Está en la página 1de 12

Appl Microbiol Biotechnol (2012) 93:7182

DOI 10.1007/s00253-011-3666-8

MINI-REVIEW

Algal fucoidan: structural and size-dependent bioactivities


and their perspectives
V. K. Morya & Jungeun Kim & Eun-Ki Kim

Received: 21 July 2011 / Revised: 3 October 2011 / Accepted: 22 October 2011 / Published online: 17 November 2011
# Springer-Verlag 2011

Abstract Fucoidan is a complex-sulfated polysaccharide


distributed in various marine organisms, and the brown algae
are reported as the major producer. The fucoidan is important
for their high bioactive properties, like antibacterial, anticoagulant, antiviral, anti-tumor, etc., and many more to be
explored. There is a strong archival support for the bioactivity
and promising properties of this molecule, which creates a
hope for this molecule as future drug against thrombosis and
some kind of cancers. Reports other than the above bioactive
properties have also been a matter of interest for the design of
signal or enzyme-arrested new class of drugs. In the past three
decades, the research on isolation, molecular characterization,
and screening of biological applications has significantly
increased. One major issue associated with this molecule is the
higher size and seasonal variation in their chemical composition; to resolve the issue and maintain its bioactivity, a
prioritized and literal hydrolysis process is required to be
developed. Here, in this mini-review, we have tried to
summarize the algal fucoidan research and the bioactivities
influenced by their molecular size.
Keywords Fucoidan . Brown algae . Antioxidant .
Anticoagulant . Antiviral . Anti-tumoral

Introduction
Diversity creates a variety of biomolecule, and these
molecules are responsible for this diversity, either directly
V. K. Morya : J. Kim : E.-K. Kim (*)
National Research Lab of Bioactive Materials, Dept. Biological
Engineering, Inha University,
Incheon, South Korea 402-751
e-mail: ekkim@inha.ac.kr

or indirectly. The secondary metabolites have been used as


a biologically active agent in different purposes like
pharmaceuticals, cosmetics, nutraceuticals, pesticide, antibiotic, etc. (Kim et al. 2007, 2010a, b). Various carbohydrates are being regarded by researchers as hypnotic
because of their diversity and respective bioactivities. The
saccharides are naturally assembled from monosaccharides
having different types of linkages, at varying positions in
the carbohydrate ring, forming straight chain or branched
polymers. Further modifications such as alkylation, phosphorylation, and sulfation provide an additional structural
complexity and peculiar activities to the already diverse
compounds. The high degree of variability among biologically relevant sulfated carbohydrates is attributed to a
number of factors. There is inconsistency among sulfated
monosaccharides (i.e., glucose (Glc), galactose (Gal), the
corresponding N-acetyl (NAc) amines GlcNAc, GalNAc,
and mannose among others), the total number of sulfate
moieties, and the hydroxyl(s) to which the sulfate group(s)
are linked (i.e., 2-O, 3-O, 4-O, and 6-O sulfates);
furthermore, the varied structure of the underlying oligosaccharide moiety. An important consequence of this
structural diversity is that each unique structure has the
potential to be recognized by an individual receptor,
making sulfated oligosaccharides ideal for carrying information in complex biological systems.
Among sulfated polysaccharide, fucoidan is extensively
explored for its medicinal properties in the past two decades
and being isolated from various species of brown seaweed.
Fucoidan can exist essentially in two distinct forms of
glycosaminoglycans: F-fucoidan which is more than 95%
of the fucoidans in seaweeds andcomposed of sulfated
esters of l-fucose and U-fucoidan which is approximately
20% glucuronic acid. Depending on the source of algae, the
sulfated polysaccharide does not only mainly consist of L-

72

fucose units but can also contain other sugars such as


galactose, mannose, xylose, or uronic acid and sometimes
proteins (Pomin et al. 2005; Venugopal 2009). Sulfated
fucan is the characteristic feature of the fucoidans. These
compounds in total comprise less than 10% in composition
(Li and Xu 2007; Pomin et al. 2005).
Fucoidan occupied about more than eight hundred citation
in PUBMED since 1918 after the first isolation, by Kylin. The
research boom in the field of fucoidan is a story of last two
decades, about 485 papers between 2000 and 2010 while 310
papers during 19902000. The figures show that the
passionate inclination of scientific community in this field is
rapid and recent. The hope with this group of polysaccharide
attracts researchers because of their numerous bioactive
properties. The new arena for green technology with a
molecule is about to come with promising fucoidans.
Various reports show that this polymer has numerous
bioactive properties like antibacterial, anticoagulant, antiviral,
and anti-tumor, etc., and many more to be explored. In recent
years, the interest in fucoidan from different sources, its
structure and its possible biological applications has increased.
For the past three decades, the bioactivities of fucoidans have
been extensively studied due to their various biological
activities, including anti-tumor, antivirus, immunomodulatory, anticoagulant, anti-inflammatory, antioxidant, antithrombotic, anticomplementary properties, activity against
hepatopathy, blood lipids reducing, anti-allergic, uropathy,
anti-adhesive, antineoplastic, renal pathy, gastric protective
effects, and therapeutic potential in surgery and hypoxic
ischemic brain injury, (Cumashi et al. 2007; Tanaka et al.
2011; Li et al. 2008). Our lab is focused on applicability of
fucoidan in cosmetics as depigmenting agent (Jin et al. 2006;
Kim et al. 2010a, b, 2011a, b). Development of non-arbutinbased skin whitening cosmetics without any side effect is a
challenge for the researcher (Jin et al. 2005; Lam et al. 2008;
Babitha et al. 2009; Lam et al. 2011). We successfully
explored the depimentation effect of fucoidan, in vitro. We
are trying to modify the molecule chemically and biologically to enhance the depigmenting properties of the fucoidan
(Jin et al. 2005, 2006; Kim et al. 2011a).
In contrast to other sulfated polysaccharides, the fucoidans are prevalently distributed among marine organisms
like brown algae and echinoderms. The demand of this
polysaccharide will increase with the increase of exploration of bioactivities. Therefore, it is required to explore
more organisms for isolation of fucoidan. The aim of this
review is to discuss the algal fucoidan and the bioactivities
influenced by their molecular weight.
Source
Among various sources of fucoidan, brown algae, sea
urchin, and sea cucumber were well explored for isolation

Appl Microbiol Biotechnol (2012) 93:7182

and evaluation of bioactivity (Holtkamp et al. 2009). In the


past two decades, different brown algae were analyzed for
their content of fucoidans including Adenocystis utricularis
(Ponce et al. 2003), Analipus japonicus (Bilan et al. 2007),
Ascophyllum nodosum (Medcalf and Larsen 1977), Chorda
filum (Bakunina et al. 2002; Chizhov et al. 1999),
Cladosiphon okamuranus (Sakai et al. 2003b), Cystoseira
indica (Mandal et al. 2007), Dictyota menstrualis
(Albuquerque et al. 2004), Fucus distichus (Bilan et al.
2005), Fucus evanescens (Bakunina et al. 2002), Fucus
serratus (Bilan et al. 2006), Fucus vesiculosus (Wu et al.
2004), Hizikia fusiforme (Li et al. 2006), Kjellmaniella
crassifolia (Bilan et al. 2006), Laminaria cichorioides
(Anastyuk et al. 2010), Laminaria japonica (Huang et al.
2010), Lessonia vadosa (Chanda and Matsuhiro 2008),
Lobophora variegate (Medeiros et al. 2008), Padina
gymnospora (Silva et al. 2005), Pelvetia canaliculata
(Descamps et al. 2006a), Saccharina latissima (Bilan et
al. 2010), Sargassum stenophyllum (Duarte et al. 2001),
Stichopus japonicas (Zhang et al. 2010), Stoechospermum
marginatum (Adhikari et al. 2006), and Undaria pinnatifida
(Chen et al. 2009). The bioactivity of fucoidan depends on
nature and extent for sulphation, which must be taken care
during extraction. Because of harshness of extraction method,
the sulphation pattern may be destroyed and the bioactivity
can thus be lost (Qiu et al. 2006). In most of the reported
research, extraction and characterization were evaluated up
to bioactivity screening while until date fucoidan only from
F. vesiculosus (Bladder wrack) are available commercially,
which embark to explore the other possibilities. The
extraction method of the supplier is adapted to the method
of Black (1954). The problem of this fucoidan is that it is
very crude and differs in color and constitution between
different batches. The bioactivity of this molecule is vast and
promising for development of new class of drug or
nutraceuticals. Therefore, there is a strong need for a
comprehensive study on its size dependent bioactivity and
precise generation of specific size low molecular weighted
fucoidan. Therefore, this molecule needs more attention for
developing better methodology for extraction and functionaries. About 1,800 species of brown algae are known
worldwide and most of them are from relatively colder
marine habitat. Very few numbers of species from the
Laminariales and Fucales are being utilized commercially
for biomass and other uses. The world production of
seaweeds was some 8 m ton in 2003. The seaweeds are
used in the production of food, feed, chemicals, cosmetics
and pharmaceutical products. Seaweeds are mainly produced
for these end uses in Asian countries such as China, the
Philippines, North and South Korea, Japan, and Indonesia
(Reith et al. 2005). The total market of the brown algae is
estimated to be approximately about $100 million. In spite of
the persistent increase in interest regarding brown algae as

Appl Microbiol Biotechnol (2012) 93:7182

sources of fucoidans, basic knowledge of mechanisms and


metabolic regulation of this group of polysaccharides is in a
juvenile period. Some studies have mentioned the species,
age of sample, thallus composition and season and method
of extraction as the probable reason of diversity of fucoidan
structure (Skriptsova et al. 2010; Usov et al. 2005). Maximal
content was found in sporophyll, which was several times
higher than any other part of the thallus. Moreover,
publications establishing a correlation between fucoidan
content and seasonality generally report that the highest
amount of the polysaccharide is obtained during the
reproductive stage (Skriptsova et al. 2010). Reproduction,
however, can be a long process involving several stages.
Within this period, the physical and morphological state of
the alga varies and physiological and biochemical processes
undoubtedly accompany such changes (Aguilar-Rosas et al.
2004). This figure may augment in the coming years because
of the wonder molecule which may revolutionize the marine
biotechnology, to meet the needs of the market. In this
review, we have tried to compile various aspects of this
wonder molecule, which may further be explored.
Structure
Since 1913, after the discovery of fucoidan by Kylin, the
structures of fucoidans from different brown seaweeds have
been investigated and deduced. The results were promising as
well as paradoxical, in terms of composition. The diversity
among distribution is not only its interesting feature but also
its divergence of structure. Fucoidans from various species of
brown seaweed has been investigated and still in progress.
One of the most studied fucoidans isolated from F.
vesiculosus, has simple chemical compositions, mainly being
composed of fucose and sulfate. However, the chemical
compositions of most fucoidans are complex. Besides fucose
and sulfate, they also contain other monosaccharides
(mannose, galactose, glucose, xylose, etc.) and uronic acids,
even acetyl groups and protein. Furthermore, the structures
of fucoidans from different brown algae vary from species to
species. Despite all that, some fucoidans structures or their
structural backbones have been elucidated.
Fucoidan prepared from F. vesiculosus is commercially
available at present. It is composed of 44.1% fucose, 26.3%
sulfate, and 31.1% ash, with a little amount of a aminoglucose (Nishino et al. 1994). Later, it was discovered that
the 1,2--fucose is the main component unit and most of the
sulfate groups were located at C-4 position of the fucose
units (Anno et al. 1970). The suggested structural models of
fucoidan of F. vesiculosus based on GC/MS data of
methylation reveal that the core region of fucoidan was
primarily a polymer of -(13) linked fucose with sulfate
groups substituted at the C-4 position on some of the fucose
residues. Within the chain at every two to three residues, the

73

fucose is attached to form branched point. The structural


variability was observed by different workers because of the
difference of preparation method and their methylation
methods for analysis (Patankar et al. 1993).
The 40-year-old structural model fucoidan was finally
revised in 1993 by Patankar, based on GC/MS analysis. In the
last two decades, research have isolated and characterized
fucoidan from various sources like F. evanescens, F. distichus,
F. serratus L., S. marginatum, P. gymnospora, etc., and
resolved the structures (Bilan et al. 2006; Adhikari et al.
2006). The fucoidan from other than F. vesiculosus was
found to be complex chemical, instead of homopolymeric
they show heteropolymer of fucose, galactose, mannose,
glucose, xylose, and glucuronic acid (Jingjing et al. 2009;
Skriptsova et al. 2010). The fucoidan from Ecklonia kurome
and C. filum is structurally 13-linked fucopyranose with
branched structure. Fucoidan isolated from C. filum shows
highly branched poly--(13)-fucopyranoside backbone
(Chizhov et al. 1999). While 13-linked fucopyranose is
dominant in nature, 12- or 14-linked fucopyranose was
also reported from some brown algae, with some unusual
branching. The structure of fucoidan from A. nodosum was
[3)--L-Fuc(2SO3-)-(14)--L-Fuc(2,3diSO3-)-(1]n with
highly branched core region with primarily -(13)-fucosyl
residues and a few -(14)-linkages, while sulfate groups
were at positions 2 and/or 4 (Chevolot et al. 2001; Marais
and Joseleau 2001). The structural properties of fucoidan
from different algal sources are summarized in Table 1.
It has been discussed that the fucoidans are a heterogeneous assemblage with regard to their monosaccharide
composition, number of sulfate, and acetyl groups and
moreover, molecular mass. Fucans can differ in structure
among algal species and can vary even within the same
species. Because of the heterogeneity in structures within
the seaweed, differing extraction conditions used by
researchers can give rise to the isolation of distinct fucan
forms, thus need to establish any systematic correspondence between structure and algal order (Li et al. 2008). All
fucoidans show a wide spectrum of biological action (Fitton
2005), which seems to be determined by their degree of
sulfatation, fine structure, monosaccharide composition,
and molecular weight. In general, Fucans have been
classified into two groups. One group includes the fucans
from Laminaria saccharina, Laminaria digitata, A. japonicus, C. okamuranus, and C. filum that have their central
chains composed of (13-L-fucopyranose residues. A
second group included fucans isolated from A. nodosum
and Fucus species that have their central chains composed
of repeating (13)- and (14 residues (Jiao et al. 2011;
Ushakova et al. 2009; Li et al. 2008). However, many
studies have revealed more complex fucans, some with
branching structures. Fucoidans of various structures are of
doubtless interest for a comparative study of differential

74

Appl Microbiol Biotechnol (2012) 93:7182

Table 1 Structural properties of some fucoidans from different sources


Sr. Source

Chemical composition

Adenocytis utricularis

Analipus japonicus

Ascophyllum nodosum

A. nodosum

Chorda filum

Dictyota menstrualis

Ecklonia kurome

Fucose, galactose, mannose,


and sulfate
Fucose, xylose, galactose, mannose,
and sulfate
Fucose (49%), xylose (10%),
GlcA (11%), and sulfate
Fucose (49%), xylose (10%),
GlcA (11%), and sulfate
Fucose/xylose/mannose/glcose/
galtose (1.0:0.14:0.15:0.40:0.10)
andsulfate
Fucose/xylose/uronic acid/
galactose/
sulfate (1/0.8/0.7/0.8/0.4) and
(1/0.3/0.4/1.5/1.3)
Fucose, galactose, mannose,
xylose, GlcA, and sulfate
Fucose/sulfate/acetate (1/1.21/0.08)

Fucoidan structure

Reference

3 (4Fucp) and 1 (2Fucp) per 10 (13)-a-L


-Fucp(2/4SO3-) residues
(13)--L-Fucp and a few (14)--L-Fucp with
(13)--L-(2and/or 4 Fucp)
3)--L-Fucp(2SO3-)-(14)--L-Fucp
(2,3-diSO3-)-(1
-[3)--L-Fucp-(1-]33)--L-Fucp
(2Fucp)-(1

Bilan et al. (2007)


Marais and Joseleau (2001)
Chevolot et al. (2001)
Chizhov et al. (1999)

Bilan et al. (2007)


3)--L-Fucp-(2,4-diSO3)-(14)--L-Fucp
-(2SO3)-(1
Bilan et al. (2006)
9 Fucus evanescens c. Ag. Fucose/sulfate/acetate (1/1.23/0.36) 3)--L-Fucp(2SO3-)-(14)--L-Fucp
(2SO3-)-(1
Bilan et al. (2006)
10 Fucus serratus L.
Fucose/sulfate/acetate (1/1/0.1)
3)--L-Fucp(2R1,4R2)-(14)--L-Fucp(2SO3)
-(1a. (50%): R1 =SO3, R2 =H b. (50%): R1 =H,
R2 =-L-Fucp-(14)--Fucp(2SO3)-(13)--L
-Fucp(2SO3)-(1
11 Fucus vesiculosus
Fucose and sulfate
8

F. distichus

12 Himanthalia lorea and


Bifurcaria bifurcate
13 Hizikia fusiforme
14 Laminaria angustata

Fucose, xylose, GlcA, and sulfate


Fucose, galactose, mannose,
xylose, GlcA, and sulfate
Fucose/galactose/sulfate (9/1/9)

(12)- and (13)-linked fucose residues with sulfation


at C-4
Li et al. (2006)
2)--D-Man(1and4)--D-GlcA(1alternately,
while a little4)--D-Gal(1

16 Lessonia vadosa

Fucose, xylose, mannose, glcose,


3)--L-Fucp(4SO3-)-(1and additionally3)
and galctose (30.8:1.4:2.1:0.9:7.9) --L-Fucp(4SO3- or2Fucp)-(1
Fucose/sulfate (1/1.12)

17 Macrocytis pyrifera

Fucose/galactose (18/1) and sulfate

18 Padina pavonia

Fucose, xylose, mannose, glucose, 12 linked fucose; 14 linked GlcA, mannose


galactose, and sulfate
and glucose
Fucose/galactose (10/1) and sulfate

15 Laminaria saccharina

19 Pelvetia wrightii

20 Sargassum stenophyllum Fucose, galactose, mannose, GlcA,


glucose, xylose, and sulfate
21 Spatoglossum
Fucose/xylose/galactose/sulfate
schroederi
(1/0.5/2/2)
22 Stoechospermum
Fucose, galacturonic acid, xylose,
marginatum
galactose, and sulfate
23 Undaria pinnatifida
(Mekabu)

3)--L-Fucp(2/4SO3)-(1and4)
--L-Fucp
(2SO3)-(1
Fucose, galactose, mannose, xylose, 1,3-linked fucose and 1,3-, 1,4-,
and sulfate
and 1,
6-linked galactose

Adhikari et al. (2006)

Lee et al. (2004) and


Kim et al. (2007)

Glc glucose, Gal galactose, NAc N-acetyl amines, Man mannose, GlcA glucuronic acid

and functional biological activities. Thus, the investigation


of content and structural variability of fucoidan, synthesized
by seaweeds at their different life stages, is essential both
for seaweed biology and fucoidan utilization by the medical
industry. Applications of polysaccharide agents in medicine
have associated problems with standardization. One method
of standardization is a definition of the season characterized
by the maximum quantity of fucoidan with a fixed structure
and the highest biological activity (Skriptsova et al. 2010).

Therefore, this wonder molecule needs more extensive


study to establish the mechanisms of complexity in
structure, to produce at the large scale.
Bioactivities of fucoidan
Various researches to have done and many have to do with
fucoidan to evaluate the medicinal and cosmeceutical
potentials (Fitton and Dragar 2006; Holtkamp et al. 2009).

Appl Microbiol Biotechnol (2012) 93:7182

75

The literatures cited on the bioactivity ranges from


anticoagulant (Silva et al. 2005; Alekseyenko et al. 2007;
Gideon and Rengasamy 2008; Kwak et al. 2010; Cho et al.
2010), antiviral (Lapshina et al. 2006; Trinchero et al.
2009), and anti-tumoral to anti-inflammatory (Aisa et al.
2005; Lapshina et al. 2006; Raghavendran et al. 2011).
Furthermore, fucoidan promotes collagen gel contraction by
probably enhancing the surface integrin 21 expression
on fibroblasts and may therefore accelerate wound-healing
processes (Holtkamp et al. 2009). In spite of the mentioned
applications fucoidan shows, some other properties like
anti-mycotoxin (Angulo and Lomonte 2003), immunomodulatory (Raghavendran et al. 2011), antioxidant (Micheline
et al. 2007; Li et al. 2008; Wang et al. 2008; Jing et al.
2009), anticomplementary properties, hypoxic ischemic
brain injury (Luo et al. 2009), cardio protective (Thomes
et al. 2010), acting as an anti-inflammatory in the brain (Del
Bigio et al. 1999), as a neuroprotectant in ischemicreperfusion injury (Uhm et al. 2003), inhibitor of proinflammatory proteins, such as TNF-alpha or nitric oxide
(Mitrasinovic and Murphy 2002), uropathy, renal pathy
(Zhang et al. 2003), and a neuroprotective effect against
dopaminergic neurodegeneration (Luo et al. 2009). The
biological activity of fucoidan depends on its structure
(Boisson-Vidal et al. 2000) and sulphation content (Qiu et
al. 2006). The structural properties are highly governed by
climatic condition and time of collection. To determine and
correlate these complexities in structure of fucoidan contents is one of the most challenging tasks for researches.
According to these activities, fucoidan exhibits a high
pharmaceutical potential with possible applications. Here,

in this review, we tried to summarize some of the biological


activity and size effect of fucoidans reported from different
sources.
Size-dependent bioactivities of fucoidan
This molecule has various biological and medicinal
properties, as discussed above which have been explored
in vitro conditions. A major obstruction to the development
of its medicinal application is the poor knowledge of its
molecular structure and, as a result, of the structural basis of
its biological activities (Mulloy et al. 2000). Since 2000,
various developments have enhanced our knowledge about
fucoidan structures and structural diversity. One major issue
associated with this molecule is its higher size and seasonal
compositional variation; thus, to resolve the issue and
maintain the bioactivity, a prcised hydrolysis process
needs to be developed. Several attempts have been made
to generate oligo-polysaccharide by free radical, mineral
acid, organic acid, and enzymatic hydrolysis of fucoidan
(Table 2). Several attempts have been done by various
groups of researchers to develop a methodology to produce
a low molecular weight fucoidan (LMWF), like physical,
chemical, and enzymatic (Holtkamp et al. 2009). As we
have discussed, the bioactivity is highly dependent on
sulfate group; enzymatic hydrolysis prevents the loss of
sulfate group than the chemical hydrolysis. One wellknown enzyme for fucoidan hydrolysis is fucoidanases or
-L-fucosidases, distributed as extracellular as well as
intracellular among several organisms (Holtkamp et al.
2009). The producer range of the fucoidan-degrading

Table 2 Various methods used to prepare low molecular weight fucoidan and their bioactivities
Applications

Method

Source

Reference

Antithrombotic

Radical

Ascophyllum nodosum

Colliec-Jouault et al. (2003) and


Chabut et al. (2003, 2004)

Radical

A. nodosum

Durand et al. (2008)

Acid

Laminaria japonica

Zhu et al. (2010)

Acid

Egg jelly

Pomin et al. (2005)

Enzyme

Sporophyll of Undaria pinnatifida

Kim et al. (2010a, b)

Anticoagulant
Antioxidant

Gamma irradiation

Sigma fucoidan (Fucus vesiculosus)

Choi et al. (2009)

Antioxidant and anticoagulant

Acid

L. japonica

Wang et al. (2009)

Anticancer

Acid

Sporophyll from U. pinnatifida

Radical

A. nodosum

Alkhatib et al. (2006)

Radical

A. nodosum

Hlawaty et al. (2011)

Radical

A. nodosum

Bachelet et al. (2009)

Radical and acid

Pheophicae cell wall

Changotade et al. (2008)

Acid

F. vesiculosus

Azofeifa et al. (2008)

Wang et al. (2010)


Yang et al. (2008)
Cho et al. (2010)

Radical
Anti-inflammatory

Acid

Lake et al. (2006)


U. pinnatifida

Park et al. (2010)

76

Appl Microbiol Biotechnol (2012) 93:7182

enzymes is wide enough, i.e., from bacteria to sea


cucumber, this enzyme is reported in higher vertebrates
also. The cleavage pattern by fucoidan-degrading enzyme
largely depends on the organism, but it has been broadly
classified in three groups: first is -L-fucosidase cleaves Lglucosyl linkage at non-reducing terminal while fucoidanases or fucansulphatehydrolases has two types of cleavage
pattern, one is an endo-cutter which produces oligomers;
another type is an exo-cutter and resulting monomers while
the exact mechanism is still unknown (Holtkamp et al.
2009; Wu et al. 2011). Most of the researcher worked on
the production study of fucosidases. The crystal structure
and extensive sequence analysis from microbial sources,
however, are limited (Rodrguez Jasso et al. 2008;
Descamps et al. 2006b; Wu et al. 2011). Looking at the
importance and perspectives of this enzyme, many more
research will be conducted in near the future. Some
important sources of fucoidan-degrading enzymes and
distribution are listed in Table 3. Here, in this review, we
have focused on important studies done on bioactivities
influenced by the molecular size of fucoidan.
Effect of LMWF as anticoagulant
As we mentioned that the prioritized research on
fucoidan was started in the 1990s, most of the paper
in the successive two decades was on screening and
structural analysis. A research by Colliec-Jouault et al.
in 2003 has pointed the size of fucoidan and their effect
on bioactivity (Colliec-Jouault et al. 2003). Chabut et al.

(2003) made a comparative study of heparin and


fucoidan on basic fibroblast growth factor (FGF-2).
They have reported that LMWF enhances the FGF-2induced tubular morphogenesis through overexpression
that was heparan sulfate dependent. Based on their
report, LMWFprevents arterial thrombus growth in
animals having low hemorrhagic propensity than heparin
and inhibits smooth muscle cell proliferation. After
myocardial infarction, endothelial cells increase expression of heparan sulfate proteoglycans, specifically at
ischemic sites. LMWF, being antithrombotic and potentially proangiogenic, could constitute an interesting drug
to initiate endothelial cell differentiation in revascularization of ischemic areas after myocardial infarction
(Chabut et al. 2004). Durand et al. (2008) have made a
comparative study of the antithrombotic and anticoagulant
activity of LMWF and LMWH in an experimental model
of arterial thrombosis due to endothelial apoptosis
(Durand et al. 2008). Deux et al. (2002) performed in
vitro growth inhibition experiments on vascular smooth
muscle cells, an in vivo pharmacokinetic study, and
locally delivered fluorescently labeled polysaccharides in
rabbit iliac arteries after angioplasty with a non-occlusive
catheter. They have reported that the low molecular weight
fraction of fucoidan as anti-in-stent restenosis after
angioplasty in rabbits (Deux et al. 2002). A similar study
done with different aspects of clinical and in vitro
evaluation of antithrombic activity of LMWF, supports
that the molecule can be exploited further (Zhu et al. 2010;
Kim et al. 2007, 2008, 2011b). The molecular weight and/

Table 3 Fucoidan-degrading enzyme sources and the nature of secretions


Sl. no.

Source organism

Nature

References

Alteromonadaceae sp.

Extracellular

Sakai et al. (2004)

Bacillus cereus

Extracellular

Miura et al. (2005)

Bifidobacterium bifidum
Lactobacillus sp.

Intracellular

Rodrguez-Daz et al. (2011) and Ashida et al. (2009)

Dendryphiella arenaria TM94

Extracellular

Wu et al. (2011)

Flavobacteriaceae sp.

Extracellular

Descamps et al. (2006a)

Flavobacterium CZ1127

Intracellular

Chang et al. (2010)

Flavobacterium sp. F-31

Intracellular

Ohshiro et al. (2011)

Fucophilus fucoidanolyticus

Intracellular

Sakai et al. (2003b)

Littorina kurila

Extracellular

Bilan et al. (2005)

10

Maribacter sp. KMM 6211


Gramella sp. KMM 6054

Intracellular

Urvantseva et al. (2006)

11

Pseudoalteromonas citrea strains KMM 3296 and KMM 3298

Bakunina et al. (2002)

12

Sphingomonas paucimobilis PF-1

Extracellular

Kim et al. (2008)

13

S. paucimobilis PF-1

Digestive

Kim et al. (2010a, b)

14

Streptomyces sp. OH11242

Extracellular

Goso et al. (2001)

15

Sulfolobus solfataricus

Intracellular

Cobucci-Ponzano et al. (2008)

16

Verrucomicrobia sp.

Digestive

Sakai et al. (2003a)

Appl Microbiol Biotechnol (2012) 93:7182

or degrees of sulfation of fucoidan are key factors for its


anticoagulating activity; therefore, enzymatically partial
digests could be a better tool for synthesis for LMWF
(Kim et al. 2010a, b)
Pomin et al. (2005) employed an apparently nonspecific
approach to cleave this polysaccharide based on mild
hydrolysis with acid and obtained low molecular weight
derivatives. They reported that the slight decrease in the
molecular size of the sulfated fucan dramatically reduces its
effect on thrombin inactivation mediated by heparin
cofactor II. Sulfated fucan with 45 tetrasaccharide
repeating units binds to heparin cofactor II but is unable
to link efficiently the plasma inhibitor and thrombin (Pomin
et al. 2005). In the same year, Matsubara et al. (2005)
have reported a comparative and comprehensive analysis
with various rages of LMWF as an inhibitor of for
inhibition human umbilical vein endothelial cells
(HUVEC) tube formation. They have reported that
LMWF with molecular weight ranges between 15 and
20 kDa, enhanced HUVEC migration but did not inhibit
HUVEC tube formation. Thus, 1520 kDa fucoidan would
have proangiogenic effect on angiogenesis. Their results
elucidated that 2030 kDa would be a critical point to
characterize the role of fucoidans on angiogenesis
(Matsubara et al. 2005).
Effect on endothelial cell migration
LMWF greatly enhances endothelial cells tube formation
in growth factor reduced Matrigel (Matsubara et al. 2005;
Lake et al. 2006). Luyt et al. (2003) have reported that a
LMWF can promote revascularization in rat critical
hindlimb ischemia. Previous studies have shown that it
can act as a potent inhibitor of vascular smooth muscle cell
proliferation in vitro (Lake et al. 2006) and prevent
neointimal hyperplasia of these cells during artery restenosis in vivo. Fucoidan is of particular pharmacological
interest because, in addition to its non-animal origin, it
exhibits anti-inflammatory activities, is a potent modulator
of connective tissue proteolysis but has low anticoagulant
activity compared with heparin. LMWF is also devoid of
direct antithrombin effect.
The vascular endothelial growth factor (VEGF) family
and its receptors are essential regulators of angiogenesis
and vascular permeability. LMWF is a strong enhancer of
VEGF165-induced EC chemotaxis but not proliferation
(Lake et al. 2006). While it was reported that LMWF was
not effective on VEGF121-induced EC migration, a VEGF
isoform that does not bind to heparan sulfate proteoglycans
(Lake et al. 2006). Surface plasmon resonance analysis
showed that LMWF binds with high affinity to VEGF165
(1.2 nM) and its receptors (520 nM) but not to VEGF121
(Lake et al. 2006). Overall, the effects of LMWF were

77

much more pronounced than those of LMWF heparin (Lake


et al. 2006).
LMWF as protective agent against alloimmune injury
The Graft vascular disease remains a major limitation to
long term organ graft survival, and conventional immunosuppressive agents fail to be effective for the treatment or
the prevention of allograft arteriosclerosis. In 2006, a study
reflected on the effect of LMWF on prevention of coronary
intimal proliferation in a rat cardiac allograft model, and it
has been reported that 5 mg kg1 day1 of LMWF prevents
coronary intimal lesion at 30-day post-transplantation. The
above model of heterotopic cardiac transplantation, allogeneic rejection is characterized by coronary intimal proliferation and myofibroblastic parenchymal remodelling
(Alkhatib et al. 2006). It was reported that on a daily basis,
injections of LMWF prevent intimal lesion, and myofibroblastic parenchymal remodelling treatment with LMWF
showed effective in the rat cardiac allograft model to
prevent arterial and parenchymal lesions occurring in
response to alloimmune injury. However, this protective
effect does not appear to depend on mobilization of bone
marrow-derived cells. It was also reported that high
molecular mass and viscous nature of these polysaccharides
has hampered their application, especially as a therapeutic
agent (Alkhatib et al. 2006). Thus, production and
optimization of single-type LMWF for proper function
and formulation is needed.
LMWF as anticancer agent
Yang et al. have studied the native and hydrolyzed fucoidan
from Undaira pinnatifida, as the anticancer agent. They
have shown that the anticancer activity was enhanced from
37.6% to 75.9% with a concentration of 1.0 mg/mL and
with native and hydrolyzed fucoidan, respectively (Yang et
al. 2008). This may be caused by the increased in molar
concentration and the enhanced mobility and diffusivity of
the partially hydrolyzed fucoidans. Yang et al. have
suggested that the anticancer activity of fucoidans could
be significantly improved by lowering their molecular
weight when they are depolymerized by mild hydrolysis
conditions without causing considerable desulfation (Yang
et al. 2008). Recently, Cho et al. have reported a
relationship between oversulfation and conformation of
LMWF and evaluation of their in vitro anticancer activity
(Cho et al. 2010).
LMWF as stumilant in 3D culture
Another interesting bioactivity of fucoidan was reported by
Changotade et al. (2008); when they have subjected the

78

LMWF (from pheophicae cell wall) on bi-dimensional


cultured normal human osteoblasts behaviors, they also
studied on, impregnation procedure of the bone biomaterials (Lubboc1) and explored the applicability of LMWF in
bone extracellular matrix context its capabilities to support
human osteoblastic behavior in 3D culture (Changotade et
al. 2008). The researchers have reported that LMWF is a
stimulant expression of osteoblastic markers differentiation
such as alkaline phosphatase activity, collagen type I,
expression, and mineral deposition; furthermore, cell
proliferation was enhanced as well (Changotade et al.
2008).
Effect of LMWF in treatment of cardiovascular desease
Inflammation of the vessel walls is a key player to be
initiated and the progression of cardiovascular diseases
(Bachelet et al. 2009). The inflammation, thrombosis, and
atherogenesis are successive phenomena, and platelets are
major components to co-ordinate the same (Gawaz et al.
2005; Bachelet et al. 2009). The activated platelets and
pathological endothelium prominently express the Pselectin, which has been described as a candidate target in
atherosclerosis (Bachelet et al. 2009). Using surface
plasmon resonance experiments, it observed that Pselectin binds to a synthetic dendrigraft polymer substituted
with sialic acid epitopes (Bachelet et al. 2009). In 1992,
Lindbom et al. reported that fucoidan can reduce the
leukocyte rolling on the vessel wall. Various reports on
LMWF as a revascularizing agent for cardiovascular
diseases (Lake et al. 2006; Bachelet et al. 2009) have been
archieved. It has been reported that in comparison to low
molecular weight heparin and dextran sulfate, the LMWF
more efficiently binds with the ligand of P-selectin. To
prove the same, they used binding assay, mass spectrometry, surface plasmon resonance, and flow cytometry on
human platelets (Bachelet et al. 2009). The inhibition of
SLex/P-selectin binding was quantified from binding assay
experiments, Bachelet et al. ranked polysaccharides as
follows (IC50): fucoidan (20 nM)>heparin (400 nM)>
dextran sulfate (25,000 nM) (Bachelet et al. 2009). Bachelet
et al. also reported that LMW fucoidan binds to activated
platelets and the level of binding was related to the degree
of platelet activation thus LMW fucoidan has an ability to
inhibit the binding of an anti-P-selectin antibody to activate
human platelets (Bachelet et al. 2009). According to
Bachelet et al., the LMWF could be used in the vascular
field as a targeting moiety in biomedical applications such
as drug-delivery systems or contrast media for molecular
imaging (Bachelet et al. 2009). Park et al. comparatively
analyze the effect of the high (HMWF; 1004 kDa),
medium (MMWF; 3.50.3), and LMWF (10.2 kDa)
molecular weight fucoidans on the progression of

Appl Microbiol Biotechnol (2012) 93:7182

collagen-induced arthritis (CIA). Their study concluded


that HMWF enhances arthritis through enhancing the
inflammatory activation of macrophages while LMWF
reduces arthritis through the suppression of Th1-mediated
immune reactions (Park et al. 2010).
Antioxidant activity of LMWF
As various studies suggest, fucoidan have been a great
potential as an antioxidant, in vitro. Fucoidan from L.
japonica can prevent the increase of lipid peroxide in
serum, liver, and spleen of diabetic mice obviously (Li et al.
2008). This fucoidan was reported as strong scavenging
effect on superoxide radical, while less effective on 1,1diphenyl-2-picryl-hydrazyl scavenging. The fucoidan can
inhabit H2O2-induced hemolysis of rat erythrocytes effectively and showed a significant protective effect on lipid
peroxidation of liver homogenate in rat induced by FeSO4ascorbic acid (Li et al. 2008). Micheline et al. reported that
fucoidan (homofucan) from F. vesiculosus and fucans
(heterofucans) from P. gymnospora had an inhibitory effect
on the formation of hydroxyl radical and superoxide radical
(Micheline et al. 2007). Similarly, various studies focus on
antioxidant properties of fucoidan in vitro (Micheline et al.
2007; Zhao et al. 2005; Li et al. 2006; Wang et al. 2008; Li
et al. 2004).
As it has been mentioned, the large size of this molecule
is a major concern with this molecule to be considered as a
drug. A little progress has been done in the direction of
sized selection of LMWF and enhancement of antioxidant
properties. A study done by Wang et al. (2009) and Choi et
al. (2009) in the direction of evaluation of antioxidant
activity of modified fucoidan is remarkable to understand
the size dependent antioxidant properties of fucoidan. They
have also explored the toxicity and activity level of
modified LMWF (Wang et al. 2009; Choi et al. 2009).
Some more studies yet to reveal for the establishment of
relation between size-dependent bioactivity of fucoidan are
still needed. Still, an intensive research is needed to develop
a safer and effective LMWF as an antioxidant.

Conclusions
In spite of their potential as biologically active compounds,
the high molecular mass and viscous natures of fucoidan
have hampered their applications, especially as a therapeutic agent. To obtain oligosaccharide with more diverse
bioactivities, LMWF can be prepared by chemical, radical,
or enzymatic means. Therefore, an extensive research
requires for boosting the generation of more and more
information regarding the LMWF as safer and enhanced
bioactivity without cross reactivity. This review is an

Appl Microbiol Biotechnol (2012) 93:7182

attempt to summarize the information regarding the


bioactivity of fucoidan and impact of molecular weight on
bioactivity of this marvel molecule. This molecule has more
hope as a medicine and may be in the future, the fabrication
of this molecule will be as smooth as required.
Acknowledgments This study was supported by a grant of the
Korea Healthcare Technology R&D Project, Ministry of Health &
welfare, Republic of Korea (Grant No. A103017). The work was also
supported in part by Inha University.

References
Adhikari U, Mateu CG, Chattopadhyay K, Pujol CA, Damonte EB, Ray
B (2006) Structure and antiviral activity of sulfated fucans from
Stoechospermum marginatum. Phytochemistry 67:24742482
Aguilar-Rosas R, Aguilar-Rosas LE, Avila-Serrano G, MarcosRamirez R (2004) First record of Undaria pinnatifida (Harvey)
Suringar (Laminariales, Phaeophyta) on the Pacific coast of
Mexico. Bot Mar 47:255258. doi:10.1515/BOT.2004.028
Aisa Y, Miyakawa Y, Nakazato T, Shibata H, Saito K, Ikeda Y, Kizaki
M (2005) Fucoidan induces apoptosis of human HS-Sultan cells
accompanied by activation of caspase-3 and down-regulation of
ERK pathways. Am J Hematol 78:714
Albuquerque IR, Queiroz KC, Alves LG, Santos EA, Leite EL, Rocha
HA (2004) Heterofucans from Dictyota menstrualis have
anticoagulant activity. Braz J Med Biol 37:167171
Alekseyenko TV, Zhanayeva SY, Venediktova AA, Zvyagintseva TN,
Kuznetsova TA, Besednova NN, Korolenko TA (2007) Antitumor and antimetastatic activity of fucoidan, a sulfated polysaccharide isolated from the Okhotsk Sea Fucus evanescens brown
alga. Bull Exp Biol Med 143(6):730732
Alkhatib B, Freguin-Bouilland C, Lallemand F, Henry JP, Litzler PY,
Marie JP, Richard V, Thuillez C, Plissonnier D (2006) Low
molecular weight fucan prevents transplant coronaropathy in rat
cardiac allograft model. Transpl Immunol 16:1419
Anastyuk SD, Shevchenko NM, Nazarenko EL, Imbs TI, Gorbach VI,
Dmitrenok PS, Zvyagintseva TN (2010) Structural analysis of a
highly sulfated fucan from the brown alga Laminaria cichorioides by tandem MALDI and ESI mass spectrometry. Carbohydr
Res 345(15):22062212
Angulo Y, Lomonte B (2003) Inhibitory effect of fucoidan on the
activities of crotaline snake venom myotoxic phospholipases A
(2). Biochem Pharmacol 66(10):19932000
Anno K, Seno N, Ota M (1970) Isolation of L-fucose 4-sulfate from
fucoidan. Carbohydr Res 13:167169
Ashida H, Miyake A, Kiyohara M, Wada J, Yoshida E, Kumagai H,
Katayama T, Yamamoto K (2009) Two distinct alpha-L-fucosidases from Bifidobacterium bifidum are essential for the
utilization of fucosylated milk oligosaccharides and glycoconjugates. Glycobiology 19(9):10101017
Azofeifa K, Angulo Y, Lomonte B (2008) Ability of fucoidan to
prevent muscle necrosis induced by snake venom myotoxins:
comparison of high- and low-molecular weight fractions.
Toxicon 3:373380
Babitha S, Shin JH, Kim EK (2009) Potential skin whitening agents of
natural origin from South Asia region. J Soc Cosmet Sci Korea
1:19
Bachelet L, Bertholon I, Lavigne D, Vassy R, Jandrot-Perrus M,
Chaubet F, Letourneur D (2009) Affinity of low molecular
weight fucoidan for P-selectin triggers its binding to activated
human platelets. Biochim Biophys Acta 1790(2):141146

79
Bakunina IIu, Nedashkovskaia OI, Alekseeva SA, Ivanova EP, Romanenko LA, Gorshkova NM, Isakov VV, Zviagintseva TN, Mikhalov
VV (2002) Degradation of fucoidan by the marine proteobacterium
Pseudoalteromonas citrea. Mikrobiologiia 71(1):4955
Bilan MI, Kusaykin MI, Grachev AA, Tsvetkova EA, Zvyagintseva TN,
Nifantiev NE, Usov AI (2005) Effect of enzyme preparation from
the marine mollusk Littorina kurila on fucoidan from the brown
alga Fucus distichus. Biochemistry (Mosc) 70(12):13211326
Bilan MI, Grachev AA, Shashkov AS, Nifantiev NE, Usov AI (2006)
Structure of a fucoidan from the brown seaweed Fucus serratus
L. Carbohydr Res 341:238245
Bilan MI, Zakharova AN, Grachev AA, Shashkov AS, Nifantev NE,
Usov AI (2007) Polysaccharides of algae: 60. Fucoidan from the
Pacific brown alga Analipus japonicus (Harv.) Winne (Ectocarpales, Scytosiphonaceae). Bioorg Khim 33(1):4453
Bilan MI, Grachev AA, Shashkov AS, Kelly M, Sanderson CJ,
Nifantiev NE, Usov AI (2010) Further studies on the composition and structure of a fucoidan preparation from the brown alga
Saccharina latissima. Carbohydr Res 345(14):20382047
Black WAP (1954) Seasonal variation in the combined L-fucose
content of the common British Laminariaceae and Fucaceae. J
Sci Food Agric 5:445448
Boisson-Vidal C, Chaubet F, Chevolot L, Sinquin C, Theveniaux J,
Millet J, Sternberg C, Mulloy B, Fischer AM (2000) Relationship
between antithrombotic activities of fucans and their structure.
Drug Dev Res 51:216224
Chabut D, Fischer AM, Colliec-Jouault S, Laurendeau I, Matou S, Le
Bonniec B, Helley D (2003) Low molecular weight fucoidan and
heparin enhance the basic fibroblast growth factor-induced tube
formation of endothelial cells through heparan sulfate-dependent
alpha6 overexpression. Mol Pharmacol 64(3):696702.
Chabut D, Fischer AM, Helley D, Colliec S (2004) Low molecular
weight fucoidan promotes FGF-2-induced vascular tube formation by human endothelial cells, with decreased PAI-1 release and
ICAM-1 downregulation. Thromb Res 113(1):9395
Chanda NP, Matsuhiro B (2008) Characterization of a fucoidan from
Lessonia vadosa (Phaeophyta) and its anticoagulant and elicitor
properties. Int J Biol Macromol 42(3):235240
Chang Y, Xue C, Tang Q, Li D, Wu X, Wang J (2010) Isolation and
characterization of a sea cucumber fucoidan-utilizing marine
bacterium. Lett Appl Microbiol 3:301307
Changotade SI, Korb G, Bassil J, Barroukh B, Willig C, ColliecJouault S, Durand P, Godeau G, Senni K (2008) Potential effects
of a low-molecular-weight fucoidan extracted from brown algae
on bone biomaterial osteoconductive properties. J Biomed Mater
Res A 87(3):666675
Chen JH, Lim JD, Sohn EH, Choi YS, Han ET (2009) Growthinhibitory effect of a fucoidan from brown seaweed Undaria
pinnatifida on Plasmodium parasites. Parasitol Res 104(2):245
250
Chevolot L, Mulloy B, Racqueline J (2001) A disaccharide repeat unit
is the structure in fucoidans from two species of brown algae.
Carbohydr Res 330:529535
Chizhov AO, Dell A, Morris HR (1999) A study of fucoidan from the
brown seaweed Chorda filum. Carbohydr Res 320:108119
Cho ML, Lee BY, You SG (2010) Relationship between oversulfation
and conformation of low and high molecular weight fucoidans
and evaluation of their in vitro anticancer activity. Molecules 30
(16):291297. doi:10.3390/molecules16010291
Choi JI, Kim HJ, Kim JH, Byun MW, Soo Chun B, Hyun Ahn D, Hwang
YJ, Kim DJ, Kim GH, Lee JW (2009) Application of gamma
irradiation for the enhanced physiological properties of polysaccharides from seaweeds. Appl Radiat Isot 67(78):12771281
Cobucci-Ponzano B, Conte F, Rossi M, Moracci M (2008) The alphaL-fucosidase from Sulfolobus solfataricus. Extremophiles 12
(1):6168

80
Colliec-Jouault S, Millet J, Helley D, Sinquin C, Fischer AM (2003)
Effect of low-molecularweight fucoidan on experimental arterial
thrombosis in the rabbit and rat. J Thromb Haemost 1(5):1114
1115
Cumashi A, Ushakova NA, Preobrazhenskaya ME, DIncecco A,
Piccoli A, Totani L, Tinari N, Morozevich GE, Berman AE,
Bilan MI, Usov AI, Ustyuzhanina NE, Grachev AA, Sanderson
CJ, Kelly M, Rabinovich GA, Iacobelli S, Nifantiev NE (2007) A
comparative study of the anti-inflammatory, anticoagulant, antiangiogenic, and antiadhesive activities of nine different fucoidans
from brown seaweeds. Glycobiology 17(5):541542
Del Bigio MR, Yan HJ, Campbell TM, Peeling J (1999) Effect of
fucoidan treatment on collagenase-induced intracerebral hemorrhage in rats. Neurol Res 21(4):415419
Descamps V, Colin S, Lahaye M, Jam M, Richard C, Potin P,
Barbeyron T, Yvin JC, Kloareg B (2006a) Isolation and culture of
a marine bacterium degrading the sulfated fucans from marine
brown algae. Mar Biotechnol (NY) 8(1):2739
Descamps V, Colin S, Marc L, Jam M, Richard C, Potin P, Barbeyron
T, Yvin J, Kloareg B (2006b) Isolation and culture of a marine
bacterium degrading the sulfated fucans from marine brown
algae. Mar Biotechnol 8:2739
Deux JF, Meddahi-Pell A, Le Blanche AF, Feldman LJ, ColliecJouault S, Br F, Boudghne F, Michel JB, Letourneur D (2002)
Low molecular weight fucoidan prevents neointimal hyperplasia
in rabbit iliac artery in-stent restenosis model. Arterioscler
Thromb Vasc Biol 22(10):16041609
Duarte ME, Cardoso MA, Noseda MD, Cerezo AS (2001) Structural
studies on fucoidans from the brown seaweed Sargassum
stenophyllum. Carbohydr Res 333(4):281293
Durand E, Helley D, Al Haj Zen A, Dujols C, Bruneval P, ColliecJouault S, Fischer AM, Lafont A (2008) Effect of low molecular
weight fucoidan and low molecular weight heparin in a rabbit
model of arterial thrombosis. J Vasc Res 45(6):529537
Fitton JH (2005) Fucoidans: healthful saccharides from the sea.
GlycoScience Nutr 6(1):16
Fitton HJ, Dragar C (2006) Method and composition for the treatment
of a viral infection. United States Patent 7056520. http://www.
freepatentsonline.com/7056520.html. Accessed 24 August 2008
Gawaz M, Langer H, May AE (2005) Platelets in inflammation and
atherogenesis. J Clin Invest 115:33783384
Gideon TP, Rengasamy R (2008) Toxicological evaluation of fucoidan
from Cladosiphon okamuranus. J Med Food 11(4):638642.
doi:10.1089/jmf.2007.0127
Goso Y, Ishihara K, Sugawara S, Hotta K (2001) Purification and
characterization of alpha-L-fucosidases from Streptomyces sp.
OH11242. Comp Biochem Physiol B Biochem Mol Biol 130
(3):375383
Hlawaty H, Suffee N, Sutton A, Oudar O, Haddad O, Ollivier V,
Laguillier-Morizot C, Gattegno L, Letourneur D, Charnaux N
(2011) Low molecular weight fucoidan prevents intimal hyperplasia in rat injured thoracic aorta through the modulation of
matrix metalloproteinase-2 expression 81(2):233243
Holtkamp AD, Kelly S, Ulber R, Lang S (2009) Fucoidans and
fucoidanasesfocus on techniques for molecular structure
elucidation and modification of marine polysaccharides. Appl
Microbiol Biotechnol 82(1):111
Huang L, Wen K, Gao X, Liu Y (2010) Hypolipidemic effect of
fucoidan from Laminaria japonica in hyperlipidemic rats. Pharm
Biol 48(4):422426
Jiao G, Yu G, Zhang J, Ewart HS (2011) Chemical structures and
bioactivities of sulfated polysaccharides from marine algae. Mar
Drugs 9:196223. doi:10.3390/md9020196
Jin YZ, Ahn SY, Hong ES, Li GH, Kim EK, Row KH (2005)
Extraction of whiteining agents from natural plants and whitening effect. J Korean Ind Eng Chem 16:348353

Appl Microbiol Biotechnol (2012) 93:7182


Jin YZ, Li GH, Ahn SY, Row KH, Kim EK (2006) Extraction and
purification of depigmenting agents from Chinese plants. Chem
Res Chin Univ 22:162167
Jing W, Bin ZQ, Shan ZZ, Jing ZJ, Cheng LP (2009) Synthesized
phosphorylated and aminated derivatives of fucoidan and their
potential antioxidant activity in vitro. Int J Biol Macromol 44
(2):170174
Jingjing Z, Quanbin Z, Jing W, Xuelian S, Zhongshan Z (2009) Analysis
of the monosaccharide composition of fucoidan by precolumn
derivation HPLC. Chin J Oceanol Limnol 27(3):578582
Kim WJ, Kim HG, Oh HR, Lee KB, Lee YK, Park YI (2007)
Purification and anticoagulant activity of a fucoidan from Korean
Undaria pinnatifida sporophyll. Algae 22(3):247252
Kim WJ, Kim SM, Lee YH, Kim HG, Kim HK, Moon SH, Suh HH,
Jang KH, Park YI (2008) Isolation and characterization of marine
bacterial strain degrading fucoidan from Korean Undaria
pinnatifida sporophylls. J Microbiol Biotechnol 18(4):616623
Kim JE, Lee HB, Nguyen HD, Yoon SA, Um JM, Kim EK (2010a)
Depigmenting effect of fucoidan isolated from sporophyll of
Undaria pinnatifida. In: KSBB fall meeting and international
symposium, Incheon, p 261
Kim WJ, Koo YK, Jung MK, Moon HR, Kim SM, Synytsya A, YunChoi HS, Kim YS, Park JK, Park YI (2010b) Anticoagulating
activities of low-molecular weight fuco-oligosaccharides prepared by enzymatic digestion of fucoidan from the sporophyll of
Korean Undaria pinnatifida. Arch Pharm 33(1):125131
Kim JE, Morya VK, Lee HB, Nguyen HD, Park SJ, Kim EK (2011a)
Depigmenting effect of different molecular weight fucoidan from
Fucus vesiculosus. In: The Korean Society for Biotechnology
and Bioengineering Spring Meeting, Jeju, p 272
Kim JE, Morya VK, Lee HB, Nguyen HD, Park SJ, Kim EK (2011b)
Depigmenting effect on melanocyte by fucoidan isolated from
sporophyll of Undaria pinnatifida. In: Asian congress of
biotechnology, p 246
Kwak KW, Cho KS, Hahn OJ, Lee KH, Lee BY, Ko JJ, Chung KH
(2010) Biological effects of fucoidan isolated from Fucus
vesiculosus on thrombosis and vascular cells. Korean J Hematol
45:5157
Lake AC, Vassy R, Di Benedetto M, Lavigne D, Le Visage C, Perret
GY, Letourneur D (2006) Low molecular weight fucoidan
increases VEGF165-induced endothelial cell migration by enhancing VEGF165 binding to VEGFR-2 and NRP1. J Biol Chem
281:3784437852
Lam DPU, Nguyen HD, Kim EK (2008) Mechanism of skin
pigmentation. Biotechnol Bioprocess Eng 13:383395
Lam DPU, Nguyen HD, Lee HB, Kim BR, Lee JD, Shin JH, Kim EK
(2011) Depigmenting effect of Sterculia lynchnophera Hance on
B16F10 Melanoma and C57BL/6 Melan-a cell. Korean J Chem
Eng 28:10741077
Lapshina L, Reunov A, Nagorskaya V, Zvyagintseva T, Shevchenko N
(2006) Inhibitory effect of fucoidan from brown alga Fucus
evanescens on the spread of infection induced by tobacco mosaic
virus in tobacco leaves of two cultivars. Russ J Plant Physiol
53:246251
Lee JB, Hayashi K, Hashimoto M , Nakano T, Hayashi T (2004)
Novel antiviral fucoidan from sporophyll of undaria pinnatifida
(mekabu). Chem Pharm Bull 52(9):10911094
Li B, Gu XH, Xu SY (2004) Determination of the position of sulphate
group in sulphated polysaccharides. Fenxi Kexue Xuebao
20:498500
Li B, Xin JW, Sun JL, Xu SY (2006) Structural investigation of a
fucoidan containing a fucose-free core from the brown seaweed
Hizikia fusiforme. Carbohydr Res 341:11351146
Li B, Xu SY (2007) Structural investigation of oligosaccharides in
partial acid hydrolyzed products of fucoidan isolated from
Hizikia fusiforme. Nat Prod Res Dev 19:550553

Appl Microbiol Biotechnol (2012) 93:7182


Li B, Lu F, Wei X, Zhao R (2008) Fucoidan: structure and bioactivity.
Molecules 13:16711695
Luo D, Zhang Q, Wang H, Cui Y, Sun Z, Yang J, Zheng Y, Jia J, Yu F,
Wang X, Wang X (2009) Fucoidan protects against dopaminergic
neuron death in vivo and in vitro. Eur J Pharmacol 617(13):3340
Luyt CE, Meddahi-Pelle A, Ho-Tin-Noe B, Colliec-Jouault S,
Guezennec J, Louedec L, Prats H, Jacob MP, Osborne-Pellegrin
M, Letourneur D, Michel JB (2003) Low-molecular-weight
fucoidan promotes therapeutic revascularization in a rat model
of critical hindlimb ischemia. J Pharmacol Exp Ther 305:2430
Mandal P, Mateu CG, Chattopadhyay K, Pujol CA, Damonte EB, Ray
B (2007) Structural features and antiviral activity of sulphated
fucans from the brown seaweed Cystoseira indica. Antivir Chem
Chemother 18:153162
Marais MF, Joseleau JP (2001) A fucoidan fraction from Ascophyllum
nodosum. Carbohydr Res 336:155159
Matsubara K, Xue C, Zhao X, Mori M, Sugawara T, Hirata T (2005)
Effects of middle molecular weight fucoidans on in vitro and ex
vivo angiogenesis of endothelial cells. Int J Mol Med 15(4):695
699
Medcalf DG, Larsen B (1977) Fucose-containing polysaccharide in
the brown algae Ascophyllum nodosum and Focus vesiculosus.
Carbohydr Res 59:531537
Medeiros VP, Queiroz KC, Cardoso ML, Monteiro GR, Oliveira FW,
Chavante SF, Guimaraes LA, Rocha HA, Leite EL (2008) Sulfated
galactofucan from Lobophora variegata: anticoagulant and antiinflammatory properties. Biochemistry (Mosc) 73(9):10181024
Micheline RS, Cybelle M, Celina GD, Fernando FS, Hugo OR, Edda
L (2007) Antioxidant activities of sulfated polysaccharides from
brown and red seaweeds. J Appl Phycol 19:153160
Mitrasinovic OM, Murphy GM Jr (2002) Accelerated phagocytosis of
amyloid-beta by mouse and human microglia overexpressing the
macrophage colony-stimulating factor receptor. J Biol Chem
277:2988929896
Miura T, Okamoto K, Yanase H (2005) Purification and characterization of extracellular 1,2-alpha-L-fucosidase from Bacillus cereus.
J Biosci Bioeng 99(6):629635
Mulloy B, Mourao PA, Gray E (2000) Structure/function studies of
anticoagulant sulphated polysaccharides using NMR. J Biotechnol 77(1):123135
Nishino T, Nishioka C, Ura H (1994) Isolation and partial characterization
of a novel amino sugar containing fucanc sulfate from commercial
Fucus vesiculosus fucoidan. Carbohydr Res 255:213224
Ohshiro T, Ohmoto Y, Ono Y, Ohkita R, Miki Y, Kawamoto H, Izumi
Y (2011) Isolation and characterization of a novel fucoidandegrading microorganism. Biosci Biotechnol Biochem 74
(8):17291732
Park SB, Chun KR, Kim JK, Suk K, Jung YM, Lee WH (2010) The
differential effect of high and low molecular weight fucoidans on
the severity of collagen-induced arthritis in mice. Phytother Res
24(9):13841391
Patankar MS, Oehninger S, Barnett T (1993) A revised structure for
fucoidan may explain some of its biological activities. J Biol
Chem 268:2177021776
Pomin VH, Pereira MS, Valente A, Tollefsen DM, Pavo MSG,
Mouro PAS (2005) Selective cleavage and anticoagulant activity
of a sulfated fucan: stereospecific removal of a 2-sulfate ester
from the polysaccharide by mild acid hydrolysis, preparation of
oligosaccharides, and heparin cofactor II-dependent anticoagulant activity. Glycobiology 15:369381
Ponce NMA, Pujol CA, Damonte EB (2003) Fucoidans from the brown
seaweed Adenocystis utricularis: extraction methods, antiviral
activity and structural studies. Carbohydr Res 338:153165
Qiu XD, Amarasekara A, Doctor V (2006) Effect of oversulfation on
the chemical and biological properties of fucoidan. Carbohydr
Polym 63:224228

81
Raghavendran HRB, Srinivasan P, Rekha S (2011) Immunomodulatory
activity of fucoidan against aspirin-induced gastric mucosal damage
in rats. Int Immunopharmacol 11(2):157163
Reith JH, BDeurwaarder BP, Hemmes K, Curvers APWM, Brandeburg
W, Zeeman G (2005) Bio-offshore. Grootschalige teelt van
zeewieren in combinatie met offshore windparken in de Noordzee.
ECN. (Rapport ECN C-05-008)
Rodrguez Jasso RM, Aguilar Gonzalez CN, Pastrana L, Teixeira JA
(2008) Identification and evaluation of fungal strains with
fucoidan degradation potential. In: Ferreira EC, Mota M (eds)
Proceedings of the 10th international chemical and biological
engineering conference CHEMPOR 2008, Universidade do
Minho, Braga, Portugal, pp 21062109
Rodrguez-Daz J, Monedero V, Yebra MJ (2011) Utilization of
natural fucosylated oligosaccharides by three novel alpha-Lfucosidases from a probiotic Lactobacillus casei strain. Appl
Environ Microbiol 77(2):703705
Sakai T, Ishizuka K, Kato I (2003a) Isolation and characterization of a
fucoidan-degrading marine bacterium. Mar Biotechnol (NY) 5
(5):409416
Sakai T, Ishizuka K, Shimanaka K, Ikai K, Kato I (2003b) Structures
of oligosaccharides derived from Cladosiphon okamuranus
fucoidan by digestion with marine bacterial enzymes. Mar
Biotechnol (NY) 5(6):536544
Sakai T, Kawai T, Kato I (2004) Isolation and characterization of a
fucoidan-degrading marine bacterial strain and its fucoidanase.
Mar Biotechnol (NY) 6(4):335346
Silva TMA, Alves LG, Queiroz KCS, Santos MGL, Marques CT,
Chavante SF, Rocha HAO, Leite EL (2005) Partial characterization and anticoagulant activity of a heterofucan from the
brown seaweed Padina gymnospora. Braz J Med Biol Res
38:523533
Skriptsova AV, Shevchenko NM, Zvyagintseva TN, Imbs TI (2010)
Monthly changes in the content and monosaccharide composition
of fucoidan from Undaria pinnatifida (Laminariales, Phaeophyta). J Appl Phycol 22:7986
Tanaka K, Ito M, Kodama M, Tomita M, Kimura S, Hoyano M,
Mitsuma W, Hirono S, Hanawa H, Aizawa Y (2011) Sulfated
polysaccharide fucoidan ameliorates experimental autoimmune
myocarditis in rats. J Cardiovasc Pharmacol Ther 16:7986
Thomes P, Rajendran M, Pasanban B, Rengasamy R (2010)
Cardioprotective activity of Cladosiphon okamuranus fucoidan
against isoproterenol induced myocardial infarction in rats.
Phytomedicine 18(1):5257
Trinchero J, Ponce NMA, Crdoba OL, Flores ML, Pampuro S, Stortz
CA, Salomn H, Turk G (2009) Antiretroviral activity of
fucoidans extracted from the brown seaweed Adenocystis
utricularis. Phytother Res 23:707712
Uhm CS, Kim KB, Lim JH, Pee DH, Kim YH, Kim H, Eun BL, Tockgo
YC (2003) Effective treatment with fucoidan for perinatal hypoxic
ischemic ncephalopathy in rats. Neurosci Lett 353:2124
Urvantseva AM, Bakunina IIu, Nedashkovskaia OI, Kim SB,
Zviagintseva TN (2006) Distribution of intracellular fucoidan
hydrolases among marine bacteria of the family Flavobacteriaceae. Prikl Biokhim Mikrobiol 42(5):552559
Ushakova NA, Morozevich GE, Ustyuzhanina NE, Bilan MI, Usov
AI, Nifantiev NE, Preobrazhenskaya ME (2009) Anticoagulant
activity of fucoidans from brown algae. Biochemistry (Moscow)
Supplement Series B: Biomedical Chemistry 3:7783
Usov AI, Smirnova GP, Klochkova NG (2005) Polisaccharidi
vodorosley. 58. Polisaccharidniy sostav buroi vodorosli Alaria
fistilosa P. et. R. (Alariaceae, Laminariales). Izvestia Academii
Nauk. Seria Khimicheskaya 5:12451249. in Russian
Venugopal V (2009) Seaweed Hydrocolloids in Marine Products for
Healthcare Functional and Bioactive Nutraceutical Compounds
from the Ocean. CRC Press Taylor & Francis Group 327329

82
Wang J, Zhang Q, Zhang Z, Zhang J, Li P (2008) Synthesized phosphorylated and aminated derivatives of fucoidan and their potential
antioxidant activity in vitro. Int J Biol Macromol 44:170174
Wang J, Wang F, Zhang Q, Zhang Z, Shi X, Li P (2009) Synthesized
different derivatives of low molecular fucoidan extracted from
Laminaria japonica and their potential antioxidant activity in
vitro. Int J Biol Macromol 44(5):379384
Wang J, Zhang Q, Zhang Z, Song H, Li P (2010) Potential antioxidant
and anticoagulant capacity of low molecular weight fucoidan
fractions extracted from Laminaria japonica. Int J Biol Macromol 46: 612
Wu Q, Zhang M, Wu KE, Liu B, Cai J, Pan R (2011) Purification and
characteristics of fucoidanase obtained from Dendryphiella
arenaria TM94. J Appl Phycol 23:197203
Wu XW, Yang ML, Huang XL, Yan J, Luo Q (2004) Effect of
Laminaria japonica polysaccharides on radioprotection and
splenic lymphocyte apoptosis. Med J Wuhan Univ 25:239241

Appl Microbiol Biotechnol (2012) 93:7182


Yang C, Chung D, Shin IS, Lee H, Kim J, Lee Y, You S (2008) Effects
of molecular weight and hydrolysis conditions on anticancer
activity of fucoidans from sporophyll of Undaria pinnatifida. Int
J Biol Macromol 43(5):433437
Zhao X, Xue CH, Cai YP, Wang DF, Fang Y (2005) The study of
antioxidant activities of fucoidan from Laminaria japonica. High
Tech Lett 11:9194
Zhang Q, Li Z, Xu Z, Niu X, Zhang H (2003) Effects of fucoidan on
chronic renal failure in rats. Planta Med 69:537541
Zhang Y, Song S, Song D, Liang H, Wang W, Ji A (2010)
Proliferative effects on neural stem/progenitor cells of a sulfated
polysaccharide purified from the sea cucumber Stichopus
japonicus. J Biosci Bioeng 109(1):6772
Zhu Z, Zhang Q, Chen L, Ren S, Xu P, Tang Y, Luo D (2010) Higher
specificity of the activity of low molecular weight fucoidan for
thrombin-induced platelet aggregation. Thromb Res 125(5):419
426

También podría gustarte