Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Appl Microbiol Biotechnol (2013) 97:2961–2970

DOI 10.1007/s00253-012-4184-z

BIOTECHNOLOGICALLY RELEVANT ENZYMES AND PROTEINS

Enzymatic production of 3,6-anhydro-L-galactose


from agarose and its purification and in vitro
skin whitening and anti-inflammatory activities
Eun Ju Yun & Saeyoung Lee & Ji Hye Kim & Bo Bae Kim &
Hee Taek Kim & Sun Hee Lee & Jeffrey G. Pelton &
Nam Joo Kang & In-Geol Choi & Kyoung Heon Kim

Received: 2 February 2012 / Revised: 19 March 2012 / Accepted: 15 May 2012 / Published online: 8 June 2012
# Springer-Verlag 2012

Abstract 3,6-Anhydro- L-galactose (L-AHG) constitutes L-AHG at a concentration of 100 or 200 μg/mL did not exhibit
50 % of agarose, which is the main component of red macro- any significant cytotoxicity. In a skin whitening assay, 100 μg/
algae. No information is currently available on the mass pro- mL of L-AHG showed significantly lower melanin production
duction, metabolic fate, or physiological effects of L-AHG. compared to arbutin. L-AHG at 100 and 200 μg/mL showed
Here, agarose was converted to L-AHG in the following three strong anti-inflammatory activity, indicating the significant
steps: pre-hydrolysis of agarose into agaro-oligosaccharides by suppression of nitrite production. This is the first report on
using acetic acid, hydrolysis of the agaro-oligosaccharides into the production of high-purity L-AHG and its physiological
neoagarobiose by an exo-agarase, and hydrolysis of neoagar- activities.
obiose into L-AHG and galactose by a neoagarobiose hydro-
lase. After these three steps, L-AHG was purified by Keywords 3,6-Anhydro-L-galactose . Agar . Red
adsorption and gel permeation chromatographies. The final macroalgae . Skin whitening . Anti-inflammation
product obtained was 95.6 % pure L-AHG at a final yield of
4.0 % based on the initial agarose. In a cell proliferation assay,
Introduction
E. J. Yun : H. T. Kim : S. H. Lee : K. H. Kim (*)
School of Life Sciences and Biotechnology, Korea University,
An anhydrosugar is synthesized from a monosaccharide
Seoul 136-713, Korea
e-mail: khekim@korea.ac.kr derivative, such as a glucopyranoside, through the forma-
tion of an intramolecular ether bond by releasing a water
S. Lee : I.-G. Choi (*) molecule. Anhydrosugars are considered rare, and their
Division of Biotechnology, College of Life Sciences and
synthesis, metabolism, and biological functions are not
Biotechnology, Korea University,
Seoul 136-713, Korea well studied (Kitamura et al. 1991; Kuhn et al. 2006;
e-mail: igchoi@korea.ac.kr Rees 1961; Yamaji et al. 2002). 3,6-Anhydro-L-galactose
(L-AHG) naturally occurs as one the monomers of aga-
J. H. Kim : B. B. Kim : N. J. Kang (*)
School of Applied Biosciences, Kyungpook National University,
rose by alternating galactose and L-AHG units; agarose
Daegu 702-701, Korea is the main component of red macroalgae (Rhodophyta)
e-mail: njkang@knu.ac.kr (Rees 1961; Sekkal et al. 1993; Su and Hassid 1962). To
date, no information is available on the biological func-
J. G. Pelton
tions and metabolic pathways of L-AHG, as is the case
Physical Biosciences Division, Lawrence
Berkeley National Laboratory, with other anhydrosugars; therefore, L-AHG is not com-
Berkeley, CA 94720, USA mercially available even as a reagent. Many marine bac-
teria are capable of degrading and metabolizing agarose,
J. H. Kim : B. B. Kim : N. J. Kang
but the metabolic pathway of L-AHG is almost complete-
School of Food Science and Biotechnology,
Kyungpook National University, ly unknown (Ekborg et al. 2006; Shin et al. 2010; Yun et
Daegu 702-701, Korea al. 2011).
2962 Appl Microbiol Biotechnol (2013) 97:2961–2970

Although the biological activity of L-AHG itself remains In this study, we produced L-AHG from agarose by a series
unknown, many biological functions of the oligomers of L- of chemical and enzymatic steps comprising mild pre-
AHG and D-galactose, namely, agaro-oligosaccharides, have hydrolysis using acetic acid and enzymatic saccharification
been reported. These functions include apoptosis induction, by using an exo-type β-agarase (Aga50D) (Kim et al. 2010a,
carcinostatic activity, antioxidation, hepatoprotection, im- b) and an NABH extracted from S. degradans 2-40
munoregulation, anti-allergy, anti-inflammation, and α- (SdNABH, formerly AgaJ) (Ha et al. 2011; Lee et al. 2009).
glucosidase inhibition (Chen et al. 2005; Chen et al. 2006; To obtain highly pure L-AHG, the reaction product of agarose
Tomono et al. 2009). Neoagarobiose (3-O-(3,6-anhydro-L- was purified through adsorption and gel permeation chroma-
galactopyranosyl)-D-galactose), the dimeric form of L-AHG tographies using silica gel and Bio-Gel P-2 columns, respec-
and galactose, can be used as a novel moisturizer with tively. The structure of purified L-AHG was verified by gas
whitening effect (Kobayashi et al. 1997). The bioactivities chromatography–mass spectrometry (GC–MS). We used this
of the agaro-oligosaccharides are presumed to be primarily purified L-AHG to perform the first biological study of its
due to the presence of L-AHG (Tomono et al. 2009). There- whitening and anti-inflammatory effects. This is the first re-
fore, it is reasonable to examine the possible skin whitening port on the enzymatic conversion of agarose into L-AHG by
and anti-inflammatory effects of L-AHG and thereby to test using necessary recombinant enzymes and the purification of
its application in the treatment of hyperpigmentation, which L-AHG for the elucidation of its possible bioactivity.
is commonly caused by inflammatory skin disorders (Urabe
et al. 1998). Although a number of whitening agents includ-
ing arbutin are currently used for the treatment of acquired Materials and methods
skin hyperpigmentation, they are only modestly effective,
and some are known to produce adverse side effects (Lee et Pre-hydrolysis of agarose into agaro-oligosaccharides
al. 2003; Penney et al. 1984).
For the industrial production of agaro-oligosaccharides, the To prepare agaro-oligosaccharides from agarose (Intron
most common method used is acid hydrolysis at high tempera- Biotechnology, Seongnam, Korea), 5 g of agarose was pre-
ture because this method is simple, cheap, and rapid and affords hydrolyzed in 100 mL of 3 M acetic acid at 80 °C for 70 min
a high yield (Chen et al. 2005; Yang et al. 2009). Acid hydrolysis with agitation at 150 rpm in a water bath. The reaction
leads mainly to the cleavage of α-1,3 glycosidic linkages of product was dried by using a rotary vacuum evaporator at
agarose to produce agaro-oligosaccharides. To further degrade 65 °C for 2 h to remove water and acetic acid. The dried
agaro-oligosaccharides into monomeric sugars (i.e., L-AHG and residues, including acetic acid, were washed with 1.5 L of
D-galactose), hydrolysis by a strong acid is required (Jol et al. 94.0 % (v/v) ethanol (Daejung, Siheung, Korea) and filtered
1999; Kim et al. 2010a, b). However, such a strong acid can through a membrane filter (pore size, 0.45 μm; Whatman,
overdegrade L-AHG and galactose to generate toxic byproducts, Dassel, Germany). The filtered solids were collected as
such as 5-hydroxy-methyl-furfural (HMF). In particular, L- hydrolyzed agaro-oligosaccharides, and they were dried in
AHG is known to be readily converted into HMF (Kim et al. a vacuum drying oven at 60 °C for 24 h.
2010a, b; Stevenson and Furneaux 1991; Yang et al. 2009).
Therefore, when producing L-AHG from agarose, agar, or red Enzymatic hydrolysis of agaro-oligosaccharides
macroalgae, it would be preferable to rely on enzymatic degra-
dation to prevent the further degradation of L-AHG. Agaro-oligosaccharides obtained from acetic acid hydrolysis
In the enzymatic hydrolysis of agarose, the β-agarase were further hydrolyzed sequentially by a neoagarobiose-
system, which contains endo-type β-agarases and exo-type producing β-agarase (i.e., Aga50D) and an NABH (i.e.,
β-agarases to produce neoagaro-oligosaccharides and neo- SdNABH). Aga50D and SdNABH both originate from S.
agarobiose, respectively, is the major pathway in marine degradans 2-40 (ATCC 43961), and their modes of action have
bacteria, such as Pseudoalteromonas atlantica and Saccha- been published previously (Kim et al. 2010a, b; Lee et al.
rophagus degradans 2-40 (Ekborg et al. 2006; Kim et al. 2009). The expression of those recombinant enzymes in
2010a, b; Vera et al. 1998). Neoagarobiose is finally hydro- Escherichia coli BL21 (DE3) and their purification by affinity
lyzed into L-AHG and D-galactose by neoagarobiose hydro- chromatography with a HisTrap HP (GE Healthcare, Piscat-
lase (NABH), which is known to be rare but was recently away, NJ, USA) were performed according to a previously
studied by our group (Ha et al. 2011; Lee et al. 2009). Thus, described method (Kim et al. 2010a, b; Lee et al. 2009). For
enzymatic hydrolysis may be a suitable method for the enzymatic hydrolysis, agaro-oligosaccharides at a concentra-
industrial production of commercially unavailable L-AHG. tion of 5 % (w/v) were first hydrolyzed by 10 mg of Aga50D in
To date, crude enzymes of cell-free lysate from S. degradans 100 mL of 50 mM Tris–HCl buffer (pH 7.4) at 30 °C for 72 h in
2-40 have been applied to hydrolyzing agarose into L-AHG a shaking incubator at 150 rpm. After the enzymatic reaction by
as the only enzymatic conversion method (Yun et al. 2011). Aga50D, the reaction mixture was centrifuged, and the
Appl Microbiol Biotechnol (2013) 97:2961–2970 2963

supernatant was then hydrolyzed by 2.5 mg of NABH under Thin-layer chromatography of the reaction products
the same reaction conditions used for the Aga50D reaction.
One hundred milliliters of the reaction product was concentrat- TLC analysis of the enzymatic reaction product was con-
ed to 20 mL using a rotary vacuum evaporator at 40 °C for 2 h ducted on a silica gel 60 (Merck, Darmstadt, Germany)
and dried with 10 g of Celite 545 (Yakuri, Tokyo, Japan) in a plate, and the plate was developed with n-butanol/ethanol/
drying oven at 40 °C for 24 h. water (3:1:1, v/v/v) for 1 h. Thereafter, the plate was dried
and visualized using a solution composed of 10 % (v/v)
Chromatographic purification of L-AHG sulfuric acid and 0.2 % (w/v) naphthoresorcinol (Tokyo
Chemical Industry, Tokyo, Japan) in ethanol at 90 °C for
To purify L-AHG from the dried enzymatic reaction 1 min.
product on Celite, we performed silica gel chromatog-
raphy. Before loading the sample, a column (I.D. 4× Preparation and purification of neoagarobiose
100 cm) packed with silica gel 60 (70-230 mesh
ASTM; Merck, Darmstadt, Germany) was equilibrated To obtain neoagarobiose as the standard for TLC and GC–
with chloroform. After the dried enzymatic reaction MS, agarose at a concentration of 10 % (w/v) was hydro-
product was directly applied on top of the column, lyzed into neoagarobiose at 30 °C for 72 h in a shaking
elution was performed with 3 L of the mobile phase incubator at 150 rpm after adding 50 mg of Aga50D to
(chloroform/methanol/H2O078:20:2, v/v/v), and 20 mL 100 mL of 50 mM Tris–HCl buffer (pH 7.4). After the
of each fraction was collected for thin-layer chromato- enzymatic reaction by Aga50D, the reaction mixture was
graphic (TLC) analysis. Only the fractions which were centrifuged, and 50 mL of the supernatant was concentrated
verified to contain only AHG without galactose by TLC to 2 mL using a rotary vacuum evaporator at 35 °C for 2 h.
were collected (Fig. 1) and dried using a rotary vacuum The concentrated reaction product was loaded onto a Bio-
evaporator at 35 °C for 2 h to remove the organic Gel P-2 column equilibrated with water for the purification
solvent. These fractions were then dissolved in water of neoagarobiose by gel permeation chromatography. Each
at a concentration of 123.0 mg/mL. For further purifi- fraction of 2 mL was obtained by elution with water as the
cation of L-AHG by size-exclusion chromatography, the mobile phase, and the fractions containing neoagarobiose
L-AHG solution obtained from the silica gel chromatog- only were collected.
raphy was loaded onto a Bio-Gel P-2 column (I.D. 2.1×
35.8 cm, Amersham Biosciences, Piscataway, NJ, USA) Quantification of L-AHG and neoagarobiose by GC–MS
equilibrated with water. Each 2 ml fraction was obtained
by elution with water as the mobile phase, and fractions To quantify L-AHG, the previously developed analytical
containing AHG only, as confirmed by TLC, were method using GC–MS was used (Yun et al. 2011). Briefly,
collected. in the range of 50–1000 μg, 3,6-anhydro-D-galactose (D-
AHG; Dextra Laboratories, Berkshire, UK) was derivatized
prior to GC–MS analysis. The aldehyde group of D-AHG
was protected by methoximation by treatment with 50 μL of
2 % (w/v) of methoxyamine hydrochloride in pyridine (Sig-
ma, St. Louis, MO, USA) at 75 °C for 30 min. To increase
the volatility of D-AHG, 80 μL of N-methyl-N-trimethyl-
silyl-trifluoroacetamide (MSTFA; Fluka, St. Louis, MO,
USA) was added to the methoxymized sample. GC–MS
analysis was performed using an Agilent 7890A GC/
5975 C MSD system (Agilent Technologies, Wilmington,
DE, USA) equipped with a DB5-MS column (I.D. 30 m×
0.25 mm, 0.25-μm film thickness; Agilent Technologies).
The GC–MS operating conditions were as follows: The
Fig. 1 TLC of the products obtained from each step of agarose
column temperature was initially set at 100 °C for 3.5 min
hydrolysis and purification: processing step 1, agarose; processing step and increased to 160 °C at 15 °C/min, at which it was
2, acetic acid pre-hydrolysis and washing and drying; processing step maintained for 20 min. It was then increased to 200 °C at
3, enzymatic hydrolysis by Aga50D; processing step 4, enzymatic 20 °C/min and held at 200 °C for 15 min. Finally, the
hydrolysis by SdNABH; processing step 5, silica gel chromatography;
and processing step 6, Bio-Gel P-2 chromatography. Neoagarobiose
temperature was increased to 280 °C at 20 °C/min and
(NAB), D-AHG, and galactose were used as the standards (first and maintained at the same temperature for 5 min. The mass
second lanes) spectra were acquired in a scan range of 50–700 m/z, and
2964 Appl Microbiol Biotechnol (2013) 97:2961–2970

the electron impact and the temperature of the ion source tetrazolium (MTT) solution was added to each well. The
were 70 eV and 230 °C, respectively. The amount of AHG cells were then incubated for 2 h at 37 °C in the CO2
was calibrated on the basis of the intensity of its peaks in the incubator, and the absorbance of the cell culture was mea-
total ion chromatogram. Neoagarobiose was also quantified sured at 570 nm. Experiments were conducted in triplicate,
by GC–MS by using the earlier method for quantifying L- and data were expressed as mean ± standard deviation.
AHG. To determine the L-AHG purity of each sample Student'st-test was performed for single statistical compar-
obtained from the production and purification steps, the isons. A probability value of p<0.05 was used as the crite-
amount of L-AHG in each sample was quantified by GC– rion for statistical significance.
MS and divided by the total dry weight of each sample.
Skin whitening activity assay
Nuclear magnetic resonance spectroscopy of L-AHG
A murine melanoma cell line, B16F10, was obtained from
Two milligrams of L-AHG, which was produced from aga- Korean Cell Line Bank (KCLB, Seoul, Korea). The cells
rose and purified in this study, was dried in a centrifugal were cultured in a Dulbecco’s modified Eagle’s medium
vacuum concentrator at 30 °C for 6 h, and the dried L-AHG (DMEM; Invitrogen, Auckland, New Zealand) supple-
was dissolved in D2O. 1 H–13 C nuclear magnetic resonance mented with 10 % (w/v) of heat-inactivated fetal bovine
(NMR) spectroscopy was performed using a Bruker Avance serum (FBS; Sigma) and 1 % (w/v) of penicillin–streptomy-
II 900 MHz NMR spectrometer equipped with a cryoprobe cin at 37 °C in a humidified atmosphere with 5 % CO2.
(Bruker, Billerica, MA, USA). Chemical shifts were RAW264.7, a mouse macrophage cell line, was obtained
referenced to 3-(trimethylsilyl)-propionic-2,2,3,3-d4 acid. from KCLB and cultured in DMEM supplemented with
The chemical structure of L-AHG was confirmed by com- 2 mM of glutamine, antibiotics such as penicillin A and
paring its 1 H and 13 C chemical shifts measured in this study streptomycin at concentrations of 100 units/mL each, and
with its literature values (Sugano et al. 1994) and agarobiose 10 % (v/v) of heat-inactivated FBS. RAW264.7 cells were
(4-O-(D-galactosyl)-3,6-anhydro-L-galactopyranose) (Miller grown at 37 °C in a fully humidified atmosphere containing
et al. 1982), respectively. 5 % CO2.
The melanin content was measured using a previously
reported method (Tsuboi et al. 1998). Briefly, confluent
Polarimetry of L-AHG
monolayers of B16F10 melanoma cells were trypsinized,
and 8×103 viable cells suspended in 3 mL of 10 % FBS
To verify that the AHG obtained in this study is L-AHG, we
DMEM were added to each well of a 96-well plate. The
have measured the specific rotation of L-AHG using a polar-
plate was incubated at 37 °C in a humidified atmosphere
imeter, and it was compared with that of D-AHG and the
containing 5 % CO2. When the cells reached 80–90 %
reported value of L-AHG. The purified L-AHG (4 mg) was
confluence, they were starved by culturing them in serum-
dissolved in 2 mL of dH2O and left to stand overnight. The
free DMEM for further 12 h. Before being exposed to
polarimetry of L-AHG was measured with a JASCO P-2000
100 nM of α-melanocyte-stimulating hormone (α-MSH;
(JASCO, Easton, MD, USA). The specific rotation, ½a25 D , was Sigma), the cells were treated with arbutin, neoagarobiose,
calculated using the following equation: ½a25 D ¼ a = lc, where α galactose, D-AHG, and L-AHG each for 1 h at concentra-
is the observed optical rotation (°), l is the length of a sample tions of 1, 10, and 100 μg/mL. After 4 days of cultivation
tube (dm), and c is the solution concentration (g/mL). The with α-MSH, the culture medium was collected to deter-
specific rotation of L-AHG was compared with that of D-AHG mine the relative melanin contents by measuring the absor-
at a concentration of 0.3 % (w/v) in dH2O as well as the bance at 475 nm with an enzyme-linked immunosorbent
previously published value of L-AHG as the reference (Araki assay (ELISA) reader (Sunrise Basic Tecan, Gröding, Aus-
and Hirase 1953). tria). These experiments were conducted in triplicate, and
data were expressed as mean ± standard deviation. For
Cell proliferation assay single statistical comparisons, Student's t-test was per-
formed, and a probability value of p<0.05 was used as the
To estimate the possible cytotoxicity of L-AHG and other criterion for statistical significance.
substances tested in this study, cell proliferation assay was
performed. B16F10 cells and RAW264.7 cells were seeded Anti-inflammatory activity assay
1×104 cells/well in 96-well plates with 10 % (v/v) FBS/
DMEM at 37 °C in a 5 % CO2 (v/v) incubator. After As an indicator of anti-inflammatory activity, nitrite concen-
culturing for 24 h, 20 μL of 3-(4,5-dimethylthiazol-2-yl)- tration in the RAW264.7 cell culture medium was measured
5-(3-carboxymethoxyphenyl)-2-(4-sulfonyl)-2 H- using Griess reaction (Ding et al. 1988). Cells were plated at
Appl Microbiol Biotechnol (2013) 97:2961–2970 2965

Table 1 Product recovery yield and L-AHG purity at the stages of Results
production from agarose and purification

Processing step Product Recovery yield Purity of Production of L-AHG from agarose
of product based L-AHG
on initial agarose (%, w/w) To obtain L-AHG from agarose, the substrate agarose was
(%, w/w)
sequentially hydrolyzed by acetic acid and enzymes
Initial substrate Agarose 100.0 NA (processing step 1 on the thin layer chromatogram shown
Acetic acid Agaro- NA NA in Fig. 1). The products obtained from each reaction and
pre-hydrolysis oligosacchar- purification step were analyzed by TLC, as shown in Fig. 1.
ides Processing step 2 on the chromatogram in Fig. 1 also
Ethanol Agaro- 76.0 NA
indicates agaro-oligosaccharides with various degrees of
washing and oligosacchar-
drying ides polymerization (DPs), which were obtained by acetic acid
Enzymatic Neoagarobiose 37.6 NA pre-hydrolysis performed at 80 °C for 70 min. After pre-
hydrolysis by hydrolysis using acetic acid, the filtered solids were washed
Aga50D
with ethanol and dried. As seen in Table 1, the total recovery
Enzymatic L-AHG 15.2 18.0
hydrolysis by yield of the filtered, ethanol-washed, and dried solids, which
SdNABH were presumably agaro-oligosaccharides, was as high as
Silica gel L-AHG 7.7 57.5 76.0 % (w/w) of the initial input agarose used in the pre-
chromatogra-
hydrolysis.
phy
Bio-Gel P-2 L-AHG 4.0 95.6 The agaro-oligosaccharides obtained as solids from the
chromatogra- acetic acid hydrolysis product were then hydrolyzed by the
phy exo-type agarase, Aga50D. The enzymatic reaction product
obtained by Aga50D showed a strong band that was pre-
NA not applicable
sumed to represent neoagarobiose in processing step 3 on
the chromatogram in Fig. 1. These results were in good
agreement with our previous finding that Aga50D was an
a density of 1×106 cells/well in a 96-well plate containing exo-type agarase, producing neoagarobiose predominantly
phenol red-free DMEM and incubated overnight. The cells from agarose (Kim et al. 2010a, b). After the enzyme reac-
were treated with 2 μg/mL of lipopolysaccharide (LPS; E. tion using Aga50D, the enzymatic conversion yield was
coli 0127:B8, Sigma) to induce nitrite production in the 49.4 %, and the final recovery yield of neoagarobiose from
absence or presence of testing samples, such as neoagaro- the initial agarose (including the pre-hydrolysis process)
biose, galactose, D-AHG, and L-AHG, at concentrations of was 37.6 % (Table 1).
50, 100, and 200 μg/mL and incubated for 24 h. After The reaction product (mainly neoagarobiose) from the
incubation, the plate was centrifuged at 3,000 rpm for enzymatic hydrolysis by Aga50D was further hydrolyzed
15 min at room temperature to obtain the supernatant of by sdNABH and analyzed by TLC, as shown in Fig. 1. The
the cell culture. The supernatant was analyzed using an monomeric sugars, AHG and galactose, showed strong
ELISA reader for measurement of nitrite concentration in bands in processing step 4 of the TLC (Fig. 1). Thus, we
terms of absorbance at 540 nm. Data were expressed as confirmed that SdNABH cleaved the glycosidic linkage of
mean ± standard deviation of three independent experi- neoagarobiose as we had previously reported (Ha et al.
ments, and Student'st-test was used for single statistical 2011; Lee et al. 2009). As shown in Table 1, the conversion
comparisons. Probability values of p<0.05 were considered yield of the reaction product of agaro-oligosaccharides by
as statistically significant. Aga50D into AHG was 40.5 %, and the purity of L-AHG in

Fig. 2 TLC of each fraction


obtained by silica gel
chromatography, where the
accumulated elution volume of
fractions from the silica gel
column was indicated. Only the
fractions containing L-AHG
were collected (i.e., 0.77–
1.77 L of elution volume) for
further chromatographic
purification
2966 Appl Microbiol Biotechnol (2013) 97:2961–2970

the reaction product by SdNABH was 18.0 %. Overall, it is


expected that, when 100 g of agarose is subjected to acid
pre-hydrolysis and enzymatic hydrolysis, 15.2 g of L-AHG
can be produced.

Purification of L-AHG by chromatography

The purity of L-AHG was only 18.0 % after pre-hydrolysis


and enzymatic hydrolysis; therefore, the obtained L-AHG
needed to be purified before being subjected to structural
analysis by GC–MS and the biological activity tests. First,
we performed silica gel chromatography, and only the frac-
tions that showed L-AHG without galactose on the TLC
(elution volume of 0.77–1.77 mL) in Fig. 2 were collected.
The original purity of L-AHG in the final enzymatic reac-
tion mixture with SdNABH, 18.0 %, increased to 57.5 % by Fig. 3 Total ion chromatograms from the GC–MS analyses of a D-
silica gel chromatography, showing a purification fold of 3.2 AHG (authentic standard) and b L-AHG produced from agarose and
(Table 1). The production and recovery yield of L-AHG purified by silica gel and Bio-Gel P2 size-exclusion chromatographies
after hydrolyses and silica gel chromatography was 7.7 %
on the basis of the initial agarose amount, but considering NMR spectra of purified L-AHG
silica gel chromatography only, the recovery yield of L-
AHG was 50.5 %, as calculated on the basis of the initial The 1 H–13 C NMR spectrum of L-AHG, which was produced
amount of L-AHG in the reaction product produced by from agarose and then purified in this study, is shown in
SdNABH. The partially purified L-AHG sample was sepa- Fig. 4. As shown in Table 2, the 1 H and 13 C chemical shifts
rated into three spots on TLC in processing step 5 of the were compared with those of L-AHG and agarobiose in the
chromatograph in Fig. 1. literature (Sugano et al. 1994; Miller et al. 1982), respectively.
The partially purified L-AHG was further purified by Although the same solvent and reference, D2O and 3-(trime-
size-exclusion chromatography by using Bio-Gel P-2 with thylsilyl)-propionic-2,2,3,3-d4 acid, respectively, were used,
a molecular cutoff size of 1,800 Da. When the fractions our measured 1 H chemical shifts differed −0.1 ppm in com-
showing only the spot of L-AHG were pooled, its purity parison with those of literature values. Since all the proton
of L-AHG was 95.6 %, with a purification fold of 1.7. The peaks exhibited the same shifts in our spectrum, and the 1 H
recovery yield of L-AHG in Bio-Gel P-2 chromatography chemical shifts matched well with the published values, the 1
was 51.8 %. As shown in processing step 6 on the TLC in H chemical shifts of −0.1 ppm were considered only as
Fig. 1, the final purified product showed only the spot of L-
AHG. Consequently, through the hydrolysis and purifica-
tion processes, 4.0 g of L-AHG with a purity of 95.6 %
could be obtained from 100 g of agarose.

GC-mass spectrum of purified L-AHG

To quantify and identify the purified L-AHG, the total ion


chromatograms (TICs) of the purified L-AHG was com-
pared with that of D-AHG, as shown in Fig. 3. Both the
mass spectra of L- and D-AHG matched exactly as they
appeared as one major peak at 20.67 min and two minor
peaks at 24.15 and 27.35 min in the TIC. The fragmentation
of the mass spectra into the major and minor peaks may be
atribtued to the structural instability of anhydrosugars (Chen
et al. 2005; Yang et al. 2009). Since AHG can also be
converted into its hydrate form, L-AHG can be detected in Fig. 4 1H-13C NMR spectrum of the purified L-AHG at 900 MHz.
hydrate form as well as in aldehyde form (Ducatti et al. The purified L-AHG was dissolved in D2O, and 3-(trimethylsilyl)
2011). propionic-2,2,3,3-d4 acid (sodium salt) was used as the standard
Appl Microbiol Biotechnol (2013) 97:2961–2970 2967

Table 2 1H- and 13 C chemical 1 13


shifts from the NMR spectros- H chemical shift (PPM) C chemical shift (PPM)
copy of the L-AHG produced
and purified in this study Measured Published Difference Measured Published Difference

H-1 5.00 5.10 −0.10 92.1 91.4 0.7


H-2 3.61 3.70 −0.10 75.0 74.2 0.8
H-3 3.94 4.02 −0.08 86.0 84.3 1.7
The 1 H and 13 C chemical shifts
of the L-AHG purified in this H-4 4.21 4.30 −0.09 78.8 87.1 −8.3
study were compared with those H-5 4.25 4.34 −0.09 80.2 76.7 3.5
of L-AHG (Sugano et al. 1994) H-6 3.84 3.93 −0.09 74.9 74.5 0.4
and agarobise (Miller et al. H-6′ 4.01 4.11 −0.10
1982) in the literature

referencing issue. Moreover, the 13 C chemical shifts of the B16F10 melanoma cells induced for melanin production
purified L-AHG showed good agreement with the previously were cultured in the presence of neoagarobiose, galac-
reported values except for C-4 assignment (Miller et al. 1982). tose, D-AHG (commercial), or L-AHG. For this experi-
ment, arbutin, a well-known skin whitening agent, was
Polarimetry of purified L-AHG used as the positive control (Maeda and Fukuda 1996).
As shown in Fig. 6, the extracellular melanin concen-
The specific rotations (½a25 tration of cells treated with 100 μg/ mL L-AHG was
D ) of the L-AHG produced and
purified in this study were measured by using a polar- significantly lower than those of the cells treated with
the same concentration of arbutin, neoagarobiose, or D-
imeter. As shown in Table 3, the ½a25 D value of L-AHG AHG. In particular, the extracellular melanin concentra-
was measured as −27.96 mL/dm/g and was distinct from
tion of the cells treated with 100 μg/mL of L-AHG was
that of D-AHG, +27.56 mL/dm/g. The ½a25 D value of the only 23.9 % that of cells treated with 100 nM α-MSH.
purified L-AHG in this study also agreed well with the Visual examination of the cell culture media revealed
previously reported value (Araki and Hirase 1953). the same results, showing the brightest color in the L-
AHG-treated sample. These results suggested that treat-
Cytotoxicity of L-AHG and other saccharides ment with L-AHG strongly suppressed melanin produc-
tion in B16F10 melanoma cells.
We evaluated the cytotoxic effects of neoagarobiose, galac-
tose, D-AHG, and L-AHG through proliferation assays us-
ing B16F10 and RAW264.7 cells. Figure 5 shows that the Anti-inflammatory activity of L-AHG
24-h treatment with neoagarobiose, galactose, D-AHG, and
L-AHG at concentrations up to 100 and 200 μg/mL did not The cellular nitrite level increases considerably under
significantly inhibit the growth of B16F10 cells and inflammatory conditions (Lee et al. 2003). To investi-
RAW264.7 cells, respectively. gate the possible anti-inflammatory effect of L-AHG,
we measured the nitrite levels in the culture media of
RAW264.7 cells that were stimulated by LPS in the
Skin whitening activity of L-AHG
presence or absence of neoagarobiose, galactose, D-
AHG, or L-AHG (all at 50, 100, and 200 μg/ mL).
To evaluate the possible skin whitening activity of the L-
Nitrite production was significantly (p<0.05) suppressed
AHG obtained and purified in this study, α-MSH-treated
by 100 and 200 μg/ mL L-AHG (Fig. 7). The nitrite
Table 3 Specific rotations of L-AHG and D-AHG levels in the culture media of the cells treated with
100 and 200 μg/ mL were, respectively, 64.5 and
Sample ½a25
D
a
Reference 38.8 % of those in the LPS-treated controls. D-AHG
showed a nitrite-suppressing effect only at 200 μg/mL,
L-AHG −27.96 −25.20b
but its effect was significantly lower than that of L-
D-AHG +27.56 NA
AHG. Other saccharides, such as neoagarobiose and
NA not applicable galactose, did not induce any significant reduction in
a
½a25
D was expressed in mL/dm/g
the nitrite production of RAW264.7 cells. These results
b
The specific rotation of L-AHG was compared with that of L-AHG in indicate that L-AHG can be used as an anti-
the literature (Araki et al. 1953) inflammatory agent.
2968 Appl Microbiol Biotechnol (2013) 97:2961–2970

Fig. 5 Cytotoxic effects of neoagarobiose (NAB), galactose, D-AHG, determined by the MTT assay. Results were expressed as cell viability
and L-AHG on a B16F10 cells and b RAW264.7 cells. Cells were relative to the untreated control. Data of three independent experiments
treated with neoagarobiose, galactose, D-AHG, or L-AHG at the indicated were expressed as mean ± standard deviation. Asterisk indicates a signif-
concentrations in 10 % FBS DMEM for 24 h. Proliferation of cells was icant difference (p<0.05) compared with the untreated control

Discussion various DPs, but Aga50D predominantly forms neoagaro-


biose (Kim et al. 2010a, b). Therefore, it can be considered
Strong acids, such as hydrochloric acid, sulfuric acid, and an effective enzyme for the production of neoagarobiose or
trifluoroacetic acid, at high temperatures (80–121 °C) are the generation of L-AHG when combined with an NABH.
widely used to prepare agaro-oligosaccharides or AHG from The combination of pre-hydrolysis by acetic acid for the
agarose or agar (Jol et al. 1999; Kim et al. 2010a, b). L- production of agaro-oligosaccharides and enzymatic hydro-
AHG from agarose is known to be converted to HMF during lysis of agaro-oligosaccharides by Aga50D was effective in
hydrolysis by a strong acid due to the instability of its producing neoagarobiose from agarose.
reducing terminal (Kim et al. 2010a, b; Quemener et al. Although β-agarases are prevalent in microorganisms, α-
1995; Stevenson and Furneaux 1991; Yang et al. 2009). In agarases that cleave α-1,3 glycosidic linkages are rare (Fu and
the present study, we conducted acid pre-hydrolysis by Kim 2010; Hassairi et al. 2001). To produce L-AHG from
using acetic acid at a relatively mild temperature (80 °C) agarose or neoagarobiose, the α-1,3 linkages between L-AHG
to minimize the degradation of L-AHG into HMF and other and galactose should be hydrolyzed by an α-agarase or a
overreaction products. Aga50D is a unique β-agarase since NABH. NABHs that preferentially or solely act on neoagar-
most β-agarases release neoagaro-oligosaccharides with obiose to produce L-AHG have been reported in Pseudomo-
nas atlantica (currently P. atlantica), Vibrio sp., and

Fig. 6 Effects of arbutin, neoagarobiose (NAB), galactose, D-AHG,


and L-AHG on α-MSH-induced melanin production in B16F10 mel- Fig. 7 Effects of neoagarobiose (NAB), galactose, D-AHG, and L-
anoma cells. Data of three independent experiments were expressed as AHG on LPS-induced production of nitrite in RAW 264.7 cells. Data
mean ± standard deviation. Asterisks indicate a significant inhibition of of three independent experiments were expressed as mean ± standard
melanin content compared to that in the cells treated with α-MSH deviation. Asterisks indicate significant differences relative to the LPS-
alone (p<0.05) treated group (p<0.05)
Appl Microbiol Biotechnol (2013) 97:2961–2970 2969

Cytophaga flevensis (Day and Yaphe 1975; Sugano et al. Chen H-M, Zheng L, Yan X-J (2005) The preparation and bioactivity
1994; Van Der Meulen and Harder 1976). However, the research of agaro-oligosaccharides. Food Technol Biotechnol
43:29–36
industrial application of these NABHs was not possible since Chen H, Yan X, Zhu P, Lin J (2006) Antioxidant activity and hepato-
their amino acid sequences were not yet fully discovered. protective potential of agaro-oligosaccharides in vitro and in vivo.
Overexpresssion of a recombinant NABH was recently per- Nutr J 5:1–12
formed for the first time, and this NABH originating from S. Day DF, Yaphe W (1975) Enzymatic hydrolysis of agar: purification
and charaterization of neoagarobiose hyrolase and ρ-nitrophenyl
degradans 2-40 was characterized to attack the α-1,3 glyco- α-galactoside hydrolase. Can J Microbiol 21:1512–1518
sidic linkage from the non-reducing end of a neoagaro- Ding AH, Nathan CF, Stuehr DJ (1988) Release of reactive nitrogen
oligosaccharide and neoagarobiose, resulting in the formation intermediates and reactive oxygen intermediates from mouse
of predominantly L-AHG (Ha et al. 2011; Lee et al. 2009). peritoneal macrophages: comparison of activating cytokines and
evidence for independent production. J Immunol 141:2407–2412
This recombinant NABH is valuable since it can be used as Dooley TP (1997) Topical skin depigmentation agents: current prod-
the key enzyme in the enzymatic saccharification of agarose ucts and discovery of novel inhibitors of melanogenesis. J Der-
into monomeric sugars (L-AHG and galactose) and in the matol Treat 8:275–279
production of L-AHG from neoagaro-oligosaccharides. Ducatti DRB, Colodi FG, Goncalves AG, Duarte MER, Noseda MD
(2011) Production of agaro- and carra-oligosaccharides by partial
Desirable skin whitening agents should inhibit the secre- acid hydrolysis of galactans. Bras J Pharmacogn 21:296–304
tion of melanin in melanosomes by acting specifically to Ekborg NA, Taylor LE, Longmire AG, Henrissat B, Weiner RM,
reduce the synthesis or activity of tyrosinase; they should also Hutcheson SW (2006) Genomic and proteomic analyses of the
exhibit low cytotoxicity and no mutagenicity (Dooley 1997). agarolytic system expressed by Saccharophagus degradans 2-40.
Appl Environ Microbiol 72:3396–3405
In particular, the melanin-suppressing effect of L-AHG was Fu XT, Kim SM (2010) Agarase: review of major sources, categories,
significantly (p < 0.05) higher than that of arbutin. The purification method, enzyme characteristics and applications. Mar
melanin-suppressing effect of neoagarobiose, which has been Drugs 8:200–218
reported previously (Kobayashi et al. 1997), was also shown Ha SC, Lee S, Lee J, Kim HT, Ko H-J, Kim KH, Choi I-G (2011)
Crystal structure of a key enzyme in the agarolytic pathway, α-
in this study. Our findings of no activity of galactose itself and neoagarobiose hydrolase from Saccharophagus degradans 2-40.
the lower activity of neoagarobiose than that of L-AHG on Biochem Biophys Res Commun 412:238–244
melanin production provide experimental evidence for the Hassairi I, Amar RB, Nonus M, Gupta BB (2001) Production and
first time that the bioactivity of neoagarobiose may mainly separation of α-agarase from Altermonas agarlyticus strain
GJ1B. Bioresour Technol 79:47–51
be attributable to the L-AHG moiety of neoagarobiose but not Jol CN, Neiss TG, Penninkhof B, Rudolph B, De Ruiter GA (1999) A
to the galactose moiety. novel high-performance anion-exchange chromatographic meth-
In conclusion, agarose, which is composed of galactose and od for the analysis of carrageenans and agars containing 3,6-
L-AHG, is the main constituent of red macroalgal biomass. In anhydrogalactose. Anal Biochem 268:213–222
Kim C, Ryu HJ, Kim SH, Yoon J-J, Kim HS, Kim YJ (2010a) Acidity
this study, agrose was pre-hydrolyzed into agaro- tunable ionic liquids as catalysts for conversion of agar into mixed
oligosaccharides by acetic acid, and the agaro-oligosaccharides sugars. Bull Korean Chem Soc 31:511–514
were hydrolyzed by an exo-type β-agarase and an NABH to Kim HT, Lee S, Lee D, Kim H-S, Bang W-G, Kim KH, Choi I-G (2010b)
obtain L-AHG. The enzyme reaction product containing L-AHG Overexpression and molecular characterization of Aga50D from
Saccharophagus degradans 2-40: an exo-type β-agarase producing
was sequentially purified by adsorption and gel permeation neoagarobiose. Appl Microbiol Biotechnol 86:227–234
chromatographies. The purified L-AHG exhibited significant Kitamura Y, Abe Y, Yasui T (1991) Metabolism of levoglucosan (1,6-
skin whitening and anti-inflammatory activities. This is the first anhydro-β-D-glucopyranose) in microorganisms. Agric Biol
report on the recombinant enzyme-based production of L-AHG Chem 55:515–521
Kobayashi R, Takisada M, Suzuki T, Kirimura K, Usami S (1997)
and its purification and biological activities. These results could Neoagarobiose as a novel moisturizer with whitening effect. Bio-
initiate the industrial production of L-AHG from red macroalgae sci Biotechnol Biochem 61:162–163
as an important health-benefiting agent. Kuhn A, Yu S, Giffhorn F (2006) Catabolism of 1,5-anhydro-D-fruc-
tose in Sinorhizobium morelense S-30.7.5: discovery, character-
ization, and overexpression of a new 1,5-anhydro-D-fructose
Acknowledgments This work was supported by the National Re-
reductase and its application in sugar analysis and rare sugar
search Foundation (NRF) Grant (2011-0015629) funded by the Korean synthesis. Appl Environ Microbiol 72:1248–1257
Government (MEST) and the Korea University Grant. Facility support
Lee AK, Sung SH, Kim YC, Kim SG (2003) Inhibition of
at Korea University Food Safety Hall for the Institute of Biomedical
lipopolysaccharide-inducible nitric oxide synthase, TNF-α and
Science and Food Safety is also acknowledged. COX-2 expression by sauchinone effects on I-κBα phosphoryla-
tion, C/EBP and AP-1 activation. Br J Pharmacol 139:11–20
Lee S, Lee JY, Ha SC, Jung J, Shin DH, Kim KH, Choi I-G (2009)
References Crystallization and preliminary X-ray analysis of neoagarobiose
hydrolase from Saccharophagus degradans 2-40. Acta Crystal-
logr Sect F-Struct Biolo Cryst Commun 65:1299–1301
Araki C, Hirase S (1953) Studies on the chemical constitution of agar- Maeda K, Fukuda M (1996) Arbutin: mechanism of its depigmenting
agar. XV. Exhaustive mercaptolyses of agar-agar. Bull Chem Soc action in human melanocyte culture. J Pharmacol Exp Ther
Jpn 26:463–467 276:765–769
2970 Appl Microbiol Biotechnol (2013) 97:2961–2970

Miller IJ, Wong H, Newman RH (1982) A 13C N.M.R. study of some Tomono J, Sagawa H, Kato I (2009) Agarase and gene thereof. US
disaccharides from algal polysaccharides. Aust J Chem 35:853– Patent 7622291 B2
856 Tsuboi T, Kondoh H, Hiratsuka J, Mishima Y (1998) Enhanced mela-
Penney KB, Smith CJ, Allen JC (1984) Depigmenting action of hy- nogenesis induced by tyrosinase gene-transfer increases boron-
droquinone depends on disruption of fundamental cell processes. uptake and killing effect of boron neutron capture therapy for
J Investig Dermatol 82:308–310 amelanotic melanoma. Pigment Cell Res 11:275–282
Quemener B, Lahaye M, Metro F (1995) Assessment of methanolysis Urabe K, Nakayama J, Hori Y (1998) Mixed epidermal and dermal
for the determination of composite sugars of gelling carageenans hypermelanoses. In: Norlund JJ, Boissy RE, Hearing VJ (eds) The
and agarose by HPLC. Carbohydr Res 266:53–64 pigmentary system: physiology and pathophysiology. Oxford
Rees DA (1961) Enzymic synthesis of 3:6-anhydro-L-galactose within University Press, New York, pp 909–911
porphyran from L-galactose 6-sulphate units. Biochem J 81:347– Van Der Meulen HJ, Harder W (1976) Characterization of neoagarotetra-
352 ase and neoagarobiase of Cytophaga flevensis. Antonie van Leeu-
Sekkal M, Huvenne J-P, Legrand P, Sombret B, Mollet J-C, Mouradi- wenhoek 42:81–94
Givernaud A, Verdus M-C (1993) Direct structural identification Vera J, Alvarez R, Murano E, Slebe JC, Leon O (1998) Identification
of polysaccharides from red algae by FTIR microspectrometry. I: of a marine agarolytic Pseudoalteromonas isolate and character-
localization of agar in Gracilaria verrucosa sections. Mikrochim ization of its extracellular agarase. Appl Environ Microbiol
Acta 112:1–10 64:4378–4383
Shin MH, Lee DY, Wohlgemuth G, Choi I-G, Fiehn O, Kim KH (2010) Yamaji K, Sarker KP, Maruyama I, Hizukuri S (2002) Antioxidant
Global metabolite profiling of agarose degradation by Saccharo- effects of 1,5-anhydro-D-fructose, a new natural sugar, in vitro.
phagus degradans 2-40. New Biotechnol 27:156–168 Planta Med 68:16–19
Stevenson TT, Furneaux RH (1991) Chemical methods for the analysis Yang B, Yu G, Zhao X, Jiao G, Ren S, Chai W (2009) Mechanism of
of sulfated galactans from red algae. Carbohydr Res 210:277–298 mild acid hydrolysis of galactan polysaccharides with highly
Su J-C, Hassid WZ (1962) Carbohydrates and nucleotides in the red ordered disaccharide repeats leading to a complete series of ex-
algal Porphyra perforata. I. Isolation and identification of carbo- clusively odd-numbered oligosaccharides. FEBS J 276:2125–
hydrates. Biochem 3:468–474 2137
Sugano Y, Kodama H, Terada I, Yamazaki Y, Noma M (1994) Purification Yun EJ, Shin MH, Yoon J-J, Kim YJ, Choi I-G, Kim KH (2011)
and characterization of a novel enzyme, α-neoagarooligosaccharide Production of 3,6-anhydro-L-galactose from agarose by agarolytic
hydrolase (α-NAOS hydrolase), from a marine bacterium, Vibrio sp. enzymes of Saccharophagus degradans 2-40. Process Biochem
strain JT0107. J Bacteriol 176:6812–6818 46:88–93

You might also like