bacterial polysaccharide levan as stabi- mentation and levan biological activities studies. Carbohydr. Polym. 96, 332–341.
lizing, non-toxic and functional coating material for microelement-nanoparticles. http://dx.doi.org/10.1016/j.carbpol.2013.03.089.
Carbohyd. Polym. 136, 710–720. http://dx.doi.org/10.1016/j.carbpol.2015.09.093. Feng, J., Gu, Y., Han, L., Bi, K., Quan, Y., Yang, C., Zhang, W., Cao, M., Wang, S., Gao, W., Sun, Y.,
Booten, K., De Soete, J., Frippiat, A. Fructan- and/or polydextrose-containing dairy powders, Song, C., 2015a. Construction of a Bacillus amyloliquefaciens strain for high purity levan
process for preparing same and their use. Eur. patent appl. EP0821885A1, 1998. production. FEMS Microbiol. Lett. 362, 1–7. http://dx.doi.org/10.1093/femsle/fnv079.
Bostan, M.S., Mutlu, E.C., Kazak, H., Sinan Keskin, S., Toksoy Öner, E., Eroglu, M.S., 2014. Com- Feng, J., Gu, Y., Quan, Y., Zhang, W., Cao, M., Gao, W., Song, C., Yang, C., Wang, S., 2015b.
prehensive characterization of chitosan/PEO/levan ternary blend films. Carbohyd. Recruiting a new strategy to improve levan production in Bacillus amyloliquefaciens.
Polym. 102, 993–1000. http://dx.doi.org/10.1016/j.carbpol.2013.09.096. Sci. Rep. 5, 13814. http://dx.doi.org/10.1038/srep13814.
Byun, B.Y., Lee, S.-J., Mah, J.-H., 2014. Antipathogenic activity and preservative effect of Finkenstadt, V.L., Côté, G.L., Willett, J.L., 2011. Corrosion protection of low-carbon steel
levan (β-2,6-fructan), a multifunctional polysaccharide. Int. J. Food Sci. Technol. 49, using exopolysaccharide coatings from Leuconostoc mesenteroides. Biotechnol. Lett.
238–245. http://dx.doi.org/10.1111/ijfs.12304. 33, 1093–1100. http://dx.doi.org/10.1007/s10529-011-0539-2.
Calazans, G.M.T., Lopes, C.E., Lima, R.M.O.C., de França, F.P., 1997. Antitumour activities of Franken, J., Brandt, B.A., Tai, S.L., Bauer, F.F., 2013. Biosynthesis of levan, a bacterial extra-
levans produced by Zymomonas mobilis strains. Biotechnol. Lett. 19, 19–21. http://dx. cellular polysaccharide, in the yeast Saccharomyces cerevisiae. PLoS One 8, e77499.
doi.org/10.1023/A:1018350617120. http://dx.doi.org/10.1371/journal.pone.0077499.
Calazans, G.M.T., Lima, R.C., de França, F.P., Lopes, C.E., 2000. Molecular weight and Fuchs, A., 1956. Synthesis of levan by pseudomonads. Nature 178, 921. http://dx.doi.org/
antitumour activity of Zymomonas mobilis levans. Int. J. Biol. Macromol. 27, 245–247. 10.1038/178921a0.
http://dx.doi.org/10.1016/S0141-8130(00)00125-2. Fuchs, A., de Bruijn, J.M., Niedeveld, C.J., 1985. Bacteria and yeasts as possible candidates
Caputi, L., Nepogodiev, S.A., Malnoy, M., Rejzek, M., Field, R.A., Benini, S., 2013. Biomolecular for the production of inulinases and levanases. Anton. Leeuw. 51, 333–343.
characterization of the levansucrase of Erwinia amylovora, a promising biocatalyst http://dx.doi.org/10.1007/BF02439942.
for the synthesis of fructooligosaccharides. J. Agric. Food Chem. 61, 12265–12273. Geier, G., Geider, K., 1993. Characterization and influence on virulence of the levansucrase
http://dx.doi.org/10.1021/jf4023178. gene from the fireblight pathogen Erwinia amylovora. Physiol. Mol. Plant Pathol. 42,
Carson, J., 2015. Available from URL: http://carsonproductdevelopment.com/. 387–404. http://dx.doi.org/10.1006/pmpp.1993.1029.
Castillo, E., López-Munguía, A., 2004. Synthesis of levan in water-miscible organic solvents. Ghareeb, M.A., Shoeb, H.A., Madkour, H.M.F., Refahy, L.A., Mohamed, M.A., Saad, A.M.,
J. Biotechnol. 114, 209–217. http://dx.doi.org/10.1016/j.jbiotec.2004.06.003. 2013. Radical scavenging potential and cytotoxic activity of phenolic compounds
Chambert, R., Gonzy-Treboul, G., 1976. Levansucrase of Bacillus subtilis: kinetic and from Tectona grandis Linn. Global J. Pharmacol. 7, 486–497. http://dx.doi.org/10.
thermodynamic aspects of transfructosylation processes. Eur. J. Biochem. 62, 55–64. 5829/idosi.gjp.2013.7.4.8263.
http://dx.doi.org/10.1111/j.1432-1033.1976.tb10097.x. Goldman, D., Lavid, N., Schwartz, A., Shoham, G., Danino, D., Shoham, Y., 2008. Two active
Chambert, R., Petit-Glatron, M.F., 1989. Study of the effect of organic solvents on the synthesis forms of Zymomonas mobilis levansucrase. An ordered microfibril structure of the en-
of levan and the hydrolysis of sucrose by Bacillus subtilis levansucrase. Carbohydr. Res. zyme promotes levan polymerization. J. Biol. Chem. 283, 32209–32217. http://dx.doi.
191, 117–123. http://dx.doi.org/10.1016/0008-6215(89)85051-7. org/10.1074/jbc.M805985200.
Chambert, R., Tréboul, G., Dedonder, R., 1974. Kinetic studies of levansucrase of Bacillus Greig-Smith, R.G., 1901. The gum fermentation of sugar cane juice. Proc. Linnean Soc.
subtilis. Eur. J. Biochem. 41, 285–300. http://dx.doi.org/10.1111/j.1432-1033.1974. NSW 26, 589–625 (Available from URL: http://biostor.org/reference/54845).
tb03269.x. Gunn, E., Gabbianelli, A., Crooks, R., Shanmuganandamurthy, K. Derivatized polysaccharide
Chen, X., Gao, H., Ploehn, H.J., 2014. Montmorillonite-levan nanocomposites with improved polymer. US patent 7,528,100. 2009.
thermal and mechanical properties. Carbohydr. Polym. 101, 565–573. http://dx.doi.org/ Gupta, S.K., Pal, A.K., Sahu, N.P., Dalvi, R., Kumar, V., Mukherjee, S.C., 2008. Microbial levan
10.1016/j.carbpol.2013.09.073. in the diet of Labeo rohita Hamilton juveniles: effect on non-specific immunity and
Choi, H.-J., Kim, C.S., Kim, P., Jung, H.-C., Oh, D.-K., 2004. Lactosucrose bioconversion from histopathological changes after challenge with Aeromonas hydrophila. J. Fish Dis. 31,
lactose and sucrose by whole cells of Paenibacillus polymyxa harboring levansucrase 649–657. http://dx.doi.org/10.1111/j.1365-2761.2008.00939.x.
activity. Biotechnol. Prog. 20, 1876–1879. http://dx.doi.org/10.1021/bp049770v. Gupta, S.K., Pal, A.K., Sahu, N.P., Dalvi, R.S., Akhtar, M.S., Jha, A.K., Baruah, K., 2010. Dietary mi-
Cordero, M.L.B. Confectionery composition, Great Britain patent GB2430344B, 2010. crobial levan enhances tolerance of Labeo rohita (Hamilton) juveniles to thermal stress.
Costa, R.R., NetoI, A.I., Calgeris, I., Correia, C.R., Pinho, A.C.M., Fonseca, J., Toksoy Öner, E., Aquaculture 306, 398–402. http://dx.doi.org/10.1016/j.aquaculture.2010.06.008.
Mano, J.F., 2013. Adhesive nanostructured multilayer films using a bacterial Gupta, S.K., Pal, A.K., Sahu, N.P., Jha, A.K., Akhtar, M.S., Mandal, S.C., Das, P., Prusty, A.K.,
exopolysaccharide for biomedical applications. J. Mater. Chem. B 1, 2367–2374. 2013. Supplementation of microbial levan in the diet of Cyprinus carpio fry (Linnaeus,
http://dx.doi.org/10.1039/c3tb20137f. 1758) exposed to sublethal toxicity of fipronil: effect on growth and metabolic
Crutz, A.M., Steinmetz, M., Aymerich, S., Richter, R., Le Coq, D., 1990. Induction of responses. Fish Physiol. Biochem. 39, 1513–1524. http://dx.doi.org/10.1007/s10695-
levansucrase in Bacillus subtilis: an antitermination mechanism negatively controlled 013-9805-7.
by the phosphotransferase system. J. Bacteriol. 172, 1043–1050 (Available from URL: Gupta, S., Ahuja, V., Gunjikar, T., Wannerberger, K. Fast dissolving pharmaceutical compo-
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC208535/). sition. US patent appl. 20150150798A1. 2015.
Dahech, I., Belghith, K.S., Hamden, K., Feki, A., Belghith, H., Mejdoub, H., 2011. Oral Haber, B., Ter Meer, H-U., Hausmanns, S. Cholesterol-reducing agent made of dietary fibre
administration of levan polysaccharide reduces the alloxan-induced oxidative stress and cholesterol-reducing substances. US patent appl. 20060062862A1. 2006.
in rats. Int. J. Biol. Macromol. 49, 942–947. http://dx.doi.org/10.1016/j.ijbiomac. Han, Y.W., 1990. Microbial levan. Adv. Appl. Microbiol. 35, 171–194. http://dx.doi.org/10.
2011.08.011. 1016/S0065-2164(08)70244-2.
Dai, Z., Nielsen, J., 2015. Advancing metabolic engineering through systems biology of Han, Y.W., Clarke, M.A., 1990. Production and characterization of microbial levan. J. Agr.
industrial microorganisms. Curr. Opin. Biotechnol. 36, 8–15. http://dx.doi.org/10. Food Chem. 38, 393–396. http://dx.doi.org/10.1021/jf00092a011.
1016/j.copbio.2015.08.006. Han, Y.W., Watson, M.A., 1992. Production of microbial levan from sucrose, sugar cane
Dawes, E.A., Ribbons, D.W., 1966. Sucrose utilization by Zymomonas mobilis: formation of juice and beet molasses. J. Ind. Microbiol. Biot. 9, 257–260. http://dx.doi.org/10.
a levan. Biochem. J. 98, 804–812. http://dx.doi.org/10.1042/bj0980804. 1007/BF01569633.
de Oliveira, M.R., da Silva, R.S.S.F., Buzato, J.B., Celligoi, M.A.P.C., 2007. Study of levan pro- Han, W.C., Byun, S.H., Kim, M.H., Sohn, E.H., Lim, J.D., Um, B.H., Kim, C.H., Kang, S.A., Jang,
duction by Zymomonas mobilis using regional low-cost carbohydrate sources. K.H., 2009. Production of lactosucrose from sucrose and lactose by a levansucrase
Biochem. Eng. J. 37, 177–183. http://dx.doi.org/10.1016/j.bej.2007.04.009. from Zymomonas mobilis. J. Microbiol. Biotechnol. 19, 1153–1160 (Available from
Diken, E., Ozer, T., Arikan, M., Emrence, Z., Toksoy Öner, E., Ustek, D., Arga, K.Y., 2015. URL: http://www.ncbi.nlm.nih.gov/pubmed/19884774).
Genomic analysis reveals the biotechnological and industrial potential of levan pro- Hernández, L., Arrieta, J., Menéndez, C., Vazquez, R., Coego, A., Suárez, V., Selman, G., Petit-
ducing halophilic extremophile, Halomonas smyrnensis AAD6T. SpringerPlus 4, 393. Glatron, M.F., Chambert, R., 1995. Isolation and enzymic properties of levansucrase
http://dx.doi.org/10.1186/s40064-015-1184-3. secreted by Acetobacter diazotrophicus SRT4, a bacterium associated with sugar
Dogsa, I., Brloznik, M., Stopar, D., Mandic-Mulec, I., 2013. Exopolymer diversity and the cane. Biochem. J. 309, 113–118. http://dx.doi.org/10.1042/bj3090113.
role of levan in Bacillus subtilis biofilms. PLoS One 8 (4), e62044. http://dx.doi.org/ Hernández, L., Arrieta, J., Betancourt, L., Falcón, V., Madrazo, J., Coego, A., Menéndez, C.,
10.1371/journal.pone.0062044. 1999a. Levansucrase from Acetobacter diazotrophicus SRT4 is secreted via periplasm
Donald, R.G.K., Kodali, S., Vino-Gradov, E. Saccharides and uses thereof. World patent by a signal-peptide-dependent pathway. Curr. Microbiol. 39, 146–152. http://dx.doi.
appl. WO2014013375A1. 2014. org/10.1007/s002849900436.
E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 841

Hernández, L., Ramírez, R., Hormaza, J.V., Madrazo, J., Arrieta, J., 1999b. Increased Biotechnol. 13, 230–235 (Available from URL: http://xn-hy1b43dy9r4rl8nb.com/
levansucrase production by a genetically modified Acetobacter diazotrophicus strain paper/view.html?no=2052199).
in shaking batch cultures. Lett. Appl. Microbiol. 28, 41–44. http://dx.doi.org/10. Kim, C.H., Kim, G.H., Ko, G.I., Lee, J.S., Lee, S.G., Park, G.S. Cosmetic composition containing
1046/j.1365-2672.1999.00470.x. levan extract having elasticity enhancing effect by cell proliferation and skin
Hernández, L., Sotolongo, M., Rosabal, Y., Menéndez, C., Ramírez, R., Caballero-Mellado, J., moisturization. Korean patent. KR100440357B1. 2004.
Arrieta, J., 2000. Structural levansucrase gene (lsdA) constitutes a functional locus Kim, K.H., Chung, C.B., Kim, Y.H., Kim, K.S., Han, C.S., Kim, C.H., 2005a. Cosmeceutical
conserved in the species Gluconacetobacter diazotrophicus. Arch. Microbiol. 174, properties of levan produced by Zymomonas mobilis. J. Cosmet. Sci. 56, 395–406.
120–124. http://dx.doi.org/10.1007/s002030000173. http://dx.doi.org/10.1111/j.1467-2494.2006.00314_2.x.
Hestrin, S., Avineri-Shapiro, S., Aschner, M., 1943. The enzymatic production of levan. Kim, C.H., Rhee, S.K., Kim, K.H., Kim, K.S., Han, C.S., Choi, B.R. Beta-fructosyl-L-ascorbic acid
Biochem. J. 37, 450–456 (Available from URL: http://www.ncbi.nlm.nih.gov/pmc/ and the preparation method thereof. World patent appl. WO2005026183A1, 2005b.
articles/PMC1257936/). Kim, Y.H., Kim, J.G., Jang, J.W., Lee, S.Y., Heo, W.B., Kim, K.H., 2012. Study on the synthesis
Hettwer, U., Gross, M., Rudolph, K., 1995. Purification and properties of an extracellular of modified levan and its properties on the hair. International Federation of Societies
levansucrase from Pseudomonas syringae pv. phaseolicola. J. Bacteriol. 177, 2834–2839 of Cosmetic Chemists, Johannesburg, South Africa, Poster Session 5, Number 302
(Available from URL: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC176956/). (Available from URL: http://www.ifscc2012.co.za/coschem/index.cfm/scientific-
Higginbotham, R.D., 1959. Participation of micellophagosis in the resistance of tissue to programme/poster-session-5/).
injurious agents. Int. Arch. Allergy Immunol. 15, 195–222. http://dx.doi.org/10. Koczan, J.M., McGrath, M.J., Zhao, Y., Sundin, G.W., 2009. Contribution of Erwinia
1159/000229046. amylovora exopolysaccharides amylovoran and levan to biofilm formation: implica-
Homann, A., Biedendieck, R., Götze, S., Jahn, D., Seibel, J., 2007. Insights into polymer versus tions in pathogenicity. Phytopathology 99, 1237–1244. http://dx.doi.org/10.1094/
oligosaccharide synthesis: mutagenesis and mechanistic studies of a novel levansucrase PHYTO-99-11-1237.
from Bacillus megaterium. Biochem. J. 407, 189–198. http://dx.doi.org/10.1042/BJ2007 Koichi, O., Masahiro, M. Antivirus agent. Japanese patent appl. JP2002293803A. 2002.
0600. Kojima, I., Saito, T., Iizuka, M., Minamiura, N., Ono, S., 1993. Characterization of levan pro-
Huang, M.-Y., Chang, C.-I., Chang, C.-C., Tseng, L.-W., Pan, C.-L., 2014. Effects of dietary duced by Serratia sp. J. Ferment. Bioeng. 75, 9–12. http://dx.doi.org/10.1016/0922-
levan on growth performance, nonspecific immunity, pathogen resistance and body 338X(93)90169-9.
composition of orange-spotted grouper (Epinephelus coioides H.). Aquac. Res. 11, Kornmann, H., Duboc, P., Marison, I., von Stockar, U., 2003. Influence of nutritional factors on
2752–2767. http://dx.doi.org/10.1111/are.12430. the nature, yield, and composition of exopolysaccharides produced by Gluconacetobacter
Idogawa, N., Amamoto, R., Murata, K., Kawai, S., 2014. Phosphate enhances levan produc- xylinus I-2281. Appl. Environ. Microbiol. 69, 6091–6098. http://dx.doi.org/10.1128/AEM.
tion in the endophytic bacterium Gluconacetobacter diazotrophicus Pal5. Bioengineered 69.10.6091-6098.2003.
5, 173–179. http://dx.doi.org/10.4161/bioe.28792. Kouvelis, V.N., Teshima, H., Bruce, D., Detter, C., Tapia, R., et al., 2014. Finished genome of
Inthanavong, L., Tian, F., Khodadadi, M., Karboune, S., 2013. Properties of Geobacillus Zymomonas mobilis subsp. mobilis strain CP4, an applied ethanol producer. Genome
stearothermophilus levansucrase as potential biocatalyst for the synthesis of levan Announc. 2 (1), e00845-13. http://dx.doi.org/10.1128/genomeA.00845-13.
and fructooligosaccharides. Biotechnol. Prog. 29, 1405–1415. http://dx.doi.org/10. Küçükaşık, F., Kazak, H., Güney, D., Finore, I., Poli, A., Yenigün, O., Nicolaus, B., Toksoy Öner,
1002/btpr.1788. E., 2011. Molasses as fermentation substrate for levan production by Halomonas sp.
Ishihara, K. Antihyperlipidemic and antiobesity agent comprising levan or hydrolysis Appl. Microbiol. Biotechnol. 89, 1729–1740. http://dx.doi.org/10.1007/s00253-010-
products thereof obtained from Streptococcus salivarius. US patent 5,527,784. 1996. 3055-8.
Jakob, F., Meißner, D., Vogel, R.F., 2012. Comparison of novel GH 68 levansucrases of Kunst, F., Ogasawara, N., Moszer, I., Albertini, A.M., Alloni, G., et al., 1997. The complete
levan-overproducing Gluconobacter species. Acetic Acid Bacteria (AAB) 1:e2, 6–14. genome sequence of the gram-positive bacterium Bacillus subtilis. Nature 390,
http://dx.doi.org/10.4081/aab.2012.e2. 249–256. http://dx.doi.org/10.1038/36786.
Jakob, F., Pfaff, A., Novoa-Carballal, R., Rübsam, H., Becker, T., Vogel, R.F., 2013. Structural Lammens, W., Le Roy, K., Yuan, S., Vergauwen, R., Rabijns, A., Van Laere, A., Streikov, S.A.,
analysis of fructans produced by acetic acid bacteria reveals a relation to hydrocolloid Van den Ende, W., 2012. Crystal structure of 6-SST/6-SFT from Pachysandra terminalis,
function. Carbohydr. Polym. 92, 1234–1242. http://dx.doi.org/10.1016/j.carbpol.2012. a plant fructan biosynthesizing enzyme in complex with its acceptor substrate 6-
10.054. kestose. Plant J. 70 (2), 205–219. http://dx.doi.org/10.1111/j.1365-313X.2011.
Jang, K.-H., Song, K.-B., Kim, C.H., Chung, B.H., Kang, S.A., Chun, U.-H., Choue, R.W., Rhee, 04858.x.
S.-K., 2001. Comparison of characteristics of levan produced by different preparations Lasseur, B., Lothier, J., Wiemken, A., Van Laere, A., Morvan-Bertrand, A., Van den Ende, W.,
of levansucrase from Zymomonas mobilis. Biotechnol. Lett. 23, 339–344. http://dx.doi. Prud'homme, M.-P., 2011. Towards a better understanding of the generation of
org/10.1023/A:1005641220946. fructan structure diversity in plants: molecular and functional characterization of a
Jathore, N.R., Bule, M.V., Tilay, A.V., Annapure, U.S., 2012. Microbial levan from Pseudomonas sucrose:fructan 6-fructosyltransferase (6-SFT) cDNA from perennial ryegrass
fluorescens: characterization and medium optimization for enhanced production. Food (Lolium perenne). J. Exp. Bot. 62, 1871–1885. http://dx.doi.org/10.1093/jxb/erq388.
Sci. Biotechnol. 21, 1045–1053. http://dx.doi.org/10.1007/s10068-012-0136-8. Leite, A.J., Silva, J.M., Ates, O., Eroğlu, M.S., Toksoy Öner, E., Mano, J.F., 2014. Bioadhesive
Jennings, L.K., Storek, K.M., Ledvina, H.E., Coulon, C., Marmont, L.S., Sadovskaya, I., Secor, and bioactive methacrylated nanocomposite levan-based beads for bone tissue applica-
P.R., Tseng, B.S., Scian, M., Filloux, A., Wozniak, D.J., Howell, P.L., Parsek, M.R., 2015. tions. Presentation at 2nd International Conference on Biological and Biomimetic Adhe-
Pel is a cationic exopolysaccharide that cross-links extracellular DNA in the Pseudomonas sives, PP01-03, p. 85 (Available from URL: www.icbba2014.org/pdf/ICBBA2014_
aeruginosa biofilm matrix. Proc. Natl. Acad. Sci. U. S. A. 112 (36), 11353–11358. http://dx. CONGRESS_BOOK.pdf).
doi.org/10.1073/pnas.1503058112. Li, J., Kim, I.H., 2013. Effects of levan-type fructan supplementation on growth perfor-
Kaimal, T.N.B., 2009. Perspective of the Week. Vitasearch (July 27, 2009). mance, digestibility, blood profile, fecal microbiota, and immune responses after lipo-
Kang, S.A., Hong, K., Jang, K.-H., Kim, S., Lee, K.H., Chang, B.-I., Kim, C.H., Choue, R., 2004a. polysaccharide challenge in growing pigs. J. Anim. Sci. 91, 5336–5343. http://dx.doi.
Anti-obesity and hypolipidemic effects of dietary levan in high fat diet-induced obese org/10.2527/jas2013-6665.
rats. J. Microbiol. Biotechnol. 14, 796–804 (Available from URL: http://agris.fao.org/ Li, H., Ullrich, M.S., 2001. Characterization and mutational analysis of three allelic lsc
agris-search/search.do?recordID=KR2005012093). genes encoding levansucrase in Pseudomonas syringae. J. Bacteriol. 183, 3282–3292.
Kang, S.A., Lee, J.C., Park, Y.M., Lee, C., Kim, S.-H., Chang, B.-I., Kim, C.-H., Seo, J.-W., Rhee, S.- http://dx.doi.org/10.1128/JB.183.11.3282-3292.2001.
K., Jung, S.J.E., Kim, S.-M., Park, S.K., Jang, K.-H., 2004b. Secretory production of Rahnella Liang, T.-W., Wang, S.-L., 2015. Recent advances in exopolysaccharides from Paenibacillus
aquatilis ATCC 33071 levansucrase expressed in Escherichia coli. J. Microbiol. spp.: production, isolation, structure, and bioactivities. Mar. Drugs 13, 1847–1863.
Biotechnol. 14 (6), 1232–1238 (Available from URL: http://www.jmb.or.kr/journal/ http://dx.doi.org/10.3390/md13041847.
list.html?mod=vol&tops=&year=2004). Liu, J., Luo, J., Ye, H., Sun, Y., Lu, Z., Zeng, X., 2009. Production, characterization and antiox-
Kang, S.A., Jang, K.-H., Seo, J.-W., Kim, K.H., Kim, Y.H., Rairakhwada, D., Seo, M.Y., Lee, J.O., Ha, idant activities in vitro of exopolysaccharides from endophytic bacterium Paenibacillus
S.D., Kim, C.-H., Rhee, S.-K., 2009. Levan: applications and perspectives. In: Rehm, B.H.A. polymyxa EJS-3. Carbohydr. Polym. 78, 275–281. http://dx.doi.org/10.1016/j.carbpol.
(Ed.), Microbial Production of Biopolymers and Polymer Precursors: Applications and 2009.03.046.
Perspectives. Caister Academic Press. ISBN: 978-1-904455-36-3, pp. 145–161. Liu, J., Luo, J., Ye, H., Sun, Y., Lu, Z., Zeng, X., 2010. Medium optimization and structural
Kang, H.K., Yun, S.I., Lim, T.Y., Xia, Y.-M., Kim, D., 2011. Cloning of levansucrase from characterization of exopolysaccharides from endophytic bacterium Paenibacillus
Leuconostoc mesenteroides and its expression in Pichia pastoris. Food Sci. Biotechnol. polymyxa EJS-3. Carbohydr. Polym. 79, 206–213. http://dx.doi.org/10.1016/j.carbpol.
20, 277–281. http://dx.doi.org/10.1007/s10068-011-0039-0. 2009.07.055.
Kazak Sarilmiser, H., Ates, O., Ozdemir, G., Arga, K.Y., Toksoy Öner, E., 2015. Effective stim- Lorenzetti, M.F.S., Moro, M.R., García-Cruz, C.H., 2015. Alginate/PVA beads for levan pro-
ulating factors for microbial levan production by Halomonas smyrnensis AAD6T. duction by Zymomonas mobilis. J. Food Process Eng. 38, 31–36. http://dx.doi.org/10.
J. Biosci. Bioeng. 119, 455–463. http://dx.doi.org/10.1016/j.jbiosc.2014.09.019. 1111/jfpe.12123.
Keith, J., Wiley, B., Ball, D., Arcidiacono, S., Zorfass, D., Mayer, J., Kaplan, D., 1991. Continuous Manandhar, S., Vidhate, S., D'Souza, N., 2009. Water soluble levan polysaccharide biopolymer
culture system for production of biopolymer levan using Erwinia herbicola. Biotechnol. electrospun fibers. Carbohydr. Polym. 78, 794–798. http://dx.doi.org/10.1016/j.carbpol.
Bioeng. 38, 557–560. http://dx.doi.org/10.1002/bit.260380515. 2009.06.023.
Kekez, B.D., Gojgic-Cvijovic, G.D., Jakovljevic, D.M., Stefanovic Kojic, J.R., Markovic, M.D., Mardo, K., Visnapuu, T., Vija, H., Elmi, T., Alamäe, T., 2014. Mutational analysis of con-
Beskoski, V.P., Vrvic, M.M., 2015. High levan production by Bacillus licheniformis served regions harboring catalytic triad residues of the levansucrase protein encoded
NS032 using ammonium chloride as the sole nitrogen source. Appl. Biochem. by the lsc-3 gene (lsc3) of Pseudomonas syringae pv. tomato DC3000. Biotechnol. Appl.
Biotechnol. 175, 3068–3083. http://dx.doi.org/10.1007/s12010-015-1475-8. Biochem. 61, 11–22. http://dx.doi.org/10.1002/bab.1129.
Kikuchi, H., Sakurai, H., Nagura, T., Aritsuka, T., Tomita, F., Yokota, A., 2010. One-pot conver- Martinez-Fleites, C., Ortiz-Lombardia, M., Pons, T., Tarbouriech, N., Taylor, E.J., Arrieta, J.G.,
sion of levan prepared from Serratia levanicum NN to difructose anhydride IV by Hernández, L., Davies, G.J., 2005. Crystal structure of levansucrase from the gram-
Arthrobacter nicotinovorans levan fructotransferase. J. Biosci. Bioeng. 109, 240–243. negative bacterium Gluconacetobacter diazotrophicus. Biochem. J. 390, 19–27.
http://dx.doi.org/10.1016/j.jbiosc.2009.09.041. http://dx.doi.org/10.1042/BJ20050324.
Kim, H.J., Park, H.-E., Kim, M.-J., Lee, H.G., Yang, J.-Y., Cha, J.H., 2003. Enzymatic characteriza- Martin-Verstraete, I., Deutscher, J., Galinier, A., 1999. Phosphorylation of HPr and Crh
tion of a recombinant levansucrase from Rahnella aquatilis ATCC 15552. J. Microbiol. by HprK, early steps in the catabolite repression signaling pathway for the Bacillus
842 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

subtilis levanase operon. J. Bacteriol. 181, 2966–2969 (Available from URL: http:// Pons, T., Naumoff, D.G., Martínez-Fleites, C., Hernández, L., 2004. Three acidic residues are
www.ncbi.nlm.nih.gov/pubmed/10217795). at the active site of a β-propeller architecture in glycoside hydrolase families 32, 43,
Marx, S.P., Winkler, S., Hartmeier, W., 2000. Metabolization of β-(2,6)-linked fructose- 62, and 68. Proteins 54, 424–432. http://dx.doi.org/10.1002/prot.10604.
oligosaccharides by different bifidobacteria. FEMS Microbiol. Lett. 182, 163–169. Porras-Domínguez, J.R., Ávila-Fernández, Á., Rodríguez-Alegría, M.E., Miranda-Molina, A.,
http://dx.doi.org/10.1016/S0378-1097(99)00584-4. Escalante, A., González-Cervantes, R., Olvera, C., López-Munguía, A., 2014. Levan-type
Masayo, F., Takayuki, T. Beautifully whitening agent, Japanese patent appl. JP2006052146A. FOS production using a Bacillus licheniformis endolevanase. Process Biochem. 49,
2006. 783–790. http://dx.doi.org/10.1016/j.procbio.2014.02.005.
Matsuhira, H., Tamura, K., Tamagake, H., Sato, Y., Anzai, H., Yoshida, M., 2014. High produc- Porras-Domínguez, J.R., Ávila-Fernández, Á., Miranda-Molina, A., Rodríguez-Alegría, M.E.,
tion of plant type levan in sugar beet transformed with timothy (Phleum pratense) 6- López-Munguía, A., 2015. Bacillus subtilis 168 levansucrase (SacB) activity affects
SFT genes. J. Biotechnol. 192, 215–222. http://dx.doi.org/10.1016/j.jbiotec.2014.09.025. average levan molecular weight. Carbohydr. Polym. 132, 338–344. http://dx.doi.
Mays, T.D., Dally, E.L. Microbial production of polyfructose. US patent 4,879,228. 1989. org/10.1016/j.carbpol.2015.06.056.
Mehmood, A., Abdallah, K., Khandekar, S., Zhurina, D., Srivastava, A., Al-Karablieh, N., Raga-Carbajal, E., Carrillo-Nava, E., Costas, M., Porras-Domínguez, J., López-Munguía, A.,
Alfaro-Espinoza, G., Pletzer, D., Ullrich, M.S., 2015. Expression of extra-cellular Olvera, C., 2016. Size product modulation by enzyme concentration reveals two
levansucrase in Pseudomonas syringae is controlled by the in planta fitness-promoting distinct levan elongation mechanisms in Bacillus subtilis levansucrase. Glycobiology
metabolic repressor HexR. BMC Microbiol. 15, 1–11. http://dx.doi.org/10.1186/ 26 (4), 377–385. http://dx.doi.org/10.1093/glycob/cwv112.
s12866-015-0349-0. Rahn, A.G., 2012. Proteolea®, a new source of strength to rebalance the signs of time
Melo, I.R., Pimentel, M.F., Lopes, C.E., Calazans, G.M.T., 2007. Application of fractional Available from URL: http://www.rahn-group.com/en/cosmetics/news/.
factorial design to levan production by Zymomonas mobilis. Braz. J. Microbiol. 38, Rahn, A.G., 2013. Slimexir®, cosmetics for the silhouette, an overview of the slimming
45–51. http://dx.doi.org/10.1590/S1517-83822007000100010. market. Presentation Available from URL: http://www.rahn-group.com/en/
Mena-Arizmendi, A., Alderete, J., Águila, S., Marty, A., Miranda-Molina, A., López-Munguía, cosmetics/news/.
A., Castillo, E., 2011. Enzymatic fructosylation of aromatic and aliphatic alcohols by Rairakhwada, D., Pal, A.K., Bhathena, Z.P., Sahu, N.P., Jha, A., Mukherjee, S.C., 2007. Dietary
Bacillus subtilis levansucrase: reactivity of acceptors. J. Mol. Catal. B Enzym. 70, microbial levan enhances cellular non-specific immunity and survival of common
41–48. http://dx.doi.org/10.1016/j.molcatb.2011.02.002. carp (Cyprinus carpio) juveniles. Fish Shellfish Immunol. 22, 477–486. http://dx.doi.
Méndez-Lorenzo, L., Porras-Domínguez, J.R., Raga-Carbajal, E., Olvera, C., Rodríguez- org/10.1016/j.fsi.2006.06.005.
Alegría, M.E., Carrillo-Nava, E., Costas, M., López-Mungia, A., 2015. Intrinsic levanase Rathsam, C., Jacques, N.A., 1998. Role of C-terminal domains in surface attachment of
activity of Bacillus subtilis 168 levansucrase (SacB). PLoS One 10 (11), e0143394. the fructosyltransferase of Streptococcus salivarius ATCC 25975. J. Bacteriol. 180,
http://dx.doi.org/10.1371/journal.pone.0143394. 6400–6403 (Available from URL: http://www.ncbi.nlm.nih.gov/pmc/articles/
Menéndez, C., Banguela, A., Caballero-Mellado, J., Hernández, L., 2009. Transcriptional PMC107731/).
regulation and signal-peptide-dependent secretion of exolevanase (LsdB) in the Rathsam, C., Giffard, P.M., Jacques, N.A., 1993. The cell-bound fructosyltransferase of
endophyte Gluconacetobacter diazotrophicus. Appl. Environ. Microbiol. 75, 1782–1785. Streptococcus salivarius: the carboxyl terminus specifies attachment in a
http://dx.doi.org/10.1128/AEM.01887-08. Streptococcus gordonii model system. J. Bacteriol. 175, 4520–4527 (Available from
Meng, G., Fütterer, K., 2003. Structural framework of fructosyl transfer in Bacillus subtilis URL: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC204894/).
levansucrase. Nat. Struct. Biol. 10, 935–941. http://dx.doi.org/10.1038/nsb974. Ritsema, T., Smeekens, S., 2003. Fructans: beneficial for plants and humans. Curr. Opin.
Meng, G., Fütterer, K., 2008. Donor substrate recognition in the raffinose-bound E342A Plant Biol. 6, 223–230. http://dx.doi.org/10.1016/S1369-5266(03)00034-7.
mutant of fructosyltransferase Bacillus subtilis levansucrase. BMC Struct. Biol. 8, Ritsema, T., Hernández, L., Verhaar, A., Altenbach, D., Boller, T., Wiemken, A., Smeekens, S.,
16–27. http://dx.doi.org/10.1186/1472-6807-8-16. 2006. Developing fructan-synthesizing capability in a plant invertase via mutations in
Molinari, M.L., Boiardi, J.L., 2013. Levans production by Gluconacetobacter diazotrophicus. the sucrose-binding box. Plant J. 48, 228–237. http://dx.doi.org/10.1111/j.1365-313X.
Electron. J. Biotechnol. 16 (3). http://dx.doi.org/10.2225/vol16-issue3-fulltext-9. 2006.02862.x.
Montana Polysaccharides Corp., 2015. Your source of levan. (Available from URL: www. Roberts, E.J., Garegg, P.J. Levan derivatives, their preparation, composition and applica-
polysaccharides.us). tions including medical and food applications. World patent appl. WO9803184. 1998.
Moosavi-Nasab, M., Layegh, B., Aminlari, L., Hashemi, M.B., 2010. Microbial production of Román-Leshkov, Y., Chheda, J.N., Dumesic, J.A., 2006. Phase modifiers promote efficient
levan using date syrup and investigation of its properties. WASET. Int. J. Biol. Biomol. production of hydroxymethylfurfural from fructose. Science 312 (5782), 1933–1937.
Agric. Food Biotechnol. Eng. 4, 603–609 (Available from URL: scholar.waset.org/1999. http://dx.doi.org/10.1126/science.1126337.
1/2614). Runyon, J.R., Nilsson, L., Ulmius, M., Castro, A., Ionescu, R., Andersson, C., Schmidt, C., 2014.
Moreno, C. Medicament-substituted dextran or levan conjugates, process for their prepa- Characterizing changes in levan physicochemical properties in different pH
ration and pharmaceutical compositions containing them. European patent appl. environments using asymmetric flow field-flow fractionation. Anal. Bioanal. Chem.
EP0000791A2. 1979. 406, 1597–1605. http://dx.doi.org/10.1007/s00216-013-7388-x.
Mu, W., Chen, Q., Wang, X., Zhang, T., Jiang, B., 2013. Current studies on physiological Saito, K., Tomita, F., 2000. Difructose anhydrides: their mass-production and physiological
functions and biological production of lactosucrose. Appl. Microbiol. Biotechnol. 97, functions. Biosci. Biotechnol. Biochem. 64, 1321–1327. http://dx.doi.org/10.1271/bbb.
7073–7080. http://dx.doi.org/10.1007/s00253-013-5079-3. 64.1321.
Nakapong, S., Pichyangkura, R., Ito, K., Iizuka, M., Pongsawasdi, P., 2013. High expression Scotti, P.A., Praestegaard, M., Chambert, R., Petit-Glatron, M.F., 1996. The targeting of
level of levansucrase from Bacillus licheniformis RN-01 and synthesis of levan nano- Bacillus subtilis levansucrase in yeast is correlated to both the hydrophobicity of
particles. Int. J. Biol. Macromol. 54, 30–36. http://dx.doi.org/10.1016/j.ijbiomac.2012. the signal peptide and the net charge of the N-terminus mature part. Yeast 12,
11.017. 953–963. http://dx.doi.org/10.1002/(SICI)1097-0061(199608)12:10b953::AID-
Oh, I.-K., Yoo, S.-H., Bae, I.-Y., Cha, J.-H., Lee, H.-G., 2004. Effects of Microbacterium YEA998N3.0.CO;2-#.
laevaniformans levans molecular weight on cytotoxicity. J. Microbiol. Biotechnol. 14, Sedgwick, A.D., Rutman, A., Sin, Y.M., Mackay, A.R., Willoughby, D.A., 1984. The effects of
985–990 (Available from URL: 210.101.116.28/W_files/kiss2/05209029_pv.pdf). levan on the acute inflammatory response. Br. J. Exp. Pathol. 65, 215–222 (Available
Oh, J.S., Lee, S.R., Hwang, K.T., Ji, G.E., 2014. The anti-obesity effects of the dietary combi- from URL: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2040958/).
nation of fermented red ginseng with levan in high fat diet mouse model. Phytother. Seibel, J., Moraru, R., Götze, S., Buchholz, K., Na'amnieh, S., Pawlowski, A., Hecht, H.-J.,
Res. 28, 617–622. http://dx.doi.org/10.1002/ptr.5042. 2006. Synthesis of sucrose analogues and the mechanism of action of Bacillus subtilis
Olvera, C., Centeno-Leija, S., López-Munguía, A., 2007. Structural and functional features of fructosyltransferase (levansucrase). Carbohydr. Res. 341, 2335–2349. http://dx.doi.
fructansucrases present in Leuconostoc mesenteroides ATCC 8293. Anton. Leeuw. 92, org/10.1016/j.carres.2006.07.001.
11–20. http://dx.doi.org/10.1007/s10482-006-9128-0. Senthilkumar, V., Rameshkumar, N., Busby, S.J.W., Gunasekaran, P., 2004. Disruption
Özcan, E., Toksoy Öner, E., 2015. Microbial production of extracellular polysaccharides of the Zymomonas mobilis extracellular sucrase gene (sacC) improves levan pro-
from biomass sources. In: Ramawat, K.G., Mérillon, J.M. (Eds.), Polysaccharides. duction. J. Appl. Microbiol. 96, 671–676. http://dx.doi.org/10.1111/j.1365-2672.
Springer, pp. 161–184 http://dx.doi.org/10.1007/978-3-319-16298-0_51. 2003.02169.x.
Ozimek, L.K., Kralj, S., van der Maarel, M.J.E.C., Dijkhuizen, L., 2006. The levansucrase and Senthilkumar, V., Rajendhran, J., Busby, S.J., Gunasekaran, P., 2009. Characterization
inulosucrase enzymes of Lactobacillus reuteri 121 catalyse processive and nonprocessive of multiple promoters and transcript stability in the sacB-sacC gene cluster in
transglycosylation reactions. Microbiology 152, 1187–1196. http://dx.doi.org/10.1099/ Zymomonas mobilis. Arch. Microbiol. 191, 529–541. http://dx.doi.org/10.1007/
mic.0.28484-0. s00203-009-0479-6.
Park, B.-S., Vladimir, A., Kim, C.H., Rhee, S.-K., Kang, H.A., 2004. Secretory production Seo, J.-S., Chong, H., Park, H.S., Yoon, K.-O., Jung, C., et al., 2005. The genome sequence of
of Zymomonas mobilis levansucrase by the methylotrophic yeast Hansenula the ethanologenic bacterium Zymomonas mobilis ZM4. Nat. Biotechnol. 23, 63–68.
polymorpha. Enzyme Microbiol. Technol. 34, 132–138. http://dx.doi.org/10. http://dx.doi.org/10.1038/nbt1045.
1016/j.enzmictec.2003.09.005. Sezer, A.D., Kazak Sarılmiser, H., Rayaman, E., Çevikbaş, A., Toksoy Öner, E., Akbuğa, J., 2015.
Pijning, T., Anwar, M.A., Boger, M., Dobruchowska, J.M., Leemhuis, H., Kralj, S., Dijkhuizen, Development and characterization of vancomycin-loaded levan-based microparticular
L., Dijkstra, B.W., 2011. Crystal structure of inulosucrase from Lactobacillus: insights system for drug delivery. Pharm. Dev. Technol. 1–8 http://dx.doi.org/10.3109/
into the substrate specificity and product specificity of GH68 fructansucrases. 10837450.2015.1116564 (Nov 25).
J. Mol. Biol. 412, 80–93. http://dx.doi.org/10.1016/j.jmb.2011.07.031. Shih, I.-L., Yu, Y.-T., 2005. Simultaneous and selective production of levan and poly
Poli, A., Kazak, H., Gürleyendağ, B., Tommonaro, G., Pieretti, G., Toksoy Öner, E., Nicolaus, (γ-glutamic acid) by Bacillus subtilis. Biotechnol. Lett. 27, 103–106. http://dx.doi.
B., 2009. High level synthesis of levan by a novel Halomonas species growing on org/10.1007/s10529-004-6936-z.
defined media. Carbohydr. Polym. 78, 651–657. http://dx.doi.org/10.1016/j.carbpol. Shih, I.-L., Yu, Y.-T., Shieh, C.-J., Hsieh, C.-Y., 2005. Selective production and characterization
2009.05.031. of levan by Bacillus subtilis (Natto) Takahashi. J. Agric. Food Chem. 53, 8211–8215.
Poli, A., Nicolaus, B., Denizci, A.A., Yavuzturk, B., Kazan, D., 2013. Halomonas smyrnensis sp. http://dx.doi.org/10.1021/jf058084o.
nov., a moderately halophilic, exopolysaccharide-producing bacterium from Camalti Shih, I.-L., Chen, L.-D., Wang, T.-C., Wu, J.-Y., Liaw, K.-S., 2010. Tandem production of levan
Saltern Area Turkey. Int. J. Syst. Evol. Microbiol. 63, 10–18. http://dx.doi.org/10. and ethanol by microbial fermentation. Green Chem. 12, 1242–1247. http://dx.doi.
1099/ijs.0.037036-0. org/10.1039/B924765C.
E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844 843

Shih, I.-L., Wang, T.-C., Chou, S.Z., Lee, G.D., 2011. Sequential production of two v. Lippmann, E.O., 1881. Ueber das Lävulan, eine neue, in der Melasse der
biopolymers-levan and poly-ε-lysine by microbial fermentation. Bioresour. Technol. Rubenzuckerfabriken vorkommende Gummiart. Chemisches Central-Blatt. 14,
102, 3966–3969. http://dx.doi.org/10.1016/j.biortech.2010.11.066. 669–671. http://dx.doi.org/10.1002/cber.188101401316.
Siddiqui, N.N., Aman, A., Silipo, A., Ul Qader, S.A., Molinaro, A., 2014. Structural analysis Van den Ende, W., Coopman, M., Clerens, S., Vergauwen, R., Le Roy, K., Lammens, W., Van
and characterization of dextran produced by wild and mutant strains of Leuconostoc Laere, A., 2011. Unexpected presence of graminan- and levan-type fructans in the
mesenteroides. Carbohydr. Polym. 99, 331–338. http://dx.doi.org/10.1016/j.carbpol. evergreen frost-hardy eudicot Pachysandra terminalis (Buxaceae). Purification, cloning
2013.08.004. and functional analysis of a 6-SST/6-SFT enzyme. Plant Physiol. 155, 603–614.
Silbir, S., Dagbagli, S., Yegin, S., Baysal, T., Goksungur, Y., 2014. Levan production http://dx.doi.org/10.1104/pp.110.162222.
by Zymomonas mobilis in batch and continuous fermentation systems. Carbohydr. Van der Meer, I.M., Ebskamp, M.J.M., Visser, R.G.F., Weisbeek, P.J., Smeekens, S.C.M., 1994.
Polym. 99, 454–461. http://dx.doi.org/10.1016/j.carbpol.2013.08.031. Fructan as a new carbohydrate sink in transgenic potato plants. Plant Cell 6, 561–570.
Sima, F., Mutlu, E.C., Eroglu, M.S., Sima, L.E., Serban, N., Ristoscu, C., Petrescu, S.M., Toksoy http://dx.doi.org/10.1105/tpc.6.4.561.
Öner, E., Mihailescu, I.N., 2011. Levan nanostructured thin films by MAPLE assembling. Van Geel-Schutten, G.H., Faber, E.J., Smit, E., Bonting, K., Smith, M.R., Ten Brink, B.,
Biomacromolecules 12, 2251–2256. http://dx.doi.org/10.1021/bm200340b. Kamerling, J.P., Vliegenthart, J.F.G., Dijkhuizen, L., 1999. Biochemical and struc-
Sima, F.N., Axente, E., Sima, L.E., Tuyel, U., Eroglu, M.S., Serban, N., Ristoscu, C., Petrescu, tural characterization of the glucan and fructan exopolysaccharides synthe-
S.M., Toksoy Öner, E., Mihailescu, I.N., 2012. Combinatorial matrix-assisted pulsed sized by the Lactobacillus reuteri wild-type strain and by mutant strains.
laser evaporation: single-step synthesis of biopolymer compositional gradient thin Appl. Environ. Microb. 65, 3008–3014. http://dx.doi.org/10.1128/AEM.69.2.
film assemblies. Appl. Phys. Lett. 101. http://dx.doi.org/10.1063/1.4769987 (233705-1 945-952.2003.
to 233705-5). van Hijum, S.A.F.T., van der Maarel, M.J.E.C., Dijkhuizen, L., 2003. Kinetic properties of an
Sinai, Y., Leibovici, J., Wolman, M., 1976. Effects of route and schedule of administra- inulosucrase from Lactobacillus reuteri 121. FEBS Lett. 534, 207–210. http://dx.doi.
tion of high-molecular levan on the growth of AKR lymphoma. Cancer Res. 36, org/10.1016/S0014-5793(02)03841-3.
1593–1597 (Available from URL: http://cancerres.aacrjournals.org/content/36/ van Hijum, S.A.F.T., Szalowska, E., van der Maarel, M.J.E.C., Dijkhuizen, L., 2004. Biochem-
5/1593). ical and molecular characterization of a levansucrase from Lactobacillus reuteri.
Sogutcu, E., Emrence, Z., Arikan, M., Cakiris, A., Abaci, N., Toksoy Öner, E., Ustek, D., Arga, Microbiology 150, 621–630. http://dx.doi.org/10.1099/mic.0.26671-0.
K.Y., 2012. Draft genome sequence of Halomonas smyrnensis AAD6T. J. Bacteriol. 194, van Hijum, S.A.F.T., Kralj, S., Ozimek, L.K., Dijkhuizen, L., van Geel-Schutten, I.G.H., 2006.
5690–5691. http://dx.doi.org/10.1128/JB.00559-12. Structure-function relationships of glucansucrase and fructansucrase enzymes from
Song, D.D., Jacques, N.A., 1999. Purification and enzymic properties of the fructosyltransferase lactic acid bacteria. Microbiol. Mol. Biol. Rev. 70, 157–176. http://dx.doi.org/10.
of Streptococcus salivarius ATCC 25975. Biochem. J. 341, 285–291. http://dx.doi.org/10. 1128/MMBR.70.1.157-176.2006.
1042/bj3410285. Velázquez-Hernández, M.L., Baizabal-Aguirre, V.M., Cruz-Vázquez, F., Trejo-Contreras,
Song, K.-B., Joo, H.-K., Rhee, S.-K., 1993. Nucleotide sequence of levansucrase gene (levU) M.J., Fuentes-Ramírez, L.E., Bravo-Patiño, A., Cajero-Juárez, M., Chávez-Moctezuma,
of Zymomonas mobilis ZM1 (ATCC10988). Biochim. Biophys. Acta 1173, 320–324. M.P., Valdez-Alarcón, J.J., 2011. Gluconacetobacter diazotrophicus levansucrase is
http://dx.doi.org/10.1016/0167-4781(93)90130-6. involved in tolerance to NaCl, sucrose and desiccation, and in biofilm formation.
Song, K.-B., Seo, J.-W., Kim, M.-G., Rhee, S.-K., 1998. Levansucrase of Rahnella aquatilis Arch. Microbiol. 193, 137–149. http://dx.doi.org/10.1007/s00203-010-0651-z.
ATCC33071: gene cloning, expression, and levan formation. Ann. N. Y. Acad. Sci. Vereyken, I.J., van Kuik, J.A., Evers, T.H., Rijken, P.J., de Kruijff, B., 2003. Structural require-
864, 506–511. http://dx.doi.org/10.1111/j.1749-6632.1998.tb10369.x. ments of the fructan-lipid interaction. Biophys. J. 84, 3147–3154. http://dx.doi.org/10.
Sonnenburg, E.D., Zheng, H., Joglekar, P., Higginbottom, S., Firbank, S.J., Bolam, D.N., 1016/S0006-3495(03)70039-3.
Sonnenburg, J.L., 2010. Specificity of polysaccharide use in intestinal Bacteroides Vigants, A., Upite, D., Scherbaka, R., Lukjanenko, J., Ionina, R., 2013. An influence of ethanol
species determines diet-induced microbiota alterations. Cell 141, 1241–1252. and temperature on products formation by different preparations of Zymomonas
http://dx.doi.org/10.1016/j.cell.2010.05.005. mobilis extracellular levansucrase. Folia Microbiol. (Praha) 58, 75–80. http://dx.doi.
Srikanth, R., Siddartha, G., Sundhar Reddy, C.H., Harish, B.S., Janaki Ramaiah, M., Uppuluri, org/10.1007/s12223-012-0185-7.
K.B., 2015. Antioxidant and anti-inflammatory levan produced from Acetobacter Viikari, L., 1984. Formation of levan and sorbitol from sucrose by Zymomonas mobilis.
xylinum NCIM2526 and its statistical optimization. Carbohydr. Polym. 123, 8–16. Appl. Microbiol. Biotechnol. 19, 252–255. http://dx.doi.org/10.1007/BF00251846.
http://dx.doi.org/10.1016/j.carbpol.2014.12.079. Vijayendra, S.V.N., Shamala, T.R., 2014. Film forming microbial biopolymers for commercial
Steinmetz, M., Le Coq, D., Aymerich, S., Gonzy-Treboul, G., Gay, P., 1985. The DNA applications—a review. Crit. Rev. Biotechnol. 34, 338–357. http://dx.doi.org/10.3109/
sequence of the gene for the secreted Bacillus subtilis enzyme levansucrase and its 07388551.2013.798254.
genetic control sites. Mol. Gen. Genet. 200, 220–228. http://dx.doi.org/10.1007/ Vijn, I., Smeekens, S., 1999. Fructan: more than a reserve carbohydrate? Plant Physiol.
BF00425427. 120, 351–359 (Available from URL: http://www.ncbi.nlm.nih.gov/pmc/articles/
Strube, C.P., Homann, A., Gamer, M., Jahn, D., Seibel, J., Heinz, D.W., 2011. Polysaccharide PMC1539216/).
synthesis of the levansucrase SacB from Bacillus megaterium is controlled by distinct Vincent, S., Brandt, M., Cavadini, C., Hammes, W.P., Neeser, J-R., Waldbuesser, S. Levan-
surface motifs. J. Biol. Chem. 286, 17593–17600. http://dx.doi.org/10.1074/jbc.M110. producing Lactobacillus strain and method of preparing human or pet food products
203166. using the same. US patent 6,932,991. 2005.
Sturzoiu, C., Petrescu, M., Galateanu, B., Anton, M., Nica, C., Simionca, G., Dinischiotu, A., Visnapuu, T., Mardo, K., Mosoarca, C., Zamfir, A.D., Vigants, A., Alamäe, T., 2011.
Stoian, G., 2011. Zymomonas mobilis levan is involved in metalloproteinases activation Levansucrases from Pseudomonas syringae pv. tomato and P. chlororaphis subsp.
in healing of wounded and burned tissues. Scientific Papers: Animal Science and Bio- aurantiaca: substrate specificity, polymerizing properties and usage of different ac-
technologies Vol. 44, pp. 453–458 (Available from URL: https://www.researchgate. ceptors for fructosylation. J. Biotechnol. 155, 338–349. http://dx.doi.org/10.1016/j.
net/publication/267951567_Zymomonas_mobilis_Levan_is_Involved_in_ jbiotec.2011.07.026.
Metalloproteinases_Activation_in_Healing_of_Wounded_and_Burned_Tissues). Visnapuu, T., Mardo, K., Alamäe, T., 2015. Levansucrases of a Pseudomonas syringae
Sukan, A., Roy, I., Keshavarz, T., 2015. Dual production of biopolymers from bacteria. pathovar as catalysts for the synthesis of potentially prebiotic oligo- and polysaccha-
Carbohydr. Polym. 126, 47–51. http://dx.doi.org/10.1016/j.carbpol.2015.03.001. rides. New Biotechnol. 32, 597–605. http://dx.doi.org/10.1016/j.nbt.2015.01.009.
Tajima, K., Uenishi, N., Fujiwara, M., Erata, T., Munekata, M., Takai, M., 1997. The produc- Waldherr, F.W., Meissner, D., Vogel, R.F., 2008. Genetic and functional characterization of
tion of a new water-soluble polysaccharide by Acetobacter xylinum NCI 1005 and Lactobacillus panis levansucrase. Arch. Microbiol. 190, 497–505. http://dx.doi.org/10.
its structural analysis by NMR spectroscopy. Carbohydr. Res. 305, 117–122. 1007/s00203-008-0404-4.
http://dx.doi.org/10.1016/S0008-6215(97)00264-4. Wang, W.-S., Wang, D.-H., 1997. Enhancement of the resistance of tilapia and grass carp
Takahashi, M., Takayama, R., Ishihara, K., 1994. Physicochemical properties and hypolipid- to experimental Aeromonas hydrophila and Edwardsiella tarda infections by several
emic function of levan and its partial hydrolysate. In: Yano, T., Matsuno, R., Nakamura, polysaccharides. Comp. Immunol. Microbiol. Infect. Dis. 20, 261–270. http://dx.doi.
K. (Eds.), Developments in Food Engineering. Springer, pp. 69–71 http://dx.doi.org/10. org/10.1016/S0147-9571(96)00035-5.
1007/978-1-4615-2674-2_15. Wu, F.-C., Chou, S.-Z., Shih, I.-L., 2013. Factors affecting the production and molecular
Támbara, Y., Hormaza, J.V., Perez, C., Leon, A., Arrieta, J., Hernández, L., 1999. Structural weight of levan of Bacillus subtilis natto in batch and fed-batch culture in fermen-
analysis and optimised production of fructo-oligosaccharides by levansucrase from ter. J. Taiwan Inst. Chem. Eng. 44, 846–853. http://dx.doi.org/10.1016/j.jtice.2013.03.
Acetobacter diazotrophicus SRT4. Biotechnol. Lett. 21, 117–121. http://dx.doi.org/10. 009.
1023/A:1005474017009. Wuerges, J., Caputi, L., Cianci, M., Boivin, S., Meijers, R., Benini, S., 2015. The crystal struc-
Tanaka, T., Oi, S., Yamamoto, T., 1980. The molecular structure of low and high molecular ture of Erwinia amylovora levansucrase provides a snapshot of the products of sucrose
weight synthesized by levansucrase. J. Biochem. 87, 297–303 (Available from URL: hydrolysis trapped into the active site. J. Struct. Biol. 191, 290–298. http://dx.doi.org/
http://www.ncbi.nlm.nih.gov/pubmed/6766925). 10.1016/j.jsb.2015.07.010.
Tonozuka, T., Tamaki, A., Yokoi, G., Miyazaki, T., Ichikawa, M., Nishikawa, A., Ohta, Y., Xiao, M., Feng, F., Lu, L. Preparation method of levan-contained yogurt, Chinese patent
Hidaka, Y., Katayama, K., Hatada, Y., Ito, T., Fujita, K., 2012. Crystal structure of a CN103190478B. 2014.
lactosucrose-producing enzyme, Arthrobacter sp. K-1 β-fructofuranosidase. Yamamoto, Y., Takahashi, Y., Kawano, M., Iizuka, M., Matsumoto, T., Saeki, S., Yamaguchi,
Enzym. Microb. Technol. 51, 359–365. http://dx.doi.org/10.1016/j.enzmictec. H., 1999. In vitro digestibility and fermentability of levan and its hypocholesterolemic
2012.08.004. effects in rats. J. Nutr. Biochem. 10, 13–18. http://dx.doi.org/10.1016/S0955-2863
Trujillo, L.E., Arrieta, J.G., Dafhnis, F., Garcia, J., Valdes, J., Tambara, Y., Perez, M., Hernández, (98)00077-1.
L., 2001. Fructo-oligosaccharides production by the Gluconacetobacter diazotrophicus Yoo, S.-H., Yoon, E.J., Cha, J., Lee, H.G., 2004. Antitumor activity of levan polysaccharides
levansucrase expressed in the methylotrophic yeast Pichia pastoris. Enzym. Microb. from selected microorganisms. Int. J. Biol. Macromol. 34, 37–41. http://dx.doi.org/
Technol. 28, 139–144. http://dx.doi.org/10.1016/S0141-0229(00)00290-8. 10.1016/j.ijbiomac.2004.01.002.
Trujillo, L.E., Gómez, R., Banguela, A., Soto, M., Arrieta, J.G., Hernández, L., 2004. Catalytical Zeng, L., Wen, Z.T., Burne, R.A., 2006. A novel signal transduction system and feed-
properties of N-glycosylated Gluconacetobacter diazotrophicus levansucrase produced back loop regulate fructan hydrolase gene expression in Streptococcus mutans.
in yeast. Electron. J. Biotechnol. 7, 115–123. http://dx.doi.org/10.2225/vol7-issue2- Mol. Microbiol. 62, 187–200. http://dx.doi.org/10.1111/j.1365-2958.2006.
fulltext-4. 05359.x.
844 E.T. Öner et al. / Biotechnology Advances 34 (2016) 827–844

Zhang, T., Li, R., Qian, H., Mu, W., Miao, M., Jiang, B., 2014. Biosynthesis of levan by Zhao, P.Y., Wang, J.P., Kim, I.H., 2013b. Effect of dietary levan fructan supplementation on
levansucrase from Bacillus methylotrophicus SK 21.002. Carbohydr. Polym. 101, growth performance, meat quality, relative organ weight, cecal microflora, and excreta
975–981. http://dx.doi.org/10.1016/j.carbpol.2013.10.045. noxious gas emission in broilers. J. Anim. Sci. 91, 5287–5293. http://dx.doi.org/10.
Zhao, P.Y., Wang, J.P., Kim, I.H., 2013a. Evaluation of dietary fructan supplementation on 2527/jas.2012-5464.
growth performance, nutrient digestibility, meat quality, fecal microbial flora, and
fecal noxious gas emission in finishing pigs. J. Anim. Sci. 91, 5280–5286. http://dx.
doi.org/10.2527/jas.2012-5393.

You might also like