Sei sulla pagina 1di 7

Polymer Degradation and Stability 59 (1998) 85-91

0 1998 Elsevier Science Limited. All rights reserved


Printed in Northern Ireland
PII: s0141-3910(97)00179-1 0141-3910/98/519.00
ELSEVIER

Biosynthesis and applications of alginates


Helga Ertesvig* & Svein Valla
Unigen Center for Molecular Biology, Norwegian University of Science and Technology, N-7005 Trondheim, Norway

(Accepted 18 June 1997)

Alginate is a family of linear polysaccharides composed of mannuronic acid (M)


and guluronic acid (G). The polymer is used as a gel-former and viscosifier in a
wide range of industrial applications. It is also used for encapsulation of cells and
enzymes. The viscosity of alginate is mainly dependent on the polymer length,
while the gel-forming and water-binding properties and the degree of immuno-
genicity are determined by the fraction and distribution of G-residues. Alginates
are currently manufactured by harvesting brown algae, but in nature the polymer
is also produced by some bacteria belonging to the genera Azotobacter and
Pseudomonas. The biosynthesis of alginate has been mostly studied in Pseudo-
monas aeruginosa, where many of the involved proteins and genes have also been
identified. In both algae and bacteria the polymer is first produced as mannur-
onan, which is then epimerized by the enzyme mannuronan C-5epimerase. A
gene encoding a periplasmic epimerase has been identified in the alginate gene
clusters of P. aeruginosa and Azotobacter vinelandii. The A. vinelandii genome
also encodes a family of at least five secreted epimerases, each of which intro-
duces different distributions of G in the alginate. These enzymes can therefore be
used to modify alginates in vitro to obtain polysaccharides with the desired con-
tent and distribution pattern of G. Such alginates may become useful in applica-
tions where reproducible and specific physical properties are required. 0 1998
Elsevier Science Limited. All rights reserved

1 STRUCTURE OF ALGINATE blocks which form the junctions, but the stability of
the gel is mostly dependent on the content of G-
Alginate is a linear copolymer composed of l-4 blocks.4 By analysing the polymer by NMR it is now
linked /3-D-mannuronic acid (M) and its C-5-epimer possible to determine the frequencies of each mono-
a-L-guluronic acid (Fig. 1). These monomers can be mer, each of the four possible dimers, and each of
organized in blocks of consecutive G-residues (G- the eight possible trimers.5,6 However, it is not pos-
blocks), consecutive M-residues (M-blocks), or sible to estimate the distribution of block-lengths for
alternating M and G (MG-blocks). The relative any alginate containing more than one type of block.
amount of each block-type varies between different
alginates. As shown in Fig. 1, there are distinct
structural differences between the block-types. MG- 2 TRADITIONAL INDUSTRIAL USES OF
blocks form the most flexible chains and are more ALGINATE
soluble at lower pH than the other two block-types.
M-blocks have been found to be strongly immunos- Alginates for commercial uses are extracted from
timulating. G-blocks form stiff chains, and two G- brown algae, and the annual production is about
blocks of more than 6 residues each can be cross- 30 000 tons.7 The composition of the poly-
linked by divalent cations (e.g. Ca2+, Ba2+, Sr2+), saccharide depends on the species used and also
leading to gel formation. 2,3 At low pH, protonized whether the blade and stipe are separated before
high-molecular-mass alginates can form soft acidic extraction (Table 1). For most applications the
gels. In these gels, it is mostly the homopolymeric polymer is sold for US$S-20/kg, but the highest
purity qualities are sold for up to US$40 OOO/kg.
*To whom correspondence should be addressed. Fax: 47 Alginates have various industrial uses as viscosi-
73598705; e-mail: helgae@unigen.ntnu.no fiers, stabilizers and gel-forming, film-forming or
85
86 H. ErtesvGg, S. Valla

a) b)

Fig. 1. Structure of a) B-D-mannuronic acid, b) cw-guluronic acid and c) alginate.

Table 1. Composition aud sequence parameters for algal 3 ALGINATES AS IMMOBILIZATION


alginatek8
MATERIALS
Source FG FM FGG FMM FGM,MG

Ascophyllum nodosum 0.10 0.90 0.04 0.84 0.06 In recent years alginates have been used for
(fruiting bodies) encapsulation of cells and enzymes.* When
Ascophyllum nodosum 0.36 0.64 0.16 0.44 0.20 droplets of a mixture of cells or enzymes and
(old tissue)
Macrocystis pyrifera 0.39 0.61 0.16 0.38 0.23
Na-alginate are mixed with a solution containing a
Laminaria hyperborea 0.55 0.45 0.38 0.28 0.17 gel-forming ion, the gel-beads will form instan-
(leaf) taneously. The encapsulated cells are protected
Laminaria hyperborea 0.68 0.32 0.56 0.20 0.12 against mechanical stress, while nutrients and
(stipe)
Laminaria hyperborea 0.75 0.25 0.66 0.16 0.09 metabolites can diffuse through the semi-permeable
(outer cortex) capsule. Gels with a high amount of G-blocks
Durvillea antartica 0.29 0.71 0.15 0.57 0.14 exhibit high porosity, low shrinkage during gel-
formation, and low swelling after drying. With an
increasing amount of M the gels become softer and
have a smaller pore size. The beads can later be
water-binding agents. These applications range coated by a polycation such as poly-L-lysin. This
from textile printing and manufacturing of cer- will stabilize and strengthen the gel network and
amics to production of welding rods and reduce the permeability.lO~ll Some examples of
water-treatment.7 The polymer is soluble in cold cells encapsulated in alginate are shown in Table 2.
water and forms thermostable gels. These properties Insulin-producing cells encapsulated in alginate
are utilized in the food industry in products are now being tested as an artificial pancreas for
like custard creams and restructured food. The treatment of Type I diabetes.25,26 The pores in the
polymer is also used as a stabilizer and thickener in capsules are large enough to permit free diffusion
a variety of beverages, ice-creams, emulsions and of glucose and insulin, while at the same time pro-
sauces. tecting the cells from the immune system.27 Since
The pharmaceutical industry7 uses alginates as mannuronan stimulates the immune system, it is
wound dressings and dental impression materials. important to use well-defined and homogenous
The polysaccharide is also used as a tablet binder or alginates for implantation purposes. On the other
disintegrant, and by carefully choosing the optimal hand, this property of mannuronan may be utilized
alginate quality one can obtain controlled release of when a stimulation of the immune system is
the drug. advantageous.28
Biosynthesis and applications of alginates 87

Table 2. Examples of cells immobilized in alginate


~____
Cells/enzymes Product/purpose Reference

Enzymes Urease Stabilization of enzyme 12


High-alkaline phosphatase Stabilization of enzyme 13
Bacteria Pseudomonas putida Removal of HzS 14
Erwinia rhapontici Isomaltulose 15
Blue green algae Anabena sp. Ammonia 16
Fungi Aspergillus niger Citric acid 17
Saccharomyces cerevisiae Ethanol 18
Algae Microalgae Cryopreservation 19
Botryococcus braunii Hydrocarbons 20
Plant cells Carrot Dry artificial seeds 21
Digitalis lanata Digitoxins 22
Mammalian cells rhCSF-1 or LM-10 CSF-1 23
Hybridoma Monoclonal antibodies 24
Islets of Langerhans Insulin 25

4 OCCURRENCE OF ALGINATES IN NATURE different,36probably reflecting the different needs


for gel-strength and water-retaining capacities
Alginate exists as a matrix polysaccharide in all (Table 3). Wild-type Pseudomonas aeruginosu does
marine brown algae. It is the most abundant not produce alginate, but mutations leading to
polysaccharide in the organisms, comprising up to derepression of the a-factor AlgU give the corre-
40% of their dry weight. The habitats of the dif- sponding mutant the ability to produce the poly-
ferent species differ with regard to exposure to mer as a response to environmental signals.37
periodic drying due to the tide and waves, and this
is reflected in different requirements for stiffness,
elasticity and water-binding capacity between 5 BIOSYNTHESIS OF ALGINATE
species. Since these properties are related to the
relative content of each block-type in the polymer, The biosynthesis of alginate was first studied by
this probably explains why different species need to Lin and Hassid,38 who in a cell-free system from
produce different alginates (Table 1). Fucus gurdnerii detected the enzyme activities
Alginate is also used as a capsular poly- necessary for the synthesis of mannuronan. A simi-
saccharide for the bacteria Azotobacter vinelandii,29 lar pathway has since been found in A. vinelundii39
Azotobacter chroococcum30 and some species and P. ueruginosdlo (Fig. 2). Starting with D-fructose-
belonging to the genus Pseudomonas.31-33 The 6-phosphate, the sugar nucleotide GDP-D-mannose
polymers from Pseudomonas do not contain G- is made and further oxidized to GDP-D-mannuronic
blocks, while alginates from A. vinelundii contain acid. This compound is then polymerized to
all three block types. 34 The main difference mannuronan. The polymer can be further modified
between algal and bacterial alginate is that some of by an acetyl transferase and by a mannuronan
the bacterial M-residues are 0-acetylated at the 2 C-5-epimerase, which epimerizes mannuronic acid
and/or 3 position. 34 A. vinefundii is able to differ- residues to guluronic acid.
entiate into a cyst, which can be viewed as a resting The genes involved in the biosynthesis of algi-
stage of the bacterium encapsulated in a Ca2+- nates were first identified in P. ueruginosa. Later,
alginate ge1.35The composition of alginates found several of the corresponding genes were identified
in the capsule of the vegetative cell and in the outer in A. vinelundii, where they are organized in the
(exine) and inner (intine) layers of the cyst coat is same manner. 41 The two genes ulgA42-43 and
ulgD,44,45 encoding the bifunctional enzyme phos-
Table 3. Alginates from different stages in tbe life-cycle of phomannoisomerase/D-mannose- 1-phosphate gua-
A. vinek&ii.36 nylyl transferase and guanosine diphosphate
Sample Distribution of diuronides mannose dehydrogenase, respectively, are flanking
a gene cluster (Fig. 3) encoding all the known bio-
FMM FMG FGG
synthetic genes except aZgC.40,46The amino acid
Capsular material 0.26 0.32 0.11 sequence of the membrane protein A1g847,48shows
Cyst exine 0.046 0.26 0.43 similarity to some processive glycosyl-transferases,49
Cyst intine 0.39 0.21 0.091
and it is probable that A1g4447,48and AlgX (also
88 H. Ertesvdg, S. Valla

D-fructose-6-phosphate mutated in mucA or mucB.37 Mutations in mucD


1 L also seem to affect the alginate biosynthesis.60 In
P. aeruginosa it has been found that AlgU is also
D-mannose-6-phosphate
necessary for transcription of AlgR, AlgZ and
2 1 AlgB,6,62 transcription-factors which bind to the
D-mannose-l -phosphate
algD-promoter. AlgR and AlgZ are necessary for
the transcription of algD, while AlgB is necessary
3 \1 for high-level-production of alginate. Several other
GDP-D-mannose genes have also been shown to have an effect on
alginate production in P. aeruginosa.40~60~63-66
4 1 The genetics of alginate biosynthesis in A. vine-
GDP-D-mannuronic acid landii have not been studied to the same extent, but
as this bacterium produces alginate constitutively,
5 L
some differences must exist. It has been shown that
mannuronan (ManA),, the expression of algL and algA, which are in the
6 L
downstream part of the alginate biosynthesis clus-
ter, are dependent on AlgU in P. aeruginosa, but
mannuronan-OAc not in A. vinelandii.43 Recently it has been reported
7 L that some strains of Pseudomonas syringae produce
alginate in response to copper.67
alginate
(-MGMMMGMGGGGMGMGGGM-)

Fig. 2. The pathway of alginate biosynthesis in bacteria. 1: 6 EPIMERIZATION, THE KEY TO


Phosphomannoisomerase, 2: phosphomannomutase, 3: D- ALGINATE STRUCTURE
mannose-l-phosphate guanylyl transferase, 4: guanosine
diphosphate mannose dehydrogenase, 5: glucosyl transferase;
The most unique feature of alginate biosynthesis is
n designates the number of uranic acid residues in the polymer
chain, 6: acetyl transferase, 7: mannuronan C-Sepimerase. that the synthesis of this heteropolymer involves
only one nucleotide-sugar (GDP-M), while the G-
designated Alg60) participate in the inner-membrane residues are introduced by epimerization at the
polymerization complex. 4o It has recently been polymer level. The first evidence of this mechanism
suggested that the periplasmic protein AlgK552 was published by Hellebust and Haug68 in 1969.
facilitates the transport of the alginate to the outer- They studied the photoassimilation of r4C03- into
membrane pore formed by AlgE53 (AlgJ54 in A. alginate by the algae Laminaria digitata, and
vinelandii). The epimerase (AlgG),4,55 lyase observed that in the presence of light the radio-
(AlgL)43,56 and acetylase (AlgF)57 are all peri- activity was mainly incorporated into M-blocks,
plasmic enzymes that modify alginate, but are while the amount of label in MG- and G-blocks
dispensable for alginate production. AlgI and increased during the night. A possible explanation
AlgJ58 have recently been shown to be necessary was found 2years later, when an enzyme-activity
for acetylation. which was able to epimerize mannuronic acid resi-
In both P. aeruginosa and A. vinelandii tran- dues on the polymer chain to guluronic acid resi-
scription of algD is dependent on the a-factor dues was demonstrated in the culture-medium of
AlgU.37,59 afgU is the first gene in the algU- A. vinelandii.69 Later mannuronan C-5-epimerases
mucABCD-operon,59,60 and its synthesis is auto- have been found both in P. aeruginosa55 and in
regulated. MucA is an antisigma-factor for AlgU, several species of brown algae.70,71 By controlling
and mucoid strains of P. aeruginosa are often the action of the C-Sepimerases, the organism can

I Iwo* \ 1
0000 5000

~r118.~~%?z?~SsleA.

Pig. 3. The alginate biosynthesis gene cluster in P. ueruginosa. Filled arrows show genes where the complementary genes have been
found in the corresponding order in A. vinelandii.The functions of the genes are described in the text.
Biosynthesis and applications of alginates 89

determine the content and distribution of G and thus Dllll


most of the properties of the alginate produced. AlgE4
The periplasmic AlgG from P. aeruginosa is
probably unable to make G-blocks, as that
block-type has never been found in alginates
from this species. A mutant with a defective algG AIgEl
has been shown to produce mannuronan.* Thus
the P. aeruginosu genome probably encodes only
one epimerase.
AlgE2
The secreted epimerase from A. vinelundii has
been shown to need calcium for activity,(j9 whereas
AlgG from A. vinelundii is independent of this
cation.41 A calcium-dependent epimerase has been AlgE3
purified and was shown to have the capacity to
incorporate G into both MG- and G-blocks.73
During the experiments aimed at cloning the gene
for the secreted epimerase from A. vinelundii, it was AlgE5
found that the bacterium encodes a family of at
least five calcium-dependent epimerases.74,75 Five Fig. 4. The modular structure of the AlgE-epimerases from A.
of these (AlgEl-5) have been cloned, sequenced vinelandii. A- and R-groups with the same hatchings belong
and expressed in Escherichiu coli. All the enzymes to the same subgroups defined by the construction of evolu-
tionary trees. The arrows indicate the locations of the putative
have been shown to be functional epimerases. The Ca*+-binding motifs. Open arrows indicate sequences that
sequences of ulgG and ulgEl-5 show little similar- deviate from the consensus sequence LXGGAGXDX. The
ity, but when the deduced sequences of the AlgE- symbol at the end of some of the R-modules indicates the
epimerases were compared they revealed a strong presence of short amino acid sequences (linkers). S refers to
the terminal 19 amino acids. These do not belong to the A- or
homology. All of them can be viewed as composed R-modules.
of one or two A-modules, being about 485 amino
acids long, and l-7 copies of a 153 amino acid-long Table 4. Epimerization of al&ate with a low (5%) content of G
module designated R (Fig. 4). The R-modules each with AIgE2 and AlgE4
contain 4-6 copies of a nine amino acid motif,
Enzyme Fo FM Distribution of diuronides
which has been identified in many secreted pro-
teins. In an alkaline phosphatase from P. uerugi- FGG FMM FMG,GM
nosu this motif has been shown to bind Ca*+ .76 AlgE274 0.40 0.60 0.34 0.54 0.06
The current hypothesis is that the A-modules are AlgE475 0.45 0.55 0.05 0.20 0.35
responsible for binding of alginate and for epimer-
ization, while the R-modules are involved in Ca* + -
binding and secretion. However, more experiments activity of the two enzymes and thus the structure of
are needed to confirm this hypothesis. the epimerized alginate. At low calcium levels the
The biological function of all these epimerases is relative amount of G-blocks will be higher, and the
not yet understood. However, comparisons of algi- polymer will thus form a stronger gel in spite of the
nate epimerized by AlgE2 and by AlgE4 gave valu- low calcium concentration. Still, this does not
able information. The experiments were performed explain why the genome encodes at least five AlgE-
by epimerizing alginate having a low content of G- epimerases. Since the bacterium produces different
residues (about 5%) with partially purified AlgE2 alginates at different stages of its life-cycle, it is
and AlgE4. The results (Table 4) show a clear dif- tempting to speculate that it regulates the degree of
ference: while AlgE2 mainly introduces G as G- epimerization by expressing different epimerases at
blocks, AlgE4 makes MG-blocks. In a paper by different times. It may also be possible that the other
Ofstad and Larsen77 it was shown that A. vinelundii epimerases make different distributions of MG- and
seemed to produce at least two epimerases, and that G-blocks or different block-lengths than AlgE2 and
the G-block-forming epimerase needed less calcium AlgE4. The function of the periplasmic epimerase
for activity than the MG-block-forming epimerase. (AlgG) in A. vinelundii is also not yet clear. Thus, a
Thus, given a mixture of AlgE2 and AlgE4 the lot of work remains before the epimerization pro-
Ca*+-concentration will determine the relative cess in this bacterium is fully understood.
90 H. Ertesvdg, S. Valla

7 FUTURE PROSPECTS REFERENCES

In most of the industrial uses of alginate, the 1. Otterlei,M., Ostgaard,K., Skjbk-Braek, G., Smidsred, O.,
properties which can be obtained by extracting Soon-Shiong, P. and Espevik, T., .I. Immunother., 1991,
10, 286.
alginate from different algae or parts of algae 2. Stokke, B. T., Smidsrerd, O., Bruheim, P. and Skjlk-Braek,
are satisfactory. But when it comes to encapsu- G., Macromolecules, 1991, 24, 4645.
lation of cells, especially for implantation, the 3. Grant, G. T., Morris, E. R., Rees, D. A., Smith, P. J. C.
requirements for gel strength and homogeneity are and Thorn, D., FEBS Lett., 1973,32, 195.
4. Draget, K. I., Skjbk-Brrek, G., Christensen, B. E.,
not so easily met. Much can be done by carefully GBsersd, 0. and Smidsrsd, O., Carbohydr. Polym., 1996,
chasing of alginates and fractionation. An alter- 29, 209.
native to fractionation would be to epimerize 5. Grasdalen, H., Larsen, B. and Smidsrnd, 0.. Carbohydr.
Res., 1981, 89, 179.
algal alginates with an initially low content of 6. Grasdalen, H., Carbohydr. Res., 1983, 118, 255.
G and G-blocks with recombinant enzymes. 7. Onserien, E., Carbohydr. Europe, 1996, 14, 26.
One might use AlgE2 to obtain G-blocks, or 8. Skjik-Braek, G. and Martinsen, A. In Seaweed Resources
in Europe: Uses and Potential, ed. M. D. Guiry and G.
AlgE4 to convert M-blocks to MG-blocks. By Blunden, 1991, p. 219.
using a mixture of the two it should be possible to 9. Martinsen, A., Skjlk-Brrek, G. and Smidsrerd, O., Bio-
obtain an alginate with the desired properties with technol. Bioengng, 1989, 33, 79.
10. Thu, B., Bruheim, P., Espevik, T., Smidsrsd, O., Soon-
respect to gel-strength, pore-size and immunogen- Shiong, P. and Skjik-Braek, G., Biomaterials, 1996, 17,
icity. Such an epimerized alginate would also be 1031.
more homogenous than that usually obtained from 11. Thu, B., Bruheim, P., Espevik, T., Smidsrerd, O., Soon-
natural sources. Shiong, P. and Skjik-Brrek, G., Biomaterials, 1996, 17,
1069.
An alternative to the use of algal alginate is to 12. Elchin, Y. M., Biomaterials, 1995, 16, 1157.
produce the polymer in bacteria. Earlier attempts 13. Roig, M. G., Rashid, D. H. and Kennedy, J. F., Appl.
to do this have mostly been hampered by the Biochem. Biotechnol., 1995, 55, 95.
14. Chung, Y. C., Huang, C. and Tseng, C. P., J. Environ. Sci.
presence of an alginate lyase in both P. aeruginosa Hlth Part A: Environ. Sci. Engng Toxic Hazar. Subst.
and A. vinelandii. Even though mucoid strains of Control, 1996, 31, 1263.
P. aeruginosa usually cease to produce alginate 15. Cheetham, P. S. J., Garretth, C. and Clark, J., Biotechnol.
Bioengng, 1985,27,471.
when cultured in the laboratory, stable alginate- 16. Musgrave, S. C., Kerby, N. W., Codd, G. A. and Stewart,
producing mutants have been isolated.78 And as a W. D. P., Biotechnol. Lett., 1982, 4, 647.
gene for alginate lyase now has been identified in 17. Eikmeier, H. and Rehm, H. J., Naturforsh. Sect. C.
Biosci., 1987, 42, 408.
both species, it should be possible to inactivate it
18. Oda, G., Sameyiuna, H. and Yameda, T. In Proceedings
and thus construct strains that are more suitable Biotechnology, London, 1983, p. 597.
for industrial production of alginate. 19. Hirata, K., Phunchindawan, M., Tukamoto, J., Goda, S.
It may also be possible to tailor the polymer in and Miyamoto, K., Cryo. Lett., 1996, 17, 321.
20. Bailliez, C., Largeau, C. and Casadevall, E., Appl. Micro-
vivo by expressing one or more of the recombi- biol. Biotechnol., 1985,23, 99.
nant AlgE-epimerases in an alginate-producing 21. Timbert, R., Barbotin, J. N. and Thomas, D.. Plant Sci.,
host. It would probably be an advantage if this 1996, 120,215.
22. Alfermann, A. W., Schuller, I. and Reinhardt, E., Planta
host produced alginates with a very low G-content. Medica, 1980, 40, 218.
The algG-mutant of P. aeruginosa meets this 23. Maysinger, D., Berezovskaya, 0. and Feodoroff, S., Exp.
requirement, but it may not be convenient to use Neurol., 1996, 141, 47.
24. Duff, R. G., TIBTECH, 1983,3, 167.
an opportunistic human pathogen for fermen-
25. Soon-Shiong, P., Feldman, E., Nelson, R., Heintz, R.,
tation. The problem with A. vinelundii is that it Yao, Q., Yao, Y., Zheng, T., Merideth, N., Skjik-Braek,
has several epimerase genes, all of which prob- G., Espevik, T., Smidsrcad, 0. and Sandford, P., Proc.
ably have to be inactivated in order to produce Nat1 Acad. Sci. USA, 1993,90, 5843.
26. Soon-Shiong, P., Heintz, R. E., Merideth, N., Yao, Q. X.,
mannuronan. Yao, Z., Zheng, T., Murphy, M., Moloney, M. K.,
When the structure-function relationships of the Schmehl, M., Harris, M., Mendez, R., Mendez, R. and
different epimerases are more fully understood, it Sandford, P. A., Lancet, 1994, 343, 950.
27. Kulseng, B., Thu, B., Espevik, T. and SkjHk-Brrek, G.,
may be possible to create new epimerases with Cell. Transplant., 1997, 6, in press.
desired technical properties by site-specific muta- 28. Skjlk-Brrek, G. and Espevik, T., Carbohyd. Europe, 1996,
genesis or by exchanging modules or parts of 14, 19.
29. Gorin, P. A. J. and Spencer, J. F. T., Can. J. Chem., 1966,
modules between different epimerase genes. These
44,993.
could then be utilized to epimerize alginate in vitro 30. Cote, G. L. and Krull, L. H., Carbohydr. Res., 1988, 181,
or in vivo. 143.
Biosynthesis and applications of alginates 91

31. Linker, A. and Jones, R. S., Nature, 1964, 204, 187. 56. Schiller, N. L., Monday, S. R., Boyd, C. M., Keen, N. T.
32. Govan, J. R. W., Fyfe, J. A. M. and Jarman, T. R., J. and Ohman, D. E., J. Bacterial., 1993, 175,478O.
Gen. Microbial., 1981, 125, 217. 57. Franklin, M. J. and Ohman, D. E., J. Bacterial., 1993,
33. Fett, W. F., Cescutti, P. and Wijey, C., J. Appl. Bacterial., 175, 5057.
1996, 81, 181. 58. Franklin, M. J. and Ohman, D. E., J. Bacterial., 1996,
34. Skjak-Brmk, G., Grasdalen, H. and Larsen, B., Carbo- 178, 2186.
hydr. Res., 1986, 154, 239. 59. Martinez-Salazar, J. M., Moreno, S., Najera, R., Boucher,
35. Sadoff, H. L., Bacterial. Rev., 1975, 39, 516. J. C., Espin, G., Sober&t-Chavez, G. and Deretic, V., J.
36. Page, W. J. and Sadoff, H. L., J. Bacterial., 1975, 122, Bacterial., 1996, 178, 1800.
145. 60. Boucher, J. C., Martinez-Salazar, J., Schurr, M. J., Mudd,
37. Deretic, V., Schurr, M. J., Boucher, J. C. and Martin, M. H., Yu, H. and Deretic, V., J. Bacterial., 1996, 178,
D. W., J. Bacterial., 1994, 176, 2773. 511.
38. Lin, T. Y. and Hassid, W. Z., J. Biol. Chem., 1966, 241, 61. Wozniak, D. J. and Ohman, D. E.. J. Batteriol., 1994,
5284. 176,6007.
39. Pindar, D. E. and Bucke, C., Biochem. J., 1975, 152, 617. 62. Baynham, P. J. and Wozniak, D. J., Mol. Microbial.,
40. May, T. B. and Chakrabarthy, A. M., Trends Microbial., 1996, 22, 97.
1994, 2, 151. 63. Delic-Attree, I., Touissant, B., Froger, A., Willison, J. C.
41. Rehm, B. H., Ertesvag, H. and Valla, S., J. Bacterial., and Vignais, P. M., Microbiology, 1996, 142, 2785.
1996, 178, 5884. 64. Hasset, D. J., Howell, M. L., Sokol, P. A., Vasil, M. L.
42. Shinabarger, D., Berry, A., May, T. B., Rothmel, R., and Dean, G. E., J. Bacterial., 1997, 179, 1442.
Fialho, A. and Chakrabarthy, A. M., J. Biol. Chem., 1991, 65. Schlichtman, D., Kubo, M., Shankar, S. and Chakra-
266, 2080. barty, A. M., J. Bacterial., 1995, 177, 2469.
43. Lloret, L., Barreto, R., Leon, R., Moreno, S., Martinez- 66. Yu, H., Mudd, M., Boucher, J. C., Schurr, M. J. and
Salazar, J., Espin, G. and Soberon-Chavez, G., Mol. Deretic, V., J. Bacterial., 1997, 179, 187.
Microbial., 1996, 21, 449. 67. Kidambi, S. P., Sundin, G. W., Palmer, D. A., Chakra-
44. Deretic, V., Gill, J. F. and Chakrabarty, A. M., Nucf. barty, A. M. and Bender, C. L., Appl. Environ. Microbial.,
Acids Res., 1987, 15,4567. 1995,61, 2172.
45. Campos, M.-E., Martinez-Salazar, J. M., Lloret, L., 68. Hellebust, J. A. and Haug, A. In Proceedings of the 6th
Moreno, S., Nuiiez, C., Espin, G. and Soberon-Chavez, International Seaweed Symposium, 1969, p. 463.
G., J. Bacterial., 1996, 178, 1793. 69. Larsen, B. and Haug, A., Carbohydr. Res., 1971, 20, 225.
46. Zielinski, N. A., Chakrabarty, A. M. and Berry, A., J. 70. Magdwick, J. C., Haug, A. and Larsen, B., Acta Chem.
Biof. Chem., 1991,266,9754. &and., 1973, 27, 3592.
47. Maharaj, R., May, T. B., Wang, S.-K. and Chakrabarty, 71. Ishikawa, M. and Nisizawa, K., Bull. Jpn. Sot. Sci. Fish.,
A. M., Gene, 1993,136, 267. 1981, 47, 889.
48. Mejia-Ruiz, H., Moreno, S., Guzman, J., Najera, R. and 72. Chitnis, C. E. and Ohman, D. E., J. Bacterial., 1990, 172,
Soberon-Chavez, G., GenBank accession no. YO8819. 2894.
49. Saxena, I. M., Brown, R. M. Jr., Fevre, M., Geremia, 73. Skjbk-Brrek, G. and Larsen, B., Carbohydr. Res., 1985,
R. A. and Henrissat, B., J. Bacterial., 1995, 177, 1419. 139, 273.
50. Monday, S. R. and Schiller, N. L., J. Bacterial., 1996, 178, 74. ErtesvPg, H., Doseth, B., Larsen, B., Skjak-Brick, G. and
625. Valla, S., J. Bacterial., 1994, 176, 2846.
51. Aarons, S. J., Sutherland, I. W., Chakrabarty, A. M. and 75. Ertesvag, H., Hoidal, H. K., Hals, I. K., Rian, A., Doseth,
Gallagher, M. P.. Microbiology, 1997, 143, 641. B. and Valla, S., Mol. Microbial., 1995, 16, 7 19.
52. Mejia-Ruiz, H., GenBank accession no. X98863. 76. Baumann, U., Wu, S., Flaherty, K. M. and McKay, D. B.,
53. Chu, L., May, T. B., Chakrabarty, A. M. and Misra, EMBO J., 1993, 12, 3357.
T. K., Gene (1991) 1. 77. Ofstad, R. and Larsen, B. In Proceedings of the 10th
54. Rehm, B. H. A., Microbiology, 1996, 142, 873. International Seaweed Symposium, 1980, p. 485.
55. Franklin, M. J., Chitnis, C. E., Gacesa, P., Sonesson, A., 78. Darzins, A. and Chakrabarty, A. M., J. Bacterial., 1984,
White, D. C. and Ohman, D. E., J. Bacterial., 1994, 1821. 159, 9.

Potrebbero piacerti anche