ArticlePDF Available

Mycobacterial Antigens Exacerbate Disease Manifestations in Mycobacterium tuberculosis-Infected Mice

American Society for Microbiology
Infection and Immunity
Authors:

Abstract and Figures

To control tuberculosis worldwide, the burden of adult pulmonary disease must be reduced. Although widely used, Mycobacterium bovis BCG vaccination given at birth does not protect against adult pulmonary disease. Therefore, postexposure vaccination of adults with mycobacterial antigens is being considered. We examined the effect of various mycobacterial antigens on mice with prior M. tuberculosis infection. Subcutaneous administration of live or heat-treated BCG with or without lipid adjuvants to infected mice induced increased antigen-specific T-cell proliferation but did not reduce the bacterial load in the lungs and caused larger lung granulomas. Similarly, additional mycobacterial antigen delivered directly to the lungs by aerosol infection with viable M. tuberculosis mixed with heat-killed Mycobacterium tuberculosis (1:1) also did not reduce the bacillary load but caused increased expression of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), which was associated with larger granulomas in the lungs. When M. tuberculosis-infected mice were treated with recombinant BCG that secreted cytokines shown to reduce disease in a preinfection vaccine model, the BCG secreting TNF-α, and to a lesser extent, IL-2 and gamma interferon (IFN-γ), caused a significant increase in granuloma size in the lungs. Moreover, treatment of M. tuberculosis-infected mice with recombinant murine TNF-α resulted in increased inflammation in the lungs and accelerated mortality without affecting the bacillary load. Taken together, these studies suggest that administration of mycobacterial antigens to mice with prior M. tuberculosis infection leads to immune activation that may exacerbate lung pathology via TNF-α-induced inflammation without reducing the bacillary load.
Content may be subject to copyright.
INFECTION AND IMMUNITY, Apr. 2002, p. 2100–2107 Vol. 70, No. 4
0019-9567/02/$04.000 DOI: 10.1128/IAI.70.4.2100–2107.2002
Copyright © 2002, American Society for Microbiology. All Rights Reserved.
Mycobacterial Antigens Exacerbate Disease Manifestations in
Mycobacterium tuberculosis-Infected Mice
Andre L. Moreira,
1
Liana Tsenova,
2
Melles Haile Aman,
3
Linda-Gail Bekker,
4
Sherry Freeman,
2
Bande Mangaliso,
2
Ulf Schro¨der,
3
Jaishree Jagirdar,
1
William N. Rom,
5
Michael G. Tovey,
6
Victoria H. Freedman,
2
and Gilla Kaplan
2
*
Department of Pathology
1
and Division of Pulmonary & Critical Care Medicine,
5
New York University School of Medicine, and
Laboratory of Cellular Physiology & Immunology, The Rockefeller University,
2
New York, New York; Swedish Institute for
Infectious Disease Control, Stockholm, Sweden
3
; Infectious Disease Clinical Research Unit, UCT Lung Institute,
Cape Town, South Africa
4
; and Laboratory of Viral Oncology, CNRS, Villejuif, France
6
Received 2 August 2001/Returned for modification 22 October 2001/Accepted 7 January 2002
To control tuberculosis worldwide, the burden of adult pulmonary disease must be reduced. Although widely
used, Mycobacterium bovis BCG vaccination given at birth does not protect against adult pulmonary disease.
Therefore, postexposure vaccination of adults with mycobacterial antigens is being considered. We examined
the effect of various mycobacterial antigens on mice with prior M. tuberculosis infection. Subcutaneous admin-
istration of live or heat-treated BCG with or without lipid adjuvants to infected mice induced increased
antigen-specific T-cell proliferation but did not reduce the bacterial load in the lungs and caused larger lung
granulomas. Similarly, additional mycobacterial antigen delivered directly to the lungs by aerosol infection
with viable M. tuberculosis mixed with heat-killed Mycobacterium tuberculosis (1:1) also did not reduce the
bacillary load but caused increased expression of tumor necrosis factor alpha (TNF-) and interleukin 6
(IL-6), which was associated with larger granulomas in the lungs. When M. tuberculosis-infected mice were
treated with recombinant BCG that secreted cytokines shown to reduce disease in a preinfection vaccine model,
the BCG secreting TNF-, and to a lesser extent, IL-2 and gamma interferon (IFN-), caused a significant
increase in granuloma size in the lungs. Moreover, treatment of M. tuberculosis-infected mice with recombinant
murine TNF-resulted in increased inflammation in the lungs and accelerated mortality without affecting the
bacillary load. Taken together, these studies suggest that administration of mycobacterial antigens to mice with
prior M. tuberculosis infection leads to immune activation that may exacerbate lung pathology via TNF--
induced inflammation without reducing the bacillary load.
The idea of immune modulation to treat tuberculosis is not
new. In 1890, at the World Congress of Medicine, Robert Koch
announced that he had prepared “substances” that completely
cured guinea pigs in the late stages of tuberculosis (TB) (14).
The substance, later called “old tuberculin,” was a glycerin
extracted filtrate of cultures of the tubercle bacillus. With
much fanfare, patients were treated with this bacterial extract.
Unfortunately, the treatment caused florid local and systemic
reactions in many of the patients with relatively mild disease.
In addition, of the 230 patients with advanced cavitary disease
who received this treatment, 30 died. This intense reaction to
extracts of Mycobacterium tuberculosis and the associated clin-
ical worsening became known as the “Koch phenomenon”
(11).
The practical implications of the Koch phenomenon should
be reconsidered in light of recent efforts to develop a new
anti-TB vaccine. The goal of vaccination is protection against
adult pulmonary TB. Neonatal Mycobacterium bovis BCG vac-
cination, which is widely used throughout the world, does not
protect against adult pulmonary disease, even when it protects
against the more severe forms of childhood TB (8). New strat-
egies have therefore been proposed, including vaccination of
adults with BCG or other mycobacterial preparations (7). The
latter approach may be problematic. In areas of high endemic-
ity, many individuals will have already been infected and may
even have subclinical disease. If they now receive a strong
immunogen, the ensuing host response may result in exacer-
bation of the occult disease leading to severe toxicities (Koch
phenomenon).
The effect of administration of mycobacterial preparations
on the outcome of disease has recently been studied by a
number of investigators (1, 10, 21). Lowrie et al. showed that
postexposure vaccination with either BCG or DNA encoding
the M. tuberculosis heat shock protein Hsp70 or ESAT6 had
little or no effect on bacillary load in the spleens or lungs of
infected mice (16). However, repeated vaccination with DNA
encoding Mycobacterium leprae Hsp65 reduced the number of
bacilli in lungs and spleen by 1 log
10
and 2 log
10
at 2 and 5
months postintervention, respectively. In contrast, Turner and
colleagues explored the use of two other new candidate vac-
cines (a subunit vaccine and a DNA vaccine, both containing
M. tuberculosis Ag85) as immunotherapeutic agents in mice
(23). When administered to mice already infected with M.
tuberculosis, neither BCG nor these two candidate vaccines
caused any improvement in the course of infection. Further-
more, repeated BCG vaccination of infected mice resulted in
exacerbation of the granulomatous response in the lungs (23).
To further examine the effect of mycobacterium-induced
immune activation on the host response during M. tuberculosis
* Corresponding author. Mailing address: Laboratory of Cellular
Physiology & Immunology, The Rockefeller University, 1230 York
Ave., New York, NY 10021. Phone: (212) 327-8375. Fax: (212) 327-
8376. E-mail: kaplang@rockvax.rockefeller.edu.
2100
infection, we infected mice by aerosol with virulent M. tuber-
culosis. Then, 5 weeks later, the infected mice were inoculated
with various preparations containing BCG or with BCG secret-
ing murine cytokines. In addition, to determine whether the
presence of additional antigen in the lungs might have an
impact on outcome, naı¨ve mice were infected by aerosol with
live M. tuberculosis or a mixture of live and dead M. tubercu-
losis. Finally, M. tuberculosis-infected mice were treated di-
rectly with recombinant cytokines via intranasal delivery. The
course of the infection was monitored by enumeration of ba-
cilli in infected tissues, histologic examination of the lungs,
quantitation of cytokine mRNA induced in the lungs, and
peripheral T-cell proliferation in response to mycobacterial
antigens.
MATERIALS AND METHODS
Mice. Seven- to 8-week-old female (B6 D2)/F
1
mice, free of common
pathogens, were obtained from Charles River Laboratories, Wilmington, Mass.,
and housed in the BSL3 (Biosafety Level 3) Animal Facility of The Rockefeller
University for the duration of these experiments. All protocols were approved by
the Animal Use and Care Committee of The Rockefeller University.
Infection of mice. Aerosol infection was carried out according to a protocol
developed in this laboratory (17). Briey, mice were inoculated via the respira-
tory route by exposure to an aerosolized suspension of M. tuberculosis (see
below) generated by a Lovelace nebulizer using a nose-only exposure system
(In-Tox Products, Albuquerque, N.M.) (22). For each experiment 24 mice were
exposed for 30 min to the aerosol. This procedure resulted in implantation of
approximately 100 organisms into the lungs of mice, as conrmed by plating lung
homogenates 3 h after infection. In some experiments M. tuberculosis was auto-
claved and then mixed 1:1 with live bacilli prior to aerosol infection.
M. tuberculosis and M. bovis strains. M. tuberculosis strain Erdman was pro-
vided as multiple stock vials by J. Belisle, Colorado State University (Fort
Collins, Colo.); M. tuberculosis H37Rv was from the Trudeau Mycobacterial
Culture Collection (Trudeau Institute, Saranac Lake, N.Y.). The M. tuberculosis
clinical isolate HN878 was provided by J. M. Musser (20). BCG was obtained
from Statens Serum Institute, Copenhagen, Denmark (BCG vaccine SSI, batch
9854). BCG strain Montreal, which was used to generate recombinant BCG
expressing murine cytokines, including BCG expressing gamma interferon
(BCGIFN-), tumor necrosis factor alpha (BCGNF-), and interleukin 2
(BCGIL-2) and the control strain of BCG, carrying the plasmid vector only
(BCG vector), was obtained from Richard Young, The Whitehead Institute,
Cambridge, Mass. (18). All mycobacteria were grown on Middlebrook 7H9
medium (Difco, Detroit, Mich.), and bacillary stocks were stored at 10
7
to 10
8
bacilli/ml and kept at 70°C until use.
BCG preparations for postinfection inoculation. Lyophilized BCG (SSI) (1.5
mg) was reconstituted in 2 ml of diluent (Sauton; Statens Serum Institute),
according to the manufacturers instructions (viable BCG). In addition, BCG was
reconstituted in 200 l of diluent as described above and was heated in a water
bath at 60°C for 10 min. No growth of heat-treated BCG was noted using culture
on egg-based solid media (Lowenstein-Jensen) or 4 weeks of growth in the
BACTEC system (Bactec 4600; Becton Dickinson, Sparks, Md.). L3 adjuvant was
prepared using 0.62 mg of solid monooleate (analytic grade; Kebo AB, Stock-
holm, Sweden), 0.48 mg of oleic acid (analytic grade; Kebo AB), and 180 mg of
soybean oil (pharmaceutical grade; Karlshamn AB, Karlshamn, Sweden) and was
liqueed by gentle heat at 30°C (manuscript in preparation). For injection, 200
l of the heat-treated BCG was mixed in the L3 adjuvant, probe sonicated for 10
to 15 s, and brought to volume (2.25 ml) with 0.1 M Tris buffer, pH 7.5 (heat-
treated BCG). For boosting of mice, 200 l of heat-treated BCG was mixed in
half the amount of L3 adjuvant (see above) and brought to volume (2.25 ml) with
0.1 M Tris buffer, pH 7.5.
Vaccination of infected mice with BCG preparations. Five weeks after the
initial infection with M. tuberculosis H37Rv, mice were divided into four groups.
Groups 1 and 2 received 0.1 ml of viable BCG subcutaneously (s.c.). Group 3
received 0.1 ml of heat-treated BCG in L3 adjuvant s.c., while group 4 received
no treatment. Three weeks later, groups 2 and 3 were boosted with heat-treated
BCG in L3 adjuvant by intranasal administration of 10 l(5l per nostril). Mice
were evaluated at 12 weeks. In another experiment, either 6 weeks before or 5
weeks after the aerosol infection, mice were vaccinated s.c. with one of the
recombinant BCG strains (BCGIFN-, BCGTNF-, BCGIL-2, or BCG vec-
tor) (18) at a dose of 10
6
organisms per mouse. Three weeks after the initial
vaccination, the mice received a boost with the same dose of recombinant BCG.
A control group of mice received no BCG. Mice were evaluated at 12 and 20
weeks postchallenge with M. tuberculosis.
Intranasal treatment of infected mice. Recombinant murine TNF-was ob-
tained from Endogen (Boston, Mass.), reconstituted in 1% bovine serum albu-
minphosphate-buffered saline to a nal concentration of 2 10
5
IU/ml, and
kept at 4°C. The cytokine was administered intranasally for 5 consecutive days
per week for 4 weeks (6). Five microliters was applied directly to the nostrils of
each mouse (nal dose of TNF-,10
3
IU per mouse per day). Recombinant
murine IFN-was obtained from Valbiotech (Paris, France) and reconstituted in
1% bovine serum albuminphosphate-buffered saline to a nal concentration of
10
6
IU/ml. Ten microliters was applied into the nostrils of each mouse as
described above (nal dose of IFN-,10
4
IU per mouse per day). Control mice
were untreated.
CFU assay. The number of viable mycobacteria in lungs, livers, and spleens of
infected mice were evaluated at designated time points. Tenfold serial dilutions
of organ homogenates were plated onto 7H11 agar (Becton Dickinson) and were
incubated at 37°C. The number of viable bacilli was evaluated by counting
individual colonies after 2 to 3 weeks of growth.
Cytokine mRNA levels in the infected lung. Total cellular RNA from lungs of
infected mice was obtained at designated time points following aerosol infection.
Tissues were homogenized in 3 ml of RNAzolB (Cinna/Biotcx Lab. Inc., Hous-
ton, Tex.), and RNA was extracted according to the manufacturers instructions.
The reverse transcription-PCR was carried out as previously described (15).
Briey,1g of RNA was reverse transcribed using a Moloney murine leukemia
virus reverse transcriptase and was amplied with Taq polymerase according to
procedures given in the GeneAmp RNA PCR kit (Perkin-Elmer, Branchburg,
N.J.). Primers for cytokines and -actin were used as described (17). Densitom-
etry of the amplied bands was carried out using a PhosphorImager (Molecular
Dynamics, Sunnyvale, Calif.). Results were normalized to the density of -actin.
Lymphocyte proliferation assay. Spleen and draining lymph nodes were re-
moved at each time point and were processed as described (4). Briey, cells
isolated from spleen and lymph nodes were cultured at 2 10
5
cells/100 lin
96-well U-bottom plates in RPMI 1640 medium (Gibco BRL, Gaithersburg,
Md.) and were supplemented with 10% fetal bovine serum, penicillin, and strep-
tomycin (nal concentration, 50 g/ml) (all obtained from Gibco BRL). The cells
were incubated with concanavalin A (nal concentration, 5 g/ml) (Sigma, St.
Louis, Mo.) or M. tuberculosis H37Ra sonicate (nal concentration, 50 g/ml) at
37°C for 3 or 5 days, respectively, and were then pulsed with [H
3
]thymidine
(1 Ci/well) for an additional 18 h. Incorporation of [H
3
]thymidine was mea-
sured by -scintillation counting. Values were expressed as mean counts per
minute in the cultures.
Histopathology. At various time points after infection, lungs were xed in 10%
neutral buffered formalin, embedded in parafn, and processed for histology.
Sections were stained with hematoxylin and eosin and Ziehl-Neelsen for histo-
logic evaluation and photography.
Morphometric evaluation of granuloma size. Morphometry of the lesions was
performed using Microcomp, a computer-based image analysis system (Southern
Micro Institute, Atlanta, Ga.) and/or Sigmascan Pro 5:0 (SPSS Science Inc.,
Chicago, Ill.). A calibration micrometer (in square micrometers) slide was used
to determine the area evaluated.
Statistical analysis. Data were analyzed using an independent Student ttest.
APof 0.05 was considered statistically signicant. Kaplan-Meier analysis was
used to determine statistical signicance of the differences in survival time of
mice.
RESULTS
The effect of postinfection exposure to mycobacterial anti-
gens. To determine whether the course of established disease
is altered by immune stimulation with mycobacterial antigens,
mice were rst infected by aerosol with 100 CFU of M. tuber-
culosis H37Rv. At 5 weeks after infection, prior to any addi-
tional treatment, the number of CFU in the lungs was approx-
imately 6.6 log
10
. At this time, infected mice were split into
four groups, and mice in groups 1 and 2 were vaccinated with
viable BCG while mice in group 3 received heat-treated BCG
in L3 adjuvant. Three weeks later mice in groups 2 and 3 were
boosted with heat-treated BCG in L3 adjuvant, as described
VOL. 70, 2002 MYCOBACTERIUM-INDUCED PATHOLOGY IN MURINE TUBERCULOSIS 2101
above (Materials and Methods). Four weeks later (at 12 weeks
after the initial infection), the numbers of CFU in lungs, livers,
and spleens of mice in all four groups were evaluated and
found to be similar (Fig. 1A). Thus, postinfection immunother-
apy with these BCG preparations had no effect on the bacillary
loads in the organs of any of the infected mice.
An evaluation of the systemic T-cell response of the infected
mice was also carried out. Cells were isolated from the draining
lymph nodes of infected and vaccinated mice at 12 weeks and
stimulated in vitro with M. tuberculosis H37Ra sonicate. Com-
pared to the control infected mice (group 4), an increased
proliferative response was noted in all treatment groups and
was signicant for group 3 (Fig. 1B). Lymph node cells from
infected animals that received heat-treated BCG in L3 adju-
vant followed by intranasal boosting with heat-treated BCG in
L3 adjuvant (group 3) showed the largest increase in prolifer-
ative responses in vitro compared to the other treatment group
and the unvaccinated controls (P0.03). Cells isolated from
the spleens of the infected, vaccinated animals showed similar
patterns of proliferative responses, although these were much
lower (not shown). To examine the immune stimulation in the
lungs of infected, vaccinated animals, cytokine mRNA was
prepared from the lungs at 12 weeks. When compared to the
infected, unvaccinated controls (group 4), the vaccinated, in-
fected animals (groups 1 to 3) had only slightly higher levels of
TNF-mRNA in the lungs (Fig. 1C). Animals in group 1 also
had somewhat higher levels of mRNAs for IL-12 and IFN-;
all these differences were not statistically signicant.
When the lungs of the mice in the four treatment groups
were examined histologically at week 12, granulomas were
larger in the infected animals that had received heat-treated
BCG in L3 adjuvant and were then boosted intranasally (group
3). Morphometric analysis revealed that 69% of the lung pa-
renchyma was occupied by granulomas, compared to 56% in
group 1, 47% in group 2, and 36% in the control animals
(group 4) (Fig. 1D). Thus, although the levels of systemic
immune activation as measured by antigen-specic T-cell pro-
liferation increased, the vaccine-induced immune stimulation
did not affect the number of CFU in the lungs. However, the
immune activation was associated with an exacerbation of pa-
thology, as indicated by the size of the granulomas in the lungs
(compare Fig. 1B and D).
Effect of additional antigen on the host response to M.
tuberculosis infection. To examine whether the presence of
additional mycobacterial antigen in the infected lung changed
the course of infection, mice were infected with either viable
M. tuberculosis strain Erdman (100 CFU) or the same dose of
viable bacilli (100 CFU) mixed with an equivalent dose of
heat-killed organisms (1:1). The presence of additional heat-
killed organisms in the infecting inoculum had no signicant
effect on the subsequent bacillary load (CFU) in the lung,
spleen, and liver of infected mice (Table 1). In mice that
FIG. 1. Effect of postinfection vaccination on bacillary load (CFU) (A), lymph node (LN) T-cell proliferation (B), lung cytokine mRNA levels
(C), and area of lung occupied by granulomas (D). (A and C) M. tuberculosis-infected mice were vaccinated according to the protocol described
in Materials and Methods. Group 1, viable BCG (black bar); group 2, viable BCG followed by boost with heat-treated BCG in L3 adjuvant (gray
bar); group 3, heat-treated BCG in L3 adjuvant and boosted as above (hatched bar); group 4, no vaccination (empty bar). (B) Lymph node cells
stimulated with H37Ra sonicate (gray bars) or unstimulated control (empty bar). (D) Percentage of lung parenchyma occupied by granuloma.
Results are from four animals per group at 12 weeks post-initial infection, expressed as means standard deviation. ,Pcompared to unvaccinated
controls (group 4).
2102 MOREIRA ET AL. INFECT.IMMUN.
received the live infection only, levels of mRNA for TNF-and
for IL-6 increased over the rst 3 weeks of infection (Table 1).
When mice were infected with the mixture of live plus dead M.
tuberculosis, even higher expression of cytokine mRNA for
TNF-and for IL-6 was noted. The difference was most pro-
nounced at week 3 (P0.05); by week 4 the difference was no
longer seen (Table 1). In the lungs of mice infected with the
mixture of viable and heat-killed bacilli, the higher cytokine
mRNA levels were associated with larger granulomas (Fig. 2).
In the mice infected with live plus dead organisms, the mean
size of lesions at 4 weeks postinfection was 6.17 mm
3
1.3
mm
3
, compared to 4.38 mm
3
0.64 mm
3
in those infected with
live organisms only. Distended alveolar spaces (edematic lung)
were also seen, suggesting a relative loss of lung function (Fig.
2). These results indicate that the additional mycobacterial
antigen in the lungs of mice receiving the mixed infection
induces a stronger local inammatory response (increased
TNF-and IL-6 production; Table 1), leading to increased
lung pathology without altering the bacillary burden.
Effect of vaccination with recombinant BCG secreting mu-
rine cytokines on the bacillary load and granuloma size in
infected organs. To determine whether particular cytokines
could enhance the BCG-induced protective immune response
to M. tuberculosis, mice were vaccinated s.c. with recombinant
BCG secreting murine cytokines (BCG vector, BCGTNF-,
BCGIFN-,orBCGIL-2) followed by M. tuberculosis
H37Rv challenge, as described (Materials and Methods). In
these experiments, the recombinant BCG used was prepared
from BCG strain Montreal, a known weak immunogen for
mice (19). By 12 weeks after M. tuberculosis challenge, the
bacilli in the lungs had grown to a concentration of 3 10
7
CFU in control (nonvaccinated) mice (Fig. 3, top). Vaccina-
tion with BCG vector resulted in a bacillary load reduced by
about fourfold. Cytokine secretion by the recombinant BCG
TABLE 1. Infection with viable and viable plus heat-killed M. tuberculosis
Type of infection and no. of wk No. of CFU
a
(log
10
) in: Amt of lung cytokine mRNA
a
(units) for:
Lung Spleen Liver TNF-IL-6
Viable bacilli
1 3.7 0.1 0 1.4 1.2 63 22 46 10
2 5.2 0.3 1.6 1.5 1.9 1.6 69 30 174 17
3 5.5 0.1 3.9 0.2 3.2 0.6 144 53 233 33
4 6.0 0.3 4.1 0.1 2.6 0.3 151 51 161 15
Viable heat-killed bacilli
1 3.5 0.4 0 1.8 1.7 33 5465
2 5.1 0.2 0.9 0.5 2.8 0.8 111 9 243 25
3 5.5 0.1 3.6 0.2 3.2 0.2 269 24
b
384 37
b
4 5.8 0.2 3.9 0.6 3.4 0.2 132 81 149 27
a
Number of mice per group per time point (n5).
b
P0.05 compared to infection with viable bacilli only.
FIG. 2. Effect of additional antigen on the granulomatous response in the lungs of mice at 3 weeks post-aerosol infection with viable M.
tuberculosis (live) or viable plus heat-killed M. tuberculosis (live dead, 1:1). G, granuloma; , distended alveolar spaces (edematic lung).
Ziehl-Neelsen stain, magnication, 10.
VOL. 70, 2002 MYCOBACTERIUM-INDUCED PATHOLOGY IN MURINE TUBERCULOSIS 2103
did not improve the control of the bacillary growth in the lungs.
At 12 weeks postinfection, the lungs of unvaccinated controls
showed an extensive inammatory inltrate composed of mac-
rophages, lymphocytes, and neutrophils within the alveolar
space consistent with a pneumonic process in addition to large,
coalescent granulomas. The size of the granulomas was re-
duced by about 50% in BCG vector-vaccinated mice compared
to unvaccinated controls (P0.05) (Fig. 3, top). Vaccination
with BCG secreting IL-2 or BCG secreting IFN-resulted in
even smaller granulomas than the BCG vector-vaccinated mice
(P0.05). However, BCGTNF-vaccination resulted in
larger granulomas than did vaccination with BCG vector (P
0.05), though the granulomas were still smaller than in non-
vaccinated mice (P0.05). Thus, of the cytokines tested, IL-2
together with BCG appeared to impart the most protection in
terms of limiting immunopathology in the lungs; the presence
of TNF-appeared to exacerbate the pathology.
Effect of postinfection vaccination with cytokine-producing
BCG. To determine whether particular cytokines could render
BCG vaccination more effective against ongoing M. tuberculo-
sis infection, recombinant strains of BCG producing various
murine cytokines were used to treat infected mice. Mice in-
fected with H37Rv were vaccinated twice (at 5 and 8 weeks)
postinfection with 10
6
organisms of viable BCGTNF-, BCG
IL-2, BCGIFN-, or BCG vector. No effect on the number of
CFU in the lungs or spleens was observed at weeks 12 and 20
postinfection compared to in the organs of unvaccinated, in-
fected mice (Fig. 3, bottom). Postinfection administration of
recombinant BCG secreting cytokines did, however, affect the
size of the granulomas. Larger granulomas involving 56.2% of
the lung were observed in the infected mice that had received
recombinant BCGTNF-, compared to 29.6% in the control
unvaccinated mice (P0.02) (Fig. 3, bottom). Infected mice
treated with BCGIL-2 or BCGIFN-had intermediate-sized
granulomas (50 or 46.4%, respectively). Thus, the combination
of the immune-stimulatory cytokines with BCG resulted in
exacerbation of lung pathology; the worst effect was seen when
BCG was combined with TNF-.
Effect of exogenous TNF-on course of M. tuberculosis in-
fection. The previous experiments suggested that the deleteri-
ous component of the immune response to mycobacterial an-
tigens is excess TNF-. To directly examine the effect of excess
TNF-on lung pathology, mice infected with 80 CFU of M.
tuberculosis were treated intranasally with recombinant murine
TNF-for the rst 4 weeks of infection. At 4 weeks and then
8 weeks after infection and treatment, the numbers of CFU in
lungs and spleens were similar (Fig. 4, top). However, survival
of the mice was substantially altered. Mice infected with M.
FIG. 3. Effect of vaccination (preinfection, top) and immunotherapy (postinfection, bottom) with cytokine-secreting BCG on number of CFU
in lung at 12 and 20 weeks post-initial infection (left panels) and on granuloma size at 12 weeks (right panels). Results are from four animals per
group per time point, expressed as means standard deviation. a, P0.05 compared to unvaccinated controls; b, P0.05, higher than for BCG
vector-vaccinated mice; c, P0.05, lower than for BCG vector-vaccinated mice.
2104 MOREIRA ET AL. INFECT.IMMUN.
tuberculosis and treated with TNF-succumbed to the infec-
tion signicantly earlier than mice that had not received the
cytokine treatment (P0.008) (Fig. 4, top). In contrast, when
infected mice (100 CFU at baseline) were treated intranasally
with IFN-, no effect on survival or number of CFU in lungs
and spleens was noted (Fig. 4, bottom).
TNF-treatment of infected mice was associated with in-
creased cellularity of the lungs, including higher numbers of
polymorphonuclear leukocytes in the granulomas and larger
numbers of foamy macrophages in the alveolar spaces at 4
weeks (Fig. 5). The macrophages did not appear to be more
activated, as there was no increase in the mRNA levels of
inammatory cytokines in the lungs (IL-6, IL-10, IL-12, and
TNF-) (not shown). The lungs of the infected, untreated mice
appeared to have higher numbers of small lymphocytes (Fig.
5). Again, the lymphocytes did not appear to be more acti-
vated, since the IFN-mRNA levels were similar or even
somewhat lower (about 40%) in the lungs of TNF--treated
mice at 4 and 8 weeks (not shown). Thus, TNF-treatment
appeared to result in the exacerbation of lung pathology with-
out signicant changes in the bacillus burden. On the other
hand, IFN-treatment did not affect these parameters.
DISCUSSION
Our results, as well as those of others, demonstrate how
difcult it is to change the course of existing M. tuberculosis
infection in the lungs by immunotherapy with vaccines. Indeed,
Koch himself did not claim that his treatment with tuberculin
lymphkilled the bacteria (5). In our postinfection experi-
ments, vaccination with BCG, BCG plus other antigens, re-
combinant BCG secreting murine cytokines, or heat-killed M.
tuberculosis did not reduce the bacterial load in the lungs of
infected mice. Even those vaccines that were shown to be
effective as preexposure vaccines (Fig. 3) failed to reduce the
number of CFU when administerred as postinfection vaccines.
Analogous results have been obtained by other investigators.
Turner et al. tested three different vaccines in infected mice
and guinea pigs as immunotherapy: (i) BCG Pasteur, (ii) a
subunit vaccine containing puried culture ltrate proteins of
M. tuberculosis emulsied in an adjuvant together with recom-
binant murine IL-2, and (iii) a DNA vaccine consisting of a
vector expressing the gene for M. tuberculosis Ag85A (23). The
candidate vaccines were selected because they had shown ef-
cacy as preexposure vaccines in protecting naı¨ve animals
against challenge with M. tuberculosis (1, 10). However, all
three of these vaccines, although protective as preexposure
vaccines, failed to reduce the bacterial load in the lungs of mice
with prior tuberculosis infection. Similarly, Lowrie et al. tested
a number of DNA vaccines encoding M. leprae Hsp65 or M.
tuberculosis Hsp70 or ESAT-6 as postinfection treatments (16).
Only the DNA vaccine encoding M. leprae Hsp65 resulted in
some reduction in bacterial load in the lungs of infected mice.
FIG. 4. Effect of exogenous recombinant cytokines on the bacillary load (right panels) and survival of infected mice (left panels). Mice were
infected with M. tuberculosis and treated intranasally with recombinant murine TNF-(top panels) or IFN-(bottom panels). Solid squares,
cytokine-treated mice; empty squares, control untreated mice. Results for survival are from 11 mice per group and for numbers of CFU from four
mice per group per time point.
VOL. 70, 2002 MYCOBACTERIUM-INDUCED PATHOLOGY IN MURINE TUBERCULOSIS 2105
Interestingly, when preexisting infection had been elimi-
nated with antibiotic therapy, repeated postexposure vaccina-
tion did protect against reactivation of disease (16). Thus, it is
possible that immune modulation in the presence of antituber-
culous therapy may facilitate bacillary clearance in the lungs
(9). Indeed, in a previous pilot study of patients with multi-
drug-resistant pulmonary TB, we observed that administration
of recombinant IL-2 as adjunctive therapy combined with an-
ti-TB chemotherapy resulted in accelerated sputum clearance
and improved radiographic appearance of the lungs (12).
As we have clearly shown here, increased immune activation
is associated with exacerbation of pathology at the sites of
preexisting TB. This had originally been noted by Koch. In
response to the treatment with tuberculin lymph,infected
tissues became inamed and necrotic and partially sloughed
often in association with clinical worsening. The English phy-
sician and writer A. Conan Doyle wrote about Kochs treat-
ment, It may also be remarked that the fever after the injec-
tion is in some cases so very high (41°C) that it is hardly safe to
use in the case of a debilitated patient(in Review of Reviews,
December 1890) (5). Our experiments in mice suggest that the
cause of the worsening pathology and fever observed by Koch
in patients may be excess TNF-at the site of infection. In our
murine experiments, the increased size and cellularity of the
granulomas were accompanied by increased expression of
TNF-mRNA in the lung. Consistent with this, vaccination of
mice with BCGTNF-resulted in an increase in the size of
lung granulomas. Also, treatment of infected mice with recom-
binant murine TNF-resulted in increased inammation in
the lung and accelerated death. In a previous study, in which
mice were infected with BCGTNF-, we noted that high
levels of TNF-at the BCG infection site resulted in much
severer lung pathology and decreased survival (2). Further-
more, TNF--associated clinical worsening has been observed
in patients with severe TB upon initiation of anti-TB chemo-
therapy (3). This was probably due to death of the organisms
and release of mycobacterial components. Mycobacteria and
their products have previously been shown to induce TNF-
production through signaling of the macrophage Toll-like re-
ceptors (24, 25).
In addition to the safety issues associated with postinfection
vaccination and the Koch phenomenon, there is also an oper-
FIG. 5. Effect of exogenous TNF-on the granulomatous response in the lungs. Mice were infected with M. tuberculosis and treated intranasally
with recombinant murine TNF-, and the lungs were examined at 4 weeks postinfection. (A to C) Untreated mice. (D to F) TNF--treated mice.
Note extensive lymphocyte inltration in arrows (A) and Ly (C). In response to TNF-treatment (E), large macrophages are present in the
alveolar spaces (arrowheads). Hematoxylin and eosin stain. (A and D) Magnication, 4. (B, C, E, and F) Magnication, 40.
2106 MOREIRA ET AL. INFECT.IMMUN.
ational consideration for any trial designed to test postexpo-
sure vaccine. Vaccination of adults in an area of high ende-
micity might reveal occult disease, which might confound
interpretation of the results, since the number of TB cases in
the vaccine arm would be selectively increased compared to
that in the untreated arm. This may have been the case in the
BCG vaccine trial carried out in the Karonga region of Malawi,
where signicantly higher rates of pulmonary TB were re-
ported among scar-positive persons who had received a second
dose of BCG (13). Thus, for both safety (immune-mediated
pathology) and study design (subclinical disease scored as a
newly acquired TB), any study of candidate postexposure vac-
cines must ensure that recruited participants do not have un-
diagnosed disease.
ACKNOWLEDGMENTS
These studies were supported by NIH grants AI22616 and AI42056
(to G.K.) and AITRP TW00231 (to L.-G.B.) and by Direct Effect (New
York, N.Y.). A.L.M. was supported by a grant from the Potts Foun-
dation.
We thank Judy Adams for preparation of the gures and Marguerite
Nulty for secretarial assistance.
L.T. and A.L.M. contributed equally to the paper.
REFERENCES
1. Baldwin, S. L., C. DSouza, A. D. Roberts, B. P. Kelly, A. A. Frank, M. A. Lui,
J. B. Ulmer, K. Huygen, D. M. McMurray, and I. M. Orme. 1998. Evaluation
of new vaccines in the mouse and guinea pig model of tuberculosis. Infect.
Immun. 66:29512959.
2. Bekker, L.-G., A. L. Moreira, A. Bergtold, S. Freeman, B. Ryffel, and G.
Kaplan. 2000. Immunopathologic effects of tumor necrosis factor alpha in
murine mycobacterial infection are dose dependent. Infect. Immun. 68:
69546961.
3. Bekker, L.-G., G. Maartens, L. Steyn, and G. Kaplan. 1998. Selective in-
crease in plasma TNF-alpha and concomitant clinical deterioration after
initiating therapy in patients with severe tuberculosis. J. Infect. Dis. 178:580
584.
4. Caruso, A. M., N. Serbina, E. Klein, K. Triebold, B. R. Bloom, and J. L.
Flynn. 1999. Mice decient in CD4 T cells have only transiently diminished
levels of IFN-gamma, yet succumb to tuberculosis. J. Immunol. 162:5407
5416.
5. Dubos, R., and J. Dubos. 1952. The white plague. Little, Brown & Co.,
Boston, Mass.
6. Eid, P., J. F. Meritet, C. Maury, A. Lasfar, D. Weill, and M. G. Tovey. 1999.
Oromucosal interferon therapy: pharmacokinetics and pharmacodynamics.
J. Interferon Cytokine Res. 19:157169.
7. Fine, P. E. 1998. Vaccines, genes and trials. Novartis Found. Symp. 217:57
69.
8. Fine, P. E. 1995. Variation in protection by BCG: implications of and for
heterologous immunity. Lancet 346:13391345.
9. Glassroth, J. 2000. Clinical considerations in designing trials of vaccines for
tuberculosis. Clin. Infect. Dis. 30:S229S232.
10. Huygen, K., J. Content, O. Denis, D. L. Montgomery, A. M. Yawman, R. R.
Deck, C. M. DeWitt, I. M. Orme, S. Baldwin, C. DSouza, A. Drowart, E.
Lozes, P. Vandenbussche, J.-P. Van Vooren, M. A. Liu, and J. B. Ulmer.
1996. Immunogenicity and protective efcacy of a tuberculosis DNA vaccine.
Nat. Med. 2:893899.
11. Iseman, M. D. 2000. A physicians guide to tuberculosis. Lippincott Williams
& Wilkins, Philadelphia, Pa.
12. Johnson, B. J., L.-G. Bekker, R. Rickman, S. Brown, M. Lesser, S. Ress, P.
Willcox, L. Steyn, and G. Kaplan. 1997. rhuIL-2 adjunctive therapy in mul-
tidrug resistant tuberculosis: a comparison of two treatment regimes and
placebo. Tuber. Lung Dis. 78:195203.
13. Karonga Prevention Trial Group. 1996. Randomised controlled trial of
single BCG, repeated BCG, or combined BCG, and killed Mycobacterium
leprae vaccine for prevention of leprosy and tuberculosis in Malawi. Lancet
348:1724.
14. Koch, R. 1890. Weitere. Mitteilungen uber ein Heilmittel gegen Tuberku-
lose. Dtsch. Med. Wochenschr. 16:10291032.
15. Laochumroonvorapong, P., J. Wang, C.-C. Liu, W. Ye, A. L. Moreira, K. B.
Elkon, V. Freedman, and G. Kaplan. 1997. Perforin, a cytotoxic molecule
which mediates cell necrosis, is not required for early control of mycobac-
terial infection in mice. Infect. Immun. 65:127132.
16. Lowrie, D. B., R. E. Tascon, V. L. Bonato, V. M. Lima, L. H. Faccioli, E.
Stavropoulos, M. J. Colston, R. G. Hewinson, K. Moelling, and C. L. Silva.
1999. Therapy of tuberculosis in mice by DNA vaccination. Nature 400:269
271.
17. Moreira, A. L., L. Tsenova-Berkova, J. Wang, P. Laochumroonvorapong, S.
Freeman, V. H. Freedman, and G. Kaplan. 1997. Effect of cytokine modu-
lation by thalidomide on the granulomatous response in murine tuberculosis.
Tuber. Lung Dis. 78:4755.
18. Murray, P. J., A. Aldovini, and R. A. Young. 1996. Manipulation and poten-
tiation of antimycobacterial immunity using recombinant bacille Calmette-
Guerin strains that secrete cytokines. Proc. Natl. Acad. Sci. USA 93:934939.
19. Orme, I. M., and F. M. Collins. 1984. Demonstration of acquired resistance
in Bcgr inbred mouse stains infected with a low dose of BCG montreal. Clin.
Exp. Immunol. 56:8188.
20. Sreevatsan, S., X. Pan, K. E. Stockbauer, N. D. Connell, B. N. Kreiswirth,
T. S. Whittam, and J. M. Musser. 1997. Restricted structural gene polymor-
phism in the Mycobacterium tuberculosis complex indicates evolutionarily
recent global dissemination. Proc. Natl. Acad. Sci. USA 94:98699874.
21. Tascon, R. E., M. J. Colston, S. Ragno, E. Stavropoulos, D. Gregory, and
D. B. Lowrie. 1996. Vaccination against tuberculosis by DNA injection. Nat.
Med. 2:888892.
22. Tsenova, L., A. L. Moreira, E. Party, V. H. Freedman, and G. Kaplan. 1997.
Aerosol infection of mice with mycobacteria using a nose-only exposure
device. J. Am. Biol. Safety Assoc. 2:2031.
23. Turner, J., E. R. Rhoades, M. Keen, J. T. Belisle, A. A. Frank, and I. M.
Orme. 2000. Effective preexposure tuberculosis vaccines fail to protect when
they are given in an immunotherapeutic mode. Infect. Immun. 68:17061709.
24. Underhill, D. M., A. Ozinsky, A. M. Hajjar, A. Stevens, C. B. Wilson, M.
Bassetti, and A. Aderem. 1999. The Toll-like receptor 2 is recruited to
macrophage phagosomes and discriminates between pathogens. Nature 401:
811815.
25. Underhill, D. M., A. Ozinsky, K. D. Smith, and A. Aderem. 1999. Toll-like
receptor-2 mediates mycobacteria-induced proinammatory signaling in
macrophages. Proc. Natl. Acad. Sci. USA 96:1445914463.
Editor: S. H. E. Kaufmann
VOL. 70, 2002 MYCOBACTERIUM-INDUCED PATHOLOGY IN MURINE TUBERCULOSIS 2107
... However, the vaccine therapy was abandoned because of side effects, and practically no studies directed at a therapeutic vaccine against TB appeared for a long time thereafter. Recently, trials using a subunit vaccine for therapy of TB also suggested that great caution needs to be paid in such strategies (Moreira et al., 2002). Coler et al., 1998, demonstrated that plasmid DNA injection could elicit an immune response in animals, which opened a new era in vaccinology. ...
... However subsequent results from other laboratories using various DNA vaccines in infected murine models produced inconsistent results (Flynn et al., 1995). When plasmid V1Jns containing a gene encoding Ag85A of M. tuberculosis (which had been shown to induce protective immunity in mice) was injected into mice previously infected with M. tuberculosis, a significant reduction in splenic bacterial load was found, but no bacterial and histological changes were detected in the lungs (Moreira et al., 2002). Later, with a view to test whether immunization with TB DNA vaccine would retard the endogenous reactivation of the latent bacilli and exogenous reinjection, Repique and colleagues injected a TB DNA vaccine combination (consisting of 10 different single DNA vaccines) into infected mice after a course of drug treatment. ...
... Similar necrosis was also seen in mice immunized with a DNA vaccine encoding the Ag85 antigen of M. tuberculosis. Such an immune-pathological reaction (known as a Koch reaction) after DNA vaccination posed a significant safety problem to the development of therapeutic DNA vaccines (Moreira et al., 2002). Fortunately, this enhanced immunopathologic reaction after DNA vaccination has not been observed in other studies, and more recently studies have provided more promising results. ...
Preprint
Full-text available
DNA vaccines are simple rings of DNA containing a gene encoding an antigen, and a promoter/terminator to express the gene in mammalian cells. Some DNA vaccines which previously showed to induce protective immunity against infection by Mycobacterium tuberculosis (M. TB) in a prophylactic manner are also surprisingly effective when used therapeutically, including persistent Mycobacterium tuberculosis and multidrug-resistant tuberculosis (MDR-TB) which are refractory to immune system and antibacterial chemotherapy alone. When used in combination with antibacterial drugs, therapeutic DNA vaccines could effectively eliminate residual bacteria in infected animals and shorten the therapy course of conventional chemotherapy. Detailed studies demonstrated that therapeutic effects of DNA vaccines may at least partly be due to the restoration of the Th1/Th2 balance. Some problems have also emerged along with these exciting results. Therapeutic DNA vaccine is a promising strategy against tuberculosis, however developing an ideal DNA vaccine for therapy of tuberculosis will require further development. The purpose of this paper is to review recent developments in therapeutic DNA vaccines against tuberculosis.
... A study employing immunohistochemical staining on granulomas from diverse TB patients revealed significant variations in immune reactivity. While macrophages and multinucleated giant cells were consistent in number across samples, T lymphocyte distribution varied [19]. Cytokine expression and release can differ among patients. ...
... For instance, tumor necrosis factor alpha (TNF-α) aids in countering TB by fostering granuloma formations. However, its premature or excessive secretion can induce an intensified inflammatory response, further damaging lung tissues [19]. A study comparing TB patient sera indicated elevated TNF-α levels in severe cases than in mild-to-moderate ones (p < 0.05) [20]. ...
Article
Full-text available
Post-tuberculosis lung disease (PTLD) is emerging as a significant area of global interest. As the number of patients surviving tuberculosis (TB) increases, the subsequent long-term repercussions have drawn increased attention due to their profound clinical and socioeconomic impacts. A primary obstacle to its comprehensive study has been its marked heterogeneity. The disease presents a spectrum of clinical manifestations which encompass tracheobronchial stenosis, bronchiectasis, granulomas with fibrosis, cavitation with associated aspergillosis, chronic pleural diseases, and small airway diseases—all persistent consequences of PTLD. The spectrum of symptoms a patient may experience varies based on the severity of the initial infection and the efficacy of the treatment received. As a result, the long-term management of PTLD necessitates a detailed and specific approach, addressing each manifestation individually—a tailored strategy. In the immediate aftermath (0–12 months after anti-TB chemotherapy), there should be an emphasis on monitoring for relapse, tracheobronchial stenosis, and smoking cessation. Subsequent management should focus on addressing hemoptysis, managing infection including aspergillosis, and TB-associated chronic obstructive pulmonary disease or restrictive lung function. There remains a vast expanse of knowledge to be discovered in PTLD. This review emphasizes the pressing need for comprehensive, consolidated guidelines for management of patients with PTLD.
... Though the relapse was demonstrated following intravenous inoculation of BCG, a lower risk from subcutaneous vaccination may depend from the vaccine dose [37] and could apply also to subsequent pathogenic Mtb infection. However, the early conditioning for reactivation is clearly different from the later mechanisms which can be alleviated by immunotherapy [38][39][40] and different from the TNFα mediated exacerbation of lung granulomas without reducing the bacillary load ('Koch phenomenon'), caused by the administration of mycobacterial antigens following Mtb infection [41]. ...
... The key importance of the sequence of antigen exposure and the potential for a deleterious impact was demonstrated by the finding that immunization with various mycobacterial antigens can aggravate lung pathology in Mtb pre-infected mice [41]. The 'original antigenic sin' (OAS) phenomenon, reflecting the predominance of immunity to the first priming antigen following booster immunization, has been described following infection and vaccination with influenza virus and several other infections [101], thus overriding the antigenic specificity of the booster antigen [102,103]. ...
Article
Current search for a new effective vaccine against tuberculosis involves selected antigens, vectors and adjuvants. These are being evaluated usually by their booster inoculation following priming with Bacillus Calmette-Guerin. The purpose of this article is to point out, that despite being attenuated of virulence, priming with BCG may still involve immune mechanisms, which are not favourable for protection against active disease. It is postulated, that the responsible ‘decoy’ constituents selected during the evolution of pathogenic tubercle bacilli may be involved in the evasion from bactericidal host resistance and stimulate immune responses of a cytokine phenotype, which lead to the transition from latent closed granulomas to reactivation with infectious lung cavities. The decoy mechanisms appear as favourable for most infected subjects but leading in a minority of cases to pathology which can effectively transmit the infection. It is proposed that construction and development of new vaccine candidates could benefit from avoiding decoy-type immune mechanisms.
... Further drug classes like fluoroquinolones, oxazolidinediones, rifamycin phenothiazine, nitroimidazole and azole derivatives which belong to different classes of chemotherapeutic agents have also emerged recently as probable and effective treatments of tuberculosis. 8,9 The plant chosen for study namely Vitex trifolia L. belongs to the genus Vitex which is a shrub about 6m in height and the plant exhibits varied pharmacological activities like thermogenic, astringent, expectorant, carminative, anthelmintic anti-inflammatory, anti-tubercular, anti-pyrogenic and anti-cancer properties. 10,11 Various diterepenes vitretifolin D, vitretifolins E have been isolated from the plant and they have shown promising results against several cell lines and promising inhibitory activity on inflammatory mediators, on in vitro cell lines and on IL-6, IL-10 and TNF-α synthesis. ...
Article
Full-text available
Background: The experiment was intended to evaluate the effect of Vitex trifolia Linn's hydroalcoholic extract to treat chronic inflammation. Materials and Methods: Chronic inflammation was induced by injecting 0.25 mL of Complete Freund's Adjuvant subcutaneously in the palmar surface of the right-hand paw of Wistar rats. The animals were treated with two different doses of the plant extract and their efficiency and efficacy was compared to standard drugs indomethacin and dexamethasone for 28days. At the end of the study the serum TNF-α and IL-10 using ELISA kits, differential leukocyte count was estimate, vital organs were subjected to histological studies. The anti-tubercular activity of the extracts of V. trifolia was also evaluated by the Microplate Alamar Blue Assay method (MABA) against Mycobacterium tuberculosis. Results: The extract has also been shown to be effective in controlling levels of chemical cytokines IL-10 and TNF-α and the anti-TB activity was evaluated using Alamar Blue Assay which also exhibited promising results. Conclusion: The obtained results implicate that hydroalcoholic extracts of V. trifolia show promising anti-inflammatory action on chronic stage of inflammation and as well as have proven to be effective as anti-tubercular drug.
... Vaccination can, in some cases, exacerbate manifestations of subsequent disease instead of mitigating it [23]. This has been observed in previous studies, under similar or different conditions (e.g., heterologous vaccination), with vaccination based on mycobacterial antigens in mice [24,25], high-dose BCG vaccination in badgers [14] and vaccination with inactivated M. paratuberculosis in pigs [26]. One possible explanation for this is a differed immune response in some individuals by shifting towards a humoral type 2 immune response, or a lymphatic dysfunction, such as lymphoma as reported in badgers after oral BCG vaccination [27]. ...
Article
Full-text available
Badgers (Meles meles) are a major tuberculosis (TB) reservoir in Europe, with the potential to transmit infection to cattle. Here we assessed whether a recently described oral tuberculosis vaccine based on heat-inactivated Mycobacterium bovis (HIMB), delivered as edible baits, can protect badgers from infection. Eight badgers were given individually five baits, each one consisting of a ball of peanut butter, natural peanut and oat flakes including a dose of the vaccine containing 5 × 107 colony-forming units. In parallel, a control group of seven badgers did not receive the vaccine. One month and a half later a second dose of the vaccine was offered to the vaccinated group. Ninety-four days after the second dose, all badgers were challenged with M. bovis (103 colony-forming units per animal) delivered endobronchially to the right middle lung lobe. Clinical, immunological, pathological and bacteriological variables were measured throughout the whole study to assess the efficacy of the vaccine. Two vaccinated animals showed high bacterial load of M. bovis and worsening of pathological lesions of TB. Conversely, the other six vaccinated animals showed slight improvement in bacterial load and pathology with respect to the control group. These results suggest that delivering the TB vaccine via food bait can partially protect wild badger populations, although vaccination can lead to either protection or tolerization, likely depending on the animal's immune status and general condition at the time of vaccination. Further optimization of the vaccination trial/strategy is needed to reduce the rate of tolerization, such as altering vaccine dose, number of doses, type of bait, use of adjuvants or route of administration.
... While adult pulmonary TB is frequently considered to involve pathological manifestations of a hyperactive anti-mycobacterial immune response, key details have been lost [26]. The Koch phenomenon today is largely a historical curiosity that has the potential for causing toxicity of therapeutic vaccines [27][28][29][30][31][32]. In the preantibiotic era, it was considered central to the pathogenesis of TB and responsible for the death of many people. ...
Article
Full-text available
Research on the pathogenesis of tuberculosis (TB) has been hamstrung for half a century by the paradigm that granulomas are the hallmark of active disease. Human TB, in fact, produces two types of granulomas, neither of which is involved in the development of adult type or post-primary TB. This disease begins as the early lesion; a prolonged subclinical stockpiling of secreted mycobacterial antigens in foamy alveolar macrophages and nearby highly sensitized T cells in preparation for a massive necrotizing hypersensitivity reaction, the Koch Phenomenon, that produces caseous pneumonia that is either coughed out to form cavities or retained to become the focus of post-primary granulomas and fibrocaseous disease. Post-primary TB progresses if the antigens are continuously released and regresses when they are depleted. This revised paradigm is supported by nearly 200 years of research and suggests new approaches and animal models to investigate long standing mysteries of human TB and vaccines that inhibit the early lesion to finally end its transmission.
Article
Full-text available
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS CoV-2) has caused a global human outbreak, making it a more serious threat to human health than any other infectious disease. Coronavirus infectious disease 2019 (COVID-19) has severely affected the lifestyles of people around the world and caused high mortality throughout the world. In both pandemic and seasonal influenza, co-infection of COVID-19 with other diseases has been linked to worse outcomes. The literature revealed that it is characteristically associated with comorbidities such as hypertension, blood pressure, obesity, cardiovascular diseases, and other microbial infections. Furthermore, microbial coinfections worsen respiratory viral infections and are a common cause of death in influenza pandemics. Deplorably, Tuberculosis (TB) is also a dreadful lung infection and attains cytokine equilibrium with host cells to maintain the latent stage. Studies showed that human coronaviruses (hCoV) activate latent TB to an active state due to unregulated cytokine production, called a cytokine storm. The present review concisely discusses the reason and status of co-infection of COVID-19 with TB based on previous case reports, cohorts, and scientific studies. COVID-19 patients are prone to be infected with TB and vice-versa in TB-prone areas. The therapeutic opportunities for overcoming the COVID-19 induced cytokine storm have also been emphasized by the present clinical trial candidates. In conclusion, we recommend categorizing the patients based on their medical history and cured or latent TB patients should be particularly closely monitored. They should be tested for Interferon Gamma Release Assay (IGRA) regularly on and after COVID-19 infection.
Article
Purpose: Ethambutol (EMB) is an important first line drug, however little information on its molecular mechanism of resistance and pathogenicity of resistant isolates is available. Present work was designed to study virulence of the EMB resistant M. tuberculosis strains and the host responses in-vivo on infection of EMB resistant M. tuberculosis using Balb/c mouse model of infection. Methods: Three groups of Balb/c mice (female, age 4-6 wk; 21 mice in each group) were infected intravenously with 106 CFU of M. tuberculosis H37Rv and two EMB resistant clinical isolates. Age and sex matched control animals were mock inoculated with Middlebrook 7H9 broth alone. At 10, 20, 30, 40, 50, 60, and 70 days post-infection three animals from each group were sacrificed by cervical dislocation and lung tissue was collected for further analysis. Results: Infection with EMB resistant M. tuberculosis led to progressive and chronic disease with significantly high bacillary load (p=0.02). Massive infiltration and exacerbated lung pathology with increased expression of IFN-γand TNF-αwas observed in lungs of mice infected with EMB resistant strains. The present study suggests that infection with EMB resistant M. tuberculosis leads to chronic infection with subsequent loss of lung function, bacterial persistence with elevated expression of TNF-αresulting in increased lung pathology. Conclusion: These findings highlight that EMB resistant M. tuberculosis regulates host immune response differentially and its pathogenicity is different from drug sensitive strains of M. tuberculosis.
Article
Full-text available
Bacille Calmette-Guérin (BCG) is a live, attenuated strain of Mycobacterium bovis used widely for tuberculosis prophylaxis and bladder cancer immunotherapy, although it has limitations in both contexts. To investigate whether BCG's immunostimulatory properties could be modified, and to gain insight into the interaction between mycobacteria and their hosts, we constructed recombinant BCG strains that secrete functional murine cytokines and studied their properties in mouse models of experimental infection. Cell-mediated immune responses to mycobacterial antigen (purified protein derivative) were assayed using splenocytes from mice inoculated with various BCG recombinants. Antigen-specific proliferation and cytokine release were found to be substantially greater with splenocytes derived from mice injected with cytokine-secreting BCG than with splenocytes from mice injected with BCG lacking cytokines. The most profound effects were induced by BCG secreting interleukin 2, interferon gamma, or granulocyte-macrophage colony-stimulating factor. Thus, cytokine-secreting BCG can enhance immune responses to mycobacterial antigens and may be improved reagents for tuberculosis prophylaxis and cancer immunotherapy.
Article
The effectiveness of an aerosol producing system (In-Tox Products, Albuquerque, NM) commonly used for toxicological studies of chemicals and radioisotopes was evaluated for experimental tuberculosis infection of mice. Previously, experimental tuberculosis of rodents has been accomplished by aerosol infection devices which exposed the entire animal to the aerosol within a chamber. We utilized the In-Tox system to infect animals via the respiratory route with Mycobacterium bovis strain Bacille Calmette-Guerin (BCG) as a model for experimental pulmonary tuberculosis. The advantage of the device tested is that exposure to the infectious aerosol is limited to the nose of the animal. The present instrument was found to yield good implantation of mycobacteria into the lungs. The infectious load was reproducible and, with the addition of some filtration, the infectious agent was well contained within the device. Our results showed that this device is easy to operate, produces an effective experimental aerosol infection and is safe for the investigator.
Article
Background Repeat BCG vaccination is standard practice in many countries for prevention of tuberculosis and leprosy, but its effectiveness has not been evaluated. The addition of Mycobacterium leprae antigens to BCG might improve its effectiveness against leprosy. A double-blind, randomised, controlled trial to evaluate both these procedures was carried out in Karonga District, northern Malawi, where a single BCG vaccine administered by routine health services had previously been found to afford greater than 50% protection against leprosy, but no protection against tuberculosis. Methods Between 1986 and 1989, individuals lacking a BCG scar were randomly assigned BCG alone (27 904) or BCG plus killed M leprae (38 251). Individuals with a BCG scar were randomly allocated placebo (23 307), a second BCG (23 456), or BCG plus killed M leprae (8102). Incident cases of leprosy and tuberculosis were ascertained over the subsequent 5-9 years. Findings 139 cases of leprosy were identified by May, 1995; 93 of these were diagnostically certain, definitely postvaccination cases. Among scar-positive individuals, a second BCG vaccination gave further protection against leprosy (about 50%) over a first BCG vaccination. The rate ratio for all diagnostically certain, definitely postvaccination cases, all ages, was 0.51 (95% CI 0.25-1.03, p=0.05) for BCG versus placebo. This benefit was apparent in all subgroups, although the greatest effect was among individuals vaccinated below 15 years of age (RR=0.40 [95% CI 0.15-1.01], p=0.05). The addition of killed M leprae did not improve the protection afforded by a primary BCG vaccination. The rate ratio for BCG plus killed M leprae versus BCG alone among scar-negative individuals was 1.06 (0.62-1.82, p=0.82) for all ages, though 0.37 (0.11-1.24, p=0 09) for individuals vaccinated below 15 years of age. 376 cases of postvaccination pulmonary tuberculosis and 31 of glandular tuberculosis were ascertained by May, 1995. The rate of diagnostically certain tuberculosis was higher among scar-positive individuals who had received a second BCG (1.43 [0.88-2.35], p=0.15) than among those who had received placebo and there was no evidence that any of the trial vaccines contributed to protection against pulmonary tuberculosis. Interpretation In a population in which a single BCG vaccination affords 50% or more protection against leprosy, but none against tuberculosis, a second vaccination can add appreciably to the protection against leprosy, without providing any protection against tuberculosis.
Article
Low-dose recombinant human interleukin 2 (rhuIL-2) adjunctive immunotherapy in multidrug resistant tuberculosis (MDR-TB) patients. Evaluation of the effects of daily versus pulse-administered rhuIL-2 compared to placebo. MDR-TB patients on best available antituberculous chemotherapy received rhuIL-2 for 30 consecutive days (daily therapy), or for 5 days followed by a 9-day 'rest', for three cycles (pulse therapy). Placebo control patients received diluent. The cumulative total dose of rhuIL-2 given to each patient in either rhuIL-2 treatment group was the same. Patient immunologic, microbiologic, and radiologic responses were compared. The three treatment schedules induced different results. Immune activation was documented in patients receiving daily rhuIL-2 therapy. Numbers of CD25+ and CD56+ cells in the peripheral blood were increased in these patients, but not in patients receiving pulse rhuIL-2 or placebo. In addition, 5/8 (62%) patients receiving daily rhuIL-2 demonstrated reduced or cleared sputum bacterial load while only 2/7 (28%) pulse rhuIL-2 treated and 2/8 (25%) controls showed bacillary clearance. Chest radiographs of 7/12 (58%) patients receiving daily rhuIL-2 indicated significant improvement over 6 weeks. Only 2/9 (22%) pulse rhuIL-2-treated patients and 5/12(42%) placebo controls showed radiologic improvement. Daily low dose rhuIL-2 adjunctive treatment stimulates immune activation and may enhance the antimicrobial response in MDR-TB.
Article
Setting: Experimental murine tuberculosis. Objective: To evaluate the effect of cytokine modulation by thalidomide on the progression of the lung granulomatous response following aerosol tuberculosis infection in mice. Design: Mice infected by the respiratory route with 200-500 viable Mycobacterium tuberculosis Erdman were treated with daily subcutaneous injections of thalidomide (30 mg/kg) or saline for 4 weeks. The bacillary load, granulomatous response and cytokine production in the lungs were evaluated. Results: Aerosol M. tuberculosis infection resulted in a progressive granulomatous response in the lungs. At 28 days after infection, large granulomata with central necrosis and no apoptosis were observed. The infection induced high serum and lung cytokine mRNA levels. Thalidomide treatment resulted in a significant reduction in tumor necrosis factor-alpha, interleukin 6 (IL-6) and IL-10 protein levels (blood) and mRNA expression (lungs). IL-12 and interferon-gamma were unaffected. The lungs of thalidomide-treated mice had smaller granulomata with apoptotic cells and no necrosis. Thalidomide treatment did not change the bacillary load. Conclusion: Thalidomide immunomodulation reduces inflammatory cytokines and concomitant lung pathology following acute aerosol M. tuberculosis infection, without increasing the bacillary load.
Article
The relationship between natural resistance to Mycobacterium bovis BCG, expressed by the Bcg gene, and the generation of acquired resistance to this infection in various selected inbred strains of mice was investigated. Consistent with previous findings, a low dose (approximately 10(4)) of BCG Montreal grew progressively in the spleens of inbred mouse strains previously designated susceptible to BCG (Bcgs), but grew poorly in resistant strains (Bcgr). In contrast, however, little difference was observed in the growth of the organism in the liver or lungs of these mice, whereas furthermore, all animals behaved as Bcgs when infected with the World Standard preparation of BCG, BCG Pasteur. Moreover, four strains tested (Bcgr; A/J, C3H/HeJ, and Bcgs; B10.A/J, BALB/c), all showed evidence of the generation of acquired resistance to a small inoculum of BCG Montreal, as demonstrated by their substantial protection against a subsequent intravenous challenge with virulent M. tuberculosis. These findings are interpreted as being inconsistent with the Bcg gene hypothesis and call into doubt the usage of the term Bcg as a gene designation.
Article
Besides being the world's most widely used vaccine, and being directed against the world's leading cause of infectious disease mortality, BCG is the most controversial vaccine in current use.1,2 Estimates of protection imparted by BCG against pulmonary tuberculosis vary from nil to 80%. This variability has been attributed to strain variation in BCG preparations, to genetic or nutritional differences between populations, and to environmental influences such as sunlight exposure, poor cold-chain maintenance, or exposure to environmental mycobacterial infections. Evidence accumulated to date indicates that regional differences in environmental mycobacteria are responsible for much of the variation observed between populations in the efficacy of BCG against pulmonary tuberculosis. This paper reviews the evidence, and notes its broader implications for the epidemiology and control of mycobacterial diseases as well as for other infections and vaccines.