Sunteți pe pagina 1din 9

BASIC IMMUNOLOGY doi: 10.1111/j.1365-3083.2011.02531.

x
..................................................................................................................................................................

Immunogenicity and Protective Efficacy of a Fusion


Protein Vaccine Consisting of Antigen Ag85B and
HspX against Mycobacterium tuberculosis Infection
in Mice
Q. Li* , H. Yu*, Y. Zhang*à, B. Wang§, W. Jiang*, Z. Da*, Q. Xian–, Y. Wang–, X. Liu  & B. Zhu*

Abstract
*Lanzhou Center for Tuberculosis Research and Subunit vaccines have the potential advantage to boost Mycobacterium bovis
Institute of Pathogenic Biology, School of Bacillus Calmette-Guérin (BCG)-primed immunity in adults. However, most
Basic Medical Sciences, Lanzhou University,
candidates are antigens highly expressed in replicating bacilli but not in dor-
Lanzhou, China;  Institute of Pathophysiology,
School of Basic Medical Sciences, Lanzhou mant or persisting bacilli, which exist during Mycobacterium tuberculosis infec-
University, Lanzhou, China; àDepartment of tion. We constructed M. tuberculosis fusion protein Ag85B-Mpt64190–198-
Molecular Microbiology and Immunology, HspX (AMH) and Ag85B-Mpt64190–198-Mtb8.4 (AMM), which consist of
Bloomberg School of Public Health, Johns Ag85B, the 190–198 peptide of Mpt64, HspX (Rv2031c) and Mtb8.4
Hopkins University, Baltimore, MD, USA;
(Rv1174c), respectively. AMH and ⁄ or AMM were mixed with adjuvants com-
§Lanzhou Institute of Biological Products,
Lanzhou, China; and –ABSL-3 Lab, Wuhan posed of dimethyl-dioctyldecyl ammonium bromide and BCG polysaccharide
University, Wuhan, China nucleic acid (DDA-BCG PSN) to construct subunit vaccines. Mice were
immunized thrice with Ag85B, AMH and AMM vaccines and the immunoge-
Received 4 October 2010; Accepted in revised nicity of the fusion protein vaccines was determined. Then, mice were primed
form 26 January 2011
with BCG and boosted twice with Ag85B, AMH, AMM and AMM + AMH
Correspondence to: Xin Liu, B. Zhu, Institute vaccines, respectively, followed by challenging with M. tuberculosis virulent
of Pathogenic Biology, School of Basic Medical strain H37Rv, and the immune responses and protective effects were mea-
Sciences, Lanzhou University, Lanzhou 730000, sured. It was found that mice immunized with AMH vaccine generated high
China. E-mail: bdzhu@lzu.edu.cn; levels of antigen-specific cell-mediated responses. Compared with the group
liux@lzu.edu.cn
injected only with BCG, the mice boosted with AMM, AMH and
AMM + AMH produced higher levels of Ag85B-specific IgG1 and IgG2a and
IFN-c-secreting T cells upon Ag85B and Mycobacterium tuberculosis purified
protein derivative (PPD) stimulation. It is interesting that only mice boosted
with AMM + AMH had significantly lower bacterial count in the lungs than
those receiving BCG, whereas mice boosted with AMH or AMM did not. The
results suggest that AMH consisting of HspX, the antigen highly expressed in
dormant bacilli, could be combined with antigens from replicating bacilli to
enhance BCG primed immunity so as to provide better protection against both
growing and non-growing bacteria that occur during the infection process.

children against disseminated TB [6, 7]. However, the


Introduction
BCG-induced protective immunity varies from 0% to
One-third of the world population is latently infected 80% in different populations and wanes in adults. More
with Mycobacterium tuberculosis, and 5–10% of which will important, BCG does not prevent reactivation of dormant
develop into active tuberculosis (TB) when the host bacilli [7, 8]. It is urgent to search for novel TB vaccines
immune system is compromised [1]. The dormant and immunization strategies. One practical way is to
M. tuberculosis, existing in active TB as well as latent boost BCG with subunit vaccines so as to improve BCG-
infection [2–4], persist in the host [5], which results in primed immunity in adult.
less facility to eradicate TB. BCG is the only TB vaccine Mycobacterium tuberculosis expresses different genes at
used in the clinic and proves to be effective to protect different conditions so as to adapt to different environ-

 2011 The Authors


568 Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
Q. Li et al. Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine 569
..................................................................................................................................................................

ments. Some genes are up-regulated in dormant phase to unique site SacI and HindIII of the previously con-
survive under suboptimal or stress conditions, such as structed pET-28a Ag85B plasmid. The correct DNA
nutrient and oxygen deprivation. It was reported that construct containing the appropriate inserts was con-
latency-associated antigens could induce antigen-specific firmed by DNA sequencing.
IFN-c production, CD4+ T cell proliferation and cyto- Expression and purification of AMH fusion protein. The
kine expression in peripheral blood mononuclear cells plasmid pET-28a AMH was transformed into the Escheri-
from persons with active and latent TB infection [9]. chia coli strain BL21 for production of the fusion protein
HspX, also known as a-crystallin, is one of genes induced AMH. E. coli BL21 expressing AMH was cultured in LB
by hypoxia. It is up-regulated significantly in non-repli- medium for 2 h at 37 C before induction with 1 mM iso-
cating conditions but is poorly expressed in replicating propylb-D-thiogalactopyranoside. After induction, growth
condition [10]. Increased HspX mRNA was detected in was continued for 4 h at 37 C when the bacterial cells
the lungs of patients with chronic TB [11]. In addition, were harvested by centrifugation at 12,000 g for 10 min at
HspX is capable of activating T cells from 80% of house- 4 C. Then, cells were suspended in buffer A without urea
hold contacts with TB patients, 90% of health care workers (sodium phosphate buffer 0.1 M, Tris–Cl 0.01 M, pH 8.0)
and 50% of controls [12]. These findings indicate that at 5 ml per gram wet weight and sonicated on ice at 200–
HspX may be an important dormancy antigen that could 300 W for 30 min with 1-s cooling period between each
be effectively recognized by human T cells [13]. 1-s bursting. The insoluble material containing AMH in
Many antigens expressed in bacterial replicating stage inclusion bodies was precipitated by centrifugation at
have been chosen as candidate antigens for new vaccines. 12,000 g for 10 min at 4 C, and AMH was solubilized
However, antigens highly expressed in dormant stage and extracted overnight at 4 C in buffer B (urea 8 M,
have not been widely evaluated until now [14]. We had sodium phosphate buffer 0.1 M, Tris–Cl 0.01 M, pH 8.0).
developed a fusion protein Ag85B-Mpt64190–198-Mtb8.4 AMH protein was subsequently purified by Ni-NTA His
(AMM) previously and showed that it could elicit strong resin-columns (Merck KGaA, Darmstadt, Germany)
humoral and cell-mediated immune responses when according to the manufacturer’s instructions. Fractions
formulated in chitosan microspheres [15] or in dimethyl- containing AMH were identified by 12% SDS-PAGE and
dioctyldecyl ammonium bromide and BCG polysaccha- pooled. Finally, the pooled fractions were dialysed against
ride nucleic acid (DDA-BCG PSN) adjuvants [16]. AMM urea concentration gradient (6, 4, 2, 1, 0.5 and 0 M urea
is composed of Ag85B, 190–198 peptide of Mpt64 and with 5 mM Tris–Cl, pH 7.9) for 12 h at each concentration
Mtb8.4, which are all expressed in replicating bacteria. at 4 C. The concentration of the pure AMH was deter-
In this study, Mtb8.4 from AMM was replaced by HspX mined by the Lowry protein assay.
antigen highly expressed in dormant bacteria, to con- Subunit vaccine preparation. AMM, Ag85B and BCG
struct a novel fusion protein Ag85B-Mpt64190–198-HspX PSN were prepared as described previously [16]. The
(AMH). And then, the immunogenicity and protective preparation of AMH and AMM + AMH vaccines is
efficacy of the AMH vaccine were evaluated. described below. AMM and AMH were suspended in
phosphate-buffered saline (PBS) (0.2 mg ⁄ ml), and BCG
PSN was suspended in saline (0.6 mg ⁄ ml). DDA (Sigma-
Materials and methods
Aldrich, St. Louis, MO, USA) was suspended in distilled
Construction of plasmid pET-28a Ag85B-Mpt64190–198- water (2.5 mg ⁄ ml), and a homogeneous dispersion of the
HspX. The DNA sequences encoding Ag85B and HspX powder was obtained by heating the suspension to 80 C
were amplified by polymerase chain reaction (PCR) from for 5–10 min. After cooling at room temperature, the
M. tuberculosis H37Rv chromosomal DNA template with suspension was mixed with diluted AMM, AMH and
the primers Ag85BF, 5¢-AATGGATCCTTCTCCCGGCC BCG PSN before use.
GGGGCT-3¢(BamHI), and HspXF1, 5¢- ATAGAG- Vaccination and challenge procedures. Female C57BL ⁄ 6
CTCATGGCCACACCCTTC-3¢(SacI), as forward primers mice aged 6 weeks old were maintained in special patho-
and the primers Ag85BR, 5¢-ATTGAGCTCGCCGGC gen-free conditions in Lanzhou Institute of Biological Prod-
GCCTAACGAACTCTGGAG-3¢(SacI), and HspXR, 5¢- ucts and ABSL-3 laboratory at Wuhan University for
ACGAAGCTTTCAGTTGGTGGACCG-3¢(HindIII), as M. tuberculosis H37Rv challenge experiment and were used
reverse primers. The PCR fragment of Ag85B was cloned for experiments at the beginning of 7 weeks of age. Mice
into the BamHI and SacI site of pET-28a to construct received free access to food and water throughout the study.
the plasmid pET-28a Ag85B. Subsequently, the fragment Vaccination with subunit vaccines alone: The mice were
of Mpt64190–198-HspX was generated by PCR from PCR inoculated subcutaneously thrice with AMM ⁄ AMH ⁄
product of HspX as template with the forward primer Ag85B vaccines with 20 lg proteins in 200 ll at week
HspXF2, 5¢-ATAGAGCTCTTCGCAGTCACGAACGAC 0, 3 and 6. Control animals were immunized with the
GGGGTGATTATGGCCACCACCCTTC-3¢(SacI), and same volume of PBS or 5 · 106 colony-forming unit
the reverse primer HspXR and was cloned into the (CFU) of BCG in 200 ll once at 0 week.

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
570 Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine Q. Li et al.
..................................................................................................................................................................

Vaccination with BCG prime and subunit vaccine boost: added to lyse the remaining cells. The plates were incu-
The mice were inoculated subcutaneously with bated on ice for 15 min, after which they were washed
5 · 105 CFU of BCG D2PB302S11First10-P4 strain 10 times with PBST. Next, biotinylated detector anti-
(Copenhagen strain) in 200 ll at 0 week followed by body solution was added and the plates were incubated
AMM (20 lg of AMM plus 250 lg of DDA and 30 lg for 1 h at 37 C. The plates were washed five times with
of BCG PSN), AMH (20 lg of AMH plus 250 lg of PBST, after which 100 ll ⁄ well streptavidin–horseradish
DDA and 30 lg of BCG PSN), Ag85B (20 lg of peroxidase was added. The plates were again incubated
Ag85B plus 250 lg of DDA and 30 lg of BCG PSN) for 1 h at 37 C and washed five times with PBST. One
and AMM + AMH (10 lg of AMM and 10 lg of AMH hundred microlitres of AEC (3-amino-9-ethylcarbazole)
plus 250 lg of DDA and 30 lg of BCG PSN) subunit substrate was added to each well. The plates were devel-
vaccines boosting twice at weeks 8 and 10, respectively. oped for 25 min at room temperature in the dark. The
Control animals were injected with PBS or BCG at wells were washed with distilled water to stop develop-
0 week followed by PBS boosting. Twelve weeks after ment when the stained cells were counted on an auto-
the last immunization, groups of mice were challenged mated ELISPOT reader and analysed with ImmunSpot
intravenously by tail vein injection with 1 · 106 CFU of software (Bio-sys, GmbH, Karben, Germany).
virulent M. tuberculosis H37Rv. Protective efficacy assay. Mice were sacrificed for bacte-
Antibody detection by ELISA. Microtiter plates were rial CFU count at 6th week post-challenge with H37Rv.
coated with 100 ll ⁄ well with specific antigens in 0.05 M The lower left lobe of the lungs from infected mice
bicarbonate buffer (pH 9.6) overnight at 4 C. The plates (n = 7) was harvested, homogenized in 0.05% PBS-
were washed five times with PBS containing 0.05% Tween 80 and planted in 10-fold dilutions (10–1000) on
Tween 20 (PBST); 4 weeks after the last vaccination, Middlebrook 7H11-OADC agar (BD, Franklin Lakes,
serum samples from four immunized mice per group NJ, USA) containing ampicillin (10 lg ⁄ ml) to prevent
were collected and diluted to 1:100 with PBS and contamination. Bacterial colonies were counted 3 weeks
applied to plates in twofold serial dilutions to 1:25,600; after incubation at 37 C.
horse radish peroxidase-conjugated rabbit anti-mouse Histopathology of the lung tissues. Each upper lobe of
IgG1 or IgG2a (Rockland Immunochemicals Inc., Rock- the left lung of infected mice (n = 5) was harvested
land, ME, USA) was used at 1:20,000 dilution as sug- 6 weeks after challenge. The lobes were fixed with 10%
gested by the manufacturer. The plates were incubated neutral buffered formalin. After 2 weeks, each lobe was
for 1 h at 37 C and washed with PBST. After washing, bisected with 5 lm thick to examine the same area of
the plates were added SureBlue tetramethyl benzidine the lung in all mice. The sections were stained with hae-
substrate with 200 ll ⁄ well and incubated at room tem- matoxylin and eosin (HE) and Ziehl–Neelsen Method.
perature for 15 min. The reaction was stopped by 50 ll Granulomas area was divided by total section area to
of 1 M H2SO4 in each well. The optical density was mea- determine the affected area in a section. Histopathology
sured at 450 nm. was evaluated by three pathologists independently.
Enzyme-linked immunspot (ELISPOT) detection for IFN-c Statistical analysis. The results were expressed as
production from splenocytes. Four weeks after the third means ± standard deviation (SD) and analysed by
immunization, spleens were aseptically harvested from SPSS10.0 software (Statistical Product and Service Solu-
four mice ⁄ group and gently ground through a 70-lm tions Company, Chicago, IL, USA). The significance of
cell strainer, and then, single-cell suspensions were pre- differences among the groups was determined by analysis
pared with Lympholyte-M density gradient centrifugation of variance (ANOVA). Independent-samples t-test was used
(Dakewe Biotech Company Limited, Shenzhen, China). for Ziehl–Neelsen stain. Probability values (P < 0.05)
The 96-well transparent polystyrene plates were coated were considered as statistically significant.
with 50 ll anti-IFN-c mAb overnight at 4 C. The
plates were then washed five times with PBST and then
Results
blocked with 200 ll blocking solution B at 37 C for
1 h. After discarding blocking solution B from the
Construction, expression and purification of AMH fusion
plates, freshly isolated spleen cells were plated in dupli-
protein
cate at 5 · 105 cell per well in 100 ll of RPMI 1640
supplemented with penicillin, streptomycin and 10% The correct DNA sequence for the recombinant fusion pro-
newborn calf serum (Shangbao Biotech Company Lt., tein, AMH was confirmed by sequencing and was found to
Shanghai, China) and stimulated with HspX, Ag85B, encode a protein with molecular weight of 54.6 kDa.
purified protein derivative and Mpt64190–198, respec- AMH was overexpressed in E. coli in inclusion bodies,
tively, with ConA and PBS as positive and negative con- which were subsequently dissolved and purified with Ni-
trols, for 36 h at 37 C, 5% CO2. The cells were then NTA His affinity chromatography. The protein was puri-
removed, and 200 ll ⁄ well ice-cold deionized water was fied, which was confirmed by SDS-PAGE analysis (Fig. 1).

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
Q. Li et al. Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine 571
..................................................................................................................................................................

A
MGSSHHHHHHSSGLVPRGSHMASMTGGQQMGRGSEFFSRPGLPVEYLQ
His Tag Ag85B
VPSPSMGRDIKVQFQSGGNNSPAVYLLDGLRAQDDYNGWDINTPAFEWY
YQSGLSIVMPVGGQSSFYSDWYSPACGKAGCQTYKWETFLTSELPQWLS
ANRAVKPTGSAAIGLSMAGSSAMILAAYHPQQFIYAGSLSACLDPSQGMG
PSLIGLAMGDAGGYKAADMWGPSSDPAWERNDPTQQIPKLVANNTRLWV
YCGNGTPNELGGANIPAEFLENFVRSSNLKFQDAYNAAGGHNAVFNFPPN
GTHSWEYWGAQLNAMKGDLQSSLGAGVDFAVTNDGVIMATTLPVQRHP
Mpt64190-198 HspX
RSLFPEFSELFAAFPSFAGLRPTFDTRLMRLEDEMKEGRYEVRAELPGVDP
DKDVDIMVRDGQLTIKAERTEQKDFDGRSEFAYGSFVRTVSLPVGADEDD
IKATYDKGILTVSVAVSEGKPTEKHIQIRSTN

Figure 1 Overexpression and purification of Ag85B-Mpt64190–198-HspX (AMH). (A) translated product of AMH; (B) purification of AMH with the
molecular weight of 54.6 kDa produced in E. coli strain BL21. Lane M, protein molecular weight standards. Lane 1, supernatant of BL-21 cells lysate.
Lane 2, resolution of inclusion bodies containing AMH. Lane 3, fraction of the purified protein AMH.

and HspX-specific IgG1 and IgG2a from AMH group


Immunogenicity of the AMH vaccine candidate
were the highest among all groups. PPD-specific IgG1
and IgG2a from the mice immunized with AMH were
Cell-mediated immune response
higher than Ag85B and BCG group. The ratio of
Splenic lymphocytes from mice immunized with AMH Ag85B-specific IgG2a ⁄ IgG1 from AMH group was lower
formulated with adjuvants DDA and BCG PSN secreted than that of BCG group but higher than that of AMM
high levels of IFN-c upon stimulation with Ag85B, and Ag85B groups. The ratio of HspX-specific
HspX, Mpt64190–198 and PPD (Fig. 2). Splenic lympho- IgG2a ⁄ IgG1 from AMH group was the highest among
cytes from mice immunized with AMH produced higher all groups. High IgG2a ⁄ IgG1 ratio reflects Th1 activity
level of IFN-c than those immunized with Ag85B, which produces IFN-c to promote intracellular killing
AMM, BCG and PBS with the stimulation of HspX, activity by activating macrophages and cytotoxic T cells
Mpt64190–198 and PPD. When stimulated with antigen [17].
Ag85B, the level of IFN-c induced by AMH vaccine was
lower than that by AMM (P < 0.05) and Ag85B
Immunogenicity assay in mice primed with BCG and boosted
(P > 0.05) vaccines, but was still higher than that receiv-
with subunit vaccines
ing BCG (P < 0.05).
The production of IFN-c from splenocytes
Humoral immune response
Cell-mediated immune responses in mice primed with
With the aid of adjuvant DDA + BCG PSN, AMH BCG and boosted by AMH, AMM, or AMM + AMH
induced higher levels of antigen-specific IgG1 and IgG2a were analysed with the stimulation of Ag85B and
than Ag85B and AMM (Table 1). Ag85B-specific IgG2a PPD. The results showed that there were higher levels of

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
572 Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine Q. Li et al.
..................................................................................................................................................................

shown). PBS control did not produce antibodies. The ti-


tres of IgG1 and IgG2a against Ag85B from mice immu-
nized with BCG and boosted with subunit vaccines were
higher than that primed with BCG alone (P < 0.05),
whereas there were no significant differences among
boosting groups.

Protective efficacy
Protective efficacy was evaluated by CFU count in mice
boosted with different protein vaccines followed by chal-
lenging with virulent M. tuberculosis H37Rv. The CFUs
from the lungs of mice boosted with the subunit vaccines
AMM + AMH and AMM were significantly lower than
PBS injection, although AMH subunit vaccine boosting
Figure 2 Number of cells secreting IFN-c in mice immunized with
did not lead to a significant decrease in CFUs. The bacilli
subunit vaccine. The mice were inoculated subcutaneously thrice with
Ag85B-Mpt64190–198-Mtb8.4 ⁄ Ag85B-Mpt64190–198-HspX ⁄ Ag85B vac- were effectively inhibited in the lungs of mice boosted
cines at week 0, 3 and 6. Control animals were injected with phos- by AMM + AMH in DDA-BCG PSN, which even
phate-buffered saline or BCG once at 0 week. Spleens were removed induced significantly lower CFU than BCG group
4 weeks after the last immunization. Splenocytes isolated from immu- (P < 0.05) (Fig. 4). These findings suggest that AMH
nized animals were plated in duplicate at 5 · 105 cells ⁄ well and stimu-
was potentially capable of enhancing the clearance of
lated with 10 lg ⁄ ml of Ag85B, 2 lg ⁄ ml of HspX, 2 lg ⁄ ml
Mpt64190–198 and 10 lg ⁄ ml of PPD. IFN-c-secreting cells were deter- tubercle bacilli in the lungs when used together with
mined by ELISPOT per 5 · 105 cells. *P < 0.05, n = 4. AMM as a boosting vaccine after BCG priming.
Sections from lungs were excised and stained by the
Ziehl–Neelsen technique for identification of acid-fast
IFN-c production in mice boosted with AMH, AMM bacilli in the tissue. AMM + AMH vaccine resulted in
and AMM + AMH vaccines than the group of BCG the lowest number of acid-fast bacteria (data not shown),
(Fig. 3). It indicated that Ag85B-, PPD-specific cell- which is consistent with the CFU data.
mediated immunity were highly induced by AMH,
AMM and AMM + AMH boosting. Unlike the fusion
HE stain of the lung tissues
proteins, single-protein Ag85B boosting did not signifi-
cantly induce high cell-mediated immunity compared HE stain showed that the granuloma areas per section of
with BCG alone. There was no significant difference the lungs from mice immunized with BCG or boosted
among AMM, AMH and AMM + AMH groups. with fusion proteins AMM, AMH or AMM + AMH
were smaller (P < 0.05) compared with PBS. There was
no difference between the three fusion protein boosting
Ag85B-specific antibody assay
groups and BCG-immunized group (Fig. 5), indicating
The boost with subunit vaccines induced a higher that boosting with the fusion protein vaccines did not
humoral immune response against Ag85B (data not aggravate pathology.

Table 1 Serological responses to antigens in subunit vaccine-immunized mice.

Anti-Ag85B Anti-HspX Anti-PPD

Group IgG1 IgG2a IgG2a ⁄ IgG1 IgG1 IgG2a IgG2a ⁄ IgG1 IgG1 IgG2a IgG2a ⁄ IgG1

AMM 12800 3200 0.25 1600 1600 1.00 12800 6400 0.50
AMH 12800 4032 0.32 2540 6400 2.52 6400 3200 0.50
Ag85B 5080 1008 0.20 800 800 1.00 400 1131 2.83
BCG 800 1008 1.26 504 635 1.26 800 1131 1.41
PBS 80 40 – 40 40 – 40 40 –

C57BL ⁄ 6 mice were subcutaneously immunized three times with fusion protein AMH and AMM and single-protein Ag85B (20 lg ⁄ mouse ⁄ time) in
adjuvant dimethyl-dioctyldecyl ammonium bromide plus BCG polysaccharide nucleic acid, respectively. Positive and negative control mice were
injected with BCG and PBS, respectively. Four weeks after the last immunization, serum samples from four immunized mice per group were col-
lected, and anti-Ag85B (5 lg ⁄ ml), PPD (20 lg ⁄ ml) and HspX (5 lg ⁄ ml) IgG1 and IgG2a were detected by ELISA.
– no calculation.
PBS, phosphate-buffered saline; AMH, Ag85B-Mpt64190–198-HspX; AMM, Ag85B-Mpt64190–198-Mtb8.4.

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
Q. Li et al. Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine 573
..................................................................................................................................................................

Figure 3 Number of cells secreting IFN-c in mice primed with BCG Figure 4 Protective efficacy of Ag85B-Mpt64190–198-Mtb8.4 + Ag85B-
and boosted with subunit vaccines. The mice were inoculated subcuta- Mpt64190–198-HspX subunit vaccine against H37Rv in mice primed
neously with BCG at 0 week followed by Ag85B-Mpt64190–198- with BCG. Twelve weeks after the last immunization, groups of mice
Mtb8.4 (AMM), Ag85B-Mpt64190–198-HspX (AMH), Ag85B and were challenged intravenously by tail vein injection with virulent Myco-
AMM + AMH subunit vaccines boosting twice at weeks 8 and 10, bacterium tuberculosis H37Rv. Six weeks post-challenge, mice were sacri-
respectively. Control animals were injected with phosphate-buffered sal- ficed for colony-forming units (CFUs) detection(n = 7) by harvesting
ine (PBS) or BCG at 0 week followed by PBS boosting. Spleens were the lower left lobe of lung and the results are represented as log10 of
removed 4 weeks after the last immunization. Splenocytes isolated from number of CFU ⁄ g of lung. *P < 0.05, versus phosphate-buffered saline;
immunized animals were plated in duplicate at 5 · 105 cells ⁄ well and **P < 0.05, versus BCG.
stimulated with 5 lg ⁄ ml of Ag85B and 10 lg ⁄ ml of PPD. IFN-c-
secreting cells were determined by ELISPOT per 5 · 105 cells.
*P < 0.05, n = 4. to non-replicating dormancy so as to have a maximum
impact on all stages of M. tuberculosis infection [2].
Different antigens are expressed in different growth
stages. Ag85B, Mtb8.4 and MPT64 are main antigens of
Discussion
bacteria in replicating stage, whereas HspX is the protein
In this research, we constructed a fusion protein AMH, that is mainly expressed in dormant phase (Fig. 6).
which included the protective antigen HspX highly Some latency antigens were detected up-regulated in
expressed in dormant stage of bacteria. Mice immunized non-replicating conditions [10]. For example, DosR, the
with AMH subunit vaccine generated high levels of anti- hypoxia-related transcriptional regulator, and its genes
gen-specific antibodies and IFN-c-producing lympho- are up-regulated under conditions closer to in vivo infec-
cytes. AMH combined with AMM could enhance the tion and prepare M. tuberculosis for dormancy [2]. Among
BCG-primed immune protection against M. tuberculosis them, HspX is the first gene to be identified as being
infection in mice. induced by hypoxia and has been identified as an impor-
Dormant bacteria exist together with replicating bac- tant latency antigen [10–13] (Fig. 6). HspX was a major
teria in vivo in human and animal infection [2–4] membrane protein in virulent M. tuberculosis [20].
(Fig. 6). Central to the success of M. tuberculosis as a M. tuberculosis and M. bovis have increased thickness of
pathogen is its ability to persist within humans for long their cell walls which contain large amounts of HspX
periods of time in a latent state [2–4]. BCG is the most under low oxygen conditions. It is likely that HspX
widely used vaccine, but it is not sufficient to prevent might stabilize cell wall structures and sustain bacterial
latent TB or prevent reactivation in adult life [18]. The viability during stress conditions [21].
antigens of subunit vaccines which were aimed to boost Mice immunized with AMH subunit vaccine generated
BCG-primed immunity were chosen frequently from high HspX-specific IgG2a and IgG1 as well as high IFN-
secreted proteins in early and log growth phase of bacte- c production with the stimulation of Ag85B and HspX.
ria based on in vitro culture and are insufficient to impart The antibodies target the extracellular mycobacteria
sufficient immunity against the latent infection where through binding to live M. tuberculosis, which can alter
some bacteria are in dormant state [19]. Therefore, it is the specific uptake pathway used for phagocytosis [22].
potentially important for the subunit vaccines to consist High IgG2a ⁄ IgG1 reflects Th1-skewing pathway that
of antigens in multiple stages from active multiplication produces IFN-c to promote intracellular microbicidal

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
574 Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine Q. Li et al.
..................................................................................................................................................................

PBS BCG

BCG+AMM BCG +AMH

BCG+AMM+AMH BCG+Ag85B

Figure 5 Pathology induced by vaccine immunization and Mycobacterium tuberculosis challenge. The bars represent the area ratio of granuloma in sec-
tions stained with HE (A). The panels depict histopathology from non-immunized controls and immunized mice (B). Six weeks post-challenge, mice
were sacrificed and lung tissue was sectioned and stained with H&E (50·). Representative pathology from one mouse per group. *P < 0.05 versus
phosphate-buffered saline, n = 5.

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
Q. Li et al. Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine 575
..................................................................................................................................................................

Ag85B 2008zx1000301104) and the National High Technology


Mtb8.4 Research and Development Program of China (863 Pro-
Replicating activity
gram) (2006AA02z420).

References
1 Young DB, Gideon HP, Wilkinson RJ. Eliminating latent tubercu-
losis. Trends Microbiol 2009;17:183–8.
2 Andersen P. Vaccine strategies against latent tuberculosis infection.
HspX Trends Microbiol 2006;15:7–13.
3 Zhang Y. Advances in the treatment of tuberculosis. Clin Pharmacol
Ther 2007;82:595–600.
4 Karakousis PC, Yoshimatsu T, Lamichhane G et al. Dormancy phe-
Figure 6 Different bacterial populations during MTB infection. White notype displayed by extracellular Mycobacterium tuberculosis within
represents bacteria in dormant stage, and black represents bacteria in artificial granulomas in mice. J Exp Med 2004;200:647–57.
replicating phase. Ag85B and Mtb8.4, and HspX are expressed in repli- 5 Flynn JL, Chan J. Immunology of tuberculosis. Annu Rev Immunol
cating and dormant stages, respectively. 2001;19:93–129.
6 Badell E, Nicolle F, Clark S et al. Protection against tuberculosis
induced by oral prime with Mycobacterium bovis BCG and intranasal
activities by activating macrophages and cytotoxic T cells subunit boost based on the vaccine candidate Ag85B-ESAT-6 does
[17]. not correlate with circulating IFN-c producing T-cells. Vaccine
AMM ⁄ AMH ⁄ AMM + AMH vaccine was designed to 2009;27:28–37.
boost BCG-primed immunity to evaluate the capability 7 Sepulveda RL, Parcha C, Sorensen RU. Case–control study of
the efficacy of BCG immunization against pulmonary tuberculosis
of generating protective immunity. The results showed
in young adults in Santiago, Chile. Tuber Lung Dis 1992;73:
that only AMM + AMH boosting resulted in a signifi- 372–7.
cant decrease in CFUs in lung tissues compared with the 8 Fine PE. Variation in protection by BCG: implications of and for
BCG group. Although AMM vaccine was found to be a heterologous immunity. Lancet 1995;346:1339–45.
promising candidate, it could not reduce markedly the 9 Govender L, Abel B, Hughes EJ et al. Higher human CD4 T cell
response to novel Mycobacterium tuberculosis latency associated anti-
bacterial load compared with BCG in BCG-primed and
gens Rv2660 and Rv2659 in latent infection compared with tuber-
subunit vaccine-boosted strategy. Although AMH alone culosis disease. Vaccine 2010;29:51–7.
could not reduce significantly CFU in lung tissues of 10 Saxena A, Srivastava V, Srivastava R, Srivastava BS. Identification of
infected mice over that of BCG, when it was combined genes of Mycobacterium tuberculosis upregulated during anaerobic per-
with AMM, interestingly, fewer CFUs were found than sistence by fluorescence and kanamycin resistance selection. Tubercu-
losis 2008;88:518–25.
the BCG group. AMM might induce immunity to
11 Timm J, Post FA, Bekker LG et al. Differential expression of iron-,
bacteria in active multiplication condition, but inclusion carbon-, and oxygen-responsive Mycobacterial genes in the lungs of
of AMH potentially induced immune protection chronically infected mice and tuberculosis patients. Proc Natl Acad
against dormant bacteria. Because of the comprehensive Sci U S A 2003;100:14321–6.
immune protection against replicating and dormant 12 Vekemans J, Ota MO, Sillah J et al. Immune responses to Mycobac-
terial antigens in the Gambian population: implications for vaccines
M. tuberculosis, the multi-stage vaccine, AMM + AMH,
and immunodiagnostic test design. Infect Immun 2004;72:
induced the most obvious protective effect among the 381–8.
BCG, BCG plus Ag85B or AMM or AMH groups 13 Caccamo N, Meraviglia S, La Mendola C et al. Characterization of
(Fig. 4). HLA-DR and TCR-binding residues of an immunodominant and
In conclusion, AMH vaccine could generate strong genetically permissive peptide of the 16-kDa protein of Mycobacte-
rium tuberculosis. Eur J Immunol 2004;34:2220–9.
antigen-specific humoral and cell-mediated immunity.
14 Gupta UD, Katoch VW, McMurray DN. Current status of TB
Only AMM + AMH boosting led to more pronounced vaccines. Vaccine 2007;25:3742–51.
M. tuberculosis clearance from the lungs of mice than 15 Zhu B, Qie Y, Wang J et al. Chitosan microspheres enhance the
BCG alone. Meanwhile, the vaccine induced higher immunogenicity of an Ag85B-based fusion protein containing mul-
immune responses and presented small lesions. The com- tiple T-cell epitopes of Mycobacterium tuberculosis. Eur J Pharm Biop-
harm 2007;66:318–26.
bination of fusion protein AMM and AMH containing
16 Luo Y, Wang B, Hu L et al. Fusion protein Ag85B-Mpt64190–198-
antigens both from replicating and dormant M. tuberculosis Mtb8.4 has higher immunogenicity than Ag85B with capacity to
may be a promising multi-stage vaccine to boost BCG boost BCG-primed immunity against Mycobacterium tuberculosis in
primed immunity for better protective efficacy. mice. Vaccine 2009;27:6179–85.
17 Maassen C, Boersma W, Holten-Neelen C, Claassen E, Laman J.
Growth phase of orally administered Lactobacillus strains differen-
Acknowledgment tially affects IgG1 ⁄ IgG2a ratio for soluble antigens: implications
for vaccine development. Vaccine 2003;21:2751–7.
This work was funded by the National Major Science and 18 Fine PE. Variation in protection by BCG: implications of and for
Technology Projects of China (2008ZX-10003-01305, heterologous immunity. Lancet 1995;346:1339–45.

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576
576 Immunogenicity and Protective Efficacy of a Fusion Protein Vaccine Q. Li et al.
..................................................................................................................................................................

19 Dietrich J, Lundberg CV, Andersen P. TB vaccine strategies – 21 Cunningham AF, Spreadbury CL. Mycobacterial stationary phase
What is needed to solve a complex problem? Tuberculosis induced by low oxygen tension: cell wall thickening and localiza-
2006;86:163–8. tion of the 16-kilodalton alpha-crystallin homolog. J Bacteriol
20 Lee BY, Hefta SA, Brennan PJ. Characterisation of the major mem- 1998;180:801.
brane protein of virulent Mycobacterium tuberculosis. Infect Immun 22 Hoft DF. Tuberculosis vaccine development: goals, immunological
1992;60:2066–74. design, and evaluation. Lancet 2008;372:164–75.

 2011 The Authors


Scandinavian Journal of Immunology  2011 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 73, 568–576

S-ar putea să vă placă și