ArticlePDF Available

Expression, Secretion, and Glycosylation of the 45- and 47-kDa Glycoprotein of Mycobacterium tuberculosis in Streptomyces lividans

American Society for Microbiology
Applied and Environmental Microbiology
Authors:

Abstract and Figures

The gene encoding the 45/47 kDa glycoprotein (Rv1860) of Mycobacterium tuberculosis was expressed in Streptomyces lividans under its own promoter and under the thiostrepton-inducible Streptomyces promoter PtipA. The recombinant protein was released into the culture medium and, like the native protein, migrated as a double band at 45 and 47 kDa in sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) gels. However, in contrast to the native protein, only the 47-kDa recombinant protein could be labeled with concanavalin A (ConA). Carbohydrate digestion with jack bean alpha-D-mannosidase resulted in a reduction in the molecular mass of the recombinant protein upper band and completely eliminated ConA binding. Two-dimensional gel electrophoresis revealed only one isoelectric point for the recombinant protein. Comparative fingerprinting analysis of the individually purified upper and lower recombinant protein bands, treated under the same conditions with specific proteases, resulted in similar peptide patterns, and the peptides had the same N-terminal sequence, suggesting that migration of the recombinant protein as two bands in SDS-PAGE gels could be due to differences in glycosylation. Mass spectrometry analysis of the recombinant protein indicated that as in native protein, both the N-terminal and C-terminal domains of the recombinant protein are glycosylated. Furthermore, it was determined that antibodies of human tuberculosis patients reacted mainly against the carbohydrate residues of the glycoprotein. Altogether, these observations show that expression of genes for mycobacterial antigens in S. lividans is very useful for elucidation of the functional role and molecular mechanisms of glycosylation in bacteria.
This content is subject to copyright. Terms and conditions apply.
APPLIED AND ENVIRONMENTAL MICROBIOLOGY, Feb. 2004, p. 679–685 Vol. 70, No. 2
0099-2240/04/$08.000 DOI: 10.1128/AEM.70.2.679–685.2004
Copyright © 2004, American Society for Microbiology. All Rights Reserved.
Expression, Secretion, and Glycosylation of the 45- and 47-kDa
Glycoprotein of Mycobacterium tuberculosis in
Streptomyces lividans
Martha Lara,
1
Luis Servı´n-Gonza´lez,
2
Mahavir Singh,
3
Carlos Moreno,
4
Ingrid Cohen,
1
Manfred Nimtz,
3
and Clara Espitia
1
*
Departamento de Inmunologı´a
1
and Departamento de Biologı´a Molecular,
2
Instituto de Investigaciones Biome´dicas, Universidad
Nacional Auto´noma de Me´xico, Me´xico D.F., Me´xico; GBF, German National Research Center for Biotechnology,
Braunschweig, 38124 Braunschweig, Germany
3
; and Department of Bacteriology, Royal Free and University
College Medical School, Windeyer Institute, London W1P 6DB, United Kingdom
4
Received 22 May 2003/Accepted 30 October 2003
The gene encoding the 45/47 kDa glycoprotein (Rv1860) of Mycobacterium tuberculosis was expressed in Strep-
tomyces lividans under its own promoter and under the thiostrepton-inducible Streptomyces promoter P
tipA
. The
recombinant protein was released into the culture medium and, like the native protein, migrated as a double
band at 45 and 47 kDa in sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) gels.
However, in contrast to the native protein, only the 47-kDa recombinant protein could be labeled with con-
canavalin A (ConA). Carbohydrate digestion with jack bean -D-mannosidase resulted in a reduction in the
molecular mass of the recombinant protein upper band and completely eliminated ConA binding. Two-
dimensional gel electrophoresis revealed only one isoelectric point for the recombinant protein. Comparative
fingerprinting analysis of the individually purified upper and lower recombinant protein bands, treated under
the same conditions with specific proteases, resulted in similar peptide patterns, and the peptides had the same
N-terminal sequence, suggesting that migration of the recombinant protein as two bands in SDS-PAGE gels
could be due to differences in glycosylation. Mass spectrometry analysis of the recombinant protein indicated
that as in native protein, both the N-terminal and C-terminal domains of the recombinant protein are glyco-
sylated. Furthermore, it was determined that antibodies of human tuberculosis patients reacted mainly against
the carbohydrate residues of the glycoprotein. Altogether, these observations show that expression of genes for
mycobacterial antigens in S. lividans is very useful for elucidation of the functional role and molecular
mechanisms of glycosylation in bacteria.
Glycosylation is an important covalent modification of pro-
teins. While eukaryotic glycoproteins have been characterized
in detail, information about the structure, function, and bio-
synthetic pathways of prokaryotic glycoproteins is scarce. The
list of known bacterial glycoproteins is growing, and the variety
of components and structures observed indicates the impor-
tance of glycosylation in cell processes, as well as the potential
role of glycosylation in pathogenesis (2, 23, 25).
Carbohydrates have been reported to be associated with
antigenic proteins of pathogenic mycobacteria (7, 10, 11). The
importance of carbohydrates attached to proteins in immune
recognition has been demonstrated by the decreased capacity
of the Mycobacterium tuberculosis 45/47 kDa recombinant pro-
tein (r45/47 kDa) to stimulate T-cell lymphocyte responses
when its mannosylation pattern is changed (12, 32).
Until now, glycosylation of M. tuberculosis proteins has been
confirmed only for the 45/47 kDa protein and for the Myco-
bacterium bovis MPB83 protein (Rv2873), in which chemical
linkage between carbohydrate and protein was demonstrated
(5, 24). The function of glycosylation of mycobacterial glyco-
proteins remains unknown. The 45/47 kDa protein corre-
sponds to the Rv1860 sequence, which is encoded by a gene
which has been annotated as modD in the M. tuberculosis
genome sequence (GenBank accession no. X99258). This sug-
gests that the 45/47 kDa protein could be part of a putative
molybdenum transport system.
Proteins homologous to the 45/47 kDa protein have been
found in M. bovis,Mycobacterium avium,Mycobacterium leprae,
and Mycobacterium vaccae. All of these proteins have the abil-
ity to bind to fibronectin (31, 33, 34, 40).
The 45/47 kDa proteins from M. tuberculosis and M. bovis
are immunodominant antigens which are secreted into the
culture medium and migrate as glycosylated double bands in
sodium dodecyl sulfate (SDS)-polyacrylamide gel electro-
phoresis (PAGE) gels (8). In the present study, the M. tuber-
culosis gene encoding the 45/47 kDa protein was expressed in
Streptomyces lividans, a gram-positive, sporulating, mycelial
bacterium which is not pathogenic. Streptomyces strains are a
well-known source of antibiotics and are characterized by their
capacity to produce secreted proteins (26, 30). In addition, like
many other eubacteria, S. lividans has the ability to glycosylate
its own proteins, as well as heterologous proteins (18, 21, 28).
The ability to glycosylate cloned gene products enhances the
usefulness of Streptomyces as a host for the production of
heterologous polypeptides, and this system is a potent tool for
studying glycosylation processes in bacteria. The existence of
vectors for inducible protein expression in S. lividans allows
production of large amounts of proteins suitable for immuno-
* Corresponding author. Mailing address: Instituto de Investigacio-
nes Biome´dicas, Departamento de Inmunologı´a, Apartado Postal 70-
228, 04510 Me´xico, D.F., Me´xico. Phone (525) 6223818. Fax: (525)
6223369. E-mail: espitia@servidor.unam.mx.
679
logical and biochemical characterization of glycoproteins. In
this study we expressed the M. tuberculosis 45/47 kDa protein
in S. lividans in order to assess the potential of the expression
system for obtaining M. tuberculosis glycoproteins with vaccine
and/or diagnostic potential.
MATERIALS AND METHODS
Bacterial strains and plasmids. Escherichia coli XL1-Blue was used as a host
for recombinant plasmids. The laboratory strain M. tuberculosis H37Rv was
obtained from the American Type Culture Collection (Rockville, Md.). Wild-
type S. lividans 1326 and the plasmid vectors pIJ486 and pIJ6021 (4) were
obtained from D. A. Hopwood, John Innes Centre, Norwich, United Kingdom.
Isolation and cloning of the DNA region carrying the 45/47 kDa protein gene.
A cosmid clone carrying the M. tuberculosis gene for the 45/47 kDa protein was
isolated by screening the Tropist3 DNA cosmid library of M. tuberculosis H37Rv
(17); a PCR product corresponding to the amplied gene was used as the probe.
DNA from the positive cosmid colony was enzyme restricted. Fragments were
separated on 8% agarose gels and transferred by blotting onto nylon lters
(Amersham). The lters were then prehybridized and probed at 42°Cin6SSC
(1SSC is 0.15 M NaCl plus 0.015 M sodium citrate, pH 7.0) containing 1 mM
sodium phosphate, 1 mM EDTA, 0.05% skim milk, and 0.5% SDS for 2 and 4 h,
respectively. After this, the lters were washed twice in 2SSC for 15 min each
time and once in 2SSC0.3% SDS for 15 min and autoradiographed by
exposing the lters to X-ray lm (Kodak). A 3.2-kb EcoRI fragment from this
cosmid clone was subcloned into the EcoRI site of pUC18 to obtain pUC18MT-
45. This plasmid carried the complete gene for the 45/47 kDa protein, together
with additional DNA on either side of the gene (1.1 kb upstream and 1.0 kb
downstream).
Cloning and expression of the gene encoding the 45/47 kDa protein in S.
lividans.The 3.2-kb EcoRI fragment obtained from pUC18MT-45 was subcloned
into the EcoRI site of the high-copy-number Streptomyces plasmid vector pIJ486
(4), resulting in plasmid pIJ486MT-45.
PCR amplication of a 983-bp fragment containing the complete 45/47 kDa
protein gene was carried out with oligonucleotides CGGATCCATATGCATC
AGGTGGACCC and GGAATTCAGGCCGGTAAGGTCC. The BamHI and
NdeI restriction sites in the sense primer and the EcoRI site in the reverse primer
(underlined) were included as extensions for further manipulation of the ampli-
ed fragments. In particular, the NdeI site (CATATG) was designed to contain
the ATG start codon of the gene in order to allow cloning into the Streptomyces
expression vector pIJ6021 (4). Amplication was carried out with Taq DNA
polymerase (Perkin-Elmer) as recommended by the manufacturer. The PCR
protocol consisted of an initial denaturation step of 5 min at 95°C, followed by 30
cycles of 1 min of denaturation at 95°C, 1 min of annealing at 50°C, and 1 min
of extension at 72°C and then a 5-min nal extension at 72°C. The PCR product
was then digested with BamHI and EcoRI and subcloned into BamHI-EcoRI-cut
pUC18 to obtain plasmid pUC18MT-45.1. To clone this fragment in the Strep-
tomyces expression vector pIJ6021, pUC18MT-45.1 was digested with NdeI and
EcoRI, and the 1-kb insert was subcloned into NdeI-EcoRI-cut pIJ6021 to obtain
pIJ6021MT-45.
Growth of Streptomyces cultures. Spores of S. lividans carrying the different
plasmids were obtained on solid R5 medium (4) with the appropriate antibiotics.
Freshly harvested spores were used to inoculate Luria-Bertani broth modied by
addition of 34% sucrose to obtain dispersed mycelial growth. For growth of S.
lividans harboring pIJ486MT-45, thiostrepton was added at a concentration of 50
g/ml, and cultures were grown at 30°C for 72 h with shaking. For cultures
carrying pIJ6021MT-45, kanamycin was added at a concentration of 100 g/ml,
and after 12 h of growth at 30°C, thiostrepton was added at a concentration of 10
g/ml and growth was continued for an additional 24 to 36 h. Subsequently,
recombinant culture ltrates (rCF) were obtained by removing the mycelium by
centrifugation at 8,000 gfor 30 min at 4°C and ltration through Whatman no.
1lter paper disks. Proteins were precipitated from the supernatant with am-
monium sulfate (73% saturation), recovered by centrifugation, dialyzed against
distilled H
2
O, and dried by lyophilization.
Growth of M. tuberculosis cultures. M. tuberculosis H37Rv was cultured on
Proskauer-Beck synthetic medium for 4 to 6 weeks. Culture ltrate (CF) proteins
and a fraction enriched with the 45/47 kDa protein (F4) were obtained from CF
as described elsewhere (6, 7).
Antibodies. Monoclonal antibody 6A3 (MAb 6A3) raised against the 45/47
kDa protein was obtained as described previously (8). Rabbit polyclonal anti-
serum against a synthetic peptide (GEVAPTPTTPTPQRTLPAC) derived from
the C-terminal sequence of the 45/47 kDa protein was produced in New Zealand
rabbits. Animals were immunized subcutaneously on days 0 and 8 with 200 gof
puried peptide in incomplete Freunds adjuvant. Two weeks later they were
boosted intraperitoneally with 100 g of puried peptide, and then they were
boosted every fortnight for 2 months. Two weeks after the last immunization the
animals were bled, and the resulting antiserum was designated anti-C45.
Human sera. Sera from patients with pulmonary tuberculosis diagnosed by
smear and/or sputum culture were obtained from Hospital General in Mexico.
Healthy control sera were obtained from laboratory workers.
SDS-PAGE and Western blotting. Electrophoresis in 12% polyacrylamide gels
containing SDS and subsequent immunoblotting procedures were carried out by
using the standard methods (19, 38). Five-microgram portions of S. lividans rCF
and M. tuberculosis F4 were electrophoresed in polyacrylamide gels. For antigen
detection, proteins were transferred to polyvinylidene diuoride Immunobilon
nylon membranes (Millipore) and incubated with MAb 6A3 and with anti-C45.
For carbohydrate detection, proteins were stained with concanavalin A (ConA)-
peroxidase (Sigma). Nonspecic binding was blocked by incubating blots with
1% (wt/vol) bovine serum albumin (BSA) in phosphate-buffered saline (PBS)
containing Tween 20 (0.05%, vol/vol); after washes with PBS-Tween 20, the
membranes were incubated for1hatroom temperature (RT) with either MAb
6A3 diluted 1/500, anti-C45 diluted 1/2,000, or ConA (2.5 g/ml). After washes
with PBS-Tween 20, the membranes were incubated with peroxidase conjugates,
anti-mouse immunoglobulin G diluted 1/2,000 (Zymed), or protein A (Sigma)
diluted 1/2,000. After incubation for 30 min at RT, the blots were stained for
peroxidase activity by adding 3,3-diaminobenzidine (Sigma) and hydrogen per-
oxide in PBS.
Two-dimensional PAGE was performed as follows. Urea was added to 10 g
of rCF from S. lividans transformed with pIJ486MT-45 at a nal concentration of
9 M, and then lysis buffer containing urea and Nonidet P-40 was added as
described by OFarrell (27). Samples were separated initially by isoelectric fo-
cusing in tube gels containing 4% ampholytes in the pH range from 3.5 to 5.0
(Pharmacia) and then by SDS-PAGE in the second dimension as described
above. Proteins were transferred to nylon membranes and incubated with MAb
6A3. Reactivity was developed as described above. Labeling of glycopropteins
with biotin-hydrazide and jack bean -D-mannosidase digestion were performed
as described previously (8).
Purication of proteins. The pH of rCF extract from S. lividans transformed
with pIJ6021MT-45 was adjusted to 5 with acetic acid at 4°C, proteins were
recovered by centrifugation at 8,000 gfor 10 min, and the pellet was resus-
pended in the appropriate buffer. The chromatography procedures were per-
formed with an AKTA-Prime system (Pharmacia Biotech). The ConA-binding
protein (47-kDa protein) was separated from the 45-kDa protein by afnity
chromatography on a ConA-Sepharose column (Pharmacia Biotech); elution
was carried out with a gradient of 0 to 0.05 M -methyl-mannopyranoside
(Sigma) (7). Flowthrough from the ConA column contained the 45-kDa protein,
which was loaded onto a HiTrap Q-Sepharose column equilibrated with 20 mM
Tris-HCl (pH 8). Elution was then carried out with a linear 0 to 1 M NaCl
gradient. Fractions were collected, dialyzed, concentrated, and analyzed by SDS-
PAGE. The r45/47 kDa protein was also puried by anion-exchange chromatog-
raphy from pH 5 rCF supernatant by using the protocol described above. Protein
quantication was carried out with a protein quantication kit (Bio-Rad).
Fingerprinting assay. Digestion of individually puried recombinant 45- and
47-kDa proteins was performed with the protein ngerprint system (Promega)
used according to the manufacturers protocol. Briey, 1 g of puried fractions
or 2.5 g of the r45/47 kDa protein was diluted in electrophoresis cocktail and
heat denatured by incubation at 95°C for 5 min. Then 5 l (0.2 g) of Lys-C and
5l (0.2 g) of Glu-C were added to each sample; after digestion, samples were
transferred to polyvinylidene diuoride nylon membranes after SDS15%
PAGE. Blots were stained with Coomassie blue, and the bands were cut out and
subjected to automated Edman degradation with a gas phase sequencer (PE-
Applied Biosystems, Weiterstadt, Germany).
Protein preparation for mass spectrometric analysis. r45/47 kDa protein pu-
ried by anion-exchange chromatography was separated by SDS-PAGE. Indi-
vidual 45- and 47-kDa bands were excised from the gel after Coomassie blue
staining. The gel slides were washed several times with 200 l of water, dehy-
drated in 50 l of acetonitrile, and dried. Then the gel pieces were washed twice
with 100 mM NH
4
HCO
3
, dehydrated with acetonitrile, and dried. Digestion of
proteins was carried out in 50 mM NH
4
HCO
3
containing 4 ng of trypsin (Pro-
mega Corp.) per lat37°C overnight (15 h). The resulting peptides were
extracted with 25 mM NH
4
HCO
3
acetonitrile and then with 5% formic acid
(HCOOH)acetonitrile. After drying, the dried peptides were reconstituted in
20 l of 0.5% HCOOH5% methanol (MeOH). The peptides were then puried
on reversed-phase C
18
ZipTip pipette tips (Millipore Corp.). Briey, ZipTips
were washed with 0.5% HCOOH65% MeOH and equilibrated with 0.5%
680 LARA ET AL. APPL.ENVIRON.MICROBIOL.
HCOOH5% MeOH. The peptides were applied to the ZipTips and eluted with
1.0% HCOOH65% MeOH.
Matrix-assisted laser desorption ionizationtime of ight (MALDI-TOF) mass
spectrometry (MS) of tryptic peptides. A 0.5- to 1-l portion of each concen-
trated peptide solution was mixed with the same volume of a saturated matrix
solution of -cyano-4-hydoxycinnamic acid (Bruker Daltonics) in 0.5%
HCOOH65%MeOH, spotted onto a 384 MTP target, and dried at RT. The
molecular masses of the tryptic peptides were determined in the positive-ion
mode with a Bruker Ultraex TOF mass spectrometer (Bruker Daltonics GmbH,
Leipzig, Germany) by using the reectron and delayed extraction facilities for
enhanced resolution, an N
2
laser (337 nm) operating with a 3-ns pulse width and
10
7
to 10
8
W/cm
2
at the surface of 0.2-mm
2
spots, and an acceleration voltage of
25 kV.
Enzyme-linked immunosorbent assay (ELISA). Polystyrene 96-well microtiter
plates (Costar 3590) were coated with 100 lof2.5g of 45-kDa protein per ml,
glycosylated 47-kDa protein, and jack bean -mannosidase-treated 47-kDa pro-
tein in 0.05 M carbonate-bicarbonate buffer (pH 9.6) at 37°C overnight. The
plates were blocked with 2% BSA in PBS0.05% Tween 20 for1hatRT.Four
human tuberculosis sera previously found to react against native 45/47 kDa
protein and four healthy sera were chosen for the assay. One hundred microliters
of a 1:100 dilution of each serum and a 1:1,000 dilution of MAb 6A3 were added
to each well in triplicate in PBS-Tween 20-BSA and incubated for1hatRT.
After the plates were washed with PBS-Tween 20-BSA, they were incubated with
the appropriate horseradish conjugates diluted in PBS-Tween 20-BSA contain-
ing protein A diluted 1:2,000 and anti-mouse immunoglobulin G diluted 1:2,000.
The plates were incubated for 1 h and then washed. Enzyme activity was assayed
by incubation for 5 min at RT with 50 lof-phenylendiamine (Sigma). The
reaction was stopped with 50 l of 3 N HCl, and the optical density at 492 nm was
determined with an automatic microtiter plate reader (Labsystem).
RESULTS
Expression of the 45/47 kDa protein gene of M. tuberculosis
from its own promoter and from the P
tipA
promoter. The M.
tuberculosis gene encoding the 45/47 kDa protein was cloned in
the Streptomyces vector pIJ486, resulting in pIJ486MT-45. To-
tal extracellular protein from S. lividans carrying either pIJ486
or pIJ486MT-45 was electrophoresed on SDS-PAGE gels. The
45/47 kDa protein bands could not be clearly distinguished
from those of S. lividans carrying only the vector (Fig. 1A, lanes
1 and 2). However, Western blot analysis showed that the gene
was expressed in cultures of S. lividans carrying pIJ486MT-45
and that the 45/47 kDa protein was released into the medium
(Fig. 1B, lane 2) (see below). Since in pIJ486MT-45 the cloned
gene is located downstream of a transcriptional terminator
(17), transcription must originate from a promoter present in
the cloned fragment. Overexpression of the 45/47 kDa protein
was achieved by cloning the gene in the expression vector
pIJ6021 under the thiostrepton-inducible promoter P
tipA
,re
-
sulting in plasmid pIJ6021MT-45 (Fig. 1A, lane 4). Total ex-
tracellular protein from S. lividans carrying either pIJ6021 or
pIJ6021MT-45 was analyzed by SDS-PAGE (Fig. 1A, lanes 3
and 4); it was evident that large amounts of the 45/47 kDa
protein were secreted into the culture medium, where it rep-
resented about 25% of the total extracellular protein (Fig. 1A,
lane 4).
Characterization of recombinant proteins by SDS-PAGE
and Western blotting. CF protein extracts from S. lividans
carrying pIJ486MT-45 were prepared and subjected to West-
ern blot analysis with MAb 6A3 and anti-C45 rabbit polyclonal
antibody. MAb 6A3 recognized a double band, which migrated
slightly above the native protein from M. tuberculosis H37Rv in
SDS-PAGE gels (Fig. 1B, lanes 1 and 2). The polyclonal anti-
C45 antibody recognized only the 47-kDa upper band of the
native protein and the 45-kDa lower band of the r45/47 kDa
protein (Fig. 1B, lanes 3 and 4). When the recombinant protein
was analyzed by two-dimensional gel electrophoresis, the up-
per and lower bands had identical acidic isoelectric points (Fig.
1C), in contrast to the native protein bands, which have been
shown to migrate as several spots having different isoelectric
points (8).
To determine whether the recombinant protein produced by
S. lividans was able to bind to ConA, as previously described
for the M. tuberculosis native 45/47 kDa protein (7, 8), rCF
proteins were transferred to a nylon membrane after SDS-
PAGE and incubated with ConA-peroxidase. Only the upper
band showed positive staining with the lectin (Fig. 2A, lane 3).
Accordingly, only this band showed an apparent reduction in
molecular mass of about 0.5 kDa after digestion with -D-
mannosidase, which caused it to migrate at the same level as
the lower band (Fig. 2A, lane 2). In addition, -D-mannosidase
digestion eliminated the ConA reactivity of the upper band
(Fig. 2A, lane 4). To assess whether the recombinant lower
band was glycosylated, recombinant and control S. lividans CF
extracts were labeled with biotin-hydrazide after periodate ox-
idation and then transferred to a nylon membrane and incu-
bated with streptavidine-peroxidase. Although several proteins
in the S. lividans control CF were labeled with biotin-hydra-
zide, the r45/47 kDa bands could be clearly distinguished when
the rCF was used. Differences in the intensity of biotin-hydra-
zide labeling between the bands were observed; the lower band
was most intensively labeled, probably due to differences in
glycosylation between the bands (Fig. 2B, lanes 1 and 2). Con-
trol membranes were developed with MAb 6A3 (Fig. 2B, lanes
3 and 4).
Protein purication. The r45/47 kDa protein was puried
from culture supernatants by anion-exchange chromatography,
and both forms of the protein eluted in one peak at 0.3 M
FIG. 1. Expression of M. tuberculosis 45/47 kDa protein in S. livi-
dans. (A) Coomassie blue-stained SDS-PAGE gel of rCF from S.
lividans carrying different plasmids. Lane 1, pIJ486 control vector with-
out insert; lane 2, pIJ486MT-45 recombinant vector; lane 3, pIJ6021
vector with thiostrepton-inducible Streptomyces promoter P
tipA
; lane 4,
pIJ6021MT-45 recombinant vector. (B) Western blot of native and
rCF proteins. Lanes 1 and 3, M. tuberculosis 45/47 kDa CF-enriched
fraction (F4) as a positive control; lanes 2 and 4, rCF from S. lividans
carrying pIJ486MT-45, which overexpressed the 45/47 kDa protein.
Lanes 1 and 2 were developed with MAb 6A3, while lanes 3 and 4 were
developed with anti-C45 polyclonal antibody. (C) Two-dimensional
PAGE analysis of rCF from S. lividans carrying pIJ486MT-45. The blot
was developed with MAb 6A3.
VOL. 70, 2004 S. LIVIDANS, GLYCOSYLATION, AND M. TUBERCULOSIS 681
NaCl. About 5 mg of protein was obtained from 1 liter of
culture. The protein was further separated into ConA-binding
and nonbinding fractions by ConA afnity chromatography.
The fraction bound to the ConA-Sepharose column was eluted
with -methyl-mannopyranoside and migrated as the 47-kDa
upper band in SDS-PAGE gels. On the other hand, the frac-
tion did not bind to the ConA-Sepharose column and was
recovered from the owthrough from a HiTrap Q-Sepharose
column, and it migrated as a 45-kDa band in SDS-PAGE gels
(Fig. 3).
Fingerprinting. Endoproteinase digestion of the 47- and 45-
kDa proteins (upper and lower bands, respectively) produced
identical proteolytic patterns. Lys-C digestion released two
peptides with apparent molecular masses of about 20 and 30
kDa, while Glu-C digestion produced two peptides with appar-
ent molecular masses of about 19 and 31 kDa (results not
shown). The N-terminal sequences of the undigested 47- and
45-kDa proteins were determined; the two sequences were
identical, and this showed that the signal peptide was cleaved
off at precisely the same position as in the native protein. This
result agrees with the presence in S. lividans of a signal pepti-
dase that cleaves at the consensus amino acid sequence AXA
in the leader peptide (29). On the other hand, the N-terminal
sequences of the proteolytic fragments generated from diges-
tion of the 47- and 45-kDa proteins were also identical. Figure
4 shows the proteolytic enzyme digestion sites based on the
N-terminal sequences of the different peptides.
The MALDI-TOF MS peptide map of the tryptically di-
gested recombinant 47-kDa protein is shown in Fig. 5. All
major peaks could be assigned. The signal comprising amino
acids 150 to 165 had a molecular ion 76 Da lower than ex-
pected. MS-MS analysis unequivocally demonstrated that
there was a change from Y to S at position 161 (data not
FIG. 2. -D-Mannosidase treatment and biotin-hydrazide labeling
of r45/47 kDa protein. (A) Effects of treatment with -D-mannosidase
of rCF extracts from S. lividans carrying the pIJ486MT-45 vector.
Lanes 1 and 3, rCF extracts not treated with -D-mannosidase; lanes 2
and 4, rCF extracts treated with -D-mannosidase. Lanes 1 and 2 were
developed with MAb 6A3, while lanes 3 and 4 were developed with
ConA-peroxidase. The arrow in lane 4 indicates the position of the
-D-mannosidase enzyme which reacted with ConA. (B) Biotin-hydra-
zide labeling. Lane 1, rCF from S. lividans carrying pIJ486; lane 2, rCF
from S. lividans carrying pIJ486MT-45. Blots were developed with
streptavidine-peroxidase. Lanes 3 and 4 were the same as lanes 1 and
2, but the blots were developed with MAb 6A3.
FIG. 3. Purication of r45/47 kDa protein: prole of ConA afnity
chromatography. The left lane of the inset shows the protein that was
retained by the ConA column after elution (47-kDa band), which was
rechromatographed by anion-exchange chromatography and analyzed
by SDS-PAGE. The right lane of the inset shows protein that was not
retained by the ConA column (45-kDa band), which was rechromato-
graphed by anion-exchange chromatography and analyzed by SDS-
PAGE. B in the yaxis is NaCl gradient from 0 to 1 M.
FIG. 4. Cleavage sites of r45/47 kDa protein with proteolytic en-
zymes. The cleavage sites for each enzyme are indicated by arrows, and
the N-terminal sequence obtained for each fragment is indicated by
boldface type. The individual glycosylation sites of native protein are
enclosed in brackets, and glycosylated recombinant peptides are un-
derlined.
FIG. 5. MALDI-TOF peptide map of the tryptically digested re-
combinant 47-kDa protein. The signal comprising amino acids 150 to
165 showed a molecular ion 76 Da lower than expected. MS-MS
analysis unequivocally demonstrated that there was a change from Y to
S at position 161. Both the N-terminal peptide T1-73 and the C-
terminal peptide T239-282 were found to be posttranslationally mod-
ied by hexose residues.
682 LARA ET AL. APPL.ENVIRON.MICROBIOL.
shown). This change could have been due to an error during
PCR amplication of the gene. Both the N-terminal peptide
T1-73 and the C-terminal peptide T239-282 were found to be
posttranslationally modied by hexose residues, which could
be identied as mannose due to the binding characteristics of
the protein to ConA. Zero to nine mannose residues were
detected in the T1-73 peptide, as indicated by a ladder of
molecular ions differing by 162 Da, which is characteristic of
hexose residues, whereas zero to four mannose residues were
found to be linked to the T239-282 peptide (Fig. 5). Glycosy-
lation on the 45-kDa protein was also found at the same po-
sition of the 47-kDa protein but with a different mannosylation
pattern, the unglycosylated N-terminal peptide had the highest
signal intensity, and peptides with one to ve mannose residues
were detectable with less than one-third the intensity of the
unglycosylated peptide. In the C-terminal peptide, the intensity
of the glycopeptides with one to three mannose residues was
less than 20% of the intensity of the unglycosylated peptide
(data not shown).
ELISA. Comparison of reactivities of human tuberculosis
and control sera by the ELISA showed that only the ConA-
binding 47-kDa protein was recognized by sera of individuals
infected with M. tuberculosis; in contrast, MAb 6A3 recognized
both the 45- and 47-kDa proteins (Fig. 6). The interaction of
antibodies with the 47-kDa protein was lost after treatment
with jack bean -D-mannosidase.
DISCUSSION
There have been several reports of mycobacterial gene ex-
pression in Streptomyces. In particular, expression of M. bovis
BCG and M. leprae genes in S. lividans has been reported
elsewhere (16, 20). More recently, two major antigens of M.
tuberculosis, the 38- and 19-kDa proteins (Rv0934 and
Rv3763), were overproduced by S. lividans as secreted extra-
cellular proteins when their genes were cloned in an engi-
neered expression-secretion vector (39). In this work, the 45/47
kDa glycoprotein of M. tuberculosis was expressed in S. livi-
dans. The gene that encodes this protein (modD) is the fourth
gene of a putative molybdenum transport operon. Even though
the fragment cloned in the vector pIJ486 does not carry the
entire operon, expression was observed. Transcriptional read-
through from the vector can be ruled out as the cause of ex-
pression, since the gene was cloned downstream of a strong
transcriptional terminator (4). Therefore, it is very likely that
the fragment cloned in pIJ486, which carried 1 kb of DNA
upstream of the gene, carried an internal promoter capable of
driving expression of the M. tuberculosis 45/47 kDa protein
gene in S. lividans. This shows that Streptomyces is able to rec-
ognize M. tuberculosis promoters, which is not surprising given
the relatedness of these organisms in the actinomycetes group,
which has been shown to extend to the genome level (1). The
presence of promoters which are internal to operons in myco-
bacteria has been described recently for the phosphate-specic
transport operon, which encodes the PstS-1 protein, and for
genes of the putative mpt70-mpt83 operon (14, 37).
Studies on glycosylation of mycobacterial proteins by S. livi-
dans are scarce. However, a few reports have shown that this
microorganism has protein glycosylation ability (18, 21, 28).
In the present work, the well-known M. tuberculosis 45/47
kDa glycoprotein was expressed in S. lividans, and we showed
that the protein is glycosylated. Like the native protein, the
recombinant protein was secreted into the medium and mi-
grated as a double band in SDS-PAGE gels; however, in con-
trast to the native mycobacterial protein, only the upper 47-
kDa band of the recombinant protein from S. lividans reacted
with ConA. This suggests either that the protein is glycosylated
in S. lividans by sugars other than mannose or that it is glyco-
sylated in a conguration not recognized by ConA. In addition,
only the 45-kDa lower band of the recombinant protein could
be recognized by the anti-C45 antibody. This difference in
reactivity between the native and recombinant proteins can be
explained by differences in glycosylation. The same explanation
can be extrapolated to the native 45/47 kDa protein, in which
the existence of the two bands can be attributed to changes in
glycosylation rather than to degradation or C-terminal modi-
cations. Our observations support the hypothesis that differ-
ences in glycosylation are the cause of the two forms of the
recombinant protein, since no differences in N-terminal se-
quences or isoelectric points could be found between the 45-
and 47-kDa proteins. In addition, evidence supporting the hy-
pothesis that the recombinant protein is glycosylated at the
same position as the native protein was obtained from the
ConA reactivity of all peptides generated by Glu-C and Lys-C
digestion of the S.lividans r45/47 kDa protein (data not shown).
This nding was further supported by the MALDI-TOF MS
analysis that showed that, as in the native protein, the glyco-
sylation sites of the r45/47 kDa protein are located in both the
N and C termini of the molecule. Interestingly, the glycosyla-
tion pattern of the recombinant N terminus from zero to nine
hexose residues was very similar to that of the native proteins
from three M. tuberculosis reference strains (12). In contrast,
differences in the degree of glycosylation were found between
recombinant 47- and 45-kDa forms of the protein, as reported
elsewhere for the native protein (12); these differences could
explain the existence of two forms of the molecule, as well as
the differences in ConA reactivity and biotin labeling. There-
fore, it is important to determine if the individual glycosylation
sites of the r45/47 kDa protein are the same as those dened
for the native protein (5). Finally, an interesting observation
FIG. 6. ELISA of puried 45- and 47-kDa proteins: reactivities of
antibodies with the ConA-binding 47-kDa protein (open bars), with
the -D-mannosidase-treated 47-kDa protein (gray bars), and with the
non-ConA-binding 45-kDa protein (solid bars). Reactivity was tested
with MAb 6A3 (6A3), with sera from human tuberculosis patients
(TB) (2), and with sera from healthy individuals (Healthy) (3). The
results are expressed as mean optical. densities and are representative
of three separate experiments.
VOL. 70, 2004 S. LIVIDANS, GLYCOSYLATION, AND M. TUBERCULOSIS 683
was the different reactivities of human tuberculosis-infected
sera with the ConA-binding 47-kDa protein and the nonbind-
ing 45-kDa protein, as analyzed by the ELISA. Antibodies
from tuberculosis patients recognized only the upper ConA-
binding fraction, suggesting that antibodies in the sera had
been generated against the carbohydrate residues. This obser-
vation was conrmed by the loss of antibody binding to degly-
cosylated 47-kDa protein.
Carbohydrates that decorate the surfaces of infectious
agents are considered pathogen-associated molecular patterns,
and they are recognized by pattern recognition receptors, such
as the mannose receptor and the dendritic cell-specic intra-
cellular adhesion molecule 3-grabbing nonintegrin receptor,
which play a key role in innate and adaptive immunity (9, 35).
ConA-binding carbohydrates have been found in lipoarabino-
mannan and other important antigens of M. tuberculosis, like
the 19- and 38-kDa antigens (7, 11, 13). The interaction of
LAM with the mannose receptor has been widely documented
(3, 15), and more recently the dendritic cell-specic intracel-
lular adhesion molecule 3-grabbing nonintegrin receptor has
been dened as the major M. tuberculosis receptor on human
dendritic cells with the capacity to discriminate between My-
cobacterium species through selective recognition of the man-
nose caps on LAM (22, 36). These observations suggest that
the presence of mannose residues in mycobacterial molecules
could be an important signal for host recognition through
mannose receptors or other carbohydrate recognition recep-
tors, and therefore, carbohydrate motifs in glycoproteins of
mycobacteria could play an important role in both the cellular
(12, 32) and humoral immune responses.
ACKNOWLEDGMENTS
This work was supported by grant IN221599 from DGAPA-Univer-
sidad Nacional Auto´noma de Me´xico and by grants 33580-M and
CONACYT-DLR from CONACYT.
The Tropist 3 cosmid library of M. tuberculosis DNA was kindly
provided by K. De Smet (Tuberculosis and Related Infections Unit,
Medical Research Council, Clinical Sciences Centre, London, United
Kingdom). We thank Rafael Cervantes and Gabriela Gonza´lez-Cero´n
for technical assistance, Rita Getzlaff for protein sequencing, and
Isabel Pe´rez Montfort for reviewing the English version of the manu-
script.
REFERENCES
1. Bentley, S. D., K. F. Chater, A. M. Cerdeno-Tarraga, G. L. Challis, N. R.
Thomson, K. D James, D. E. Harris, M. A. Quail, H. Kieser, D. Harper, A.
Bateman, S. Brown, G. Chandra, C. W. Chen, M. Collins, A. Cronin, A.
Fraser, A. Goble, J. Hidalgo, T Hornsby, S. Howarth, C. H. Huang, T. Kieser,
L. Larke, L. Murphy, K. Oliver, S. ONeil, E. Rabbinowitsch, M. A. Rajan-
dream, K. Rutherford, S. Rutter, K. Seeger, D. Saunders, S. Sharp, R.
Squares, S. Squares, K. Taylor, T. Warren, A. Wietzorrek, J. Woodward,
B. G. Barrell, J. Parkhill, and D. A. Hopwood. 2002. Complete genome
sequence of the model actinomycete Streptomyces coelicolor A3(2). Nature
417:141147.
2. Benz, I., and M. A. Schmidt. 2002. Never say never again: protein glycosy-
lation in pathogenic bacteria. Mol. Microbiol. 45:267276.
3. Bernardo, J., A. M. Billingslea, R. L. Blumenthal, K. F. Seetoo, E. R. Simons,
and M. J. Fenton. 1998. Differential responses of human mononuclear
phagocytes to mycobacterial lipoarabinomannans: role of CD14 and the
mannose receptor. Infect. Immun. 66:2835.
4. De Smet, K. A., S. Jamil, and N. G. Stoker. 1993. Tropist3: a cosmid vector
for simplied mapping of both GC rich and AT rich genomic DNA.
Gene 136:215219.
5. Dobos, K. M., K. H. Khoo, K. M. Swiderek, P. J. Brennan, and J. T. Belisle.
1996. Denition of the full extent of glycosylation of the 45-kilodalton gly-
coprotein of Mycobacterium tuberculosis. J. Bacteriol. 178:24982506.
6. Espitia, C., I. Cervera, R. Gonzalez, and R. Mancilla. 1989. A 38-kD Myco-
bacterium tuberculosis antigen associated with infection. Its isolation and
serologic evaluation. Clin. Exp. Immunol. 77:373377.
7. Espitia, C., and R. Mancilla. 1989. Identication, isolation and partial char-
acterization of Mycobacterium tuberculosis glycoprotein antigens. Clin. Exp.
Immunol. 77:378383.
8. Espitia, C., R. Espinosa, R. Saavedra, R. Mancilla, F. Romain, A. Laqueyre-
rie, and C. Moreno. 1995. Antigenic and structural similarities between
Mycobacterium tuberculosis 50- to 55-kilodalton and Mycobacterium bovis
BCG 45- to 47-kilodalton antigens. Infect. Immun. 63:580584.
9. Feinberg, H., D. A. Mitchell, K. Drickamer, and W. I. Weis. 2001. Structural
basis for selective recognition of oligosaccharides by DC-SIGN and DC-
SIGNR. Science 294:21632166.
10. Fis, T., C. A. J. Costopoulos, A. Bacic, and P. R. Wood. 1991. Purication
and characterization of major antigens from a Mycobacterium bovis culture
ltrate. Infect. Immun. 59:800807.
11. Garbe, T., D. Harris, M. Vordemeier, R. Lathigra, J. Ivanyi, and D. Young.
1993. Expression of the Mycobacterium tuberculosis 19-kilodalton antigen in
Mycobacterium smegmatis: immunological analysis and evidence of glycosy-
lation. Infect. Immun. 61:260267.
12. Horn, C., A. Namane, P. Pescher, M. Riviere, F. Romain, G. Puzo, O. Barzu,
and G. Marchal. 1999. Decreased capacity of recombinant 45/47-kDa mol-
ecules (Apa) of Mycobacterium tuberculosis to stimulate T lymphocyte re-
sponses related to changes in their mannosylation pattern. J. Biol. Chem.
274:3202332030.
13. Hunter, S. W., H. Gaylord, and P. J. Brennan. 1986. Structure and antige-
nicity of the phosphorylated lipopolysaccharide antigens from the leprosy
and tubercle bacilli. J. Biol. Chem. 261:1234512351.
14. Juarez, M. D., A. Torres, and C. Espitia. 2001. Characterization of the
Mycobacterium tuberculosis region containing the mpt83 and mpt70 genes.
FEMS. Microbiol. Lett. 203:95102.
15. Kang, B. K., and L. S. Schlesinger. 1998. Characterization of mannose
receptor-dependent phagocytosis mediated by Mycobacterium tuberculosis
lipoarabinomannan. Infect. Immun. 66:27692777.
16. Kieser, T., M. T. Moss, J. W. Dale, and D. A. Hopwood. 1986. Cloning and
expression of Mycobacterium bovis BCG DNA in Streptomyces lividans.J.
Bacteriol. 168:7280.
17. Kieser, T., M. J. Bribb, M. J. Buttner, K. F. Chater, and P. A. S. Hopwood.
2000. Practical Streptomyces genetics. The John Innes Foundation, Norwich,
United Kingdom.
18. Kluepfel, D., S. Vats-Mehta, F. Aumont, F. Shareck, and R. Morosoli. 1990.
Purication and characterization of a new xylanase (xylanase B) produced by
Streptomyces lividans 66. Biochem. J. 267:4550.
19. Laemmli, U. K. 1970. Cleavage of structural proteins during the assembly of
the head of bacteriophage T4. Nature 277:680685.
20. Lamb, F. I., A. E. Kingston, I. Estrada, and M. J. Colston. 1988. Heterolo-
gous expression of the 65-kilodalton antigen of Mycobacterium leprae and
murine T-cell responses to the gene product. Infect. Immun. 56:12371241.
21. MacLeod, A. M., N. R. Gilkes, L. Escote-Carlson, R. A. Warren, D. G.
Kilburn, and R. C. Miller, Jr. 1992. Streptomyces lividans glycosylates an
exoglucanase (Cex) from Cellulomonas mi. Gene 121:143147.
22. Maeda, N., J. Nigou, J. L. Herrmann, M. Jackson, A. Amara, P. H. Lagrange,
G. Puzo, B. Gicquel, and O. Neyrolles. 2003. The cell surface receptor
DC-SIGN discriminates between Mycobacterium species through selective
recognition of the mannose caps on lipoarabinomannan. J. Biol. Chem.
278:55135516.
23. Messner, P. 1997. Bacterial glycoproteins. Glycocon. J. 14:311.
24. Michell, S. L., A. O. Whelan, P. R. Wheeler, M. Panico, R. L. Easton, T.
Etienne, S. M. Haslam, A. Dell, H. R. Morris, A. J. Reason, L. Herrmann,
D. B. Young, and G. Hewinson. 2003. The MPB83 antigen from Mycobacte-
rium bovis contains O-linked mannose and (133)-mannobiose moieties.
J. Biol. Chem. 278:1642316432.
25. Moens, S., J. F. Vanderleyden, and A. Janssens. 1997. Glycoproteins in
prokaryotes. Arch. Microbiol. 168:169175.
26. Morosoli, R., F. Shareck, D. Kluepfel. 1997. Protein secretion in streptomy-
cetes. FEMS Microbiol. Lett. 146:167174.
27. OFarrel, P. H. 1975. High resolution two-dimensional electrophoresis of
proteins. J. Biol. Chem. 250:40074021.
28. Ong, E., D. G. Kilburn, R. C. Miller, Jr., and R. A. Warren. 1994. Strepto-
myces lividans glycosylates the linker region of a beta-1,4-glycanase from
Cellulomonas mi. J. Bacteriol. 176:9991008.
29. Page, N., D. Kluepfel, F. Shareck, and A. R. Morosoli. 1996. Effect of signal
peptide alterations and replacement on export of xylanase in Streptomyces
lividans. Appl. Environ. Microbiol. 62:109114.
30. Pozidis, C., E. Lammertyn, A. S. Politou., J. Anne, A. S. Tsiftsoglou, G.
Sianidis, and A. Economou. 2001. Protein secretion biotechnology using
Streptomyces lividans: large-scale production of functional trimeric tumor
necrosis factor alpha. Biotechnol. Bioeng. 72:611619.
31. Ratliff, T. L., R. McCarthy, W. B. Telle, and E. J. Brown. 1993. Purication
of a mycobacterial adhesin for bronectin. Infect. Immun. 61:18891894.
32. Romain, F., C. Horn, P. Pescher, A. Namane, M. Riviere, G. Puzo, O. Barzu,
and G. Marchal. 1999. Deglycosylation of the 45/47 kilodalton antigen com-
plex of Mycobacterium tuberculosis decreases its capacity to elicit in vivo or in
vitro cellular immune responses. Infect. Immun. 67:55675572.
33. Schorey, J. S., Q. Li, D. W. McCourt, M. Bong-Mastek, J. E. Clark-Curtiss,
684 LARA ET AL. APPL.ENVIRON.MICROBIOL.
T. L. Ratliff, and E. J. Brown. 1995. Mycobacterium leprae gene encoding a
bronectin binding proteins is used for efcient invasion of epithelial cells
and Schwann cells. Infect. Immun. 63:26522657.
34. Schorey, J. S., M. A. Holsti, T. L. Ratliff, P. M. Allen, and E. J. Brown. 1996.
Characterization of the bronectin-attachment protein of Mycobacterium
avium reveals a bronectin-binding motif conserved among mycobacteria.
Mol. Microbiol. 21:321329.
35. Stahl, P. D., and R. A. Ezekowitz. 1998. The mannose receptor is a pattern
recognition receptor involved in host defense. Curr. Opin. Immunol. 10:50
55.
36. Tailleux, L., O. Schwartz, J. L. Herrmann, E. Pivert, M. Jackson, A. Amara,
L. Legres, D. Dreher, L. P. Nicod, J. C. Gluckman, P. H. Lagrange, B.
Gicquel, and O Neyrolles. 2003. DC-SIGN is the major Mycobacterium
tuberculosis receptor on human dendritic cells. J. Exp. Med. 197:121127.
37. Torres, A., M. D. Juarez, R. Cervantes, and C. Espitia. 2001. Molecular
analysis of Mycobacterium tuberculosis phosphate specic transport system in
Mycobacterium smegmatis. Characterization of recombinant 38 kDa (PstS-1).
Microb. Pathog. 5:289297.
38. Towbin, H., T. Staehelin, and J. Gordon. 1979. Electrophoretic transfer of
proteins from polyacrylamide gels to nitrocellulose sheets: procedure and
some applications. Proc. Natl. Acad. Sci. 76:43504354.
39. Tremblay, D., J. Lemay, M. Gilbert, Y. Chapdelaine, C. Dupont, and R.
Morosoli. 2002. High-level heterologous expression and secretion in Strep-
tomyces lividans of two major antigenic proteins from Mycobacterium tuber-
culosis. Can. J. Microbiol. 48:4348.
40. Wieles, B., M. V. Agterveld, A. Janson, J. E. Clark-Curtiss, T. Rinke De Wit,
M. Harboe, and J. Thole. 1994. Characterization of Mycobacterium leprae
antigen related to the secreted Mycobacterium tuberculosis protein MPT 32.
Infect. Immun. 62:252258.
VOL. 70, 2004 S. LIVIDANS, GLYCOSYLATION, AND M. TUBERCULOSIS 685
... In the present work, we studied the immune response to recombinant S. lividans Apa secreted into the culture medium as a complex formed by two bands, one of them modified with mannoses in the same amino acid positions as the native protein [17][18][19] and a second band non-glycosylated. It should be noted that, recombinant M. tuberculosis proteins with post-translational modifications such as glycosylation or methylation, outside of Mycobacterium genus, have been only produced in bacteria phylogenetically related to M. tuberculosis such as S. lividans and Rhodococcus erythropolis, with the advantage that recombinant proteins expressed in these bacteria can be obtained on a large scale and under controlled conditions [19][20][21][22]. ...
... In the present work, we studied the immune response to recombinant S. lividans Apa secreted into the culture medium as a complex formed by two bands, one of them modified with mannoses in the same amino acid positions as the native protein [17][18][19] and a second band non-glycosylated. It should be noted that, recombinant M. tuberculosis proteins with post-translational modifications such as glycosylation or methylation, outside of Mycobacterium genus, have been only produced in bacteria phylogenetically related to M. tuberculosis such as S. lividans and Rhodococcus erythropolis, with the advantage that recombinant proteins expressed in these bacteria can be obtained on a large scale and under controlled conditions [19][20][21][22]. Since members of the genus Streptomyces do not synthesize lipoglycan structures like Mycobacterium it constitutes an advantage for the purification of glycosylated mycobacterial proteins produced in this bacterium [23]. ...
... Recombinant Apa expressed in S. lividans 66 strain 1326 (rS.lividansApa) was recovered from the culture supernatant as described before with some modifications [19]. To obtain the rS.lividansApa, bacterial culture supernatant was precipitated with Ammonium sulfate (J.T. Baker, USA) 45% saturation at 4 °C overnight (ON) and centrifuged at 9700×g for 30 min at 4 °C. ...
Article
Full-text available
Identifying and evaluating potential vaccine candidates has become one of the main objectives to combat tuberculosis. Among them, mannosylated Apa antigen from Mycobacterium tuberculosis and the non-mannosylated protein expressed in Escherichia coli, have been studied. Although both proteins can induce a protective response in mice, it has been considered that native protein can be dispensed. In this work, we study the protective response induced by Apa expressed in E. coli and in Streptomyces lividans. The latter, like native is secreted as a double band of 45/47 kDa, however, only its 47 kDa band is mannosylated. Both antigens and BCG were intranasal administrated in mice, and animals were then challenged by aerosol with M. tuberculosis H37Rv. The results showed that both, Apa from S. lividans and E. coli conferred statistically significantly protection to animals compared to controls. The cytokine immune response was studied by an immunoassay after animals’ immunization, revealing that Apa from S. lividans induced a statistically significant proliferation of T cell, as well as the expression of IFN-γ, IL-1β, IL-17 and IL-10. In contrast, non-proliferation was obtained with non-mannosylated protein, but induction of IL-12 and IL-17 was observed. Together, these results demonstrate that both proteins were able to modulate a specific immune response against M. tuberculosis, that could be driven by different mechanisms possibly associated with the presence or not of mannosylation. Furthermore, stimulation of cells from BCG-vaccinated animals with the proteins could be an important tool, to help define the use of a given subunit-vaccine after BCG vaccination. Supplementary Information The online version contains supplementary material available at 10.1007/s00284-024-03697-7.
... The first evidence of the involvement of these two enzymes in glycosylation in Streptomyces came from studies of the glycosylation of two phage receptor proteins in S. coelicolor [50,51]. The enzymes encoded by the pmm and pmt1 genes of S. lividans were able to glycosylate antigens and extracellular enzymes of M. tuberculosis [52,53] confirming the similarity of the glycosylation systems in both actinobacteria. In addition to its role in the synthesis of glycoproteins that serve as receptor of phages in S. coelicolor, Wehmeier et al. [31] found that these two enzymes were also involved in the glycosylation of the PstS phosphate binding protein. ...
Article
Full-text available
: Phosphorous, in the form of phosphate, is a key element in the nutrition of all living beings. In nature, it is present in the form of phosphate salts, organophosphates, and phosphonates. Bacteria transport inorganic phosphate by the high affinity phosphate transport system PstSCAB, and the low affinity PitH transporters. The PstSCAB system consists of four components. PstS is the phosphate binding protein and discriminates between arsenate and phosphate. In the Streptomyces species, the PstS protein, attached to the outer side of the cell membrane, is glycosylated and released as a soluble protein that lacks its phosphate binding ability. Transport of phosphate by the PstSCAB system is drastically regulated by the inorganic phosphate concentration and mediated by binding of phosphorylated PhoP to the promoter of the PstSCAB operon. In Mycobacterium smegmatis, an additional high affinity transport system, PhnCDE, is also under PhoP regulation. Additionally, Streptomyces have a duplicated low affinity phosphate transport system encoded by the pitH1–pitH2 genes. In this system phosphate is transported as a metal-phosphate complex in simport with protons. Expression of pitH2, but not that of pitH1 in Streptomyces coelicolor, is regulated by PhoP. Interestingly, in many Streptomyces species, three gene clusters pitH1–pstSCAB–ppk (for a polyphosphate kinase), are linked in a supercluster formed by nine genes related to phosphate metabolism. Glycerol-3-phosphate may be transported by the actinobacteria Corynebacterium glutamicum that contains a ugp gene cluster for glycerol-3-P uptake, but the ugp cluster is not present in Streptomyces genomes. Sugar phosphates and nucleotides are used as phosphate source by the Streptomyces species, but there is no evidence of the uhp gene involved in the transport of sugar phosphates. Sugar phosphates and nucleotides are dephosphorylated by extracellular phosphatases and nucleotidases. An isolated uhpT gene for a hexose phosphate antiporter is present in several pathogenic corynebacteria, such as Corynebacterium diphtheriae, but not in non-pathogenic ones. Phosphonates are molecules that contains phosphate linked covalently to a carbon atom through a very stable C–P bond. Their utilization requires the phnCDE genes for phosphonates/phosphate transport and genes for degradation, including those for the subunits of the C–P lyase. Strains of the Arthrobacter and Streptomyces genera were reported to degrade simple phosphonates, but bioinformatic analysis reveals that whole sets of genes for putative phosphonate degradation are present only in three Arthrobacter species and a few Streptomyces species. Genes encoding the C–P lyase subunits occur in several Streptomyces species associated with plant roots or with mangroves, but not in the laboratory model Streptomyces species; however, the phnCDE genes that encode phosphonates/phosphate transport systems are frequent in Streptomyces species, suggesting that these genes, in the absence of C–P lyase genes, might be used as surrogate phosphate transporters. In summary, Streptomyces and related actinobacteria seem to be less versatile in phosphate transport systems than Enterobacteria.
... This result is in accordance with that reported by Lara et al. demonstrating glycosylation-dependant peptide epitope by ELISA using mannosidase-treated recombinant Mtb Apa antigen produced in Streptmyces lividans. Indeed, it has been shown that immunoreactivity of patients' sera towards rApa was significantly reduced after mannosidase treatment [4]. Furthermore, it has been reported that glycans play a major role in the modulation of immune responses and several studies have shown that the deglycosylation of the antigen may decrease its immunoreactivity [24e26]. ...
Article
We previously reported that immunoreactivity of recombinant CFP32 (Rv0577), a virulence factor of Mycobacterium tuberculosis, was higher when produced in transformed Pichia pastoris as compared to transformed E. coli. In this study, we show that this difference is partly due to the N-glycosylation of the recombinant CFP32 (rCFP32) by the yeast Pichia pastoris. In addition, SDS-PAGE and western blotting analysis of Mycobacterium bovis BCG and yeast-produced rCFP32 showed the presence of a band corresponding to a homodimeric state of the protein, unlike that of rCFP32 produced in E. coli. Computational modeling indicates that a single cysteine residue at position 193 of each monomer might bond to stabilize the homodimeric state of CFP32. Computational study showed that this residue is buried inside the protein core of E. coli-produced rCFP32 suggesting that rCFP32 may adopt a different folding in P. pastoris and BCG, in which C193 is solvent exposed. Surprisingly, an enzyme-linked immunosorbent assay using a generated monoclonal antibody (14D4) reveals the presence of a differential epitope that appears to be the consequence of the protein dimerization of the yeast- and BCG-, but not E.coli- produced, CFP32 recombinant form. We conclude that, in addition to N-glycosylation, homodimeric folding significantly enhances the immunoreactivity of rCFP32 and may these post-translational modifications may factor into the structure and function of native M. tuberculosis CFP32.
... Native APA induces a potent, delayed-type hypersensitivity response, and stimulates the priming of T-cells in vitro and in vivo, in contrast to the non-glycosylated recombinant APA produced in E. coli [28,29]. Moreover, antibodies derived from human TB patients react strongly with the O-glycosylated form of APA, whereas the non-glycosylated protein is unable to bind to these antibodies [30]. In addition, it has been suggested that the protective properties of the anti-tuberculosis vaccine are related to the pattern of O-mannosylation generated by M. bovis bacillus Calmette-Guérin [27][28][29]31]. ...
Article
Full-text available
Background Pichia pastoris (syn. Komagataella phaffii) is one of the most highly utilized eukaryotic expression systems for the production of heterologous glycoproteins, being able to perform both N- and O-mannosylation. In this study, we present the expression in P. pastoris of an O-mannosylated recombinant version of the 38 kDa glycolipoprotein PstS-1 from Mycobacterium tuberculosis (Mtb), that is similar in primary structure to the native secreted protein. Results The recombinant PstS-1 (rPstS-1) was produced without the native lipidation signal. Glycoprotein expression was under the control of the methanol-inducible promoter pAOX1, with secretion being directed by the α-mating factor secretion signal. Production of rPstS-1 was carried out in baffled shake flasks (BSFs) and controlled bioreactors. A production up to ~ 46 mg/L of the recombinant protein was achieved in both the BSFs and the bioreactors. The recombinant protein was recovered from the supernatant and purified in three steps, achieving a preparation with 98% electrophoretic purity. The primary and secondary structures of the recombinant protein were characterized, as well as its O-mannosylation pattern. Furthermore, a cross-reactivity analysis using serum antibodies from patients with active tuberculosis demonstrated recognition of the recombinant glycoprotein, indirectly indicating the similarity between the recombinant PstS-1 and the native protein from Mtb. Conclusions rPstS-1 (98.9% sequence identity, O-mannosylated, and without tags) was produced and secreted by P. pastoris, demonstrating that this yeast is a useful cell factory that could also be used to produce other glycosylated Mtb antigens. The rPstS-1 could be used as a tool for studying the role of this molecule during Mtb infection, and to develop and improve vaccines or kits based on the recombinant protein for serodiagnosis.
... In the absence of outer membranes, the Gram-positive bacteria Streptomycetes, secrete proteins directly into the medium. Streptomyces lividans, a useful Gram-positive secretion model, can secrete several heterologous polypeptides of bacterial and eukaryotic origin (Pozidis et al., 2001;Hong et al., 2003;Lara et al., 2004;Ogino et al., 2004;Sianidis et al., 2006;Hamed et al., 2017;Kashiwagi et al., 2017). The absence of lipopolysaccharides, the availability of advanced genetic tools (Kieser et al., 2000;Kashiwagi et al., 2017), low protease activity, established industrial bioprocessing as a major producer of antibiotics (Ndlovu et al., 2015), synthetic biology tools (Phelan et al., 2017) and the avoidance of inclusion body formation, renders S. lividans secretion an attractive biotechnology platform. ...
Article
Full-text available
Fluorescent proteins are a major cell biology tool to analyze protein sub-cellular topology. Here we have applied this technology to study protein secretion in the Gram-positive bacterium Streptomyces lividans TK24, a widely used host for heterologous protein secretion biotechnology. Green and monomeric red fluorescent proteins were fused behind Sec (SP Sec) or Tat (SP Tat) signal peptides to direct them through the respective export pathway. Significant secretion of fluorescent eGFP and mRFP was observed exclusively through the Tat and Sec pathways, respectively. Plasmid over-expression was compared to a chromosomally integrated sp Sec-mRFP gene to allow monitoring secretion under high and low level synthesis in various media. Fluorimetric detection of SP Sec-mRFP recorded folded states, while immuno-staining detected even non-folded topological intermediates. Secretion of SP Sec-mRFP is unexpectedly complex, is regulated independently of cell growth phase and is influenced by the growth regime. At low level synthesis, highly efficient secretion occurs until it is turned off and secretory preforms accumulate. At high level synthesis, the secretory pathway overflows and proteins are driven to folding and subsequent degradation. High-level synthesis of heterologous secretory proteins, whether secretion competent or not, has a drastic effect on the endogenous secretome, depending on their secretion efficiency. These findings lay the foundations of dissecting how protein targeting and secretion are regulated by the interplay between the metabolome, secretion factors and stress responses in the S. lividans model.
Chapter
Actinomycetes are an incredibly prolific source of therapeutic secondary metabolites. Indeed, over 50% of currently used commercial antibiotics are of actinomycetal origin. However, discovery has slowed significantly since the so-called golden age of antibiotics in the 1940s–1960s. The expansion of genome sequencing has revealed many more undiscovered natural products which are often cryptic, or not produced under standard laboratory conditions due to their finely tuned ecological roles in their natural contexts. Advances in synthetic biology approaches have revealed new strategies to manipulate actinomycetes and perform screens to uncover some of these cryptic natural products. Additionally, such strategies provide the tools to generate analogs of natural products through manipulation of precursor flux and biosynthetic enzymes.KeywordsSynthetic biologyCryptic natural productsHeterologous expression Streptomyces
Article
With the emergence of multidrug‐resistant bacteria, insufficiency of the established chemotherapy, and the existing vaccine BCG, tuberculosis (TB) subsists as the chief cause of death in different parts of the world. Thus, identification of novel target proteins is urgently required to develop more effective TB interventions. However, the novel vaccine and drug target knowledge based on the essentiality of the pathogen cell envelope components such as glycoconjugates; glycans and the peptidoglycan layer of the lipid‐rich capsule are limited. Furthermore, most of the genes encoding proteins are characterized as hypothetical and functionally unknown. Correspondingly, some researchers have shown that the lipid and sugar components of the envelope glycoconjugates are largely in charge of TB pathogenesis and encounter many drugs and vaccines. Therefore, in this review we provide an insight into a comprehensive study concerning the importance of cell envelope glycoconjugates and hypothetical proteins, the impact of posttranslational modification, and the bioinformatics based implications for better antitubercular intervention development.
Article
Full-text available
Enzymes are increasingly applied as biocatalysts for fulfilling industrial needs in a variety of applications and there is a bursting of interest for novel therapeutic proteins. Consequently, developing appropriate expression platforms for efficiently producing such recombinant proteins represents a crucial challenge. It is nowadays widely accepted that an ideal ‘universal microbial host’ for heterologous protein expression does not exist. Indeed, the first-choice microbes, as Escherichia coli or yeasts, possess known intrinsic limitations that inevitably restrict their applications. In this scenario, bacteria belonging to the Streptomyces genus need to be considered with more attention as promising, alternative, and versatile platforms for recombinant protein production. This is due to their peculiar features, first-of-all their natural attitude to secrete proteins in the extracellular milieu. Additionally, streptomycetes are considered robust and scalable industrial strains and a wide range of tools for their genetic manipulation is nowadays available. This mini-review includes an overview of recombinant protein production in streptomycetes, covering nearly 100 cases of heterologous proteins expressed in these Gram-positives from the 1980s to December 2019. We investigated homologous sources, heterologous hosts, and molecular tools (promoters/vectors/signal peptides) used for the expression of these recombinant proteins. We reported on their final cellular localization and yield. Thus, this analysis might represent a useful source of information, showing pros and cons of using streptomycetes as platform for recombinant protein production and paving the way for their more extensive use in future as alternative heterologous hosts.
Article
Full-text available
The filamentous morphology of Streptomyces lividans depends on the culture conditions, a�ecting the production, secretion and post-translational modifications of recombinant glycoproteins. In this work, the previously reported volumetric power input (P/V) in conventional (NF) and coiled (CF) shake flasks were scaled-up to a stirred bioreactor. The e�ects on the growth and morphology of S. lividans were analyzed, as well as, the production and O-mannosylation of the recombinant APA glycoprotein from Mycobacterium tuberculosis. Specific growth rates of S. lividans and similar recombinant glycoprotein (rAPA) yields were observed between NF and bioreactor cultures. In addition, we have found up to seven mannose residues attached to the Cterminal of the rAPA in bioreactor cultures, one more than in NF and CF. However, at similar P/V values, morphological and kinetic di�erences were found. Our data indicate that P/V as scale-up criteria in the production of recombinant glycoproteins in S. lividans can be successful in some, but not all the kinetic and stoichiometric parameters, suggesting that the metabolic cell responses can be a�ected by aeration/hydrodynamics between bioreactor and shake flasks. Keywords: S. lividans, power input, morphology, rheology, scale-up, shake flasks
Article
Post-translational modifications represent a key aspect of enzyme and protein regulation and function. Post-translational modifications are involved in signaling and response to stress, adaptation to changing environments, regulation of toxic and damaged proteins, proteins localization and host-pathogen interactions. Glycosylation in Mycobacterium tuberculosis (Mtb), is a post-translational modification often found in conjunction with acylation in mycobacterial proteins. Since the discovery of glycosylated proteins in the early 1980's, important advances in our understanding of the mechanisms of protein glycosylation have been made. The number of known glycosylated substrates in Mtb has grown through the years, yet many questions remain. This review will explore the current knowledge on protein glycosylation in Mtb, causative agent of Tuberculosis and number one infectious killer in the world. The mechanism and significance of this post-translational modification, as well as maturation, export and acylation of glycosylated proteins will be reviewed. We expect to provide the reader with an overall view of protein glycosylation in Mtb, as well as the significance of this post-translational modification to the physiology and host-pathogen interactions of this important pathogen. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD011081 and 10.6019/PXD011081.
Article
Full-text available
Mycobacterium tuberculosis andMycobacterium bovis, the causative agents of human and bovine tuberculosis, have been reported to express a range of surface and secreted glycoproteins, although only one of these has been subjected to detailed structural analysis. We describe the use of a genetic system, in conjunction with lectin binding, to characterize the points of attachment of carbohydrate moieties to the polypeptide backbone of a second mycobacterial glycoprotein, antigen MPB83 fromM. bovis. Biochemical and structural analysis of the native MPB83 protein and derived peptides demonstrated the presence of 3 mannose units attached to two threonine residues. Mannose residues were joined by a (1 → 3) linkage, in contrast to the (1 → 2) linkage previously observed in antigen MPT32 from M. tuberculosisand the (1 → 2) and (1 → 6) linkages in other mycobacterial glycolipids and polysaccharides. The identification of glycosylated antigens within the M. tuberculosis complex raises the possibility that the carbohydrate moiety of these glycoproteins might be involved in pathogenesis, either by interaction with mannose receptors on host cells, or as targets or modulators of the cell-mediated immune response. Given such a possibility characterization of mycobacterial glycoproteins is a step toward understanding their functional role and elucidating the mechanisms of mycobacterial glycosylation.
Article
Full-text available
A technique has been developed for the separation of proteins by two-dimensional polyacrylamide gel electrophoresis. Due to its resolution and sensitivity, this technique is a powerful tool for the analysis and detection of proteins from complex biological sources. Proteins are separated according to isoelectric point by isoelectric focusing in the first dimension, and according to molecular weight by sodium dodecyl sulfate electrophoresis in the second dimension. Since these two parameters are unrelated, it is possible to obtain an almost uniform distribution of protein spots across a two-diminsional gel. This technique has resolved 1100 different components from Escherichia coli and should be capable of resolving a maximum of 5000 proteins. A protein containing as little as one disintegration per min of either 14C or 35S can be detected by autoradiography. A protein which constitutes 10 minus 4 to 10 minus 5% of the total protein can be detected and quantified by autoradiography. The reproducibility of the separation is sufficient to permit each spot on one separation to be matched with a spot on a different separation. This technique provides a method for estimation (at the described sensitivities) of the number of proteins made by any biological system. This system can resolve proteins differing in a single charge and consequently can be used in the analysis of in vivo modifications resulting in a change in charge. Proteins whose charge is changed by missense mutations can be identified. A detailed description of the methods as well as the characteristics of this system are presented.
Article
Full-text available
A method has been devised for the electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets. The method results in quantitative transfer of ribosomal proteins from gels containing urea. For sodium dodecyl sulfate gels, the original band pattern was obtained with no loss of resolution, but the transfer was not quantitative. The method allows detection of proteins by autoradiography and is simpler than conventional procedures. The immobilized proteins were detectable by immunological procedures. All additional binding capacity on the nitrocellulose was blocked with excess protein; then a specific antibody was bound and, finally, a second antibody directed against the first antibody. The second antibody was either radioactively labeled or conjugated to fluorescein or to peroxidase. The specific protein was then detected by either autoradiography, under UV light, or by the peroxidase reaction product, respectively. In the latter case, as little as 100 pg of protein was clearly detectable. It is anticipated that the procedure will be applicable to analysis of a wide variety of proteins with specific reactions or ligands.
Article
The mannose receptor recognizes the patterns of carbohydrates that decorate the surfaces and cell walls of infectious agents. This macrophage and dendritic cell pattern-recognition receptor mediates endocytosis and phagocytosis. The mannose receptor is the prototype of a new family of multilectin receptor proteins (membrane-spanning receptors containing eight-ten lectin-like domains, which appear to play a key role in host defense) and provides a link between innate and adaptive immunity. Recent advances include the identification of three new members of the mannose receptor family, additional work on defining the molecular requirements for sugar binding, a role for the mannose receptor in antigen presentation of lipoglycan antigens and evidence that the mannose receptor is associated with a signal transduction pathway leading to cytokine production.
Article
Exoglucanase Cex from Cellulomonas fimi is a glycoprotein [Langsford et al., J. Gen. Microbiol. 130 (1984) 1367-1376]. Cex produced by Streptomyces lividans from the cloned cex gene is also glycosylated. The extent and nature of glycosylation are similar for Cex from both organisms. The glycosylation affords protection against proteolysis for the enzymes from both organisms when they are bound to cellulose, but not in solution. The ability to glycosylate cloned gene products enhances the utility of Streptomyces as a host for the production of heterologous polypeptides.
Article
Ten major antigens from Mycobacterium bovis culture filtrate of 39, 32, 30, 25, 24, 22 (a and b forms), 19, 15, and 12 kDa have been purified and characterized by classical physicochemical methods. With monoclonal antibodies and/or N-terminal amino acid sequencing data, it was found that the antigens of 32, 30, 24, 22 (a), 19, and 12 kDa are related to M. bovis or M. tuberculosis antigens P32, MPB59, MPB64, MPB70, 19 kDa, and 12 kDa, respectively. The 39-, 25-, 22 (b)-, and 19-kDa antigens showed concanavalin A-binding properties and were positive in a glycan detection test, suggesting that they are glycoproteins. The 25- and 22 (b)-kDa proteins were found to be glycosylated forms of MPB70.
Article
A third extracellular xylanase produced by Streptomyces lividans 66 was isolated from a clone obtained by shotgun cloning through functional complementation of a xylanase- and cellulase-negative mutant using the multicopy vector pIJ702. This enzyme, designated xylanase C, has a relative molecular mass of 22000 and acts on xylan similarly to xylanase B as an endo-type xylanase producing short-chain oligoxylosides. Its specific activity determined at 1100 IU·mg−1 of protein corresponds on a molecular basis to that of xylanase B and is about three times that of xylanase A. The enzyme shows optimal activity at pH 6.0 and 57°C, values that correspond closely to those observed previously for xylanase A and B. Xylanase C appears not to be glycosylated and has a pI > 10.25. Its K m and V max on birchwood xylan are 4.1 mg·ml−1 and 3.0 μmol·min−1·mg−1 of enzyme respectively. Whereas specific antibodies raised against xylanase A show no cross-reaction with either xylanase B or with xylanase C, the anti-(xylanase C) antibodies react slightly with xylanase B but not with xylanase A. A comparison of hydrolysis products obtained by reacting individually the three enzymes with birchwood xylan showed characteristic endo-activity patterns for xylanases B and C, whereas xylanase A hydrolysed the substrate preferentially into xylobiose and xylotriose. Sequential xylanase action on the same substrates showed synergistic hydrolysis only when endo-xylanase activity was followed by that of xylanase A.
Article
In Mycobacterium tuberculosis culture filtrates, three concanavalin A (ConA)-binding bands of 55, 50 and 38 kilodaltons (kD) were identified by labelling blotted proteins with a ConA-peroxidase conjugate. Binding was inhibited by the competitor sugar alpha-methyl mannoside and by reduction with sodium m-periodate. Bands of 55, 50 and 38 kD stained with Coomasie blue were sensitive to digestion with proteases, thus indicating that they are proteins. Glycoproteins were isolated by lectin affinity chromatography or by elution from nitrocellulose membranes. On the isolated form, the 55-50 kD doublet glycoprotein was 65.4% protein and 34.6% sugar. The purified 38 kD molecule was 74.3% protein and 25.7% carbohydrate. By immunoblot, antibodies against mycobacterial glycoproteins were demonstrated in immunized rabbits and in patients with pulmonary tuberculosis, but not in healthy individuals. Treatment with sodium m-periodate abolished binding of rabbit antibodies to the 38 kD glycoprotein. Reactivity of the 55-50 kD doublet glycoprotein was not altered by reduction. By immunoblot with monoclonal antibodies TB71 and TB72, a carbohydrate-dependent and a carbohydrate-independent epitope could be identified on the 38 kD glycoprotein.
Article
To identify antigens that could be specifically associated with tuberculosis infection, the antibody response to Mycobacterium tuberculosis antigens of patients with pulmonary tuberculosis and of healthy individuals were compared by immunoblot. In healthy individuals, serum antibodies were found in the majority of cases. Bands of 60 and 32-31 kilodaltons (kD) were the antigens more frequently recognized by antibodies of normal sera (55.8 and 64.7%, respectively). In patients with pulmonary tuberculosis, the number and intensity of the developed antigen bands were much higher than in normal individuals. Antigens reacting preferentially with tuberculosis sera were also identified. Furthermore, a unique disease-associated protein antigen of 38 kD was found to react with 57% of patients' sera but with none of the controls. This antigen was isolated by elution from nitrocellulose membranes and tested as an ELISA reagent in the serodiagnosis of pulmonary tuberculosis. A specificity of 0.96 and sensitivity of 0.68 were obtained.