El-Enshashy Et Al 2003
El-Enshashy Et Al 2003
El-Enshashy Et Al 2003
net/publication/8902571
CITATIONS READS
35 710
2 authors:
Some of the authors of this publication are also working on these related projects:
Bacterial chitinases, and proteases - biochemical properties and regulation of synthesis View project
All content following this page was uploaded by Urszula Jankiewicz on 26 November 2014.
2003
POLSKIE TOWARZYSTWO MIKROBIOLOGÓW
EDITORS
K.I. Wolska (Editor in Chief)
K. Kote³ko, L. Sedlaczek, E. Strzelczyk, A. Kraczkiewicz-Dowjat
E.K. Jagusztyn-Krynicka (Scientific Secretary)
EDITORIAL BOARD
President: Zdzis³aw Markiewicz (Warsaw, Poland)
Ryszard Chróst (Warsaw, Poland), Miros³aw Kañtoch (Warsaw, Poland), Donovan Kelly (Warwick, UK),
Tadeusz Lachowicz (Wroc³aw, Poland), Wanda Ma³ek (Lublin, Poland), Andrzej Piekarowicz (Warsaw,
Poland), Anna Podhajska (Gdañsk, Poland), Gerhard Pulverer (Cologne, Germany), Geoffrey Schild
(Potters, Bar, UK), Torkel Wadström (Lund, Sweden), Jadwiga Wild (Madison, USA),
Miros³awa W³odarczyk (Warsaw, Poland)
EDITORIAL OFFICE
Miecznikowa 1, 02-096 Warsaw, Poland
tel. 48 (22) 55 41 302, Tuesday and Thursday from 10 A.M. till 2 P.M.
fax 48 (22) 55 41 402
e-mail izabelaw@ biol.uw.edu.pl
Front cover: Bacillus subtilis Hly entering Int 407 epithelial cells
(courtesy of Jaros³aw Winiewski, M.Sc. and Magdalena Sobolewska, Ph.D.)
Sk³ad i druk:
Zak³ad Wydawniczy Letter Quality
Warszawa, ul. Brylowska 35/38, tel. 631 45 18, 607 217 879
Nak³ad 500 + 15, Drukarnia PUIP Jachranka 94
Acta Microbiologica Polonica
2003, Vol. 52, No 3
CONTENTS
MINIREVIEW
ORIGINAL PAPERS
Abstract
Aminopeptidases are enzymes that release N-terminal amino residues from oligopeptides,
polypeptides and proteins. The classification of aminopeptidases has often been based on mecha-
nism of catalysis, structure of active site, substrate specificity kinetic and molecular properties. In
terms of catalytic mechanism bacterial aminopeptidases can be divided into three main catalytic
groups: metallo-, cysteine- and serine aminopeptidases. According to their substrate specificity
the enzymes can be ordered into two sub-groups: having broad or narrow specificity. Almost half
of the characterized aminopeptidases show a subunit structure. Enzymes having a quaternary
structure are most often built of a combination of 2, 4, 6 subunits. Bacterial aminopeptidases may
be localised in the cytoplasm, on membranes, associated with the envelope or secreted into the
extracellular media. Regulation of the synthesis of aminopeptidases is assumed to take place
mainly at the level of transcription. Most genes encoding the enzymes are monocistronic and
contain a promotor characteristic for the genes transcribed by RNA polymerase associated with
the factor F70. Aminopeptidases play an important role in the initial and final steps of protein
turnover and they are involved in several specific regulatory functions.
Introduction
Because of the mechanism of catalysis and the structure of their active site, bacte-
rial aminopeptidases may be sub-divided into three main catalytic groups: metallo-
aminopeptidases, cysteine aminopeptidases and serine aminopeptidases.
Metallo-aminopeptidases are the most numerous group comprising 60% of these
enzymes. These are enzymes whose activity is inhibited by metal chelating compounds
e.g. EDTA, hydroxyquinoline, and 1, 10 phenanthroline and also by 3-amino-2-hy-
droxy-4-phenylbutanoyl-L-leucine (bestatin) and by 3-amino-2-hydroxy-5-methyl-
hexanoyl-L-valil- L-valil-L-aspartic acid (amastatin). The metal ion is most often
combined through coordination bonds with two residues of histidine and with one
residue of glutamic acid. The catalysis requires, apart from metal ligands, also at least
one residue of an amino acid, often glutamic acid, arginine or lysine. Mechanism of
reactions catalysed by these enzymes is not yet fully explained. It is assumed that the
cation of a metal markedly increases the reactivity of combined water molecule mak-
ing easier the nucleophilic attack on carbonyl carbon of the hydrolysed peptide bond
and in addition it stabilises the transitional stage of the reaction (R a w l i n g s, 1998;
H o l z, 2002). In spite of the fact that the identification of the metal ion participating
in catalysis is not always possible, it was found that the most numerous group are zinc
dependent aminopeptidases (G o n z a l e s and R o b e r t - B a u d o u y, 1996). Two
sub-groups have been distinguished within this group. The first comprises aminopep-
tidases with two zinc ions in the catalytic site of the enzyme. Here belong bacterial
intracellular leucine aminopeptidases called Pep A showing a homology of amino
acid sequences with the animal leucine aminopeptidase (S t i r l i n g et al., 1989;
B u r l e y et al., 1992; T a y l o r, 1993a) and extracellular bacterial leucine amino-
peptidases (P r e s c o t t and W i l k e s, 1966; S p u n g i n and B l u m b e r g,
1989; C a h a n et al., 2001; H o l z, 2002). Aminopeptidases of the second group
contain at least one zinc ion in the catalytic site. Aminopeptidases of the N and A type
belong to this subgroup (G e i s s et al., 1985; N i v e n et al., 1995; K l e i n, 1998;
B u t l e r, 1998). A group of enzymes containing in the active site of each subunit two
Co+2 ions was also distinguished from among metal dependent aminopeptidases. Such
mechanism of catalysis was described for methionine aminopeptidase from Escherichia
coli (R o d e r i c k and M a t t h e w s, 1993, L o w t h e r and M a t t h e w s, 2000),
from Lactobacillus plantarum (Macedo et al., 2003) and for extracellular aminopep-
tidase from Bacillus sp. N2 (L e e et al., 1998). Type T aminopeptidases synthesised
3 Minireview 219
only by thermophilic and extremophilic strains of bacteria were also included among
metallo-aminopeptidases containing zinc or cobalt in their active sites (M o t o s h i m a
and K a m i n a g a w a, 1998, F e r n a n d e z - E s p l a and R u l, 1999).
Additional types of catalysis were also found in aminopeptidases dependent on
other than the mentioned metal ions. Mn+2 and Mg+2 focused particular interest since
they are able to activate enzymes or to remove the activity of some metal dependent
aminopeptidases. In the active site of aminopeptidase P isolated from E. coli there
are two ions of Mn+2 (Y o s h i m o t o et al., 1989; Z h a n g, 1998) while aminopep-
tidase isolated from the strain of Thermatoga maritime was classified as magnesium
dependent (R a t n a y a k e et al., 2003).
Cysteine aminopeptidases contain SH groups of cysteine and most often histidine,
asparagine and glutamic acid residues in the active site. In contrast to metallo-ami-
nopeptidases, they do not contain an ionic co-factor in their structure. The reaction
starts with the nucleophilic attack of the sulfur of the sulphydril group on the carbo-
nyl carbon of the peptide bond. The enzymes are specifically inhibited by
iodoacetamide, iodoacetate, N-ethyl-maleimide, p-chloromercuribenzoate (pCMB),
trans-epoxysuccinyl-L-leucylamido-4-guanidino-butane (E-64). The activity of this
group of enzymes is decreased by some serine inhibitors: TLCK, TPCK, leupeptin,
antipain. Activators of this group are: cysteine, dithiothreitol (DTT) and some chelat-
ing agents like EDTA. The group comprises enzymes, whose mechanism of catalysis
is much better understood than that of metal-dependent aminopeptidases. Type C ami-
nopeptidases and other cysteine aminopeptidases belong to this group. The former
have been purified only from lactic acid fermentation bacteria. This is a family of
cytoplasmic aminopeptidases, closely related in structure and activity to bleomycin
hydrolase (N e v i a n i et al., 1989; C h a p o t - C h a r t i e r et al., 1993; C h a p o t -
C h a r t i e r et al., 1994; M i s t o u and G r i p o n, 1998; d e P a l e n c i a et al.,
2000). Thiol aminopeptidases that do not show homology of amino acid sequences
with aminopeptidases type C are distinguished in the literature as other cysteine ami-
nopeptidases. Here belong acrylamidase from the strain of Neisseria catarrhalis
(B e h a l and C o x, 1968) and pyrrolidone carboxyl peptidases (A w a d é et al.,
1994; P a t t i et al., 1995; L e S a u x and R o b e r t - B a u d o u y, 1997). Ami-
nopeptidase specifically inhibited by cysteine enzyme inhibitors was also isolated
from the Pseudomonas sp. (J a n k i e w i c z and B i e l a w s k i, 2002a).
Bacterial serine aminopeptidases are the least numerous group of aminopeptidases
that do not belong to any of the known families of serine proteolytic enzymes repre-
sented by trypsin and subtilisin (R a w l i n g s and B a r r e t t, 1994). Diagnostic
inhibitors for serine aminopeptidases are diisopropyl fluorophosphate (DFP) and
phenylmethylsulfonyl fluoride (PSMF). Serine aminopeptidases are also inhibited by
ketones: N-p-Tosyl-L-phenylalanine chloromethyl ketone (TPCK) and N-p-Tosyl-L-
lysine-chloromethyl ketone (TLCK) but these are not specific inhibitors since they
act also on other cysteine aminopeptidases.
Among the serine aminopeptidases are proline aminopeptidases which have also
been called prolineiminopeptidases (PIP), containing a triad of serine, histidine and
aspartic acid in the active site (K i t a z a n o et al., 1994; M o r e l et al., 1999) and
220 Jankiewicz U., Bielawski W. 3
D-aminopeptidase, in which four amino acid residues (3 of Ser and 1 of Lys) are found
in the active site. Lysine plays a role of proton acceptor during the nucleophilic attack of
the serine hydroxyl group on carbonyl carbon of the peptide bond (A s a n o, 1998;
F a n u e l et al., 1999, A s a n o and L u b b e h u s e n 2000, K o m e d a et al., 2003).
Substrate specificity
aminopeptidases specific to N-end alanine, arginine, leucine and lysine were also pu-
rified from strains of and Pseudomonas fluorescens ATCC 948 and Pseudomonas sp.
(G o b b e t t i et al., 1995; J a n k i e w i c z and B i e l a w s k i, 2001; J a n k i e w i c z
and B i e l a w s k i, 2002b).
Type C aminopeptidases belongs also to those of broad substrate specificity. They
release N-terminal residues of Ala, Lys, Arg, Met and Phe most easily. Enzymes of
that group show a lack or very low activity with substrates containing proline in the
P1 and P1 position (Ve s a n t o et al., 1994; W o l h r a b and B o c k e l m a n n,
1993, d e P a l e n c i a et al., 2000). From among enzymes of broad substrate speci-
ficity there are also cysteine aminopeptidases specifically inhibited by p-CMB and
showing no homology in amino acid sequences with aminopeptidases C.
Aminopeptidases of a narrow substrate specificity are divided into 6 sub-groups,
with the kind of cleaved N-end amino acid being the criterion for distinction.
1. Methionine aminopeptidases called aminopeptidases M. These are enzymes pre-
ferring substrates that contain methionine in the N-end position of peptide chain.
Aminopeptidase M was characterised from e.g. strains of E. coli and Bacillus
subtilis (N a k a m u r a et al., 1990; R o d e r i c k and M a t t h e w s, 1993).
2. Aspartate aminopeptidases A called also glutamyl aminopeptidases. Enzymes of
this group show substrate specificity for peptides containing residues of aspartic
of glutamic acid (E x t e r k a t e and d e Ve e r, 1987; B a c o n et al., 1994).
3. Pyrrolidone carboxyl peptidase (pyrase) prefers substrates containing pyroglutamic
acid which forms as a result of spontaneous, intracellular cyclization of glutamic
acid (D o o l i t t l e, 1970).
4. Arginine aminopeptidases enzymes of this group demonstrate specificity to sub-
strates with the N-end arginine. Arginine aminopeptidases were isolated from only
several strains of Streptococcus and E. coli (I s h i n o et al., 1987; F l o d e r u s,
1990, G o l d s t e i n et al., 2002; J o b i n and G r e n i e r, 2003)
5. Aminopeptidases P detach N-end amino acid from substrates containing proline
in the P1 position. Aminopeptidase P was obtained from Streptomyces lividans
and Salmonella typhimurium (M c H u g h and M i l l e r, 1974; B u t l e r et al.,
1993; B u t l e r et al., 1994, M c D o n n e l et al., 1997).
6. Proline aminopeptidases are specific for substrates that contain proline at the
N-end. Such aminopeptidases were isolated e.g. from E. coli, bacteria of the
genus Lactobacillus, and from Arthrobacter nicotianae (K u n j i et al., 1996;
S m a c c h i et al., 1999).
The activity of aminopeptidases of narrow specificity is often determined not only
by amino acid situated in the last position at the N-end but also by that in the last
but one or further positions. Proline iminopeptidase hydrolyses peptide bonds in
substrates containing N-end proline on condition that neither Lys nor Phe is the next
in polypeptide chain (Y o s h i m o t o et al., 1983). Similarly, methionyl aminopepti-
dase shows activity against substrates with N-terminal methionine only if P1 posi-
tion is occupied by alanine, glycine, proline, serine or threonine. The presence of
arginine, leucine, lysine or phenylalanine totally inhibits the activity of this aminopep-
tidase (B e n - B a s a d et al., 1987). Amino acids in positions P1, P1, P2, P3 and
222 Jankiewicz U., Bielawski W. 3
P4 affect the kinetic constant of the reaction of aminopeptidase P from E. coli
(Y o s h i m o t o et al., 1994). Aminopeptidases of a narrow specificity e.g. proline
or glutamine aminopeptidase can hydrolyse substrates with N-end amino acids unaf-
fected by aminopeptidases of broad specificity.
Subunit structure
Almost half of the already known aminopeptidases show a subunit structure. En-
zymes having a quaternary structure are most often built of a combination of 2, 4, 6
subunits. Most frequent is the homomeric subunit system. Only several yet described
aminopeptidases have the heteromultimeric structure. Subunit structure possess intra-
cellular bacterial leucine aminopeptidases i.a. enzymes isolated from Pseudomonas
putida of a mass of 400 kDa built of 8 identical subunits (H e r m e s et al., 1993),
those isolated from Brevibacterium linens SR 3 built of 12 subunits 18 kDa each
(F e r n a n d e z et al., 2000) or aminopeptidase A from E. coli combined of
6 homomeric subunits 55 kDa each (Vo g t, 1970). Aminopeptidase isolated from
Mycoplasma salivarium has a heteromeric subunit structure. It consists of subunits
with a mass of 50 and 47 kDa (S h i b a t a et al., 1987). Aspartate aminopeptidases
3 Minireview 223
or aminopeptidases type C from bacteria of lactic acid fermentation also show subunit
structure (N i v e n, 1991; B a a n k r e i s, 1992; d e P a l e n c i a et al., 2000).
Interesting results were presented by S t o l l et al. (1973) from their studies on
metallo-aminopeptidase from Bacillus stearothermophilus. The enzyme was built
of 12 identical subunits, 36 kDa each. Analysis of the amino acid sequence of this
enzymatic protein proved the existence of two distinct subunits types. The enzymes
of monomeric structure involve also aminopeptidases type N of a molecular mass
between 78 and 99 kDa (T a n et al., 1990; R u l et al., 1994; C h a v a g n a t et al.,
1999). The so far recognised extracellular aminopeptidases belong also to monomeric
enzymes with the exception of dimeric (2×33 kDa) aminopeptidase excreted by
Alteromomonas B-207 (M e r k e l et al., 1981). The mass of extracellular leucine
aminopeptidases is small and usually varies between 20 and 30 kDa (S p u n g i n
and B l u m b e r g, 1989; V i t a l e et al., 1986; T o m a and H o n m a, 1996) while
those of arginine aminopeptidase and prolineiminopeptidase are 70 and 53 kDa,
respectively (G o l d s t e i n et al., 2002, S m a c c h i et al., 1999).
Though the amino acid sequence is known and the homology of encoding genes is
established e.g. for leucine, C and N aminopeptidases from various bacterial strains,
information on the regulation of these genes expression is still incomplete. Regula-
tion of the synthesis of enzymatic proteins in Procaryotes is assumed to take place
mainly at the level of transcription. Most genes encoding bacterial aminopeptidases
are monocistronic and contain a promotor characteristic for the genes transcribed
by RNA polymerase associated with the factor F70 (G o n z a l e s and R o b e r t -
B a u d o u y, 1996). Regulation of aminopeptidase encoding genes is most often stud-
ied in Enterobacteria and particularly in E. coli and Salmonella typhimurium. The
relatively best characterised enzyme in E. coli is aminopeptidase N. The enzyme
is synthesised during the whole life cycle of the bacteria but the expression of the
gene encoding this enzyme increases in the case of phosphorus or oxygen deficit
even four-fold while carbon and nitrogen deficiencies do not affect the activity
(L a z d u ñ s k i et al., 1975, G h a r b i et al., 1985). The molecular base of this
regulation is not known. The synthesis of pyroglutamic aminopeptidase in Pseudo-
monas fluorescens is induced under iron deficiency and in the presence of the product
pyroglutamic acid (L e S a u x and R o b e r t - B a u d o u y, 1997). In spite of the
fact that catabolic repression is the common model of regulation of the proteolytic
enzymes biosynthesis, only one gene regulated in that way was characterised in bac-
teria. The expression of a gene encoding aminopeptidase E (dipeptide aminopepti-
dase) with the specificity for N-end Asp in S. typhimurium is controlled by catabolic
repression (C o n l i n et al., 1994). The physiological basis of such type of regula-
tion is not fully explained. The authors suggest that this might be a way of adaptation
of bacteria to limited carbon resources. Released aspartic acid can serve as a source of
carbon or energy for the bacteria end may enable the synthesis of other amino acids
224 Jankiewicz U., Bielawski W. 3
like Asn, Lys, Met. Protein molecules of extracellular aminopeptidases undergo post-
translational modifications. Extracellular aminopeptidases are synthesised in the form
of inactive precursors (preproenzymes) built of four or three domains: typical signal
sequence, proteolytic domain, N-end propeptide and often of C-end propeptide. N-end
propeptide is given a function of an intracellular chaperon responsible for proper fold-
ing of the particle of enzymatic protein. Moreover, it was found that N-end propeptides
are often inhibitors of the activity of the proteolytic domain. C-end propeptide partici-
pates in the secretion of this protein. After cleavage off the signal sequence and usu-
ally the double maturation at the N- and C-terminus they appear as active enzymes
(N i r a s a w a et al., 1999; Z h a n g et al., 2000; T a n g et al., 2002).
Another mechanism of regulation of the enzyme activity was described for E. coli,
where an endogenous competitive inhibitor of N-aminopeptidase was found in the
cell (Y a n g and S o m e r w i l l e, 1976).
Many studies on the effect of environmental factors on the production of amino-
peptidases have been carried out. However, the obtained results concerned enzyme
activities only and therefore they could not be used to determine of the level of regu-
lation, i.e. transcriptional, translational or post-translational.
Both constitutive and induced enzyme synthesis take place in various growth
phases of bacteria and are affected by various factors. Induced expression of the gene
encoding aminopeptidase synthesised by Bacillus stearothermophilus takes place at
a high temperature (M o s e r et al., 1970; S t o l l et al., 1972). It is known that the
presence of peptides in culture medium induces the expression of dipeptidase in bac-
teria of the lactic acid fermentation (A t l a n et al., 1989), which might suggest that
the enzyme plays a role in supplying the cell with free amino acids. In a strain of
Lactococcus lactis, however, the activity of aminopeptidase N decreased in the pres-
ence of dipeptides at high concentrations in medium (M e i j e r et al., 1996). Results
of experiments on the effect of medium composition on the biosynthesis of several
aminopeptidases of different substrate specificity in Pseudomonas fluorescens proved
the presence of both induced and constitutive aminopeptidases in this strain
(G o b e t t i and R o s s i, 1992). C h o i et al., (1996) presented results on the regu-
lation mechanism of the synthesis of extracellular aminopeptidase. The experiment
was carried out on two strains of L. casei cultured on various media. Casein contain-
ing medium induced the synthesis of extracellular aminopeptidases while medium
with glucose did not bring such effect. Most experiments carried out so far did not
give a clear picture of the regulation of aminopeptidase synthesis in bacteria and there-
fore, the problem still focuses much attention.
Apart from the mentioned functions, bacterial aminopeptidases play some other
roles in the cell. S t i r l i n g et al. (1989) reported on the particular structural role of
aminopeptidase A in E. coli in stabilising plasmid COL E. An additional role is played
also by D aminopeptidase, which participates in the synthesis and degradation of pep-
tidoglycan (A s a n o et al., 1992). It is expected that some aminopeptidases take
place in the activation and transport of antibiotics into the cell. Aminopeptidase N in
E. coli and aminopeptidases A and N in S. typhimurium are responsible for the activa-
tion of the antibiotic albomycin in the cytoplasm. Mutation of genes encoding amino-
peptidase N in E. coli and aminopeptidases A and N in S. typhimurium resulted in
the insensitivity of the bacteria to this antibiotic (B r a u n et al., 1983). The partici-
pation of aminopeptidases in the degradation of toxic peptides and inactivation of
physiologically important proteins has also been suggested (M i l l e r et al., 1975).
Literature
Abstract
In the environment horizontal DNA transfer between various bacterial species and genera takes
place by transformation, transduction, but mainly by conjugation. Conjugation is responsible for
the spread of genes coding for antibiotic resistance and xenobiotic degradation. Transfer events
are reported in animal, rhizosphere and phylloplane ecosystems and in non polluted and polluted
water and soil. Genetic exchange between Bacteria and Archaea is also observed. Evaluation of
the extent of interspecies gene transfer is crucial in view of the deliberate release of a variety
of unmodified and genetically modified microorganisms into the natural environments.
Introduction
Horizontal DNA transfer between various bacterial species and genera is quite
common in the environment. Of the three mechanisms of genes transfer transfor-
mation, transduction and conjugation, the last one facilitated by conjugative plas-
mids and transposons is the most often observed (D a v i d s o n, 1999). Conjugation
is responsible for the spread of the antibiotic resistance genes (D a v i e s, 1996;
M a n z e l and D a v i e s, 1999) and xenobiotic degradation genes (V a n d e r
M e e r et al., 1992). The presence of efficient donors in heterologous bacterial
populations can accelerate plasmid transfer and spread by several orders of magni-
tude (D i o n i s i o et al., 2002).
The extent of horizontal transfer can be estimated by an analysis of microbial
genome sequences and codon usage patterns (B u s h m a n, 2002a). It was shown
that even different strains of the same species display remarkable diversity. 17.6%
of Escherichia coli genes have been received by horizontal transfer and the ge-
nomes of the laboratory K-12 strain and pathogenic O157:H7 strain are remarkably
* phone (48 22) 554 1302; e-mail: [email protected]
234 Wolska K.I. 3
different. The two genomes differ by more than a megabase, the pathogenic strain
being larger. Differences are also observed in various Helicobacter pylori and
Chlamydia trachomatis strains (after B u s h m a n, 2002a). The discovery of exten-
sive lateral transfer in bacteria causes that their tree of life appears to be highly
reticulated rather than branched in an orderly way (B u s h m a n, 2002b).
The detection of environmental gene transfer creates severe problems. Only when
easily selectable phenotypes are available, genetic transfer experiments may be per-
formed under natural conditions and the relevant phenotype selected. The use of
green fluorescent protein, GFP (D a h l b e r g et al., 1998) removes the need to
select the phenotype and to cultivate transconjugants. It should be mentioned here
that less than 1% of bacteria are cultivable using the available techniques (A m a n n
et al., 1995). The ability to detect gene transfer may be interfered with restriction
systems present in the recipient cells, the inability of a plasmid to replicate in a new
recipient, the inability of chromosomal DNA to recombine with the recipient chro-
mosome and the lack of incoming transposon integration (D a v i d s o n, 1999). Also
mismatch repair systems (M a t i c et al., 1996) negatively influence recombination
and therefore also the detection of gene transfer. Most studies on environmental
gene transfer have been performed in microcosms designed to represent natural
environmental conditions but permitting their manipulation (D a v i d s o n, 1999).
The knowledge about the extent of horizontal gene transfer is extremely important
in view of the deliberate release of a variety of nonrecombinant and recombinant
microorganisms into the environment for such purposes as nitrogen fixation, phos-
phate solubilization, control of phytopathogenic fungi and bacteria, plant growth stimu-
lation, insect and weed control and bioremediation (W i l s o n and L i n d o w, 1993).
In this review gene transfer events between various bacteria reported in different
environments are described with special emphases on conjugation. Transfer between
Bacteria and Archaea is also noticed. Transfer between Bacteria and Eukarya e.g.
from Agrobacterium tumefaciens to plants (Z u p a n and Z a m b r y s k i, 1995),
to yeast (B u n d o c k and H o o y h a a s, 1996), from E. coli to Saccharomyces
cerevisiae (H e i n e m a n and S p r a g u e, 1989), from transgenic plants to
phytosphere (D r o g e et al., 1998) and terrestial bacteria (N i e l s e n et al., 1998),
DNA transfer to animal cells by intracellular bacteria (G r i l l o t - C o u r v a l i n
et al., 1998) and from Bacteria to Eukarya via endosymbiosis (D o o l i t t l e, 1998)
are beyond the scope of this minireview.
Conjugation
The vast majority of bacterial gene transfer in the environment involves conjuga-
tion. Important, however arbitrarily chosen, examples of conjugational events in the
environment are summarized in Table I. Only in some cases DNA is transferred
from a known bacterial donor to known recipient, more often the donor or the recipi-
ent may be unknown indigenous bacteria. Conjugative events were reported in ani-
mal ecosystems, rhizosphere, plant leaves, nonpolluted and polluted water and soil.
3 Minireview 235
Table I
Bacterial conjugation in various ecosystems
MR-P, heavy metal resistant plasmid; AR-P, antibiotic resistant plasmid; cat-P, catabolic plasmid; cat-Tn, catabolic
transposon; sym-P, nitrogen fixing symbiotic plasmid ; sym-I, chromosomal, nitrogen fixing symbiotic island;
Bt-P, B. thuringiensis insect toxin plasmid; gfp-P, green fluorescent protein gene; ?, possible or unknown
From D a v i d s o n, 1999, modified
Conjugative transfer of genes coding for antibiotic resistance among the bacteria
in the human and animal intestine tracts has taken place quite often as was proved
by the analysis of plasmid profiles in different gram-positive and gram-negative
bacteria (B a l i s et al., 1996; B r a t o e v a and J o h n, 1994; P r o d i n g e r
et al., 1996; T a u x e et al., 1989). It was shown that, in spite of peristaltic move-
ment in the gut, the intestinal environment displays transfer kinetics different from
those expected of the mixed, liquid culture, but quite similar to those of a biofilm
236 Wolska K.I. 3
indigenous water and soil bacterial population. Plasmid transfer to indigenous soil
bacteria is highly stimulated by the activity of earth worms which act as a vector for
dispersal of bacteria (D a a n e et al., 1996). Efficient dissemination of catabolic
plasmids among desiccation-tolerant bacteria in soil microcosms was reported
(W e e k e r s et al., 2001). Recently it was shown that gene transfer occurs with
enhanced efficiency in biofilms and induces enhanced stabilization of the biofilm
structure (M o l i n and T o l k e r - N i e l s e n, 2003).
Bacteria have been isolated that are able to degrade most man-made pollutants
and most of the degradative genes are part of the operons localized at wild host-
range, conjugative or mobilizable plasmids (V a n d e M e e r et al., 1992). Bacte-
rial genes for degradation of pollutants have been studied for their potential use in
bioremediation of the contaminated sites. For example, an operon comprising genes
for degradation of chlorobenzoate (cbaAB) was found in a conjugative plasmid of
Alcaligenes isolated in contaminated soil. The cba genes were subsequently found
to be associated with transposon Tn5271, a part of this plasmid. In an environment
simulating a chlorobenzoate-contaminated river the initial host bacteria did not
survive well but the plasmid was found to transfer to a wide range of indigenous
species (F u l t h o r p e and W y d h a m, 1989; 1991; 1992). PCR analysis, using
primers located within the catabolic genes showed that transposon Tn5271-like
sequences were present in diverse bacterial species from a bioremediation system
treating contaminated water from a chemical landfill site (W y n d h a m et al.,
1994). The transfer of degradative plasmids was subsequently demonstrated between
various bacterial species in polluted soil and activated sludge (D i G i o v a n n i
et al., 1999; R a v a t n et al., 1998; H a l l i e r - S o u l i e r et al., 1999).
Studies of a river in Estonia contaminated from a subterranean oil-shale fire demon-
strated the horizontal spread of pheBA, genes encoding catehol 1,2-dioxygenase and
a single-component phenol monooxygenase, a part of plasmid PaW85 originally
present in phenol-degrading P. putida to indigenous bacteria P. corrugata, P. fragi,
P. stuzeri and P. fluorescent (P e t e r s et al., 1997).
Transformation
Table II
Bacterial transformation in the environment
particles, thereby greatly increasing the local concentration of both, and therefore
the frequency of transformation (B a u r et al., 1996).
DNA transfer by transformation has been demonstrated in several natural set-
tings (Table II). S t e w a r t and S i n i g a l l i a n o (1991) demonstrated transfor-
mation of Pseudomonas stutzeri to rifampicin resistance by chromosomal DNA in
sterile and nonsterile marine sediments and P a u l et al. (1991) detected the trans-
formation by a broad host-range plasmid in a marine Pseudomonas sp. Studying the
efficiency of P. stutzeri transformation in soil, it was shown that highly different
levels of natural transformation are associated with the genomic subgroups within
a local population (S i k o r s k i et al., 2002). W i l l i a m s and coworkers (1996)
assayed biofilms of Acinetobacter calcoaceticus growing on river stones for incor-
poration of a DNA marked with a his gene and documented the transfer of this
marker. Environmental conditions, such as soil type, soil moisture and nutrient
accessibility affect the efficiency of A. calcoaceticus transformation (N i e l s e n
et al., 1997a; b). Recently genetic transformation of clinical isolates of E. coli under
naturally occurring conditions in oligotrophic, aquatic environments containing
physiologic concentrations of calcium was described. In contrast, transformation
was suppressed in a nitrogen-rich body fluid like urine, a common habitat of
uropathogenic strains (W o e g e r b u n e r et al., 2002).
Transduction
Table III
Transfer of genes by phage transduction under environmental conditions
Bacterial
Bacterial donor Phage Environment Reference
recipient
Marine bacteria Marine bacteria T-phiHSIC Seawater-nutrient mix J i a n g and P a u l, 1998
P. aeruginosa P. aeruginosa F116 Fresh water M o r r i s o n et al., 1978
P. aeruginosa P. aeruginosa F116 Leaf surface S a y e et al., 1987
E. coli E. coli P1 Soil Z e p h and S t o t z k y, 1989
Using a mathematical model, the rate of transduction in Tampa Bay Estuary was
estimated at about 1.3×1014 events per year. Moreover, it was shown that 43% of
bacterial population isolated from this bay lysed after treatment with mitomycin C,
yielding phage particles or particle-like structures (B u s h m a n, 2002a).
Transduction of both chromosomal and plasmid markers by P. aeruginosa phage
F116 was seen in fresh-water reservoir (M o r r i s o n et al., 1978) and on the leaf
surface (K i d a m b i et al., 1994). Another phage, UT1, able to promote trans-
duction in P. aeruginosa and as well as in members of the indigenous population of
natural lake-water environments was also isolated (R i p p et al., 1994).
It should be mentioned here that bacteriophages encoding virulence factors
expressed upon lysogenization can also be transferred to a new hosts. This phenom-
enon explain the distribution of pyrogenic exotoxin C determinants among Strepto-
coccus pyogenes (K a p u r et al., 1992), cholera toxin determinants among Vibrio
cholerae (W a l d o r and M e k a l a n o s, 1996) and shiga toxin determinants in
pathogenic E. coli O157::H7 (M u n i e s a and J o f r e, 1998).
The recent focus on the potential for life in extreme environments has generated
a great deal of interest in the Archaea because of their adaptation to extremes of
temperature, salinity and anaerobicity. The discovery of genetic transfer systems for
the Archaea allows the understanding of their genetic mechanisms allowing to study
their unique adaptive strategies (S o w e r s and S c h r e i e r, 1999). In such stud-
ies special focus is on hyperthermophilic Archaea (N o l l and Va r g a s, 1997).
Among the best characterized archaeal representatives all mechanisms of transfer
such as transduction, conjugation and transformation have been discovered (L u o
and W a s s e r f a l l e n, 2001). Conjugative transfer of ING family of conjugative
plasmids from the extremely thermophilic archaeon Sulfolobus islandicus was
proved (S t e d m a n et al., 2000) and transfer of pyr chromosomal marker was
described between the various strains of Sulfolobus acidocaldarius, another
hyperthermophilic archaeon (R e i l l y and G o r g a n, 2001).
240 Wolska K.I. 3
The comparison of genome sequences proved the presence in archaeal and bacte-
rial genomes of a comparable amount of mobile DNA (L a w r e n c e and
O c h m a n, 1998). Moreover there are many bacterial and archaeal genes that have
the strongest similarities to genes from another domain, suggesting lateral transfer.
Aquifex and Thermatoga, both extremeophile bacteria contain 16% and 24% archaeal
genes, respectively; an archaeon Archaeoglobus fulgidis contains a large set of
bacterial genes for fatty acids metabolism (B u s h m a n, 2002b).
Literature
L u o Y. and A. W a s s e r f a l l e n. 2001. Gene transfer systems and their applications in Archaea. Sys.
Appl. Microbiol. 24: 1525
M a n z e l D. and J. D a v i e s. 1999. Antibiotic resistance in microbes. Cell Mol Life Sci. 56: 742754
M a t i c I., F. T a d d e i and M. R a d m a n. 1996. Genetic barriers among bacteria. Trends Microbiol.
4: 6972
M o l i n S. and T. T o l k e r - N i e l s e n. 2003. Gene transfer occurs with enhanced efficiency in
biofilms and induces enhanced stabilization of the biofilm structure. Curr. Opin. Biotechnol. 14:
255261
M o r r i s o n W.D., R.V. M i l l e r and G.S. S a y l e r. 1978. Frequency of F116-mediated transduction
of Pseudomonas aeruginosa in a freshwater environment. Appl. Environ. Microbiol. 36: 724730
M u n i e s a M. and J. J o f r e. 1998. Abundance in sewage of bacteriophages that infect Escherichia
coli O157:H7 and that carry the Shiga toxin 2 gene. Appl. Environ. Microbiol. 64: 24432448
N i e l s e n K.M., A.M. B o n e s and J.D. v a n E l s a s. 1997a. Induced natural transformation of
Acinetobacter calcoaceticus in soil microcosms. Appl. Environ. Microbiol. 63: 39723977
N i e l s e n K.M., M.D. v a n W e e r e l t, T.N. B e r g, A.M. B o n e s, A.N. H a g l e r and J.D. v a n
E l s a s. 1997b. Natural transformation and availability of transforming DNA to Acinetobacter
calcoaceticus in soil microcosms. Appl. Environ. Microbiol. 63: 19451952
N i e l s e n K.M., A.M. B o n e s, K. S m a l l a and J.D. v a n E l s a s. 1998. Horizontal gene trans-
fer from transgenic plants to terrestrial bacteria a rare event? FEMS Microbiol. Rev. 22: 79103
N i k o l i c h M.P., G. H o n g, N.B. S h o e m a k e r and A.A. S l a y e r s. 1994. Evidence for natural
horizontal transfer of tetQ between bacteria that normally colonize humans and bacteria that nor-
mally colonize livestock. Appl. Environ. Microbiol. 60: 32553260
N o l l K.M. and M. Va r g a s. 1997. Recent advances in genetic analyses of hyperthermophilic archaea
and bacteria. Arch. Microbiol. 168: 7380
N o r m a n d e r B., B.B. C h r i s t e n s e n, S. M o l i n and N. K r o e r. 1998. Effect of bacterial dis-
ruption on conjugal gene transfer on the phylloplane of the bush bean (Phaseola vulgaris). Appl.
Environ. Microbiol. 64: 19021909
O p p r e g a a r d H., T.M. S t e i n u m and Y. W a s t e s o n. 2001. Horizontal transfer of a multi-drug
resistance plasmid between coliform bacteria of human and bovine origin in a farm environment.
Appl. Environ. Microbiol. 67: 37323734
P a u l J.H., J.M. T h u r m o n d and M.E. F i s h e r. 1991. Gene transfer in marine water column and
sediment microcosms by natural plasmid transformation. Appl. Environ. Microbiol. 57: 15091515
P e t e r s M., E. H e i n a r u, E. T a l p s e p, H. W a n d, U. S t o t t e m e i s t e r, A. H e i n a r u and
A. N u r k. 1997. Acquisition of a deliberately introduced phenol degradation operon, pheBA, by
different indigenous Pseudomonas species. Appl. Environ. Microbiol. 63: 48994906
P r o d i n g e r W.M., M. F i l l e, A. B a u e r f e i n d, I. S t e m p l i n g e r, S. A m a n n, B. P f a u s l e r,
C. L a s s - F l o r l and M.P. D i e r i c h. 1996. Molecular epidemiology of Klebsiella
pneumoniae producing SHV-5 b lactamase: parallel outbreaks due to multiple plasmid transfer.
J. Clin. Microbiol. 34: 564568
R a v a t n R., A.J.B. Z e h n d e r and J.R. v a n d e r M e e r. 1998. Low-frequency horizontal trans-
fer of an element containing the chlorocatechol degradation genes from Pseudomonas sp. Strain
B13 to Pseudomonas putida F1 and to indigenous bacteria in laboratory-scale activated sludge
microcosms. Appl. Environ. Microbiol. 64: 21262132
R a v e l J., W e l l i n g t o n E.M. and R.T. H i l l. 2000. Interspecific transfer of Streptomyces giant
linear plasmids in sterile amended soil microcosms. Appl. Environ. Microbiol. 66: 529534
R e i l l y M.S. and D.W. G r o g a n. 2001. Characterization of intragenic recombination in a hyper-
thermophilic archaeon via conjugational DNA exchange. J. Bacteriol. 183: 29432946
R i p p S., O.A. O g u n s e i t a n and R.V. M i l l e r. 1994. Transduction of freshwater microbial com-
munity by a new Pseudomonas aeruginosa generalized tarnsducing phage, UTI. Mol. Ecol. 3:
121126
R o m a n o w s k i G., M.G. L o r e n z and W. W a c k e r n a g e l. 1993. Plasmid DNA in a ground-
water aquiter microcosms Adsorption, DNAase resistance and natural genetic transformation of
Bacillus subtilis. Mol. Ecol. 2: 171181
3 Minireview 243
Abstract
Many bacterial genomes are under asymmetric mutational pressure which introduces composi-
tional asymmetry into DNA molecule resulting in many biases in coding structure of chromo-
somes. One of the processes affected by the asymmetry is translocation changing the position of
the coding sequence on chromosome in respect to the orientation on the leading and lagging
DNA strand. When analysing sets of paralogs in 50 genomes, we found that the number of
observed genes which switched their positions on DNA strand is lowest for genomes with the
highest DNA asymmetry. However, the number of orthologs which changed DNA strand
increases with the phylogenetic distance between the compared genomes. Nevertheless, there is
a fraction of coding sequences that stay on the leading strand in all analysed genomes, whereas
there are no sequences that stay always on the lagging strand. Since sequences diverge very fast
after switching the DNA strand, this bias in mobility of sequences is responsible, in part, for
higher divergence rates among some of coding sequences located on the lagging DNA strand.
Introduction
Experimental
Data for analysis. Prokaryotic genomic sequences and gene annotations have been downloaded
from the Genbank (ftp://www.ncbi.nlm.nih.gov). Boundaries between leading and lagging strands (posi-
tions of origins and termini of replication) and decisions concerning the location of genes on one of
these strands were set on the basis of experimental results or on the basis of the results of DNA walks
describing nucleotide compositional bias of differently replicating DNA strands (M a c k i e w i c z et al.,
1999b, see also: http://smorfland.microb.uni.wroc.pl). The asymmetry of the genomes was measured
by the absolute value of the difference between the GC3 skews of the genes in the leading strand and the
ones in the lagging strand:
)GC3 skew = |(GdCd)/(Gd+Cd) (GgCg)/(Gg+Cg)|
where: Gd and Cd numbers of guanine and cytosine in the third codon positions of the leading strand
genes; Gg and Cg numbers of guanine and cytosine in the third codon positions of the lagging strand
genes. The AT skew and GC skew values proved to be good parameters describing asymmetry of DNA
strands (L o b r y , 1996).
248 Mackiewicz D. et al. 3
Paralogs for 50 genomes (listed in Table I) showing leading/lagging strand asymmetry were ex-
tracted from the TIGR database (http://www.tigr.org). In the analysis only paralogs with minimum 50%
identity were chosen.
Classification of genes to orthologous groups and their amino acid sequences were extracted from
Clusters of Orthologous Groups (COGs) downloaded from ftp://www.ncbi.nlm.nih.gov/pub/COG in Sep-
tember 2001. COGs contain protein sequences which are supposed to have evolved from one ancestral
protein (K o o n i n et al., 1998; T a t u s o v et al., 2001). In the analyses only the best matches for
each ortholog (the closest orthologs) have been chosen.
Analyses of all orthologous sequences have been done on the two sets of bacterial genomes showing
evident compositional asymmetry between leading and lagging strands.
7 genomes belonging to (-subdivision of Proteobacteria group compared with each other: E. coli
K12-MG1655 (EcK), E. coli O157:H7 EDL933 (EcE), H. influenzae (Hi), P. multocida (Pm),
P. aeruginosa (Pa), V. cholerae (Vc), X. fastidiosa (Xf);
14 genomes compared with E. coli O157:H7 EDL933 (EcE): E. coli K12-MG1655 (EcK),
V. cholerae (Vc), P. multocida (Pm), P. aeruginosa (Pa), X. fastidiosa (Xf), N. meningitidis MC58
(Nm), B. subtilis (Bs), R. prowazekii (Rp), M. tuberculosis H37Rv (Mt), C. jejuni (Cj), T. pallidum
(Tp), H. pylori 26695 (Hp), C. pneumoniae CWL029 (Cp), P. horikoshii (Ph).
Moreover, from the 7 genomes of the (-Proteobacteria group, the 7 sets of 1521 orthologs present in
all the genomes, being the best hits for E. coli EDL933 sequences (the closest orthologs), were with-
drawn. Similarly, from the set of 14 genomes compared with E. coli EDL933, the 14 sets of 233 orthologs
present in all the genomes, being the best hits for E. coli EDL933 sequences, were extracted.
For each pair of genomes, orthologs and paralogs were classified into three groups according to their
strand location: pairs of sequences lying on the leading strands, pairs of sequences lying on lagging
strands, and pairs of sequences of which one is lying on the leading and the other on the lagging strand.
For each case fractions of the three groups of sequences have been counted.
Phylogenetic analysis. The amino acid sequences of each COG were aligned by the CLUSTAL
W 1.8 v. program (T h o m p s o n et al., 1994). Pairwise evolutionary distances (expressed by the mean
number of amino acid substitutions per site) between sequences of each COG were calculated using the
WAG model of amino acid substitution (W h e l a n and G o l d m a n, 2001) as implemented in the
TREE-PUZZLE program version 5.0 (S c h m i d t et al., 2002). The analyses of divergence of the three
groups of orthologs were shown for the sets of 1521 orthologs present in all 7 (-Proteobacteria genomes.
For each of the three groups of orthologs a mean value of the evolutionary distances was calculated.
Nonparametric analyses by Mann-Whitney U, Kolmogorov-Smirnov and ANOVA Kruskal-Wallis tests
(S o k a l and R o h l f, 1995) were carried out to assess statistical significance of differences between
these groups.
Evolutionary distances between 16S rRNA sequences (measured by the number of substitutions per
site) were calculated by the MEGA 2.1 program (K u m a r et al., 1993) assuming Tamura-Nei model of
nucleotide substitutions (T a m u r a and N e i, 1993).
in Fig. 1 we show the relation between the fraction of trans-paralogs in the genome
and the asymmetry of chromosomes measured by )GC3 skew. The observed negative
correlation (Spearman correlation coefficient, r = 0.715) is statistically significant
with high confidence (p = 5.6×109). There are two possible explanations for the ob-
served negative correlation. One, assuming a higher mutation rate and in consequence
higher elimination rate of gene copies translocated to the other DNA strand in highly
asymmetric genomes. The second, to us less plausible, refers to the influence of fre-
quency of rearrangements on the maintenance of chromosomal asymmetry. If a global
frequency of rearrangements in a genome is low, it does not disturb chromosomal
asymmetry established by the mutational pressure. On the contrary, high frequency of
rearrangements should diminish this asymmetry.
We have performed a pairwise analysis of orthologs found in compared genomes
belonging to (-Proteobacteria. For each pair of genomes, the orthologs were divided
into three groups: i/ pairs of orthologs which are in both compared genomes on the
leading strand, ii/ pairs of orthologs which are in both genomes on the lagging strand
and iii/ pairs of orthologs of which one is located on the leading and the second on the
60
y = -73.61x + 47.34
r = -0.715
50 p = 5.6E-09
fraction of trans-paralogs
40
30
20
10
0
0 0.1 0.2 0.3 0.4 0.5 0.6 0.7
∆ GC3 skew
Table I
Number of all paralogs, the fraction of trans-paralogs and DGC3 skew for 50 analysed genomes
number fraction
genome )GC3 skew
of all paralogs of trans-paralogs
Agrobacterium tumefaciens C58 Cereon 1096 46.8 0.08
Agrobacterium tumefaciens C58 Uwash 1117 46.6 0.08
Bacillus halodurans C-125 2421 39.2 0.17
Bacillus subtilis 168 558 31.4 0.15
Borrelia burgdorferi B31 11 9.1 0.62
Brucella melitensis 16M 314 38.2 0.13
Campylobacter jejuni NCTC 11168 79 30.4 0.41
Caulobacter crescentus CB15 511 44.2 0.05
Chlamydia muridarum Nigg 19 0.0 0.49
Chlamydia pneumoniae AR39 111 6.3 0.29
Chlamydia pneumoniae CWL029 112 6.3 0.30
Chlamydia pneumoniae J138 100 7.0 0.29
Chlamydia trachomatis serovar D 6 16.7 0.45
Clostridium perfringens 13 217 29.0 0.41
Deinococcus radiodurans R1 282 46.5 0.01
Escherichia coli O157:H7 EDL933 3604 26.9 0.10
Escherichia coli K12-MG1655 919 47.0 0.09
Escherichia coli VT2-Sakai 4020 24.1 0.10
Haemophilus influenzae KW20 73 15.1 0.16
Helicobacter pylori 26695 198 51.5 0.12
Helicobacter pylori J99 109 41.3 0.12
Lactococcus lactis IL1403 811 42.9 0.22
Listeria innocua CLIP 11262 349 18.9 0.18
Listeria monocytogenes EGD-e 255 31.4 0.20
Mesorhizobium loti MAFF303099 1414 42.9 0.04
Mycobacterium leprae TN 121 47.1 0.13
Mycobacterium tuberculosis CDC1551 2417 43.7 0.09
Mycobacterium tuberculosis H37Rv 2279 45.1 0.08
Neisseria meningitidis MC58 595 35.0 0.20
Neisseria meningitidis Z2491 874 42.3 0.22
Pasteurella multocida PM70 86 23.3 0.23
Pseudomonas aeruginosa PAO1 786 44.8 0.11
Pyrococcus abyssi GE5 118 48.3 0.04
Pyrococcus horikoshii shinkaj OT3 147 42.2 0.07
Ralstonia solanacearum GMI1000 784 50.8 0.08
Rickettsia conorii Malish 7 478 40.4 0.21
3 Rearrangements in asymmetric bacterial genomes 251
Table I continued
number fraction
genome )GC3 skew
of all paralogs of trans-paralogs
Salmonella enterica Typhi CT18 679 35.3 0.13
Salmonella typhimurium LT2 SGSC1412 1280 41.0 0.12
Sinorhizobium meliloti 1021 460 48.0 0.05
Staphylococcus aureus Mu50 228 16.2 0.28
Staphylococcus aureus N315 482 15.1 0.29
Streptococcus pneumoniae R6 759 28.5 0.30
Streptococcus pneumoniae TIGR4 479 38.2 0.31
Streptococcus pyogenes SF370 M1 130 26.2 0.26
Thermoplasma acidophilum DSM 1728 41 39.0 0.03
Thermotoga maritima MSB8 216 49.5 0.06
Treponema pallidum Nichols 72 43.1 0.34
Vibrio cholerae El Tor N16961 868 16.0 0.17
Xylella fastidiosa 9a5c 779 23.7 0.33
Yersinia pestis CO92 8551 49.1 0.09
The set of paralogs (with minimum 50 % identity) was extracted from TIGR database.
A 60
50
fraction of orthologs
40
30
20
10
0
0.0 0.1 0.2
phylogenetic distance
B 60
50
fraction of orthologs
40
30
20
10
0
0.0 0.1 0.2
phylogenetic distance
C 60
50
fraction of orthologs
40
30
A 60
50
fraction of orthologs
40
30
20
10
0
0 0.1 0.2 0.3 0.4 0.5
phylogenetic distance
B 60
50
fraction of orthologs
40
30
20
10
0
0 0.1 0.2 0.3 0.4 0.5
phylogenetic distance
C 60
50
fraction of orthologs
40
Fig. 3. Relation between the fractions
of orthologs and the phylogenetic dis- 30
tance measured by 16S rRNA per-
formed for three groups of orthologs: 20
lying on the leading strand (A), lying
on the lagging strand (B) and which
10
changed DNA strand (C).
Data obtained from comparison of the
E. coli EDL933 genome with 14 other ge- 0
nomes belonging to different taxonomic 0 0.1 0.2 0.3 0.4 0.5
groups. phylogenetic distance
254 Mackiewicz D. et al. 3
This observation implies also that there are some sequences which are used to
staying on the leading DNA strand and they have lower probability of being inverted
than sequences which are used to staying on the lagging strand. As a consequence,
the set of coding sequences found on the leading strand should be not uniform. It
should consist of a set of sequences which permanently or preferentially stay on the
leading strand and a set of mobile sequences which are only transiently transferred
from the lagging strand. To test this hypothesis we analysed the sets of 233 orthologs
represented in all 15 genomes. In the first step we compared the most closely related
genomes in the analysed set two E. coli strains and we counted the fractions of
orthologs which stayed at the same DNA strands (leading or lagging) and the fraction
of orthologs which switched their strands. In the next step we added to the compari-
son the third genome (the closest to the E. coli EDL933 genome according to the 16S
rRNA phylogenetic distance) and again counted sequences which stayed at the same
DNA strand in all the three genomes and sequences which switched their strand at
least in one genome and so on, adding new, more distant genome to the analysed
group. In Fig. 4, in the diagram, we have presented the results of analysis; values on
y-axis correspond to the fraction of sequences of a given group of orthologs, while at
the bottom the name of a new genome added to the comparison is shown. The fraction
100
80
fraction of orthologs
60
20
0
t
p
f
h
a
p
m
s
cK
p
j
m
+M
+X
+C
+H
+B
+V
+P
+P
+R
+C
+T
+N
+P
+E
Fig. 4. The fractions of three groups of orthologs counted for the comparisons of E. coli EDL933
with successively added genomes to the comparison.
The group of the leading strand orthologs contains sequences which stay on the leading strand in all analysed
genomes in a given comparison. Analogously for the lagging strand orthologs. The third group of orthologs
includes sequences which switched their strand at least in one genome in a given comparison. Data were obtained
for the sets 233 orthologs present in all 15 genomes. For genomes name abbreviations see Materials and Methods.
3 Rearrangements in asymmetric bacterial genomes 255
Table II
Orthologs found in all 15 analysed genomes
on the leading strand
of sequences which stay in all analysed genomes on the lagging strand drops very fast
and after adding the eighth genome it reaches zero, which means that there are no
orthologous coding sequences located on the lagging strands in all compared genomes.
For this group of compared genomes, there are still some orthologs which stay on the
leading strand in all the genomes and this fraction seems to approximate asymptoti-
cally about 7% of all compared coding sequences, even after adding the most distant
genome belonging to Archaea. These orthologs code for ribosomal proteins commonly
considered highly conserved (Table II). The position of these genes on the leading
strand seems to be conserved even across the two kingdoms (Bacteria and Archaea).
It was observed that their operons are well preserved even in divergent species
(W a t a n a b e et al., 1997; I t o h et al., 1999; N i k o l a i c h i k and D o n a c h i e,
2000; T a m a m e s, 2001). Moreover, it was found that ribosomal genes are prefer-
entially located in many genomes on the leading strand (M c L e a n et al. 1998) prob-
ably (what is important for highly expressed genes) to avoid head-on collisions
between replication and transcription complexes (B r e w e r, 1988; F r e n c h, 1992).
In the next studies we have analysed the divergence measured by the mean num-
ber of amino acid substitutions per site in groups of sequences classified according to
their mobility between differently replicating DNA strands. Analyses were performed
with the sets of 1521 orthologs present in all 7 genomes belonging to (-Proteobacteria.
We compared the E. coli EDL933 genome with six other genomes.
256 Mackiewicz D. et al. 3
3
orthologs in all genomes on the leading strand
orthologs in all genomes on the lagging strand
2.5 orthologs on the leading strand in a given pair
orthologs on the lagging strand in a given pair
orthologs on different DNA strands in a given pair
2
1.5
0.5
0
EcE-EcK EcE-Vc EcE-Pm EcE-Hi EcE-Pa EcE-Xf
Fig. 5. The divergence measured by the mean number of amino acid substitutions per site according to WAG
model (Whelan and Goldman, 2001) in five groups of sequences classified according to their mobility
between differently replicating DNA strands for comparisons of E. coli EDL933 with other genomes.
Analyses were performed with the sets of 1521 orthologs present in all 7 genomes belonging
to (-Proteobacteria. For genomes name abbreviations see Materials and Methods.
We divided all orthologs into five sets: 1 genes staying in all analysed genomes
on the leading strand, 2 genes staying in all analysed genomes on the lagging strand,
3 genes which are located in E. coli EDL933 and in the compared genome on
the leading strand but can be found in at least one of the other genomes of
(-Proteobacteria on the lagging strand, 4 genes which are located in E. coli EDL933
and in the compared genome on the lagging strand but can be found in at least one
of the other genomes on the leading strand and, 5 sequences which are located on
different DNA strands in the compared genomes. The divergence values between
genes of the E. coli EDL933 genome and other genomes of (-Proteobacteria are
shown in Fig. 5. We have found that there are statistically significant differences
in the relative divergence between genes classified according to their position and
mobility. The differences between set 1 and set 5 are statistically significant (with
p < 0.01) for all comparisons. It is clear that the divergence of the orthologs which
switched strand (set 5) is especially high for the closest genomes, which was already
reported (T i l l i e r and C o l l i n s, 2000c; S z c z e p a n i k et al., 2001; R o c h a
and D a n c h i n, 2001) and decreases for pairs of distant genomes. Differences in
divergence between set 5 and all other sets are statistically significant (with p <0.01)
for pairs: EcE-EcK, EcE-Vc and EcE-Pa.
In all compared pairs of genomes the lowest divergence is observed for the
orthologs which permanently stay at the leading strand and do not change their strand
3 Rearrangements in asymmetric bacterial genomes 257
even at long evolutionary distances (set 1). If we eliminate this set of conserved genes
from the set of all orthologs found on the leading strand (receiving set 3), the rest still
seems to be less prone to accumulate substitutions than the genes from the lagging
strand. However, we have found only one statistically significant difference in diver-
gence (5.6% of all comparisons) when we compared sets 2, 3 and 4 with each other
for all pairs of genomes. Furthermore, the divergence in these three sets is signifi-
cantly different (with p < 0.01) when analysed by the ANOVA Kruskal-Wallis test
only for one pair EcE-Pa. It indicates that these three sets form rather uniform group.
Conclusions
Acknowledgments. The work was supported by the grant number 1016/S/IMi/03. M.K. was sup-
ported by Foundation for Polish Science.
Literature
Abstract
Siderophore activity as the feature of microorganisms enabling colonization of human body and
the survival in inanimate environment was investigated in 108 strains of Staphylococcus cohnii:
S. cohnii ssp. cohnii (50 strains) and S. cohnii ssp. urealyticus (58 strains). Strains were isolated
from people, hospital and non-hospital environment. Highest siderophore activity was noted in
strains S. cohnii ssp. urealyticus particularly from the inanimate environments origin. In 86%
analyzed strains siderophores of hydroxamate class were detected. Larger amounts of these com-
pounds were synthesized in strains S. cohnii ssp. urealyticus. Strains belonging to both subspe-
cies from human origin showed lower activity of siderophores (total pool) and did not produce
hydroxamate class chelators or produced very small amounts of these compounds.
Introduction
Experimental
Bacterial strains. The study comprised 108 strains of Staphylococcus cohnii (50 strains of S. cohnii
ssp. cohnii and 58 of S. cohnii ssp. urealyticus) from the collection of the Department of Pharmaceutical
Microbiology, Medical University of £ód. S. cohnii strains were isolated in 19971999 in the Intensive
Care Unit of the clinical hospital from material samples (skin swab) taken from patients and personnel
40 strains, and hospital environment (swab from floors, walls and equipment surfaces) 26 strains,
as well as material samples (swab from floors, walls and furniture surfaces in private houses) in non-
hospital environment 42 strains. These strains were isolated and identified by S z e w c z y k and
R ó ¿ a l s k a (2000). Strains were stored at 70°C in 50% glycerol broth medium and cultivated on agar
plates suplemented with sheep blood for the experiment use.
Media. The NB with dipyridyl medium (iron poor medium with iron chelator added used for iron
starvation): Nutrient Broth (Difco) 0.8g, Agar No1 (Oxoid) 1.5g, ","-bipyridyl (Sigma) 3.04 mg and
deionized water 100 mL. Nutrient Broth solutions and Agar No1 were combined and after sterilization
(20 min at 121°C) a sterile solution of ","-bipyridyl was added. The BM-3 medium used for determin-
ing siderophore production was: stock solution of trace elements according to Lankford et al. (1966)
supplemented with 1% glucose, 0,2% casamino acids (Difco) and 0,05% yeast extract (Difco). The
solutions were sterilized separately (20 min at 121°C) and combined in appropriate proportions.
Siderophore production. For enhancement of siderophore production and reduction of intracellular
iron reserves strains were inoculated on NB medium with dipyridyl and incubated for 24 hours at
a temperature of 37°C (double passage). Next a small portion of bacterial mass (material from some
colonies was taken and inoculated in BM-3 medium),was incubated for 48 hours at 37°C with constant
shaking (75 strokes/min) and centrifuged (10000 rpm for 15 min) and after siderophores were deter-
mined in the supernatant culture.
Siderophore determination. Total siderophore chelating activity was tested by the Schwyn and
Neilands method with standard reagent containing Chrome Azurol S (CAS) (Sigma) (S c h w y n and
N e i l a n d s, 1987). Siderophore activity was expressed as :moles of desferrioxamine mesylate
(Desferal, Ciba-Geigy) per mL of culture supernatant; the standard curve for Desferal was used.
Hydroxamate class siderophores production was estimated by the periodic acid method (H o l z b e r g
and A r t i s, 1983) and also expressed as :moles of Desferal per mL of supernatant (standard curve for
Desferal). Phenol-catechol class siderophores production was assessed by the colorimetric assay of
A r n o w (1937) and siderophore activity was expressed as :moles of 2.3-dihydroxybenzoic acid
DHBA (Fluka) per mL of culture supernatant (standard curve for DHBA). Obtained results were
recounted to mg bacterial protein contained in 1 mL of culture. Protein concentration was estimated
by Ehresmann et al. method (1973) in cell extract obtained after lysis (with 1 M NaOH) of cell sedimen-
tation after culture centrifuging. The standard curve for bovine albumin (Sigma) was used.
All solutions and media were prepared with deionized water. Glassware was washed with deionized
water, kept for 24 hours in 2 N hydrochloric acid (to remove remains of iron) and rinsed three times with
deionized water.
3 Synthesis of siderophores by S. cohnii 263
Table I
Distribution of Staphylococcus cohnii strains isolated from various environments
(percentage of investigated environment) in groups of total siderophore activity
regularity. Strains isolated from human were obviously different from strains isolated
from the environment but no distinct differences between strains isolated from hospi-
tal and non-hospital environment (Table I), especially in Staphylococcus cohnii
ssp. cohnii strains were observed. Staphylococcus cohnii ssp. urealyticus strains
were rarely isolated from human; most of them were from the environment and were
characterized by high siderophore activity (Table I, Figure 1). The differences
in siderophore activity in strains relating to human and environment could be
explained by lower iron accessibility in the environment, which leads to intensive
siderophore synthesis. There is a high supply of iron in human body, even though it is
binded with proteins, bacteria are capable to obtain it from such sources (M i k u c k i
and L i s i e c k i, 1998, W a l d o n et al., 2002).
Besides total siderophore activity determination, level of chelators synthesis in
two main siderophore classes: hydroxamate and catecholate was assayed. Catecholate
siderophores were not detected in supernatants culture of all investigated strains. That
result was not surprising, because rare strains of staphylococci are able to synthetize
catecholate siderophores. L i s i e c k i et al. (1994) found catecholate siderophores
only in 7,8% of 180 studied strains of staphylococci belonging to 26 species.
Hydroxamate synthesis is common among staphylococci but some strains also could
not produce these compounds (L i s i e c k i et al., 1994).
In our study hydroxamate siderophores were not detected in 15 (13,9%) of
108 analyzed Staphylococcus cohnii strains. Twelve of them belonged to Staphylo-
coccus cohnii ssp. cohnii (ten of them were isolated from human) and only three
belonged to Staphylococcus cohnii ssp. urealyticus (all isolated from non-hospital
environment). Staphylococcus cohnii ssp. urealyticus strains produced hydroxamate
in significant amount in contrast to Staphylococcus cohnii ssp. cohnii (Table II,
Figure 2). That regularity was also observed in environmental strains of Staphylo-
coccus cohnii ssp. urealyticus comparing to strains of other subspecies.
Analyzing the obtained results concerning the total siderophore activity in super-
natants culture and hydroxamates presence we observed that both the values were not
3 Synthesis of siderophores by S. cohnii 265
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
<20 20-40 40-60 60<
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
<20 20-40 40-60 60<
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
<20 20-40 40-60 60<
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
<20 20-40 40-60 60<
Fig. 1. Percentage of strains Staphylococcus cohnii ssp. cohnii and Staphylococcus cohnii ssp. urealyticus in
groups of total siderophore activity expressed in :moles of Desferal per mg of protein.
A all studied strains in subspecies, B strains isolated from human, C strains from hospital environment,
D strains from non-hospital environment.
266 Szarpiñska-Kwaszewska J., Farkas £.I. 3
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
0 0-0,5 0,5-1,0 1,0-1,5 1,5<
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
0 0-0,5 0,5-1,0 1,0-1,5 1,5<
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
0 0-0,5 0,5-1,0 1,0-1,5 1,5<
100,0%
80,0%
S. cohnii ssp. cohnii
60,0%
0,0%
0 0-0,5 0,5-1,0 1,0-1,5 1,5<
Fig. 2. Percentage of strains Staphylococcus cohnii ssp. cohnii and Staphylococcus cohnii ssp. urealyticus in
groups of hydroxamate siderophore class activity expressed in :moles of Desferal per mg of protein.
A all studied strains in subspecies, B strains isolated from human, C strains from hospital environment,
D strains from non-hospital environment.
3 Synthesis of siderophores by S. cohnii 267
Table II
Distribution of Staphylococcus cohnii strains isolated from various environment
(percentage of investigated environment) in groups of hydroxamate siderophore class activity
always correlated. The occurence of siderophore activity and the simultaneous lack of
hydroxamates and catecholates compounds could be due to the presence of other
siderophores classes, e.g. staphyloferrin A and staphyloferrin B produced commonly
by staphylococci (D r e c h s e l et al., 1993; H a a g et al., 1994; K o n e t s c h n y -
R a p p et al., 1990) or ketoacids (H e u c k et al., 1995) or also not yet described
chelators. H e u c k et al. (1995) examined iron assimilation by coagulase negative
staphylococci by ketoacids mediation. They noted a great amount of pyruvic and
"-ketoglutaric acid in Staphylococcus cohnii cultures growing in poor iron environ-
ment. W a l d o n et al. (2002) showed that S. cohnii strains from the same collection
were able to utilize numerous oxo-acids in iron assimilating system.
Strains, in which no hydroxamates have been detected, were characterized by low
total siderophore activity (except two). That proves the significant participation of
hydroxamates in all Staphylococcus cohnii siderophores, though it was not always the
same in all strains. Nor is it known if all hydroxamates issued from both subspecies
have the same structure. They could be different compounds with different chelating
activity. That may explain the differences between hydroxamate amounts and total
siderophore activity in some strains.
High correlation level between high siderophore activity and high hydroxamate
production was noted in strains of Staphylococcus cohnii ssp. urealyticus, especially
in those isolated from the environment; in a group of 19 strains which had over
1 :mole hydroxamates per mg of protein in supernatants cultures, 13 strains were
with high and 4 strains with the highest total siderophore activity, while in a group of
six strains which had under 0.5 :mole hydroxamates per mg of protein no strain with
high total siderophore activity was observed.
Most of Staphylococcus cohnii ssp. cohnii strains, which mainly synthesized
hydroxamates was characterized with low total siderophore activity. We may presume
that siderophore activity of these Staphylococcus cohnii ssp. cohnii strains depends
268 Szarpiñska-Kwaszewska J., Farkas £.I. 3
Acknowledgements. This study was supported by the grant 503-406/2002 from Medical Univer-
sity of £ód.
Literature
Abstract
The purpose of the present study was to determine whether there is the relation between the
virulence of used HSV-1 strains and inhibition of apoptosis. HEp-2 cells were induced to
apoptosis by osmotic shock after infection by HSV-1 strains. HSV-1 ts, earlier described as less
virulent for mice inhibited apoptosis in smaller degree than native strain and HSV-1 tr. We sug-
gest that this is due to the hyperproduction of IFN alpha by human cells after the stimulation by
this strain. All strains of HSV-1 didnt inhibit apoptosis in the presence of IFN alpha and apoptosis
was inhibited by anti IFN alpha antibodies. We confirm that IFN alpha plays an important func-
tion in controlling acute HSV-1 infection.
Introduction
Recent studies indicate that HSV-1 also prevents apoptosis of infected cells
and that it is able to protect cells against apoptosis by various inducers (A u b e r t
and B l a h o, 1999; A u b e r t et al., 1999; G a l v a n et al., 1999; G a l v a n and
R o i z m a n, 1998; K o y a m a and M i w a, 1997; Z a c h o s et al., 2001). Several
other viral genes have been proposed to play antiapoptotic roles during HSV-1 infec-
tion. Studies indicate that apoptosis is inhibited at early times postinfection in Jurkat
cells by the action of two immediate-early genes, Us5 and Us3 (J e r o m e et al.,
1999). Furthermore, cultured human epithelial cells infected with an ICP27 deletion
virus exhibited the characteristic signs of apoptosis (A u b e r t and B l a h o, 1999).
Finally, it has been suggested that the viral glycoproteins gD (Us6) and gJ (Us5) are
involved in blocking the apoptotic pathway during productive infections in neuron-
like SK-N-SH cells (Z h o u et al., 2000). These studies suggest that several HSV-1
factors may have antiapoptotic functions that contribute to maintaining the viability
of the virus during its life cycle.
Furthermore, in cells infected with several mutant viruses, HSV-1 can activate
apoptosis (A u b e r t et al., 1999; G a l v a n and R o i z m a n, 1998; K o y a m a
and A d a c h i, 1997). However, the specific mechanisms by which HSV activates or
suppresses apoptotic pathways and the relative importance or contribution of these
pathways to the outcome of the infection are still unclear.
We induced apoptosis in HEp-2 cells by osmotic shock after infection by HSV-1
strains characterised by different pathogenicity to determine the effect of virulence on
inhibition of apoptosis. We used temperature sensitive mutant in 28°C (HSV-1 ts) and
temperature resistant in 39°C (HSV-1 tr) isolated by L i t w i ñ s k a et al., 1991, from
McIntyre strain. It was defined earlier, that HSV-1 ts is less virulent for mice and
establish latent infections rarely and HSV-1 tr is more virulent for mice than native
strain (L i t w i ñ s k a et al., 1996; L i t w i ñ s k a et al., 2001).
Experimental
Cell lines. CV1, a monkey kidney cell line, obtained from National Bacteriological Laboratory,
Department of Virology in Stockholm, was grown in MEM (Gibco BRL) supplemented with 10% FCS
(Sigma), 100 U/ml penicillin, and 100 :g/ml streptomycin (MEM 10% FCS). HEp-2, a human epider-
moid carcinoma cell line was grown in MEM 5% FCS.
Viruses. HSV-1 strain McIntyre, obtained from Institute of Hygiene in Freiburg, temperature sensi-
tive mutant in 28°C (HSV-1 ts), and temperature resistant in 39°C (HSV-1 tr) were grown and titered on
CV1 cells.
Induction of apoptosis. HEp-2 cells were seeded 1 day before infection in 24-well plates at 1×105
cells per well. Sheep antiserum to human leukocyte interferon alpha (aIFN alpha) and human leukocyte
interferon alpha (IFN alpha) (National Institute of Allergy and Infectious Diseases) were added to some
wells at concentration: 88 :g/ml (1 :500) the first one, 500 IU/ml the second one. The next day,
HEp-2 cells were exposed to 1×105 pfu of HSV-1, HSV-1ts, HSV-1 tr, and incubated at 37°C, 28°C, or
39°C for 1 h. Then culture medium was replaced with fresh MEM 5% FCS, in some cases with aIFN
alpha and IFN alpha, and incubated at 37°C, 28°C, or 39°C for 4 h. Next, the cells were incubated in
medium containing 1 M sorbitol for 1 h at 37°C, 28°C, or 39°C and then maintained in sorbitol-free
medium (in some cases with IFN or aIFN) overnight.
3 Apoptosis and virulence of HSV-1 273
DNA fragmentation assay. The medium was removed, and attached cells were resuspended with
0.05% trypsin and added back to the removed medium. Cells were pelleted at 2000 xg for 20 min and
resuspended in 100 :l of phosphate buffered saline. Fragmented DNA was extracted by the Hirt method
(Hirt, 1967) with minor modifications. Briefly, the cell suspension was lysed by adding 400 :l of TE
buffer (10 mM Tris-HCl pH 7,4; 10 mM EDTA) containing 0,6% sodium lauryl sulfate. The cell lysate
was gently mixed with 125 :l of 5 M NaCl and kept at 4°C overnight. The mixture was centrifuged at
14000 xg for 30 min and chromatin pellet was then removed. After treatment with RNase (0,1 mg/ml
1 h at 37°C) and proteinase K (0,1 mg/ml 1 h at 50°C), DNA in the supernatant was precipitated with
ethyl alcohol and resuspended in TE buffer. Samples were analysed for a nucleosomal DNA ladder by
electophoresis on a 1,5% agarose gel.
Results
1 2 3 4 5 6 7 1 2 3 4 5 6 7 1 2 3 4 5 6 7 1 2 3 4 5 6 7
A B C D
Fig. 2. Agarose gel containing electrophoretically separated, ethidium bromide-stained low-molecular-
weight DNA from HEp-2 cultures that were infected different strains HSV-1: A non-infected,
B HSV-1 ts, C HSV-1, D HSV-1 tr. Line 1: molecular ruler (Fermentas); line 2, 3, 4: culture
exposed to sorbitol; line 5, 6, 7: culture not exposed to sorbitol; line 3, 6: culture exposed to IFN alpha;
line 4, 7: culture exposed to anti-IFN alpha antibodies.
Lysates from cells infected by HSV-1, HSV-1ts, and HSV-1 tr and exposed to IFN
alpha and sorbitol showed extensive DNA fragmentation (Fig. 2: A, B, C, D, lane 3),
whereas cells infected by HSV-1, HSV-1ts, and HSV-1 tr and exposed to aIFN alpha
and sorbitol showed no degradation of DNA (Fig. 2: A, B, C, D, lane 4). Lysates from
cells infected by HSV-1, HSV-1 tr and HSV-1 ts and exposed to IFN alpha but not
exposed to sorbitol showed no degradation DNA (Fig. 2: A, B, C, D, line 5, 6, 7). These
results indicate that HSV-1 doesnt inhibit apoptosis in the presence of IFN alpha. We
also observed that apoptosis was inhibited by anti-IFN alpha antibodies.
Discussion
We studied how virulence of the used HSV-1 strains affects the inhibition of
apoptotic death. The temperature sensitive (ts) and temperature resistant (tr) HSV-1
mutants from the collection of Department of Virology, National Institute of Hygiene
were used. Current research has implemented characteristics of HSV-1 ts and HSV-1
tr. HSV-1 ts was described earlier as less virulent for mice but manifesting high im-
munogenic potency. Moreover, HSV-1 ts showed a significantly lower possibility to
cause the latent infection but the state of immunosuppression increased the frequency
3 Apoptosis and virulence of HSV-1 275
Literature
A l f o n s o C.L., J.G. N e i l a n, G.F. K u t i s h and D.L. R o c k. 1996. An African swine fever virus
Bcl-2 homolog, 5-HL, suppresses apoptotic cell death. J. Virol. 70: 48584863.
A u b e r t M. and J. B l a h o. 1999. The herpes simplex virus type 1 regulatory protein ICP27 is re-
quired for the prevention of apoptosis in infected human cells. J. Virol. 73: 28032813.
A u b e r t M.J. O T o o l e and J. B l a h o. 1999. Induction and prevention of apoptosis in human
HEp-2 cells by herpes simplex virus type 1. J. Virol. 73: 1035910370.
C r o o k N.E., R.J. C l e m and R.A. M i l l e r. 1993. An apoptosis-inhibiting baculovirus gene with a
zinc finger-like motif. J. Virol. 67: 21682174.
D e r f u s s T., H. F i c k e n s c h e r, M.S. K r a f t, G. H e n n i n g, D. L e n g e n f e l d e r, B.
F l e c k e n s t e i n and E. M e i n l. 1998. Antiapoptotic activity of the herpesvirus saimiri-en-
coded Bcl-2 homolog: stabilization of mitochondria and inhibition of caspase 3-like activity. J.
Virol. 73: 58975904.
G a l v a n V. and B. R o i z m a n. 1998. Herpes simplex virus type 1 induces and blocks apoptosis at
multipe steps during infection and protects cells from exogenous inducers in a cell-type-depen-
dent manner. Proc. Natl. Acad. Sci. USA 95: 39313936.
G a l v a n V., R. B r a n d i m a r t i and B. R o i z m a n. 1999. Herpes simplex virus type 1 blocks
caspase-3-independent and caspase-dependent pathways to cell death. J. Virol. 73: 32193226.
G u m i e n n y T.L., E. L a m b i e, E. H a r t w i e g, H.R. H o r v i t z, M.O. H e n g a r t n e r. 1999.
Genetic control of programmed cell death in the Caenorhabditis elegans hermaphrodite germline.
Development 126: 10111022.
H e n d e r s o n S., D. H u e n, M. R o w e, C. D a w s o n, G. J o h n s o n and A. R i c k i n s o n.
1993. Epstein-Barr virus-coded BHRF1 protein, a viral homologue of Bcl-2, protects human
B-cells from programmed cell death. Proc. Natl. Acad. Sci. USA 90: 84798483.
H i r t B. 1967. Selective extraction of polyoma DNA from infected mouse cell culture. J. Mol. Biol. 26:
365369.
276 Rechnio M., Litwiñska B. 3
Abstract
Twenty Aeromons caviae isolates from stool of children with diarrhea symptoms were examined
for virulence-associated properties: production of cytotoxic and cytotonic toxins, and invasive
ability. Most of A. caviae strains were cytotoxic to Vero and CHO cells and produced cytotonic
toxins which caused elongation of CHO cells. Moreover, five of A. caviae strains revealed inva-
sive ability towards HEp-2 cells.
Introduction
Experimental
Bacterial strains. A total of 20 strains of A. caviae were used in this study. They were recovered
from fecal samples of patients suffering from diarrhea: 13 strains were isolated in Poland
(K a z n o w s k i, 1995), and seven isolates cultured in a hospital in Hong Kong were obtained from
dr R. Kong (Hong Kong City University). All strains were stored at 75°C in brain heart infusion broth
(BHI, Difco) containing 50% (v/v) glycerol.
Cell cultures. Chinese hamster ovary cells (CHO), African monkey kidney cells (Vero) and human
laryngeal epithelial cell line (HEp-2) were cultured in Minimum Essential Medium Eagle (MEM, Sigma)
supplemented with 5% fetal calf serum (FCS, Sigma), 2 mM glutamine, 80 IU penicillin per ml, 80 :g
streptomycin per ml, and 1 mg/ml of nystatin. The cell cultures were incubated at 37°C in atmosphere
containing 5% CO2 (G r a y et al., 1990; S c h i a v a n o et al., 1998).
Cytotoxic and cytotonic activity. Culture supernatants were prepared as described previously by
S c h u l t z and M c C a r d e l l (1998). A. caviae strains were grown on tryptic soy broth (TSB, Difco)
supplemented with 0.6% yeast extract at 37°C for 24 h in a water bath with agitation of 150 rpm. The
bacterial cultures were centrifuged at 700 g for 20 minutes and the supernatants were sterilized through
3 Enteropathogenic activity of A. caviae 279
0.22 :m-pore size filters Millex-GV (Millipore) of low protein binding. Sterile culture supernatants
were heated at 56°C for 20 min to destroy the activity of heat-labile toxins.
Cytotoxic activity of the supernatants was measured on Vero and CHO cells whereas cytotonic
activity was examined on CHO cells. The cells were seeded into 96-wells microtitre trays (Nunc) at
a concentration of 3×103 cells per well (CHO) and 1×104 cells per well (Vero cells). A 100-:l volume of
serial twofold dilutions of culture supernatants was added to the monolayer and the plate was incubated
at 37°C for 24 hours in atmosphere containing 5% CO2. The plates were read under an inverted micro-
scope. Cytopathic effects were identified as rounding and detachment of 50% of Vero or CHO cells
(S c h i a v a n o et al., 1998). Cytotonic activity was identified as elongation of CHO cells. Cytotonic
and cytotoxic titre was expressed as the reciprocal of the highest dilution yielding a positive result
(G r a y et al., 1990).
Invasion assay of HEp-2 cells. Invasion assay was performed according to W a t s o n et al. (1985)
with minor modifications. The cells were seeded at a concentration of 1×103 per well in 96-wells
microtitre tissue plates and incubated for 24 h in MEM supplemented with 5% FCS and antibiotics.
Then the cell culture medium was removed and replaced with MEM without serum and antibiotics, and
the plate was incubated for further 24 hours. A. caviae strains were cultured on BHI at 37°C for 24 h.
The bacterial inoculum was made to 1 McFarland standard and diluted 1:10 in Eagle MEM to give
a concentration of about 1×105 CFU per ml. After MEM removing, 100 :l of bacterial suspension was
added to each well and the monolayer was incubated at 37°C for 3 h. The cells were washed three times
with 200 :l of phosphate-buffered saline (PBS, Biomed), and 200 :l of MEM containing gentamycin
(0.1 mg/1 ml) was added to each well and the samples were incubated for 2 h at 37°C. After washing
three times with PBS, the integrity of the monolayer was checked and 200 :l of lysing solution contain-
ing 0.01 M NaH2PO4, 0.1% Tween 20 (v/v) and 0,025% trypsin (w/v) pH 8.0 was added to each well,
and incubation was continued for further 30 min at 37°C. Bacterial CFUs were determined by plating
100 :l of the lysates onto tryptic soy agar (TSA, Difco).
Invasion index was expressed as CFU×100 per 1×103 of HEp-2 cell according to L a w s o n et al.
(1985). The monolayer was infected separately with the invasive strain of Yersinia enterocolitica O:3 as
the positive control and E. coli K12C600 as the negative one.
Results
The results of the study concerning virulence factors like cytotoxic and cytotonic
toxins, and the invasive ability are listed in Table I.
Cytotoxic and cytotonic activity. Nineteen of twenty A. caviae strains were
found to be cytotoxic to Vero cells with cytotoxic titres ranging from 1 to 8, and
cytotoxic to CHO cells with titres ranging from 1 to 16. Preheating (56°C for
20 min) of the supernatants caused a decrease in cytotoxic activity of the isolates
to Vero cells and CHO cells. Cytotonic activity was observed in case of 15 strains
with titres ranging from 1 to16.
Invasion assay. Invasion of HEp-2 cells by A. caviae isolates was investigated in
a quantitative assay. Five strains (25%) gave invasion index greater than 10 CFU per
HEp-2 and these strains might be classified as invasive according to the criteria
of L a w s o n et al. (1985). However, the invasion index of Y. enterocolitica ser-
ving as a positive control was 68 CFU per HEp-2 cell. Twelve strains of A. caviae
demonstrated invasion index ranging from 1.1 to 6.8 CFU per HEp-2 cell. Strain
AK 390 showed invasion index 0.6 CFU per HEp-2 cell whereas AK 388 was
negative in this test. E. coli K12C600 used as negative control did not show invasion
ability towards HEp-2 cells.
280 Krzymiñska S. et al. 3
Table I
Cytotoxic, cytotonic activity, and invasion of HEp-2 cells by 20 clinical isolates of A. caviae
Discussion
models have been developed. One approach to this problem is the use of cell lines.
Studies have been conducted on various cell types including Vero, CHO, HEp-2 and
Y1. These systems resembling in vivo-like conditions are now being used to elucidate
mechanisms of bacterial enteropathogenesis (T h o r n l e y et al., 1997 ).
This study focused on explaining the contribution of cytotoxic, cytotonic toxins,
and invasiveness in gastrointestinal disease caused by A. caviae isolates. The analysis
of the incidence of virulence markers in A. caviae isolates revealed that 14 A. caviae
strains (70%) produced both cytotoxic and cytotonic toxins (Tab. I). G r a y et al.
(1990) reported that 2 of 22 A. caviae strains isolated from pig feces produced both
cytotoxic and cytotonic toxins with cytotoxic titre 32 and 1024, and cytotonic titre 1.
Moreover, four of environmental isolates of A. caviae produced cytotonic toxins with
a titre of 1. S c h i a v a n o et al. (2000) noticed that 1 of 3 A. caviae strains isolated
from stool of diarrheal patients was found to be cytotoxic to CHO cells with cytotoxic
titre equal 4. In our study, five isolates of A. caviae (25%) produced only cytotoxins
whereas one strain produced only cytotonic toxin. Previously, G r a y et al. (1990)
noted that 13.6% of A. caviae strains isolated from the environment produced
cytotoxin and 27% of these isolates possesed a heat-stabile cytotonic factor that
caused elongation of CHO cells. In research of G r e y and K i r o v (1993), none of
A. caviae isolates from clinical specimens produced cytotoxins. These differences may
be dependent on bacterial genetic determinants. C h a k r a b o r t y et al. (1984) noted
that the cytotoxic, cytotonic enterotoxin and hemolytic activities of Aeromonas spp.
strains were due to three different proteins resulting from the expression of three
distinct genes. However, R o s e et al. (1988) stated that hemolytic, cytotoxic and
enterotoxic activities as well as mice lethality were associated with only one protein.
C h o p r a et al. (1993) isolated cytotoxic enterotoxin gene from human diarrheal
isolate of A. hydrophila.
Gastrointestinal symptoms (dysentery, colitis) associated with some cases of
Aeromonas-linked diarrhea may be connected with an invasive mechanism. However,
there is little experimental evidence supporting this hypothesis. In our study, 5 of 20
A. caviae strains (25%) were invasive to HEp-2 cells with invasion index lower than
that of Y. enterocolitica positive control. This result is in agreement with the observa-
tions of S h a w et al. (1995) who reported that the majority of A. caviae strains
showed little or no invasive ability. W a t s o n et al. (1985), expressing invasion
index as CFU per 1 ml of lysate, suggested that strains which showed 5×106 CFU
per ml of lysate were invasive. They found that 26% of Aeromonas spp. isolates were
invasive but only 1 of 22 A. caviae strains was classified as invasive.
We compared our results of production concerning cytotoxic, cytotonic entero-
toxins and invasive ability of 13 A. caviae isolates with clinical manifestations of
infections previously described by M o k r a c k a et al. ( 2001). We found that
strains AK 375, AK 376 and AK 390, which caused intermittent diarrhea for 5 days
without dehydratation symptoms, revealed cytotoxic activity and low invasion
index. Strains AK 378 and AK 384, which produced cytotoxic enterotoxin and
possessed low invasion index, provoked hemorrhagic diarrhea in young children. The
isolate AK 388 which demonstrated cytotoxic and cytotonic activity without invasion
282 Krzymiñska S. et al. 3
ability was an etiological agent of diarrhea with toxicant course with vomiting and
dehydratation of the body.
The results of this study demonstrate that A. caviae strains isolated from fecal
samples produce cytotoxic and cytotonic toxins which may contribute to gastrointes-
tinal disease. Some of these strains showed invasive ability although the invasion
index was lower than that of Y. enterocolitica.
Literature
Abstract
Ten cardiovascular drugs were procured in pure form from their manufacturers in India and
screened for antimicrobial property against fifteen known bacteria belonging to both gram-positive
and gram-negative types. These bacteria were inhibited by the common antibiotics at 15 mg ml 1
level through our earlier studies. Since most of the bacteria were moderate to highly responsive
to amlodipine, this compound was further tested in vitro against 504 bacteria comprising 4 genera
of gram-positive and 15 genera of gram-negative bacteria. Most of these were inhibited by
the drug at 50200 :g ml1 level and few strains were sensitive even at lower concentrations
(10 :g ml1). The bacteria could be arranged in the decreasing order of sensitivity towards
amlodipine in the following manner: Staphylococcus aureus, Vibrio cholerae, Vibrio para-
hemolyticus, Shigella spp., Salmonella spp., Bacillus spp., whereas Escherichia coli, Klebsiella
spp. and Pseudomonas aeruginosa were found to be resistant to the lower concentrations of the
drug. Amlodipine was found to be bactericidal in nature when its mode of action was studied
against S. aureus 6571, V. cholerae 14035 and Sh boydii 8 NCTC 254/66. The antibacterial activ-
ity of amlodipine could also be confirmed in vivo. When it was given to Swiss strain of white
mice at different dosages (30 and 60 :g/mouse), it could significantly protect the animals chal-
lenged with 50 MLD of Salmonella typhimurium NCTC 74. According to Chi square test the
in vivo data were highly significant (p < 0.001).
Introduction
Antibiotics are one of our most important weapons in fighting bacterial infections
and have greatly benefited the health-related quality of human life since their introduc-
tion. However, over the past few decades these health benefits are under threat as many
commonly used antibiotics have become less and less effective against certain illness
not only because many of them produce toxic reactions but also due to emergence of
drug resistant bacteria. It is essential to investigate newer drugs with lesser resistances.
Systematic studies among various pharmaceutical compounds have revealed that any
drug may have the possibility of possessing diverse functions and thus may have useful
activity in completely different spheres of medicine. Drugs belonging to different phar-
macological classes such as antihistamines like diphenhydramine and bromodiphen-
hydramine (D a s t i d a r et al., 1976), methdilazine (C h a t t o p a d h y a y et al.,
1988) and promethazine (C h a k r a b a r t y et al., 1989), psychotropics, e.g., pro-
mazine (D a s h et al., 1977), chlorpromazine (M o l n a r et al., 1976), fluphenazine
(D a s t i d a r et al., 1995) and trifluoperazine (M a z u m d e r et al., 2001), anti-
hypertensives such as methyl-DOPA (D a s t i d a r et al., 1986), local anaesthetics
like procaine (D a s t i d a r et al., 1988) and antiinflammatory drugs e.g., diclofenac
(A n n a d u r a i et al.,1998), possess powerful antibacterial activity. Such chemo-
therapeutic agents have been grouped together and are now entitled as Non- anti-
biotics (C h a k a r b a r t y et al., 1998; K r i s t i a n s e n, 1992). The present paper
describes the detailed in vitro and in vivo activity of such a non antibiotic the cardio-
vascular drug amlodipine.
Experimental
Drugs. The cardiovascular drugs clonidine and dipryridamole were obtained from German Rem-
edies, enalapril from Nicholas Piramol, lacidipine from Glaxo Pharma, nifidipine (Torrent), nitrendipine
(Concept), felodipine from Cipla, digoxin (Cadila) Pharma, benidipine from Stancare and amlodipine
from Pfizer Pharmaceuticals. All these drugs were obtained in pure dry powder form and dissolved in
either distilled water or DMSO depending on their solubility, and kept at 4°C upto 15 days.
Bacteria. A total of 504 strains of bacteria belonging to 19 genera comprising 172 gram-positive
and 332 gram-negative types were tested (Table I). These were of human origin, identified as described
by B a r r o w and F e l t h a m (1993) and preserved in freeze-dried state.
Media. Liquid media used for this study were peptone water (PW, Oxoid brand bacteriological
peptone 1% (w/v) plus Analar NaCl 0.5% (w/v), nutrient broth (NB, Oxoid), Mueller Hinton broth
(MHB; Difco). Solid media were: peptone agar (PA), bromothymol blue lactose agar media (BLA),
nutrient agar (NA) and Mueller Hinton agar (MHA), obtained by solidifying the liquid media with 1.2%
(w/v) agar (Oxoid No. 3). In case of BLA, bromothymol blue indicator 1.2% (w/v) and lactose 1% (w/v)
are added. The pH is maintained at 7.27.4 for all the media. NA was used for tests with gram-positive
bacteria and PA and BLA were used for the rest of the bacteria as needed.
Determination of minimum inhibitory concentration (MIC) of different drugs. The MIC of
clonidine, dipyridamole, enalapril, digoxin, benidipine, nitrendipine, nifidipine, lacidipine, felodipine and
amlodipine with respect to different test bacteria was accurately determined both by broth and agar dilution
methods. For broth dilution, 0.1 ml of standardized suspension of a strain (106 CFU/ml) was added to each
tube containing amlodipine at concentrations of 0 (control), 2, 5, 10, 25, 50, 100 and 200 :g ml1 in
MHB.The tubes were incubated at 37°C for 24 h, and looked for, visible growth after vortexing the tubes
gently. For agar dilution the drug was added at concentrations of 0 (control), 10, 25, 50, 100, 200, 400 and
800 :g ml1 in molten NA and poured in Petridishes (NCCLS, 1993). The organisms were grown in
PW, and the overnight culture was spot-inoculated on the NA plates such that each inoculum contained
2×106 CFU. The plates were incubated at 37°C, examined after 24 h and incubated further for 72 h, if
3 Antimicrobial activity of amlodipine 287
Table I
Source of bacterial strains
Bacteria Source
Bacillus pumilus NCTC 8241 S.P. Lapage, London
Staphylococcus aureus NCTC 6571, 8530,8531, 8532 S.P. Lapage, London
Escherichia coli K12 Row J.D. Abbott, U.K.
E. coli pBR 322 S. Palchaudhuri, USA
Salmonella typhimurium NCTC 11, 74, S. viballerup, J. Taylor, London
S. choleraesuis 37, S. uganda 101, S. paratyphi 85, S.typhi 57,59
Shigella boydii 5 NCTC 541/60, Sh. boydii 8 NCTC 254/66, K. Patricia Carpenter, London
Sh. boydii 9 NCTC 304/67, Sh. dysenteriae 3 NCTC 102/65,
Sh. dysenteriae 7 NCTC 519/66, Sh. dysenteriae 8 NCTC 599/52,
Sh. sonnei NCTC 5/59
Vibrio cholerae ATCC 14033, 14035 S. Mukerjee, Calcutta
V. cholerae 80, 540, 546, 566, 590, 738,7 64, 824, 838, 906, 1003, National Institute of Cholera
1021, 1023. & Enteric Diseases, Calcutta.
V. parahaemolyticus 4750, 9369, 72001, 72006 Y. Miyamoto, Japan
All the remaining organisms were available in the Department. They were clinical isolates collected from different
hospitals in Calcutta and identified by the methods described by Barrow and Feltham.
necessary. Since one solid agar medium containing amlodipine can be used for inoculation of a large
number of bacteria at a time, the results of this method are being presented here, as the total number of
test bacteria was 504. The lowest concentration of amlodipine in a tube or a plate that failed to show any
visible macroscopic growth was considered as its MIC. The MIC determination was performed in dupli-
cate for each organism, and the experiment was repeated where necessary. The MIC values for a given
isolate were either identical, or within ± one dilution with respect to different test bacteria.
Determination of mode of action of amlodipine. For this purpose S. aureus NCTC 6571,
V. cholerae ATCC 14035, Sh. boydii 8 NCTC 254/66 were grown in NB overnight at 37°C. 2 ml from
each of these were added to 4 ml of fresh NB and incubated for 2 h so that the cultures could attain the
logarithmic growth phase. To determine the number of colony forming units (CFU), 0.5 ml of each
culture was individually added to 4.5 ml fresh NB. This culture was subjected to serial dilutions in test
tubes containing 4.5 ml NB to produce 8-fold dilutions. A 0.025 ml aliquot was then removed from each
tube and pipetted onto a NA plate (K r o g s t a d and M o e l l e r i n g, 1980). Amlodipine was then
added to each culture at a concentration of 2×MIC value (50 :g ml1) of the test bacterium. The CFU
counts were similarly determined upto 6 h at intervals of 2 h and then after 18 h. The number of colonies
appearing in each plate was counted after 24 h incubation for determination of CFU.
In vivo tests. Swiss strain of male white mice weighing 1820 g were used for the in vivo studies.
Animals were maintained at standard conditions at 21 ±1°C and 5060% relative humidity with a photo-
period of 14 : 10 h of light darkness. Water and a dry pellet diet were given ad libitum. The virulence of
the test strain S. typhimurium NCTC 74 was enhanced in the following manner: 0.1 ml of an overnight
grown NB culture of S. typhimurium NCTC 74 was given intraperitoneally as the challenge to 3 mice.
Next day, the animals were anaesthetized and dissected. Heart blood was aseptically collected from
them, added to fresh NB and incubated at 37°C. From this, the pure culture of S. typhimurium NCTC 74
was cultivated and given as challenge to another set of mice in the same manner. After 5 such passages,
the organism was administered as challenge to 4 batches of mice. The median lethal dose (MLD or
LD50) of the passaged strain corresponding to 0.95×109CFU/mouse suspended in 0.5 ml NB served as
the challenge dose for all the groups of animals (C r u i c k s h a n k et al., 1975). Reproducibility of the
288 Kumar K.A. et al. 3
challenge dose was ensured by standardization of its optical density in a Klett Summerson colorimeter at
640 nm and determination of the CFU count in NA.
Four batches of 20 mice each were taken for the study. The first two batches (constituting one group)
were administered 30 :g of amlodipine (by injecting i.p. 0.2 ml from a stock solution containing 150 :g
ml1 of the drug) and the next two batches received 60 :g of amlodipine (0.2 ml from a stock solution of
300 :g ml1 of the drug). After 3 h, one batch from each of the above two groups was challenged with
50 MLD of S. typhimurium 74. A control group comprising 60 animals was also injected with the same
organism and 0.1 ml of sterile saline in place of the drug. The number of animals dying upto 100 h was
recorded in each group to determine the protective capacity of amlodipine (Table IV).
In another in vivo experiment, the CFU counts in blood and organ homogenates of amlodipine treated
and untreated mice were determined. Two groups of mice (10 animals per group) were taken. All the
animals were given a 50 MLD challenge dose; of these 50% received the drug (60 :g/mouse) 3 h before
the challenge and the rest received saline (Table V). After 18 h all mice were sacrificed, blood was
collected individually from heart, and livers and spleens were removed aseptically and homogenized in
tissue homogenisers. CFU counts of individual organs were determined separately. Statistical analysis
of the data was performed using Students t-test.
Results
Table II
Primary screening of cardiovascular drugs in vitro for presence of antibacterial action
Table III
In vitro activity of amlodipine on gram-positive and gram-negative bacteria
to the drug. The MIC of 58 out of 165 V. cholerae were found to be 25 :g ml1
(highly sensitive), 45 had MIC at 50 :g ml1 (markedly sensitive), 37 at 100 :g ml1,
18 at 200 :g ml1 and 7 at 400 :g ml1 (moderately sensitive). Similarly, of
29 V. parahemolyticus, 10 were inhibited at 25 :g ml1, 8 strains at 50 :g ml1,
9 could not grow at100 :g ml1 and 2 at 200 :g ml1 of the drug (Table III).
The drug also showed good inhibitory action (2550 :g ml1) against strains of
Pasturella and Hafnia, while Arizona (200 :g ml1) and Bordetella bronchiseptica
(400 :g ml1), were much less sensitive to this compound. Proteus, Citrobacter,
Providencia and Enterobacter cloaca were resistant to amlodipine.
9
8
Log viable cells in ml1
7
6
5
4
3
2
1
0
0 2 4 6 8
Time in hours
Table IV
Determination of protective capacity of amlodipine in vivo
* Received a challenge dose of 0.95 × 109 CFU in 0.5 ml NB of S. typhimurium NCTC 74.
None of the animals died when 30 :g or 60 :g amlodipine was injected to 2 separate groups
of mice (20 mice in each), i.e., amlodipine was found to be non-toxic to mice.
p < 0.001, according to Chi-square test
Table V
Reduction in CFU/ml of S. typhimurium NCTC 74 in organ homogenates
of mice treated with amlodipine
Discussion
Literature
Abstract
Pure crystals of seven Bacillus thuringiensis field isolates from the Lower Silesia region (Poland)
were tested against larvae of Aedes aegypti L. and Culex pipiens L. (Culicidae, Diptera). The crys-
tals of OpQ3 phylloplane isolate (belonging to the first biochemical type of B. thuringiensis subsp.
japonensis, yoso, jinghongiensis) killed from 68 ± 7% to 84 ± 7% of the fourth instar larvae of
A. aegypti. The crystals of two other strains (KpF3 and KpC1) of this group caused mortality
between 3 ± 2% and 70 ± 7%. The LC50 ranged from 3.2 ± 0.4 to 34.1 ± 4.8 :g/ml. The effect of
B. thuringiensis wratislaviensis H-47 crystals was the lowest with larval mortality from 0% to
17 ± 3%. No significant (0% 37 ± 6%) effect of B. thuringiensis crystals on the larvae of C. pipiens
was observed. Our results show that the delta-endotoxins of B. thuringiensis act very specifically.
Introduction
Experimental
Bacillus thuringiensis strains. The susceptibility of mosquitoes (A. aegypti L. and C. pipiens L.) is
assessed against seven B. thuringiensis strains derived from the bacterial collection of the Microbiologi-
cal Institute of the Wroc³aw University. Two of them, designated as PO12 and PO13, were the only
Polish isolates (amongst 69 B. thuringiensis subspecies, each including several hundred strains from
more than 22 countries) which have been classified according to the H serotype based on flagellar
antigens registered at the International Entomopathogenic Bacillus Centre (IEBC) Collection at Institut
Pasteur, Paris, France (L o n c et al., 1997, L e c a d e t et al., 1999). They were isolated from soil
samples from garden, flower-bed and park areas in Wroc³aw, and designated as a new serovar Bacillus
thuringiensis wratislaviensis H-47. The remaining five strains (KpC1, KpF3, KsS1, OpQ1 and OpQ3)
were selected for bioassays from 26 phylloplane and 33 soil isolates originating from the Karkonosze
National Park, a part of the Sudety Mts, and Osola, vicinity of Wroc³aw (D o r o s z k i e w i c z and
L o n c 1999). The tree names and strains origins were used to designate isolates: K = Karkonosze
National Park, O = Osola (sampling sites for leaf and soil samples); sample source: p = phylloplane,
s = soil; C = Corylus avellana, F = Fagus silvatica, Q = Quercus robur, S = Sorbus aucuparia. The
numbers reflect the number of successive isolates. On the basis of their biochemical activities these five
strains, have been classified (L e c a d e t et al., 1999) into the first, third and fourth biochemical types
comprising the subspecies: japonensis, yoso, jinghongiensis (strains: KpC1, KpF3, OpQ3) I type,
finitimus (KsS1) III and tochigiensis (OpQ1) IV.
Stock bacterial cultures were maintained on brain-heart infusion agar (Difco) slants and stored
at 4°C.
Separation of parasporal crystals. B. thuringiensis strains were grown overnight on nutrient
agar plates. The inoculum was introduced with a sterilized bent wire into 40 ml of sporulation medium
containing: glucose 10 g, casamino acids 7.5 g, KH2PO4 6.8 g, MgSO4 × 7H2O 123 mg, MnSO4 ×
4H2O 2.23 mg, ZnSO4 × 7H2O 14 mg, Fe2SO4 20 mg, H2O 1000 ml; pH 7.5 (K a e l i n et al.,
1994). Such cultures were incubated at 28°C for 144 hours with shaking, giving a spore content of about
108 ml1. Crystals and spores were harvested by centrifugation (5000 rpm, 30 min). The pellet was
washed twice with cold double destilled water and finally resuspended in 3 ml of 50 mmol/l1 Tris-Cl
buffer, pH 7.5, containing 10 mmol/l of KCl (N i c h o l s et al., 1989). From this suspension crystals
were separated by centrifugation (14 000 rpm, 30 min) through a discontinuous sucrose gradient (67, 72,
79 and 87%). The crystal-containing band was collected, observed under a phase contrast microscope
(S h a r i f and A l a e d d i n o g l u 1988), pelleted by centrifugation (14 000 rpm, 30 min), washed
several times with double distilled water and solubilized in 50 mmol l1 Na2CO3HCl, pH 10,5 for 1 h at
37°C (G i l l and F e d e r i c i 1987). The crystal concentrations were measured by the B r a d f o r d
(1976) assay with bovine serum albumin as the standard.
Mosquito culture. Larvae of A. aegypti were obtained from the laboratory culture of the Institute of
Organic Industry in Warsaw, Pszczyna Branch (Poland). The insects were reared in the laboratory at the
room temperature of 28°C and 70% RH. They were maintained in porcelain cups and fed an artifical diet
medium (product of Pszczyna) according to B y r d y (1965) and ¯ ó ³ t o w s k i (1976). Larvae of
Culex pipiens were collected from water bodies of the Wroc³aw area.
Mosquito bioassay. The activity of B. thuringiensis strains against mosquitoes was assessed accord-
ing to the International Entomopathogenic Bacillus Centres formula (T h i é r y et al., 1997). Twenty
specimens of fourth instar larvae (in plastic cups with 10 ml of diet) were exposed to each of four
concentrations: 6.25 :g/ml, 12.5 :g/ml, 25 :g/ml or 50 :g/ml of pure crystals. Three replicates were
made at each concentration including two controls: one with the distilled water and another with
B. subtilis B003 suspensions. The percentage of larvae mortality was corrected according to A b o t t s
(1928) formula. The concentrations of toxic proteins at which 50% of larvae were killed (LC50) were
determined according to the logaritmic-probit method (F i n n e y 1971).
3 Delta-endotoxins of B. thuringiensis 295
Results
Table I
LC50 values and corrected mortality (%) of Aedes aegypti and Culex pipiens larvae treated with
parasporal crystals
Corrected Corrected
Symbol of
Subspecies (serovar)/ Crystal LC50 mortality of mortality of
Bacillus
No Biochemical type dosage (:g/ml) A. aegypti C. pipiens
thuringiensis
(:g/ml) %±s %±s
strain
24 h 48 h 24 h 48 h
1. OpQ3 japonensis, yoso, 6.25 68 ± 7 79 ± 9 0 0
jinghongiensis / I 12.5 3.2 ± 0.4 74 ± 4 81 ± 7 5±4 11 ± 8
25 77 ± 9 84 ± 4 15 ± 4 17 ± 6
50 82 ± 7 84 ± 7 34 ± 6 37 ± 6
2. KpF3 japonensis, yoso, 6.25 4±2 35 ± 3 0 0
jinghongiensis / I 12.5 11.5 ± 2.2 14 ± 4 55 ± 4 0 0
25 37 ± 6 60 ± 7 5±4 5±3
50 56 ± 6 70 ± 7 22 ± 5 23 ± 6
3. KpC1 japonensis, yoso, 6.25 4±3 3±2 0 0
jinghongiensis / I 12.5 34.1 ± 4.8 10 ± 6 10 ± 5 2±3 2±3
25 32 ± 6 32 ± 6 4±2 4±2
50 66 ± 11 68 ± 7 8±4 8±4
4. KsS1 finitimus (H2) / III 6.25 0 1±2 0 0
12.5 3±3 4±3 0 0
25 7±4 6±4 3±3 3±2
50 13±4 17 ± 5 6±2 7±3
5. OpQ1 tochigiensis (H19) / IV 6.25 1±2 3±2 1±2 1±1
12.5 4±4 6±5 2±1 3±2
25 6±5 9±3 9±5 11 ± 5
50 19 ± 3 22 ± 5 12 ± 5 15 ± 6
6. PO12 wratislaviensis (H47) 6.25 0 0 0 0
12.5 0 2±3 0 0
25 5±3 7±4 1±2 1±1
50 11 ± 5 12 ± 5 1±2 1±2
7. PO13 wratislaviensis (H47) 6.25 0 1±1 0 0
12.5 0 0±1 0 0
25 1±2 6±4 0 0
50 5±6 17 ± 3 0 0
Table II
Comparision of A. aegypti and C. pipiens corrected mortality (PS), Student t-test (" = 0.05)
Mortality Mortality
Dosage Symbol
Subspecies of of
of crystals of t p
(serovar) / Biochemical type A. aegypti C. pipiens
:g/ml strain
PS ± s PS ± s
6.25 OpQ3 japonensis, yoso, jinghongiensis / I 79±9* 0* 42.0 0.00
KpF3 japonensis, yoso, jinghongiensis / I 35±3 0 33.5 0.00
KpC1 japonensis, yoso, jinghongiensis / I 3±2 0 2.6 0.03
KsS1 finitimus (H2) / III 1±2 0 1.6 0.15
OpQ1 tochigiensis (H19) / IV 3±2 1±1 1.4 0.19
PO12 wratislaviensis (H47) 0 0
PO13 wratislaviensis (H47) 1±1 0 3.2 0.01
12.5 OpQ3 japonensis, yoso, jinghongiensis / I 81±7 11±8 16.4 0.00
KpF3 japonensis, yoso, jinghongiensis / I 55±4 0 30.2 0.00
KpC1 japonensis, yoso, jinghongiensis / I 10±5 2±3 3.3 0.01
KsS1 finitimus (H2) / III 4±3 0 3.6 0.01
OpQ1 tochigiensis (H19) / IV 6±5 3±2 1.4 0.20
PO12 wratislaviensis (H47) 2±3 0 1.6 0.15
PO13 wratislaviensis (H47) 0±1 0 1.6 0.15
25 OpQ3 japonensis, yoso, jinghongiensis / I 84±4 17±6 24.2 0.00
KpF3 japonensis, yoso, jinghongiensis / I 60±7 5±3 17.4 0.00
KpC1 japonensis, yoso, jinghongiensis / I 32±6 4±2 10.2 0.00
KsS1 finitimus (H2) / III 6±4 3±2 1.5 0.17
OpQ1 tochigiensis (H19) / IV 9±3 11±5 0.9 0.39
PO12 wratislaviensis (H47) 7±4 1±1 3.5 0.01
PO13 wratislaviensis (H47) 6±4 0 3.4 0.01
50 OpQ3 japonensis, yoso, jinghongiensis / I 84±7 37±6 13.0 0.00
KpF3 japonensis, yoso, jinghongiensis / I 70±7 23±6 12.2 0.00
KpC1 japonensis, yoso, jinghongiensis / I 68±7 8±4 17.6 0.00
KsS1 finitimus (H2) / III 17±5 7±3 4.5 0.01
OpQ1 tochigiensis (H19) / IV 22±5 15±6 2.4 0.04
PO12 wratislaviensis (H47) 12±5 1±2 4.8 0.01
PO13 wratislaviensis (H47) 17±3 0 13.2 0.00
active strain was OpQ3 (68 ± 7% 84 ± 7%). LC50 values were 3.2 ± 0.4 :g/ml,
11.5 ± 2.2 :g/ml and 34.1 ± 4.8 :g/ml, respectively. Toxins of the remaining isolates
B. thuringiensis finitimus KsS1, B. thuringiensis tochigiensis OpQ1 and B. thurin-
giensis wratislaviensis PO12 and PO13 caused mortality from 0% to 22 ± 5%.
3 Delta-endotoxins of B. thuringiensis 297
The mortality of C. pipiens larvae was significantly lower than that of A. aegypti.
The most toxic crystals (0% 37 ± 6%) were isolated from the OpQ3 strain. Crystals
of the other two isolates of the first biochemical type were less active (0% 23 ± 6%).
No significant effect (0% 1 ± 2%) was observed after exposure of C. pipiens larvae
to B. thuringiensis wratislaviensis PO12 and PO13 toxins.
In most cases, the insecticidal activity of B. thuringiensis crystals was positively
correlated with their concentration. Some toxins were pathogenic only at higher dos-
ages. High mortality (66 ± 11% after 24 hours and 68 ± 7% after 48 hours) of
A. aegypti larvae was observed only after exposure to 50 :g/ml of the toxins of iso-
late KpC1. However, they were not susceptible ( 3 ± 2% 32 ± 6%) to lower concen-
trations. Only the crystals of OpQ3 strain were highly toxic (68 ± 7% 84 ± 7%) for
A. aegypti at all four dosages.
The differences in mosquito larval mortality were evaluated with Student t-test
(" = 0.05). In most cases they proved to be statistically significant (Table II). At the
highest concentrations, the crystals of all B. thuringiensis strains were much (p < 0.05)
more toxic to A. aegypti than to C. pipiens. Less significant differences were observed
at the two lowest dosages of toxins of isolates B. thuringiensis finitimus KsS1,
B. thuringiensis tochigiensis OpQ1 and B. thuringiensis wratislaviensis PO12 and
PO13. C. pipiens larvae proved to be much less susceptible to parasporal crystals of
all B. thuringiensis strains examined.
Discussion
Table III
Anti-dipteran activity of the Wroc³aw isolates of B. thuringiensis
concentrations examined) of isolates OpQ3 and KpC1, because over 90% of A. aegypti
larvae died within 24 hour exposure time.
L o n c et al. (2001b) examined susceptibility of A. aegypti to spore/crystal
formulations of these B. thuringiensis strains (Table III). The most insecticidal iso-
lates to A. aegypti were OpQ3 (5566%) I biochemical type (representing sub-
species: japonensis, yoso, jinghongiensis) and B. thuringiensis finitimus KsS1
(4866%). High mortality was observed also after exposure of larvae to three other
isolate suspensions: KpC1 (3061%), KpF3 (1760%) I group and B. thuringiensis
tochigiensis OpQ1 (4154%). The pure crystals of B. thuringiensis finitimus KsS1
and B. thuringiensis tochigiensis OpQ1 were not toxic to A. aegypti larvae; they
caused mortality from 0% to 17 ± 5% (KsS1) and from 1 ± 2% to 22 ± 5% (OpQ1).
According to E l l a r (1994) spores of B. thuringiensis kurstaki HD-1 significantly
increased the potency of crystals. The mixture of spores and crystals killed lepi-
dopteran larvae, Plutella xylostella sooner than the toxins alone, with 2.8-fold
decrease in the time required to reach 50% mortality. LC50 value decreased 146-fold
after the addidion of 108 ml1 B. thuringiensis kurstaki HD-1 spores. Such results
suggest that although viable spores play little or no role in the action of lethal doses
of toxin against highly susceptible insects, they are important in enhancing the action
of sublethal doses of delta-endotoxin. This observation would explain the significant
differences in A. aegypti mortality caused by spore/crystal and pure crystals of isolate
B. thuringiensis finitimus KsS1 and B. thuringiensis tochigiensis OpQ1. The pure
crystals of B. thuringiensis wratislaviensis H-47 (PO12 and PO13) were not active
against A. aegypti. Mixture of spores and crystals of these isolates caused a higher
(730%) mortality (L o n c et al., 2001b).
No significant effect (0% 37 ± 6%) on C. pipiens treated with crystals of the
Wroc³aw B. thuringiensis isolates was observed. The highly alkaline mosquito gut pH
3 Delta-endotoxins of B. thuringiensis 299
plays the principal role in delta-endotoxin activation. After ingestion by the insect,
crystals dissolve in the gut juice and gut proteases clip the protoxins off. The resulting
activated toxin binds to receptors on the epithelial cell membrane, inserts into the
membrane, and forms pores that disturb ion stability and finally cause the death of the
insect. For this reason, slightly alkaline pH of C. pipiens (7.2) could have influenced
the low mosquitocidal activity of B. thuringiensis crystals. At such pH they were only
partially or not dissolved. The activity of spore and crystal suspensions of the Wroc³aw
B. thuringiensis isolates against C. pipiens larvae was examined by R y d z a n i c z
(2001). She observed 045% mortality which in most cases was higher than that
caused by crystals alone (0% 37 ± 6%). Larvae of the other dipteran, Musca
domestica, proved to be the most susceptible (4181%) to B. thuringiensis spore/
crystal formulas (L o n c et al., 2001a). However, these suspensions were signifi-
cantly less toxic (1045%) to the fruit fly, Drosophila melanogaster.
Literature
A b o t t S.W. 1928. A method of computing the effectiveness of an insecticide. J. Econ. Entomol. 18:
265267.
A r o n s o n A.I. and Y. S h a i. 2001. Why Bacillus thuringiensis insecticidal toxins are so effective:
unique features of their mode of action. FEMS Microbiol. Lett. 195:18.
B e c k e r N. and J. M a r g a l i t. 1993. Use of Bacillus thuringiensis israelensis against mosquitoes
and blackflies. p. 147170. In: P.F. Enwistle, J.S. Cory, M.J. Bailey, S. Higgs (eds). Bacillus
thuringiensis, an environmental biopesticide: theory and practice. New York, John Wiley & Sons.
B r a d f o r d M.N. 1976. A rapid and sensitive method for the quantification of microgram quantities of
proteins utilizing the principle of protein-dye binding. Anal. Biochem. 72: 248254.
B y r d y S. 1965. Biological investigations on the efficacy of zoocides in the laboratory conditions (in
Polish). Prace naukowe IOR w Poznaniu 1: 3438.
D o r o s z k i e w i c z W. and E. L o n c. 1999. Biodiversity of Bacillus thuringiensis Strains in the
Phylloplane and Soil of Lower Silesia Region (Poland). Acta Microbiol. Pol. 48: 355361.
E l l a r D.J. 1994. Molecular genetics of Bacillus thuringiensis *-endotoxins and toxin receptors.
VIth International Colloquium on Invertebrate Pathology and Microbial Control 1: 1015.
F i n n e y D.J. 1971. Probit analysis. 3rd ed. Cambridge Univ. Press, Cambridge.
G i l l S.S. and B.A. F e d e r i c i. 1987. Cytolytic activity and immunological similarity of the
B. thuringiensis subsp. israelensis and B. thuringiensis subsp. morrisoni isolate PG-14. Appl.
Environ. Microbiol. 53: 12511256.
K a e l i n P., P. M o r e l and F. G a d a n i. 1994. Isolation of Bacillus thuringiensis from stored to-
bacco and Lasioderma serricorne (F.). Appl. Environ. Microbiol. 19: 16.
L e c a d e t M.M., V. C o s m a o - D u m a n i o r and E.E. F r a c h o n. 1994. Collection of Bacillus
thuringiensis and Bacillus sphaericus (classified by H-serotypes). Catalogue of strains No 1.
International Entomopathogenic Bacillus Centre.
L e c a d e t M.M., E.E. F r a c h o n, V. C o s m a o D u m a n i o r, H. R i p o t e a u, P. H a m o n,
P. L a u r e n t and I. T h i e r y. 1999. Updating the H-antigen classification of Bacillus
thuringiensis. J. Appl. Microbiol. 86: 660672.
L o n c E., M.M. L e c a d e t, T.M. L a c h o w i c z and E. P a n e k. 1997. Description of Bacillus
thuringiensis wratislaviensis (H-47), a new serotype originating from Wroc³aw (Poland), and
other Bt soil isolates from the same area. Lett. Appl. Microbiol. 24: 467473.
L o n c E., W. D o r o s z k i e w i c z, M.J. K l o w d e n, K. R y d z a n i c z and A. G a ³ g a n. 2001a.
Entomopathogenic activities of environmental isolates of Bacillus thuringiensis against dipteran
larvae. J.Vector Ecol. 26: 1520.
300 Lonc E. et al. 3
Alexandria, Egypt
Abstract
Introduction
Experimental
Microorganism. Amycolatopsis mediterranei ATCC 21789 was obtained from The American Type
Culture Collection, USA. It was maintained on BENNET´s medium containing (g/l): 10.0 glucose, 1.0 yeast
extract, 1.0 beef extract, 2.0 N-Z amine, 20.0 agar. The pH was adjusted to 6.8 before sterilization. Agar
slants were incubated at 28°C for 57 days.
Inoculum preparation. Inoculum was prepared in vegetative culture medium contained (g/l): glu-
cose, 20.0; KH2PO4, 3.0; K2HPO4, 1.5; MgSO4 × 7H2O, 0.016; Zinc acetate, 0.001 and yeast extract, 5.0.
The pH was adjusted to 7.0 with 1 M NaOH. Cultivation was carried out for 48 h at 30°C on an incuba-
tor shaker at 200 rpm (Infors Co., Switzerland). The obtained vegetative cells were used to inoculate
either the shake flask or the bioreactor with a concentration of 8% (v/v).
3 Rifamycins production by A. mediterranei 303
Medium for rifamycins production. Unless otherwise mentioned, the medium used in both shake
flask and bioreactor experiments was composed of (g/l): glucose, 40.0; KH2PO4, 3.0; K2HPO4, 1.5;
MgSO4 × 7H2O, 0.016; Zinc acetate, 0.001 and yeast extract, 5.0. The pH was adjusted to 7.0 before
sterilization. Glucose was sterilized separately and added to the cultivation medium before inoculation.
Cultivation conditions. In case of shake flask, cultivation was carried out in 250 ml Erlenmeyer
flasks containing 50 ml liquid medium. Inoculum was in the form of 48 h old vegetative culture as
described previously and the inoculated flasks were incubated at 30°C on a rotary shaker at 200 rpm for
144 h. In case of bioreactor experiments, cultivation was carried out in a 3 l stirred tank bioreactor
Bioflow III (New Brunswick Scientific Co., New Brunswick, NJ, USA) with a working volume of 2.2 L.
Agitation was performed using a three 4-bladed rushton turbine impellers (di(impeller diameter) = 65 mm;
dt(tank diameter) = 135 mm, didt1 = 0.48) at 600 rpm. Aeration was performed by filtered sterile air
[1 v/v × m]. Dissolved oxygen concentrations were analyzed by polarographic electrode (Ingold, Germany).
Foam was suppressed, when necessary, by the addition of silicon antifoam reagent (Fluka, Switzerland).
Analysis
Sample preparation and cell dry weight determination. During cultivation in shake flask, samples
in form of three flasks each were withdrawn intermittently for analysis. In case of bioreactor cultivation,
aliquots (in form of 20 ml) of the culture were taken from the bioreactor vessel through a sampling system.
Samples were filtered using dry and pre-weighed filter paper (Whatman filter paper No. 1). The superna-
tant was taken for determination of antibiotic activity and glucose concentration. The filtered biomass was
washed twice by distilled water and subsequently dried in an oven at 100°C for a constant weight.
Determinations of rifamycins activity. Rifamycins were determined spectrophotometrically ac-
cording to the method of P a s q u a l u c c i et al. (1970).
Determination of glucose. Glucose was determined in the fermentation media by enzymatic
method using a glucose determination kit (Glucose kit Cat. No. 4611, Biocon Diagnostic GmbH,
Burbach, Germany).
110
100
YRifaSV/X
90
80
[mg/g] 70
60
YRifaB/X ,
50
40
30
20
10
0
rifamycin SV
1000
800
[mg/L]
600
rifamycin B,
400
200
0
18
16
14
CDW [g/L]
12
10
8
6
4
2
0
0 20 40 60 80 100 120
Glucose [g/L]
Fig. 1. Effect of initial glucose concentration on cell growth, rifamycins production
and yield coefficients of rifamycins based on cell dry weight.
Error bars correspond to samples taken from two independent shake flask experiments.
et al. (1999) reported that glucose was the best carbon source for production of
rifamycins by A. mediterranei MTCC14, but the maximal antibiotic production, about
1080 mg/l, obtained at 25 g/l glucose. D e m a i n et al. (1980) also reported that
carbon source, which support high specific growth, such as glucose, lead to the sup-
pression of antibiotic synthesis. Therefore, the development of fed-batch cultivation
strategy is necessary to increase the antibiotic yield to overcome the glucose inhibi-
tion effect in batch culture.
3 Rifamycins production by A. mediterranei 305
110
YRifaSV/X
100
90
80
[mg/g]
YRifaB/X, 70
60
50
40
30
20
10
0
rifamycin SV
1000
800
600
[mg/L]
rifamycin B,
400
200
0
18
16
14
CDW [g/L]
12
10
8
6
4
2
0
0 1 2 3 4 5 6 7 8 9
Yeast extract [g/L]
Fig. 2. Effect of initial yeast extract concentration on cell growth, rifamycins production
and yield coefficients of rifamycins based on cell dry weight.
100
90 50
YrifaSV/X
YrifaSV/S
80
70 40
60
[mg/g]
[mg/g]
30
50
YrifaB/X ,
YrifaB/S ,
40 20
30
20 10
10
0 0
rifamycin SV
1000
800
[mg/L]
600
rifamycin B,
400
200
0
18 45
16 40
14 35
glucose [g/L]
CDW [g/L]
12 30
10 25
8 20
6 15
4 10
2 5
0 0
0 20 40 60 80 100 120 140 160 180
time [h]
Fig. 3. Growth, glucose consumption and rifamycins production during batch cultivation
of A. mediterranei in shake flask.
this growth phase the dissolved oxygen decreased reaching the minimal of about 40%
and increased again during the cell degradation phase. Glucose was consumed during
this phase with a rate of about 0.35 g/ l × h and the concentration was about 5 g/l after
96 h. On the other hand, the production of rifamycins reached 650 mg/l and 1190 mg/l
for rifamycins B and SV, respectively. The specific production of antibiotic was higher
in the bioreactor compared to the corresponding batch cultivation in shake flask
cultures. This was due to the higher mixing and better oxygenation in the bioreactor
308 El-Endhasy H.A. et al. 3
120 50
YrifaSV/X
YrifaSV/S
100
40
80
30
[mg/g]
[mg/g]
60
YrifaB/X ,
YrifaB/S ,
20
40
20 10
0 0
1400
rifamycin SV
1200
1000
800
[mg/L]
600
rifamycin B,
400
200
0
14
50
12
glucose [g/L]
10 40
CDW [g/L]
8 30
6
20
4
10
2
0 0
0 50 100 150 200
time [h]
which reflect on the cell physiological activities and secondary metabolite production
since also the production process of rifamycins could be increased with higher oxy-
gen supply (C h u n g et al., 1987). Based on this data of batch culture, fed-batch
cultivation strategy was developed with a constant feeding rate of 0.35 g glucose/l × h
after 90 h to keep the glucose concentration at the level of 57 g/l (Figure 6). As
feeding started, cells grew exponentially again with a specific rate of 0.054 [h1] and
the cell dry weight reaching its maximum of about 14.4 g/l after 132 h. After
3 Rifamycins production by A. mediterranei 309
220
200
YrifaSV/X 180
[mg/g] 160
140
YrifaB/X ,
120
100
80
60
40
20
0
rifamycin SV
1400
1200
1000
[mg/L]
800
rifamycin B,
600
400
200
0
100
90 10 50
80
70 8 40
glucose [g/l]
CDW [g/L]
DO [%]
60
50 6 30
40
4 20
30
20 2 10
10
0 0 0
0 20 40 60 80 100 120 140 160 180 200
time [h]
Fig. 5. Cell growth, glucose consumption and rifamycins production during batch cultivation
of A. mediterranei in stirred tank bioreactor.
Data are the average of values taken from two bioreactor cultivations. The standard error based
on these two cultivations was calculated and expressed as the error bar in the figure.
the feeding phase, the cell concentration decreased again with a specific degradation
rate of 0.035 [h1]. The cell degradation rate was almost the same in both of batch and
fed-batch cultures after the growth phase. This indicates that, the cell degradation
310 El-Endhasy H.A. et al. 3
100
YrifaB/X ,
80
60
40
20
0
rifamycin SV
2500
2000
[mg/L]
1500
rifamycin B ,
1000
500
0
100 18 45
90 16 40
80 14 35
CDW [g/L]
glucose [g/L]
70 30
12
DO [%]
60
10 25
50
8 20
40
30 6 15
20 4 10
10 2 5
0 0 0
0 50 100 150 200
time [h]
Fig. 6. Cell growth, glucose consumption and rifamycins production during fed-batch cultivation
of A. mediterranei in stirred tank bioreactor.
was mainly as a result of mechanical shear stress in the stirred tank bioreactor more
than substrate deficiency effect. This phenomenon of cell autolysis as a result of
shear stress was also reported by other authors in case of cultivation of filamentous
microorganisms producing antibiotics in stirred tank bioreactor such as in case of
3 Rifamycins production by A. mediterranei 311
Table I
Kinetic parameters of cell growth and rifamycins production by A. mediterranei
during shake flask and bioreactor cultivations in batch and fed-batch cultures
glucose feeding was not due to the specific production, but to the increased cell
concentration. Supplemental glucose in the fed-batch culture prevented glucose
deficiency for certain extent and thus increased the cell mass, resulting in an increase
in the volumetric production.
In general, the different stages of bioprocess development for the production of
rifamycins are summarized in Table I. This show clearly the significant increase in
the total volumetric antibiotic production through the process transfer from shake
flask to bioreactor culture and switching the process from batch to fed-batch manner.
Literature
H e y d a r i a n S.M, M.D. L i l l y and A.P. I s o n. 1996. The effect of culture conditions on the pro-
duction of erythromycin by Saccharopolyspora erythraea in batch culture. Biotechnol. Lett. 18:
11811186.
L e e J.G., C.Y. C h o i, B.L. S e o n g and M.H. H a n. 1983. Optimal pH profile in rifamycin B
fermentation. J. Ferm. Technol. 61: 4953.
K a w a g u c h i T., T. A s a h i, T. S a t o h, T. U o z u m i and T. B e p p u. 1984. B-factor an essen-
tial regulatory substance including the production of rifamycin in a Nocardia sp. J. Antibiotic. 37:
15871595.
K a w a g u c h i T., M. A z u m a, S. H o r i n o u c h i and T. B e p p u. 1988. Effect of B-factor and its
analogues on rifamycin biosynthesis in Nocardia sp. J. Antibiotic. 41: 360365.
K r i s h n a P.S.M., G. Ve n k a t e s w a r l u and L.V. R a o. 1998. Studies on fermentative production
of rifamycin using Amycolatopsis mediterranei. World J. Microbiol. Biotechnol. 14: 689691.
K r i s h n a M.P.S., G. Ve n k a t e s w a r l u and L.V. R a o. 1999. Production of rifamycin SV using
mutant strains of Amycolatopsis mediterranei. MTCC17. World J. Microbiol. Biotechnol. 15:
741743.
M a r t i n J.F. and A.L. D e m a i n. 1980. Control of antibiotic biosynthesis. Microbiol. Rev. 44:
230251.
P a s s q u a l u c c i C.R., A. V i g e v e n i, P. R a d a e l l i and C.G. G a l l o. 1970. Improved differ-
ential spectrophotometric determination of rifamycins. J. Pharm. Sci. 59: 685687.
R o l a n d C.L., K. P r e m, K.S. N a r a n s, J. S h a i, W. C a m e r a n, R. J o s e p h and A.
B i a n c h i n e. 1997. Rifabutin absorption in the gut unaltered by concomitant administration of
didinoline in AIDS patients. Antimicrob. Agent Chemother. 41: 15661570.
S e p k o w i t z K.A., J. R a f a l l i, L. R i l e y, T.E. K i e h n and D. A r m s t r o n g. 1995. Tubercu-
losis in the AIDS era. Clin. Microbiol. Rev. 8: 180199.
V e n k a t e s w a r l u G., P.S. M u r a l i K r i s h n a and V.L. R a o. 1999. Production of rifamycin
using Amycolatopsis mediterranei (MTCC14). Bioprocess Eng. 20: 2730.
V e n k a t e s w a r l u G., P.S.M. K r i s h n a, A. P a n d e y and L.V. R a o. 2000. Evaluation of
Amycolatopsis mediterranei VA18 for production of rifamycin-B. Process Biochem. 36: 305309.
V i r g i l i o A., E. M a r c e l l i and A. A g r i m i n o. 1964. Aeration-agitaion studies in rifamycin
fermentation. Biotechnol. Bioeng. 6: 271278.
Acta Microbiologica Polonica
2003, Vol. 52, No 3, 315316
BOOK REVIEW
side effect and with the pharmacokinetics and pharmacodynamics (Zhanel and
Noreddin) of this group of drugs.
The following nine chapters of the book are devoted to the use of the fluoro-
quinolones in specific infections, such as urinary tract infections (Johnson), sexually
transmitted infections (Wyllie and Ridgway), pneumonia and other respiratory tract in-
fections (Marrie and Low, respectively). These chapters should be of particular interest
to physicians dealing with these particular infections since besides general information
they also present specific data frequently collected from various in-depth studies.
Finally, the last, very short chapter by the editors of the volume (Ronald and Low)
discusses the future prospects of the quinolones, with special emphasis on the fact
that these antibiotics are not readily destroyed biologically in the environment and
their persistence may therefore generate additional resistance. The authors predict
that by 2010 there should be 56 additional fluoroquinolones that will augment cur-
rent therapeutic regimens.
The book brings a lot of important and up-to-date information on this interesting
group of antibiotics and is highly recommended to clinicians as well as researchers in
pharmacology, clinical medicine or epidemiology. I am not sure whether researchers
in molecular biology will find it equally useful, this in view of the mentioned scant
data on the mechanism of action of these drugs as well as the mechanism of bacterial
resistance to them.
Zdzis³aw Markiewicz
Acta Microbiologica Polonica
2003, Vol. 52, No 3, 317
Instructions to authors
I. General information
AMP publishes descriptions of all aspects of basic and applied research that focuses on
topics of basic research of practical value in microbiology. Topics that are considered
include microbiology in relation to genetics and molecular biology, foods, agriculture,
industry, biotechnology, microbial ecology, public health and basic biological properties of
bacteria, viruses, and simple eukaryotic microorganisms.
Submit manuscripts directly to the Editorial Office, Acta Microbiologica Polonica. The
manuscript should be accompanied by a covering letter stating the address, fax number, e-mail
of the corresponding author and running head of the manuscript (no longer than 47 charac-
ters). Submit two complete copies of each manuscript, including figures and tables. The manu-
script should be either the original typescript from jet or laser printer (not dot matrix).
Accepted papers are copy-edited as word-processor files, so authors are asked to provide
their paper in this form on a disk when they submit the revised version. The text should
be edited in Word 7 or ASCII format (Mazovia or Latin II standard). Submission of figures
in TIF or CorelDraw Format is very appreciated.
All manuscripts are subjected to peer review by the editors, by members of the editorial
board and by qualified outside reviewers. When a manuscript is returned to the authors
for modification, it should be returned to the editor within 2 months; otherwise it may
be considered withdrawn.
15 reprints are sent free to the first author.