ArticlePDF Available

Role of the chemokine decoy receptor D6 in balancing inflammation, immune activation, and antimicrobial resistance in Mycobacterium tuberculosis infection

Rockefeller University Press
Journal of Experimental Medicine (JEM)
Authors:

Abstract and Figures

D6 is a decoy and scavenger receptor for inflammatory CC chemokines. D6-deficient mice were rapidly killed by intranasal administration of low doses of Mycobacterium tuberculosis. The death of D6(-/-) mice was associated with a dramatic local and systemic inflammatory response with levels of M. tuberculosis colony-forming units similar to control D6-proficient mice. D6-deficient mice showed an increased numbers of mononuclear cells (macrophages, dendritic cells, and CD4 and CD8 T lymphocytes) infiltrating inflamed tissues and lymph nodes, as well as abnormal increased concentrations of CC chemokines (CCL2, CCL3, CCL4, and CCL5) and proinflammatory cytokines (tumor necrosis factor alpha, interleukin 1beta, and interferon gamma) in bronchoalveolar lavage and serum. High levels of inflammatory cytokines in D6(-/-) infected mice were associated with liver and kidney damage, resulting in both liver and renal failure. Blocking inflammatory CC chemokines with a cocktail of antibodies reversed the inflammatory phenotype of D6(-/-) mice but led to less controlled growth of M. tuberculosis. Thus, the D6 decoy receptor plays a key role in setting the balance between antimicrobial resistance, immune activation, and inflammation in M. tuberculosis infection.
This content is subject to copyright.
The Journal of Experimental Medicine
ARTICLE
The Rockefeller University Press $30.00
J. Exp. Med. Vol. 205 No. 9 2075-2084
www.jem.org/cgi/doi/10.1084/jem.20070608
2075
The formation of granulomas at the site of my-
cobacterial infection is an essential component
of host immunity for controlling infection. This
process is dependent on the activation of myco-
bacteria-reactive T lymphocytes ( 1 ), particularly
IFN- secreting CD4 and CD8 T cells ( 2, 3 ).
Granuloma formation, however, is a complex
process that requires not only the activation of
lymphocytes, but also their recruitment with
monocytes to the site of the infection, migra-
tion into the tissues, and juxtaposition around
mycobacteria-infected macrophages ( 4 ). This
colocalization facilitates the activation of bacte-
ricidal mechanisms in infected macrophages by
T cell derived cytokines ( 1 ). Some mycobacte-
ria, however, survive within macrophages, and
persistent antigenic stimulation perpetuates the
process, leading to chronic granuloma forma-
tion characterized by dense accumulations of
infected macrophages, epithelioid cells, and T
lymphocytes ( 5 ). These granulomas contain the
mycobacterial infection and prevent dissemina-
tion to other organs, but they are also responsi-
ble for lung immunopathology, as the granulomas
displace and destroy parenchymal tissue ( 6 ). One
of the major roles of the granuloma is to local-
ize and contain not only the bacteria but also
the in ammatory response to the bacteria itself.
CORRESPONDENCE
Francesco Dieli:
dieli@unipa.it
Abbreviations used: ALT, alanine
aminotransferase; AST, aspartate
aminotransferase; BAL, bron-
choalveolar lavage; BCG, bacille
Calmette-Gu é rin; BUN, blood
urea nitrogen; i.n., intranasal;
MCP, monocyte chemotactic
protein; MIP, macrophage in-
ammatory protein; RANTES,
regulated on activation, normal
T cell expressed and secreted.
D. Di Liberto and M. Locati contributed equally to this paper.
The online version of this article contains supplemental material.
Role of the chemokine decoy receptor D6
in balancing in ammation, immune
activation, and antimicrobial resistance
in Mycobacterium tuberculosis infection
Diana Di Liberto ,
1 Massimo Locati ,
2,3 Nadia Caccamo ,
1
Annunciata Vecchi ,
2 Serena Meraviglia ,
1 Alfredo Salerno ,
1 Guido Sireci ,
1
Manuela Nebuloni ,
4 Neus Caceres ,
5 Pere-Joan Cardona ,
5 Francesco Dieli ,
1
and Alberto Mantovani
2,3
1 Dipartimento di Biopatologia e Metodologie Biomediche, Universit à di Palermo, 90134 Palermo, Italy
2 Istituto Clinico Humanitas IRCCS, 20089 Rozzano, Italy
3 Istituto di Patologia Generale and
4 Pathology Unit, L. Sacco Institute of Medical Sciences, University of Milan,
20133 Milan, Italy
5 Unitat de Tuberculosi Experimental, Department of Microbiology, Fundaci ó Institut per a la Investigaci ó en Ci è ncies de la
Salut Germans Trias i Pujol and Universitat Aut ò noma de Barcelona, 08916 Badalona, Spain
D6 is a decoy and scavenger receptor for in ammatory CC chemokines. D6-de cient mice
were rapidly killed by intranasal administration of low doses of
Mycobacterium tuberculosis
.
The death of D6
/
mice was associated with a dramatic local and systemic in ammatory
response with levels of
M. tuberculosis
colony-forming units similar to control D6-pro cient
mice. D6-de cient mice showed an increased numbers of mononuclear cells (macrophages,
dendritic cells, and CD4 and CD8 T lymphocytes) in ltrating in amed tissues and lymph
nodes, as well as abnormal increased concentrations of CC chemokines (CCL2, CCL3, CCL4,
and CCL5) and proin ammatory cytokines (tumor necrosis factor , interleukin 1 , and
interferon ) in bronchoalveolar lavage and serum. High levels of in ammatory cytokines in
D6
/
infected mice were associated with liver and kidney damage, resulting in both liver
and renal failure. Blocking in ammatory CC chemokines with a cocktail of antibodies re-
versed the in ammatory phenotype of D6
/
mice but led to less controlled growth of
M. tuberculosis
. Thus, the D6 decoy receptor plays a key role in setting the balance between
antimicrobial resistance, immune activation, and in ammation in
M. tuberculosis
infection.
© 2008 Di Liberto et al. This article is distributed under the terms of an Attribu-
tion–Noncommercial–Share Alike–No Mirror Sites license for the  rst six months
after the publication date (see http://www.jem.org/misc/terms.shtml). After six
months it is available under a Creative Commons License (Attribution–Noncom-
mercial–Share Alike 3.0 Unported license, as described at http://creativecommons
.org/licenses/by-nc-sa/3.0/).
2076 ROLE OF THE CHEMOKINE RECEPTOR D6 IN TUBERCULOSIS | Di Liberto et al.
and has suggested a role for D6 in controlling the resolution
of CC chemokine driven in ammatory responses. Accord-
ingly, D6
/
mice display exaggerated in ammatory responses
after skin application of phorbol esters ( 15 ) and subcutaneous
injection of complete Freund s adjuvant ( 16 ). In this last
model, D6
/
mice developed granulomas characterized by
angiogenesis, extensive leukocyte in ltration, and prominent
areas of necrosis more rapidly and severely than WT coun-
terparts. However, whether D6 plays a similar role in in am-
matory responses caused by infectious microorganisms, or
has a role in the induction of a protective response to infec-
tious agents, is presently unknown. To test this possibility,
we have studied the course of M. tuberculosis infection in
D6
/
mice.
RESULTS
Expression of the D6 receptor during
M. tuberculosis
infection
Preliminarily, we investigated the expression of the D6 receptor
during M. tuberculosis infection. In human lungs and lymph nodes
from a patient with pulmonary tuberculosis, D6 expression
was observed in lymphatic endothelial cells and only occasion-
ally in scattered leukocytes (Fig. S1, available at http://www
.jem.org/cgi/content/full/jem.20070608/DC1). In the mouse,
qualitative RT-PCR experiments indicated that D6 transcripts
in the liver, spleen, and lungs do not undergo signi cant
changes at 4, 8, and 12 wk after infection with M. tuberculosis
(Fig. S2). These data suggest that D6 is mainly expressed in
lymphatic endothelial cells, in agreement with previous re-
ports ( 17 ), and that its expression does not change appreciably
during M. tuberculosis infection.
M. tuberculosis
infection in D6
/
mice
To determine whether the D6 receptor plays a role in the
control of M. tuberculosis infection, WT and D6
/
mice
were infected via the intranasal (i.n.) route with 2,000 CFU.
This dose did not cause mortality in C57BL/6 mice in our
study ( Fig. 1 A ). However, M. tuberculosis infection in D6
/
mice resulted in increased mortality, with 20% of mice suc-
cumbing by week 8, 50% of mice succumbing by week 12,
and 100% of mice succumbing at week 16, at which time the
experiments were terminated ( Fig. 1 A ). The phenotype of
D6
/
was dramatic and was found highly reproducible over
a period of 3 yr in two additional experiments performed on
a total of 17 mice per group. Despite the exaggerated suscep-
tibility of D6
/
mice to M. tuberculosis infection, there was
no di erence between the two groups of mice in bacterial
loads, as determined by CFU counts in the lung, liver, and
spleen at 2, 4, 8, and 12 wk after infection ( Fig. 1 B ).
Of note, CFU data at 4, 8, and 12 wk after M. tuberculosis
infection were rechecked in two additional independent
experiments with three mice per group performed over a
period of 2 yr, con rming the results reported in Fig. 1 B .
We then examined by histology the e ect of D6 de ciency
on the in ammatory response in the lungs, liver, and spleen.
Although in WT mice a moderate cellular in ltration was
Indeed, if immune cells are not tightly controlled within the
lungs, this could lead to excess in ammation. Thus, rigorous
control of the organization of granulomas is likely necessary to
prevent immunopathology. In most cases, after the repair of
an in amed or damaged tissue, in ammation subsides and
the tissue returns to its homeostatic norm ( 7 ). However, if the
resolution phase of in ammation is ine ective, chronic in-
ammatory pathologies may develop ( 7, 8 ). Failure to resolve
ongoing in ammation is an invariable key feature of pathol-
ogies, which are typically characterized by the high-level
expression of in ammatory cytokines and chemokines ( 9 ).
Therefore, understanding the resolution phase of transient in-
ammatory responses will probably yield insights into some of
these chronic in ammatory pathologies.
TNF- and the related cytokine lymphotoxin- ( 10 13 )
are potent proin ammatory cytokines with a wide range
of activities in both the in ammatory and immune re-
sponses, and they play an essential role in host resistance
against infection with Mycobacterium tuberculosis and other
mycobacteria ( 10 12 ). TNF- de cient mice infected by
aerosol with M. tuberculosis develop normal T cell responses
to mycobacterial antigens, but because of the failure of gran-
uloma formation in the infected organs they are profoundly
susceptible to the infection, succumbing with extensive ne-
crosis in the lungs and infected organs ( 12 ). Other than
TNF- , the role of other soluble mediators in regulating
granuloma formation and persistence is poorly understood.
Chemokines and their receptors are involved in cell migra-
tion and are logical candidates for a role in granuloma for-
mation, although their expression has been studied to a limited
degree in M. tuberculosis infection (for review see reference
14 ). In general, the production of chemokines at a certain
level may be a factor in preventing cell movement out of
the granuloma.
Recently, studies of the D6 chemokine receptor (for re-
view see reference 9 ) have provided novel insights regarding
the mechanism of chemokine removal from in amed sites.
D6 is structurally similar to the other chemokine receptors,
and is most homologous to CCR4 and CCR5. However,
several properties set D6 apart from other CCRs. First, D6 is
extremely promiscuous, recognizing 15 chemokines. Inter-
estingly, all D6 ligands are in ammatory CC chemokines,
whereas constitutive CC chemokines, as well as chemokines
of other subfamilies, are not recognized. Thus, D6 is a pro-
miscuous receptor with selectivity for in ammatory CC che-
mokines. Second, the D6 expression pattern is unusual, with
the predominant expression sites being trophoblasts in the
placenta and lymphatic endothelial cells in the skin, gut, and
lung. Third, and most crucially, D6 does not signal in re-
sponse to the binding of any of its ligands. Conversely, the
only apparent function of D6 is to convey the entire ligand
receptor complex intracellularly, target the chemokine for
degradation, and recycle the receptor to the membrane. This
observation, together with the ability of D6 to bind most in-
ammatory CC chemokines, has resulted in the proposal that
D6 functions as a scavenging or decoy receptor ( 9, 15 ),
JEM VOL. 205, September 1, 2008
ARTICLE
2077
showed a di use liver necrosis in D6
/
mice when com-
pared with WT mice ( Fig. 1 C ). In agreement with the his-
tology results, at 12 wk after M. tuberculosis infection, serum
seen at 12 wk after infection and most of the lung airspace
remained intact, an exaggerated response was evident in sur-
viving D6
/
mice (see Fig. 5 A ). Histological analysis also
Figure 1.
M. tuberculosis
infection in D6
/
mice. (A) Survival curve of WT (closed squares) and D6
/
(open squares) mice infected i.n. with
2 × 10 3 CFU M. tuberculosis ( n = 10). Data indicate the percentage of surviving mice over time. The difference in the survival rates of WT and D6
/
mice was statistically signi cant (P < 0.001). (B) The course of M. tuberculosis infection in WT (closed squares) and D6
/
(open squares) mice in-
fected i.n. with 2 × 10
3 CFU M. tuberculosis and followed against time in the lungs, liver, and spleen. Data shown are the mean bacterial counts ± SD.
Differences between WT and D6
/
mice were not statistically signi cant at any time point. (C) Liver and kidney sections from WT and D6
/
mice at
12 wk after infection with M. tuberculosis . Hematoxylin and eosin staining is shown. Bars, 250 μ m. Insets show magni ed structures. Original magni-
cation was 100 × for the main images and 400 × for the insets. (D) Transaminase (ALT and AST) levels are shown. (E) BUN and proteinuria values are
shown. Similar results were obtained in  ve different experiments (shaded bars, WT mice; open bars, D6
/
mice). Differences between WT and D6
/
mice
were highly statistically signi cant (P < 0.001). Data are the mean ± the SD.
2078 ROLE OF THE CHEMOKINE RECEPTOR D6 IN TUBERCULOSIS | Di Liberto et al.
35% of CD8 T cells expressed the same activation markers in
WT mice, although di erences were not statistically signi -
cant (unpublished data). Similar  ndings were also detected
in the liver, although the di erences between D6
/
and WT
mice were lower than in the lungs and generally reached sig-
ni cance at 8 and 12 wk after infection ( Fig. 2 B ).
Cell migration to lymph nodes
Because of the  nding that the lungs of D6
/
mice con-
tained more macrophages and T lymphocytes than the lungs
of WT mice, we asked whether a greater number of these
cells migrated from the lungs to the draining lymph nodes.
To investigate this possibility,  ow cytometric analysis was
performed on the mediastinal lymph nodes of D6
/
and
WT mice after M. tuberculosis infection. As shown in Fig. 2 C ,
the number of macrophages showed a signi cant increase in
the lymph nodes of D6
/
mice at 4 wk after infection. At
4 and 8 but not 12 wk after infection, there were also statisti-
cally signi cant di erences in the total number of DCs in the
lymph nodes of D6
/
mice. The numbers of CD4 and CD8
T lymphocytes were signi cantly higher in the lymph nodes
of D6
/
mice at 4 and 8 wk after infection, but although the
latter constantly increased, the former peaked at week 4 and
then decreased. The increased migration of DCs and T lym-
phocytes to the lymph nodes was expected to result in increased
numbers of primed T lymphocytes, followed by migration to
the lungs. ELISPOT analysis con rmed this hypothesis, show-
ing that signi cantly more CD4 and CD8 T lymphocytes
from the lungs of D6
/
mice were primed to produce IFN-
when stimulated by bacille Calmette-Gu é rin (BCG) infected
DCs ( Fig. 3 ).
transaminases were abnormally increased in D6
/
compared
with WT mice ( Fig. 1 D ). Areas of focal necrosis and mono-
nuclear cell in ltrates were also evident in the kidneys ( Fig.
1 C ), and consistent with this, at 12 wk after M. tuberculosis
infection renal function was also severely compromised
in D6
/
mice, which had signi cantly higher blood urea
nitrogen (BUN) levels and frequency of severe proteinuria
as compared with WT mice ( Fig. 1 E ). Of note, although
M. tuberculosis growth occurred in the liver ( Fig. 1 B ), we
failed to detect M. tuberculosis CFU in kidneys of both WT
and D6
/
mice over a 12-wk period (not depicted). Thus,
despite the successful control of the growth of the infect-
ing mycobacteria, D6 de ciency led to increased mortality
associated with lung in ammation and combined renal and
liver failure.
Leukocyte in ltration in tissues during
M. tuberculosis
infection in D6
/
mice
We examined the cellular in ltrate in the lungs and liver of
D6
/
mice after M. tuberculosis infection. At predetermined
time points, organs were removed, the total number of cells
was calculated, and  ow cytometric analysis was performed to
determine cell populations ( Fig. 2 ). Numbers of DCs (F4/80
and CD11c
+ ) in the lungs ( Fig. 2 A ) were not signi cantly
di erent between D6
/
and WT mice. The numbers of
macrophages and CD4 and CD8 lymphocytes were signi -
cantly higher in D6
/
mice at all time points after infec-
tion. Approximately 45% of CD4 and CD8 T cells in ltrating
the lungs of M. tuberculosis infected D6
/
mice at 8 and
12 wk after infection expressed activation markers (CD25
high ,
CD44
high , and CD62L
low ), whereas only 30% of CD4 and
Figure 2. Leukocyte recruitment to tissues and lymph nodes after infection with
M. tuberculosis
. Leukocytes were isolated from the lungs
(A), livers (B), and mediastinal lymph nodes (C) of WT (closed squares) and D6
/
(open squares) mice and analyzed by  ow cytometry at different
times after infection with M. tuberculosis . The cells were stained with  uorescent antibodies to F4/80 (macrophages), CD11c (bright; DCs), CD4,
and CD8. Shown are the mean cell numbers ± SD for each cell type. Similar results were obtained in  ve different experiments. *, P < 0.001;
**, P < 0.01.
JEM VOL. 205, September 1, 2008
ARTICLE
2079
and CCL5/regulated on activation, normal T cell expressed
and secreted (RANTES) were also signi cantly increased in
the serum of D6
/
mice at 12 wk after infection ( Fig. 4 B ).
Control of M. tuberculosis infection depends on the activation
of macrophages by IFN- and TNF- , leading to the induc-
tion of inducible NO synthase and the production of reactive
nitrogen intermediates such as NO. TNF- is reported to be
essential for the early expression of mRNA encoding CC and
CXC chemokines, the initial recruitment of CD4 T lympho-
cytes, and the formation and maintenance of the granulomas
( 12, 19, 20 ). To determine whether these components of the
Chemokine and cytokine levels in D6
/
mice
Previous studies have shown that exacerbated in ammation in
mice lacking the D6 receptor is associated with higher levels
of chemokines in blood and tissues ( 15, 16, 18 ). We therefore
examined chemokine levels in the bronchoalveolar lavage
(BAL) and serum of M. tuberculosis infected mice. As shown
in Fig. 4 A , the concentrations of all tested chemokines were
signi cantly higher in the BAL of D6
/
than of WT mice at
12 wk after infection. Similarly, concentrations of CCL2/
monocyte chemotactic protein 1 (MCP-1), CCL3/macro-
phage in ammatory protein 1 (MIP-1 ), CCL4/MIP-1 ,
Figure 3. IFN- producing CD4 and CD8 T cells in the lungs after infection with
M. tuberculosis
. Lung leukocytes were isolated from WT
(closed squares) and D6
/
(open squares) mice at the indicated time points after infection and were restimulated in vitro with irradiated BCG-infected
WT DCs for 36 40 h at 37 ° C. The numbers of IFN- producing CD4 and CD8 T cells were quanti ed by ELISPOT at the indicated weeks. The mean
numbers ± SD of IFN- producing CD4 and CD8 T cells from WT and D6
/
mice at each time point are shown. Similar results were obtained in  ve
different experiments. *, P < 0.005; **, P < 0.01.
Figure 4. Chemokine and cytokine levels in the BAL and serum of
M. tuberculosis
infected WT and D6
/
mice. WT (shaded bars) and D6
/
(open bars) mice were infected i.n. with M. tuberculosis , and BAL or sera were obtained 12 wk later. The levels of CC chemokines (A and B) and cytokines
(C and D) were measured by ELISA. Shown are the mean values ± SD. Similar results were obtained in three different experiments. *, P < 0.001; **, P < 0.01.
2080 ROLE OF THE CHEMOKINE RECEPTOR D6 IN TUBERCULOSIS | Di Liberto et al.
ing the e cacy of the blocking. Conversely, D6
/
mice
treated with control antibody had very high levels of serum
chemokines, as did untreated D6
/
mice. TNF- , IL-1 ,
and IFN- levels in serum were also consistently reduced by
treatment with antibodies to CC chemokines (Table S1), and
cellular in ux to the infected tissues signi cantly decreased
(unpublished data). Additionally, both renal and liver func-
tion appeared improved by this treatment, as suggested by
the low levels of serum transaminases and BUN, and by the
low proteinuria detected in these mice (Table S1). Over-
all, the most impressive consequence of the neutralization
of CC chemokines in D6
/
mice was a signi cantly pro-
longed survival, as demonstrated by the only 10% mortality
at 12 wk and 30% mortality at 16 wk after infection with
M. tuberculosis ( Table I ). However, in spite of the ability of
chemokine-blocking antibodies to reduce the overall in am-
matory response ( Fig. 5 A ), the ability of D6
/
mice treated
in this manner to control the growth of M. tuberculosis in the
lungs was impaired, with higher CFU counts found at 8 and
12 wk ( Fig. 5 B ).
This result therefore indicates that the D6
receptor plays a crucial role in M. tuberculosis infection, and in
the balance between immunopathology and protective im-
mune responses.
DISCUSSION
A better understanding of the immunological mechanisms of
pathogenesis and protection is of essential importance for the
design of novel vaccines and immunotherapies against tuber-
culosis. It has been proposed that the protective response to
M. tuberculosis infection requires CD4 and CD8 lymphocytes,
the Th1-type cytokines IFN- and TNF- , and activated
macrophages (for review see reference 21 ). The cooperation
between the cells and cytokines requires close interaction,
which is achieved after migration and granuloma formation
in the lungs. The hallmark of infection in the lung is granuloma
formation, consisting of clusters of macrophages, lympho-
cytes, and DCs, which physically contains the mycobacteria
and creates a microenvironment for immune cell interaction,
limiting M. tuberculosis growth and dissemination. Chemo-
kines are potent leukocyte activators and chemoattractants
aiding granuloma formation and thought to be critical for the
immune response to M. tuberculosis ( 14 ). In vitro experiments
demonstrated that M. tuberculosis infection of bone marrow
derived mouse macrophages results in the expression of
TNF- , as well as several chemokines, including ligands for
the chemokine receptors CXCR3, CCR5, and CCR2. Neu-
tralization of TNF- by using antibody or TNFR1-de cient
macrophages demonstrated that expression of certain chemo-
kines (CXCL9/monokine induced by IFN- , CXCL10/
IFN-inducible protein 10, CXCL11/IFN-inducible T cell
chemoattractant, CCL5/RANTES, and CCL2/MCP-1) af-
ter M. tuberculosis infection was dependent, at least in part, on
TNF- . However, the lack of TNF- did not completely
abrogate chemokine expression, indicating that there are
other factors, induced as a result of infection, that stimulate
chemokine production ( 22 ).
antimycobacterial immune response were a ected by D6 de-
ciency, ELISA was performed on the BAL and serum of
M. tuberculosis infected mice at 8 (unpublished data) and 12 wk
after infection. Concentrations of TNF- , IL-1 , and IFN-
were signi cantly higher in the BAL ( Fig. 4 C ) and serum
( Fig. 4 D ) of M. tuberculosis infected D6
/
mice at all tested
time points, as compared with infected WT mice. Thus,
M. tuberculosis infection in the presence of D6 de ciency
causes a local and systemic in ammatory response charac-
terized by increased levels of proin ammatory chemokines
and cytokines.
Neutralization of chemokines in D6
/
mice in vivo reverses
pathology but increases susceptibility to infection
The results in the previous paragraph clearly show that
M. tuberculosis infection in the absence of the D6 receptor is
characterized by an exaggerated in ammatory response and
increased mortality, despite the successful control of bacterial
growth. To investigate whether the highly increased levels
of chemokines observed in D6
/
mice were involved in
the pathogenesis, we attempted to block in ammatory che-
mokines in vivo. For this purpose, mice were treated with a
mixture of antibodies to the CC chemokines CCL2/MCP-
1, CCL3/MIP-1 , CCL4/MIP-1 , and CCL5/RANTES,
as previously described ( 15, 18 ), or with antibodies to each
individual chemokine. On the same days, control mice re-
ceived i.p. injections of an equivalent amount of irrelevant
antibodies. Mice were injected weekly with M. tuberculosis ,
starting from the third week after infection. Following this
treatment schedule, CCL2/MCP-1, CCL3/MIP-1 , CCL4/
MIP-1 , and CCL5/RANTES production in the serum of
D6
/
mice was consistently reduced (Table S1, available at
http://www.jem.org/cgi/content/full/jem.20070608/DC1),
even below levels detected in WT mice, thus demonstrat-
Table I. Effects of treatment with anti CC chemokine
antibodies on the survival of D6
/
mice
to M. tuberculosis infection
Weeks after infection
Mice Treatment 8 12 16
WT Nil 0/10 0/10 0/10
D6
/
Nil 3/10 5/10 9/10
D6
/
Control antibody 2/10 4/10 9/10
D6
/
Anti-CCL2/MCP-1 3/10 3/10 8/10
D6
/
Anti-CCL3/MIP-1 4/10 6/10 9/10
D6
/
Anti-CCL4/MIP-1 2/10 4/10 8/10
D6
/
Anti-CCL5/RANTES 2/10 4/10 9/10
D6
/
Mix antibody 0/10* 1/10* 3/10**
WT mice and D6
/
mice either untreated or treated with monoclonal antibodies
to single individual CC chemokines, or with a cocktail of anti CC chemokines (mix
antibody) or irrelevant control antibodies, were infected with M. tuberculosis .
Mice were scored for survival over time. Data shown are the numbers of deaths
out of the number of treated mice ( n = 10 per group). *, P < 0.001; and **, P <
0.01 when compared with values in D6
/
mice either untreated (Nil) or treated
with control antibody.
JEM VOL. 205, September 1, 2008
ARTICLE
2081
The action of chemokines is tightly controlled by decoy
receptors ( 9 ), which internalize and target chemokines for
intracellular degradation, thus avoiding excessive in amma-
tory responses and the consequent tissue damage. D6 is a
decoy receptor that recognizes most in ammatory CC che-
mokines, implying that it may play a key role in resolving
CC chemokine driven in ammatory responses. The results
reported in this paper con rm this possibility in a mouse
model of M. tuberculosis infection through the mucosal (i.n.)
route and show that D6
/
mice have an exaggerated in am-
matory response leading to death, despite their ability to e -
ciently control bacterial load. Upon i.n. delivery of low-dose
M. tuberculosis , 20% of mice succumbed at week 8, 50% of
mice succumbed at week 12, and 100% of mice succumbed
at week 16, despite the successful control of the growth of
M. tuberculosis bacilli. The phenotype of D6
/
was dramatic and
was found highly reproducible over a period of 3 yr in two
additional experiments performed on a total of 17 mice per
group. Moreover, CFU data at 4, 8, and 12 wk after M. tu-
berculosis infection were found highly reproducible in two ad-
ditional independent experiments performed over a period of
2 yr. Mortality in D6
/
mice coincided with an overwhelm-
ing local and systemic in ammatory response that mainly
compromised liver and kidney functions in the host, charac-
terized by increased and sustained numbers of macrophages,
DCs, and CD4 and CD8 T lymphocytes, and increased
production of in ammatory CC chemokines and cytokines
(TNF- , IL-1 , and IFN- ). However, we failed to detect
M. tuberculosis CFU in kidneys of both WT and D6
/
mice
over a 12-wk period. This might re ect either M. tuberculosis
growth below the CFU detection limit or the development
of kidney damage in the absence of kidney infection. A simi-
lar increase in macrophage, DC, and T cell numbers was
detected in the mediastinal lymph nodes, suggesting the
possibility that more T lymphocytes were being primed and
migrating to the lungs. Such increased priming likely ac-
counts for the substantially increased numbers of T lympho-
cytes migrating to the lungs of D6
/
mice after M. tuberculosis
infection, as con rmed by ELISPOT analysis showing that
signi cantly more CD4 and CD8 T lymphocytes from the
lungs of D6
/
mice were primed to produce IFN- when
stimulated in vitro by BCG-infected DCs. The  nding that
D6
/
mice had more in ammation is intriguing and con-
vincingly supports an immunoregulatory role for the D6 re-
ceptor. Recent data in other systems have shown that D6
/
mice display exaggerated responses to in ammatory stimuli,
resulting in an aggressive cutaneous in ammatory pathology
after the application of phorbol esters ( 15 ). In another study
( 16 ), D6
/
mice displayed an exaggerated response to the
subcutaneous injection of complete Freund s adjuvant. Un-
fortunately, none of these studies has been able to de ne
a mechanism for the heightened immune response in the
D6
/
mice. Although there are several di erent possibilities
for the increased in ammatory response in M. tuberculosis
infected D6
/
mice, the one we favor is that lack of the D6
receptor fails to remove CC chemokines from many di erent
In mouse models, gene expression of CXC and CC che-
mokines has been detected in the lungs after M. tuberculosis
infection ( 22 24 ). CXCR3-de cient mice ( 25 ) have an im-
paired granuloma formation after aerosol infection with M.
tuberculosis , although this e ect is transient, occurring at the
early stages of infection. CCR2-de cient mice are extraordi-
narily susceptible to moderate- or high-dose M. tuberculosis
administered i.v. ( 26 ), and susceptibility is dose-dependent
( 27 ). Conversely, CCR5 ( 28 ) may not be essential to the de-
velopment of a protective response to M. tuberculosis infection.
CCL2/MCP-1 de cient mice did not demonstrate an in-
creased susceptibility to M. tuberculosis infection, but whether
cell in ltration or histology was a ected in these mice was not
reported ( 29, 30 ). However, transgenic mice overexpressing
CCL2/MCP-1 were more susceptible to tuberculosis ( 31 ).
Figure 5.
M. tuberculosis
growth in D6
/
mice treated with anti
CC chemokine antibodies. (A) WT mice and D6
/
mice either untreated
(Nil), or treated with a cocktail of anti CC chemokine (Anti-CC Ab) or irrel-
evant control antibodies (Control Ab), were infected with M. tuberculosis .
Granulomatous in ltration in the lungs of M. tuberculosis – infected mice
was assessed by analyzing two lung lobes from each mouse. Data are ex-
pressed as the percentage ± SD of the lung area involved, calculated by
dividing the granuloma-involved area by the total tissue area. A representa-
tive of two different experiments is shown. (B) The course of M. tuberculosis
infection followed against time in lungs. WT mice and D6
/
mice either
untreated (Nil), or treated with antibodies against individual chemokines,
with a cocktail of anti CC chemokine (Mix Ab), or with irrelevant control
antibodies (Control Ab), were infected with M. tuberculosis . Data shown are
the mean bacterial counts ± SD. *, P < 0.01 when compared with CFU val-
ues in D6
/
mice either untreated (Nil) or treated with control antibody.
2082 ROLE OF THE CHEMOKINE RECEPTOR D6 IN TUBERCULOSIS | Di Liberto et al.
in amed tissue, may favor resolution of chronic in amma-
tory responses and overall provide a  ne mechanism for the
control of the balance between protective immune responses
and immunopathology.
MATERIALS AND METHODS
Mice. D6
/
mice were generated as previously described ( 15 ). Homoge-
neous populations were established by backcrossing heterozygous mice to
C57BL/6 mice for more than eight generations. The resultant heterozygous
mice were bred to obtain homozygotes. WT C57BL/6J and D6
/
mice
were bred in a speci c pathogen-free/viral antibody free barrier facility and
obtained from Charles River Laboratories. 8 14-wk-old male and female
mice were used in accordance with institutional guidelines in compliance
with national (D.L. N.116, Gazzetta U ciale della Repubblica Italiana, sup-
plement 40, 18-2-1992) and international law and policies (European Eco-
nomic Community Council, 1987, Directive 86/609, O cial Journal of
European Communites L 358,1; and Institute of Laboratory Animal Re-
sources, Committee on Life Sciences, National Research Council, 1996,
Guide for the Care and Use of Laboratory Animals). Each experimental
group consisted of 7 10 mice. All infected mice were maintained under
germ-free conditions and were routinely monitored for mouse pathogens.
Experiments were performed in speci c pathogen-free facilities.
Chemicals and reagents. All chemicals were purchased from Sigma-
Aldrich, unless otherwise noted. Middlebrook 7H9 liquid medium and 7H10
agar were obtained from Difco Laboratories. Antibodies used in  ow cytom-
etry were obtained from BD Bioscience or R & D Systems.
M. tuberculosis and infection of mice. Mice were infected via the i.n.
route with M. tuberculosis H37Rv (2 × 10
3 CFU in 20 μ l). This resulted in
reproducible delivery of 50 100 viable CFU M. tuberculosis , as con rmed
by CFU determination on the lungs of infected mice 1 d after infection. In
fact, in a total of 10 mice per group from four independent experiments,
day 1 M. tuberculosis CFU ranged from 50 to 100 (with only one exception
in one WT mouse of 110 CFU), and in any case no statistical signi cant
di erences between day 1 CFU in WT (72.5 ± 21) and D6
/
(75 ± 19)
mice were detected (P = 0.9658 by the Student s t test). Moreover, using
the i.n. infection route, we failed to detect M. tuberculosis CFU in nasal tis-
sues in two di erent experiments. The tissue bacillary load was quanti ed
by plating serial dilutions of the lung, liver, and spleen homogenates into
7H10 agar, as described previously ( 34 ).
Flow cytometric analysis of tissue cells. To determine cellular in ltrate,
the lungs, liver, and mediastinal lymph nodes were removed at di erent
weeks after infection and digested in the presence of 200 U/ml of collage-
nase, and mononuclear cell suspensions were obtained through Lympholyte
M (Cederlane Laboratories) gradient centrifugation. The viability of cells, as
determined by Trypan blue exclusion, was > 90%. A single-cell suspension
was prepared by pushing the tissue through a cell strainer. In some experi-
ments, lung mononuclear cells were enriched in T cells by passage through
a nylon wool column, and CD4 and CD8 T cells were then sorted by anti-
CD4 or -CD8 immunomagnetic beads (Miltenyi Biotec), according to the
manufacturer s instructions ( 34 ). The cells were incubated for 24 h at 37 ° C
in complete medium to allow cells and beads to dissociate. Single-cell sus-
pension were counted. The samples were triple stained with  uorochrome-
conjugated anti-CD3, -CD4, -CD8, -CD44, -CD62L, -CD25, -CD11c,
and -F4/80, in FACS bu er (0.1% Na azide, 0.1% BSA, and 20% mouse se-
rum). Puri ed or PE-, FITC-, CyChrome-, or allophycocyanin-conjugated
isotype control Igs were used as controls. After washes, the cells were  xed
in 4% paraformaldehyde for 1 h and collected on a FACSCalibur (BD Bio-
sciences). Analysis was performed on CellQuest software (BD Biosciences).
ELISPOT analysis for IFN- . The ELISPOT method ( 35 ) was used to
detect IFN- secretion by individual CD4 or CD8 T cells from infected
organs and tissues, determining their increased concentration.
Such increased expression/circulation of CC chemokines
may, in turn, lead to signaling through their receptors and
could result in the in ux of immune cells and abundant, un-
controlled production of proin ammatory cytokines, which
ultimately mediate tissue damage. However, the increased
expression may also have been a reaction to the numbers of
cells in infected organs. Whatever the case, the concentra-
tions of CCL2/MCP-1, CCL3/MIP-1 , CCL4/MIP-1 ,
and CCL5/RANTES were found to be highly increased in
the BAL and serum of M. tuberculosis infected D6
/
mice
and sustained over time; moreover, blocking all four of them
in D6
/
mice in vivo (CCL2/MCP-1, CCL3/MIP-1 ,
CCL4/MIP-1 , and CCL5/RANTES) by repeated injec-
tion of a cocktail of neutralizing antibodies signi cantly re-
duced cell recruitment, production of proin ammatory CC
chemokines and cytokines, liver and kidney damage, and,
most importantly, the mortality of D6
/
mice. However,
despite controlling in ammatory responses, these mice be-
came more susceptible to infection with M. tuberculosis , as
demonstrated by higher CFU counts in the lungs at 8 and 12
wk after infection. The data reported in Table I clearly show
that none of the neutralized chemokines alone accounts for
the reduced in ammatory response and mortality, and for the
increased susceptibility to infection.
CCL3/MIP-1 , CCL4/MIP-1 , and CCL5/RANTES
are all ligands of CCR5, which can attract and activate mac-
rophages and Th1 lymphocytes. However studies in CCR5
/
mice have found that although CCR5 plays a role in the mi-
gration of DCs to and from lymph nodes ( 28, 32 ), it is not in-
dispensable for granuloma formation and immune protection
against M. tuberculosis infection ( 32 ). CCL5/RANTES has
been associated with the generation of type 1 cytokine pro-
ducing granulomas ( 33 ). CCR2-de cient mice are extraordi-
narily susceptible to moderate- or high-dose M. tuberculosis
administered i.v. but not to low aerosol doses ( 27 ), and CCL2/
MCP-1 de cient mice do not demonstrate an increased sus-
ceptibility to M. tuberculosis infection ( 29 ). However, there is
redundancy in the chemokine system. Speci c to this study, the
chemokines CCL2/MCP-1, CCL3/MIP-1 , CCL4/MIP-1 ,
and CCL5/RANTES are ligands of CCR5, but CCL3/MIP-
1 and CCL5/RANTES can also signal through CCR1 and
CCR3. This  nding makes it di cult to dissect the precise
roles of individual chemokines and their receptors. In this
study, enhancing the type 1 T cell response or the ability of
T lymphocytes to enter the lungs over the WT level does not
have an obvious bene cial e ect on the control of infection
and in ammation, indicating that there are additional factors
that must be induced or enhanced to increase the ability of the
host to eliminate M. tuberculosis infection.
In summary, our study suggests an important role for
the D6 receptor in immune regulation. D6 controls chemo-
kine accumulation and activity, migration, and possibly, the
maintenance of DCs and T cells in infected organs, as well
as the production of in ammatory cytokines. Therefore D6,
through its ability to avoid an excess of chemokines in the
JEM VOL. 205, September 1, 2008
ARTICLE
2083
determine the statistical signi cance of the di erences in survival times of
WT and D6
/
mice. The signi cance of di erences of log
10 CFU counts
between groups was determined using one-way analysis of variance
(ANOVA), with Sche e tests for post-ANOVA individual comparisons. P <
0.05 was considered signi cant.
Online supplemental material. Fig. S1 shows lung and draining lymph
nodes sections from M. tuberculosis patients stained for D6 and the macro-
phage marker CD68. Fig. S2 shows expression levels of the D6 transcript in
mouse tissues during M. tuberculosis infection. Table S1 reports the e ects
of treatment of D6
/
mice with anti CC chemokine antibodies. Online
supplemental material is available at http://www.jem.org/cgi/content/full/
jem.20070608/DC1.
This study was carried out with  nancial support from the European Union
FP6 contracts LSHP-CT-2003-503240 (MUVAPRED), LSHB-CT-2005-518167
(INNOCHEM), and LSHG-CT-2005-005203 (MUGEN); the CARIPLO Foundation
(NOBEL project); the Ministero dell Istruzione, Universit à e Ricerca (FIRB project);
and the Universities of Palermo and Milan (FIRST projects). This work was
conducted with the support of the Fondazione Humanitas per la Ricerca and Italian
Association for Cancer Research.
The authors have no con icting  nancial interests.
Submitted: 26 March 2007
Accepted: 3 July 2008
REFERENCES
1 . Flynn , J.L. , and J. Chan . 2001 . Immunology of tuberculosis. Annu. Rev.
Immunol. 19 : 93 129 .
2 . Cooper , A.M. , D.K. Dalton , T.A. Stewart , J.P. Gri n , D.G. Russell ,
and I.M. Orme . 1993 . Disseminated tuberculosis in interferon gene-
disrupted mice. J. Exp. Med. 178 : 2243 2247 .
3 . Feng , C.G. , A.G.D. Bean , H. Hooi , H. Briscoe , and W.J. Britton .
1999 . Increase in interferon-secreting CD8
+ , as well as CD4
+ , T cells
in lungs following aerosol infection with Mycobacterium tuberculosis. Infect.
Immun. 67 : 3242 3247 .
4 . Feng , C.G. , W.J. Britton , U. Palendira , N.L. Groat , H. Briscoe , and A.G.
Bean . 2000 . Up-regulation of VCAM-1 and di erential expansion of
integrin-expressing T lymphocytes are associated with immunity to pul-
monary Mycobacterium tuberculosis infection. J. Immunol. 164 : 4853 4860 .
5 . Orme , I.M. , and F.M. Collins . 1994 . Mouse model of tuberculosis. In
Tuberculosis: Pathogenesis, Protection and Control. B.B. Bloom, edi-
tor. American Society for Microbiology, Washington, DC. 113 134.
6 . Orme , I.M. 1998 . The immunopathogenesis of tuberculosis: a new
working hypothesis. Trends Microbiol. 6 : 94 97 .
7 . Nathan , C. 2002 . Points of control in in ammation. Nature . 420 : 846 852 .
8 . Mantovani , A. , R. Bonecchi , and M. Locati . 2006 . Tuning in amma-
tion and immunity by chemokine sequestration: decoys and more. Nat.
Rev. Immunol. 6 : 907 918 .
9 . Mantovani , A. 1999 . The chemokine system: redundancy for robust
outputs. Immunol. Today . 20 : 254 257 .
10 . Kindler , V. , A.P. Sappino , G.E. Grau , P.F. Piguet , and P. Vassalli . 1989 .
The inducing role of tumor necrosis factor in the development of bacte-
ricidal granulomas during BCG infection. Cell . 56 : 731 740 .
11 . Flynn , J.L. , M.M. Goldstein , J. Chan , K.J. Triebold , K. Pfe er , C.J.
Lowenstein , R. Schreiber , T.W. Mak , and B.R. Bloom . 1995 . Tumor
necrosis factor- is required in the protective immune response against
Mycobacterium tuberculosis in mice. Immunity . 2 : 561 572 .
12 . Bean , A.G.D. , D.R. Roach , H. Briscoe , M.P. France , H. Korner , J.D.
Sedgwick , and W.J. Britton . 1999 . Structural de ciencies in granuloma
formation in TNF gene-targeted mice underlie the heightened suscepti-
bility to aerosol Mycobacterium tuberculosis infection, which is not compen-
sated for by lymphotoxin. J. Immunol. 162 : 3504 3511 .
13 . Roach , D.R. , H. Briscoe , B. Saunders , M.P. France , S. Riminton , and
W.J. Britton . 2001 . Secreted lymphotoxin is essential for the control of
an intracellular bacterial infection. J. Exp. Med. 193 : 239 246 .
14 . Algood , H.M. , J. Chan , and J.L. Flynn . 2003 . Chemokines and tubercu-
losis. Cytokine Growth Factor Rev. 14 : 467 477 .
mice, after stimulation with BCG-infected DCs in vitro prepared as previ-
ously described ( 36 ). In brief, ELISPOT plates (BD Biosciences) were
coated with IFN- capture antibody overnight at 4 ° C. The capture anti-
body was discarded, and the plates were washed and blocked with com-
plete media for 2 h at room temperature. 150,000 puri ed CD4 or CD8 T
cells per well were cultured with irradiated BCG-infected WT DCs for
36 40 h at 37 ° C. The cells were discarded, and plates were washed with
deionized water and PBS/Tween 20. Secondary biotinylated antibody was
added for 2 h and incubated at room temperature, followed by washing
with PBS/Tween 20. Streptavidin-alkaline phosphatase was added to the
plates for 1 h, followed by washing and development of a color reaction
using the AEC substrate reagent kit (BD Biosciences). The reaction was
stopped when spots developed by running the plate under water. The spots
were enumerated using an ELISPOT reader (Bioreader 3000; BioSys).
ELISA for cytokines and chemokines. BAL was obtained by  ushing
2 ml PBS into the lungs of killed mice ( 37 ). The concentrations of chemo-
kines (CCL2/MCP-1, CCL3/MIP-1 , CCL4/MIP-1 , CCL5/RANTES,
CCL11/eotaxin, and CCL22/MDC) and cytokines (TNF- and IFN- )
were quanti ed by ELISA in BAL and sera using commercially available
kits (R & D Systems).
Determination of serum transaminases, BUN, and proteinuria.
Serum alanine aminotransferase (ALT; EC 2.6.1.2) and aspartate amino-
transferase (AST; EC 2.6.1.1) were measured by the standard photometric
method using an automatic analyzer (type 7350; Hitachi Chemical Diagnos-
tics) and a commercial kit (Sigma-Aldrich) adapted to small-sample volumes,
as previously described ( 38 ). The normal ranges of ALT and AST were ob-
tained testing sera from 10 mice, age- and sex-matched with animals used in
experimental groups ( 38 ). Proteinuria was measured on a 0 4+ scale using
a colorimetric assay strip ( 38 ) for albumin (Albustix; Bayer): 0, absent; 1+,
30 mg/100 ml (mild); 2+, 100 mg/100 ml (moderate); 3+, 300 mg/100 ml
(severe); and 4+, > 2,000 mg/100 ml (severe). BUN levels were measured by
impregnating Azostix strips (Bayer) with a drop of fresh blood ( 38 ) and using
the following scale: 1+, 5 15 mg/100 ml (normal); 2+, 15 26 mg/100 ml
(mild); and 3+, 30 mg/100 ml (severe).
Neutralization of chemokines in vivo. To block in ammatory chemo-
kines, mice were treated with a mixture of goat antibodies to the CC che-
mokines CCL2/MCP-1, CCL3/MIP-1 , and CCL4/MIP-1 ), and a
monoclonal antibody to CCL5/RANTES purchased lyophilized from R & D
Systems, resuspended in sterile PBS, and mixed as previously described ( 16,
18 ). Mice were given i.p. injections of 200 μ l of the mixture, equivalent to
100 μ g of each antibody. Alternatively, mice were treated with 400 μ
g of in-
dividual antibodies speci c for a single CC chemokine. On the same days,
control mice received i.p. injections equivalent to 400 μ g of irrelevant anti-
bodies (normal goat IgG; Sigma-Aldrich) in 200 μ l PBS. Mice were injected
weekly, starting from the third week after infection with M. tuberculosis .
Histology and morphometry. Tissue samples for histological studies
were  xed in 10% normal bu ered formalin, followed by para n embed-
ment. 5 6- μ m sections were stained with hematoxylin and eosin. For
histometry, 5- μ m-thick sections from each specimen were stained with
hematoxilin and eosin and photographed at 6 × using a stereoscopic zoom
microscope (SMZ800; Nikon) and a digital camera (Coolpix 990; Nikon).
Sections of at least eight lung lobes were studied in each case. A sequence of
appropriate software programs were used (Scion Image [Scion Corporation];
Photoshop 5.0 [Adobe Systems Inc.]) to determine the area of each single
lesion and the total tissue area on photomicrographs at each time point. Mea-
surements were done blindly by two independent pathologists (P.J. Cardona
and N. Caceres). Data are percentages calculated by dividing the granuloma-
involved area by the global tissue area and multiplying by 100.
Statistics. The double-tailed Student s t test was used to compare the sig-
ni cance of di erences between groups. Kaplan-Meier analysis was used to
2084 ROLE OF THE CHEMOKINE RECEPTOR D6 IN TUBERCULOSIS | Di Liberto et al.
15 . Jamieson , T. , D.N. Cook , R.J.B. Nibbs , A. Rot , C. Nixon , P. Mclean ,
A. Alcami , S.A. Lira , M. Wiekowski , and G.J. Graham . 2005 . The che-
mokine receptor D6 limits the in ammatory response in vivo. Nat.
Immunol. 6 : 403 411 .
16 . Martinez de la Torre , Y. , M. Locati , C. Buracchi , J. Dupor , D.N. Cook ,
R. Bonecchi , M. Nebuloni , D. Rukavina , L. Vago ,
A. Vecchi , et al .
2005 . Increased in ammation in mice de cient for the chemokine decoy
receptor D6. Eur. J. Immunol. 35 : 1342 1346 .
17 . Nibbs , R.J. , E. Kriehuber , P.D. Ponath , D. Parent , S. Qin , J.D. Campbell ,
A. Henderson , D. Kerjaschki , D. Maurer , G.J. Graham , and A. Rot .
2001 . The beta-chemokine receptor D6 is expressed by lymphatic endo-
thelium and a subset of vascular tumors. Am. J. Pathol. 158 : 867 877 .
18 . Martinez de la Torre , Y. , C. Buracchi , E.M. Borroni , J. Dupor , R.
Bonecchi , M. Nebuloni , F. Pasqualini , A. Doni , C. Agostinis , R. Bulla ,
et al . 2007 . Protection against in ammation- and autoantibody-caused
fetal loss by the chemokine decoy receptor D6. Proc. Natl. Acad. Sci.
USA . 104 : 2319 2324 .
19 . Adams , L.B. , C.M. Mason , J.K. Kolls , D. Scollar , J.L. Krahenbuhl , and
S. Nelson . 1995 . Exacerbation of acute and chronic murine tuberculosis
by administration of a tumor necrosis receptor-expressing adenovirus.
J. Infect. Dis. 171 : 400 405 .
20 . Algood ,
H.M. , P.L. Lin , and J.L. Flynn . 2005 . Tumor necrosis factor and
chemokine interactions in the formation and maintenance of granulomas
in tuberculosis. Clin. Infect. Dis. 41 : S189 S193 .
21 . Kaufmann , S.H.E. 2001 . How can immunology contribute to the con-
trol of tuberculosis? Nat. Rev. Immunol. 1 : 20 30 .
22 . Algood , H.M. , and J.L. Flynn . 2004 . TNF in uences chemokine
expression of macrophages in vitro and CD11b
+ cells in vivo during
Mycobacterium tuberculosis infection. J. Immunol. 172 : 6846 6857 .
23 . Sadek , M.I. , E. Sada , Z. Toossi , S.K. Schwander , and E.A. Rich . 1998 .
Chemokines induced by infection of mononuclear phagocytes with my-
cobacteria and present in lung alveoli during active pulmonary tubercu-
losis. Am. J. Respir. Cell Mol. Biol. 19 : 513 521 .
24 . Rhoades , E.R. , A.M. Cooper , and I.M. Orme . 1995 . Chemokine re-
sponse in mice infected with Mycobacterium tuberculosis. Infect. Immun.
63 : 3871 3877 .
25 . Seiler , P. , P. Aichele , S. Bandermann , A.E. Hauser , B. Lu , N.P. Gerard ,
C. Gerard , S. Ehlers , H.J. Mollenkopf , and S.H.E. Kaufmann . 2003 .
Early granuloma formation after aerosol Mycobacterium tuberculosis infec-
tion is regulated by neutrophils via CXCR3-signaling chemokines . Eur.
J. Immunol. 33 : 2676 2686 .
26 . Peters , W. , H.M. Algood , H.F. Chambers , J.L. Flynn , I.F. Charo , and J.D.
Erns . 2001 . Chemokine receptors 2 serve an early and essential role in resis-
tance to Mycobacterium tuberculosis . Proc. Natl. Acad. Sci. USA . 98 : 7958 7963 .
27 . Scott , H.M.
, and J.L. Flynn . 2002 . Mycobacterium tuberculosis in chemo-
kine receptor 2-de cient mice: in uence of dose on disease progression.
Infect. Immun. 70 : 5946 5954 .
28 . Algood , H.M. , and J.L. Flynn . 2004 . CCR5-de cient mice control
Mycobacterium tuberculosis infection despite increased pulmonary lympho-
cytic in ltration. J. Immunol. 173 : 3287 3296 .
29 . Lu , B. , B.J. Rutledge , L. Gu , J. Fiorillo , N.W. Lukacs , S.L. Kunkel , R.
North , C. Gerard , and B.J. Rollins . 1998 . Abnormalities in monocyte
recruitment and cytokine expression in monocyte chemoattractant pro-
tein 1-de cient mice.
J. Exp. Med. 187 : 601 608 .
30 . Kipnis , A. , R.J. Basaraba , I.M. Orme , and A.M. Cooper . 2003 . Role of
chemokine ligand 2 in the protective response to early murine pulmo-
nary tuberculosis. Immunology . 109 : 547 551 .
31 . Rutledge , B.J. , H. Rayburn , R. Rosenberg , R.J. North , R.P. Gladue ,
C.L. Corless , and B.J. Rollins . 1995 . High-level monocyte chemoat-
tractant protein-1 expression in transgenic mice increases their suscepti-
bility to intracellular pathogens. J. Immunol. 155 : 4838 4843 .
32 . Badewa , A.P. , L.J. Quinton , J.E. Shellito , and C.M. Mason . 2005 .
Chemokine receptor 5 and its ligands in the immune response to mu-
rine tuberculosis. Tuberculosis (Edinb.) . 85 : 185 195 .
33 . Chensue , S.W. , K.S. Warmington , E.J. Allenspach , B. Lu , C. Gerard ,
S.L. Kunkel , and N.W. Lukacs . 1999 . Di erential expression and cross-
regulatory function of RANTES during mycobacterial (type 1) and
schistosomal (type 2) antigen-elicited granulomatous in ammation.
J. Immunol. 163 : 165 173 .
34 . Dieli , F. , J. Ivanyi , P. Marsh , A. Williams , I. Naylor , G.
Sireci , N.
Caccamo , C. Di Sano , and A. Salerno . 2003 . Characterization of lung
T cells following intranasal infection with Mycobacterium bovis bacillus
Calmette-Guerin. J. Immunol. 170 : 463 469 .
35 . Lazarevic , V. , A.J. Myers , C.A. Scanga , and J.L. Flynn . 2003 . CD40,
but not CD40L, is required for the optimal priming of T cells and con-
trol of aerosol M. tuberculosis infection. Immunity . 19 : 823 835 .
36 . Dieli , F. , N. Caccamo , S. Meraviglia , J. Ivanyi , G. Sireci , C.T. Bonanno ,
V. Ferlazzo , C. La Mendola , and A. Salerno . 2004 . Reciprocal stimula-
tion of T cells and dendritic cells during the anti-mycobacterial im-
mune response. Eur. J. Immunol. 34 : 3227 3235 .
37 . Buccheri , S. , R. Reljic , N. Caccamo , J. Ivanyi , M. Singh , A. Salerno ,
and F. Dieli . 2007 . IL-4 depletion enhances host resistance and passive
IgA protection against tuberculosis infection in BALB/c mice. Eur. J.
Immunol. 37 : 729 737 .
38 . Sireci , G. , D. Russo , F. Dieli , S.A. Porcelli , M. Taniguchi , M.P. La
Manna , D. Di Liberto , F. Scarpa , and A. Salerno . 2007 . Immunoregulatory
role of J 281 T cells in aged mice developing lupus-like nephritis. Eur.
J. Immunol. 37 : 425 433 .
... ACKR2 was shown to limit inflammatory processes and tissue injury in several disease models, including Mycobacterium tuberculosis infection, skin inflammation, cardiac remodeling, and immune complex-mediated glomerulonephritis (12)(13)(14)(15). In contrast, in the context of autoimmune disease, ACKR2 expression on lymphatic endothelium reduces accumulation of inflammatory chemokines around lymphatic capillaries, which facilitates fluid flow and migration of activated dendritic cells (DCs) into regional lymph nodes to prime autoreactive T cells (16,17). ...
... Animal studies Ackr2-deficient (Ackr2-/-) mice on the C57BL/6J background were established as previously described (13,20). After generating Ackr2-deficient B6lpr/lpr (Ackr2-/-B6lpr) mice by crossing Ackr2deficient with Fas-mutated (lpr/lpr) C57BL/6J mice, the phenotype of female mice at 28 weeks of age was compared to WT B6lpr littermates. ...
Article
Full-text available
Introduction The atypical chemokine receptor 2 (ACKR2) is a chemokine scavenger receptor, which limits inflammation and organ damage in several experimental disease models including kidney diseases. However, potential roles of ACKR2 in reducing inflammation and tissue injury in autoimmune disorders like systemic lupus erythematosus (SLE) and lupus nephritis are unknown, as well as its effects on systemic autoimmunity. Methods To characterize functional roles of ACKR2 in SLE, genetic Ackr2 deficiency was introduced into lupus-prone C57BL/6lpr (Ackr2-/- B6lpr) mice. Results Upon inflammatory stimulation in vitro, secreted chemokine levels increased in Ackr2 deficient tubulointerstitial tissue but not glomeruli. Moreover, Ackr2 expression was induced in kidneys and lungs of female C57BL/6lpr mice developing SLE. However, female Ackr2-/- B6lpr mice at 28 weeks of age showed similar renal functional parameters as wildtype (WT)-B6lpr mice. Consistently, assessment of activity and chronicity indices for lupus nephritis revealed comparable renal injury. Interestingly, Ackr2-/- B6lpr mice showed significantly increased renal infiltrates of CD3+ T and B cells, but not neutrophils, macrophages or dendritic cells, with T cells predominantly accumulating in the tubulointerstitial compartment of Ackr2-/- B6lpr mice. In addition, histology demonstrated significantly increased peribronchial lung infiltrates of CD3+ T cells in Ackr2-/- B6lpr mice. Despite this, protein levels of pro-inflammatory chemokines and mRNA expression of inflammatory mediators were not different in kidneys and lungs of WT- and Ackr2-/- B6lpr mice. This data suggests compensatory mechanisms for sufficient chemokine clearance in Ackr2-deficient B6lpr mice in vivo. Analysis of systemic autoimmune responses revealed comparable levels of circulating lupus-associated autoantibodies and glomerular immunoglobulin deposition in the two genotypes. Interestingly, similar to kidney and lung CD4+ T cell numbers and activation were significantly increased in spleens of Ackr2-deficient B6lpr mice. In lymph nodes of Ackr2-/- B6lpr mice abundance of activated dendritic cells decreased, but CD4+ T cell numbers were comparable to WT. Moreover, increased plasma levels of CCL2 were present in Ackr2-/- B6lpr mice, which may facilitate T cell mobilization into spleens and peripheral organs. Discussion In summary, we show that ACKR2 prevents expansion of T cells and formation of tertiary lymphoid tissue, but is not essential to limit autoimmune tissue injury in lupus-prone B6lpr mice.
... These findings support recent observations that ACKR2 is mostly expressed in LECs and does not alter much during Mtb infection. Despite insignificant expression of ACKR2 during Mtb infection, it has been shown that Mtb infection increased mortality in Ackr2 -/mice, with 20% dying by week 8, 50% by week 12, and 100% by week 16 (153). In spite of their increased vulnerability to Mtb infection, Ackr2-deficient mice had similar bacterial burdens (Figure 4). ...
... In Ackr2-deficient mice, at all time points following infection, the population of macrophages, CD4 + and CD8 + lymphocytes, and other immune cells was considerably more significant in lungs and liver, leading to higher production of IL-1b, IFN-g, CCL2, CCL3, CCL5, and TNF-a in the bronchoalveolar lavage of infected mice. Therefore, the elevated levels of proinflammatory chemokines and cytokines induced by Mtb infection in ACKR2 deficiency lead to a local and systemic inflammatory response (153). Collectively, ACKR2 has been shown to regulate the production of inflammatory chemokines and cytokines, DC, and T-cell migration, and presumably their maintenance in infected tissues. ...
Article
Full-text available
Chemokines exert crucial roles in inducing immune responses through ligation to their canonical receptors. Besides these receptors, there are other atypical chemokine receptors (ACKR1–4) that can bind to a wide range of chemokines and carry out various functions in the body. ACKR2, due to its ability to bind various CC chemokines, has attracted much attention during the past few years. ACKR2 has been shown to be expressed in different cells, including trophoblasts, myeloid cells, and especially lymphoid endothelial cells. In terms of molecular functions, ACKR2 scavenges various inflammatory chemokines and affects inflammatory microenvironments. In the period of pregnancy and fetal development, ACKR2 plays a pivotal role in maintaining the fetus from inflammatory reactions and inhibiting subsequent abortion. In adults, ACKR2 is thought to be a resolving agent in the body because it scavenges chemokines. This leads to the alleviation of inflammation in different situations, including cardiovascular diseases, autoimmune diseases, neurological disorders, and infections. In cancer, ACKR2 exerts conflicting roles, either tumor-promoting or tumor-suppressing. On the one hand, ACKR2 inhibits the recruitment of tumor-promoting cells and suppresses tumor-promoting inflammation to blockade inflammatory responses that are favorable for tumor growth. In contrast, scavenging chemokines in the tumor microenvironment might lead to disruption in NK cell recruitment to the tumor microenvironment. Other than its involvement in diseases, analyzing the expression of ACKR2 in body fluids and tissues can be used as a biomarker for diseases. In conclusion, this review study has tried to shed more light on the various effects of ACKR2 on different inflammatory conditions.
... ACKR2 recognizes inflammatory CC and CXC chemokines and is distributed on lymphatic endothelial cells where it controls leukocyte influx into the lymph node [39]. It is expressed in mucosal sites (skin, lung, intestine) and leukocytes in humans and mice [32,39,45,47,48,[51][52][53][54][55]. Unlike ACKR1 and ACKR2, ACKR3 and ACKR4 are scavenger receptors of homeostatic CXC and CC chemokines [34,39,44,45,56]. ...
... ACKR1 expressed on erythrocytes scavenges a variety of chemokines, but also transcytoses chemokines across endothelial cells (3,5,6). ACKR2 internalizes and degrades multiple inflammatory chemokines of the CC family and plays a critical role in the dissolution of inflammation (7)(8)(9). ACKR4 binds the T-cell chemoattractants CCL19, CCL20, CCL21, CCL22, CCL25 (10)(11)(12). The third member, ACKR3, also known as CXCR7, binds and scavenges CXCL12, and with lower affinity, CXCL11 (13,14). ...
Article
Full-text available
Immune responses highly depend on the effective trafficking of immune cells into and within secondary lymphoid organs (SLOs). Atypical chemokine receptors (ACKRs) scavenge chemokines to eliminate them from the extracellular space, thereby generating gradients that guide leukocytes. In contrast to canonical chemokine receptors, ACKRs do not induce classical intracellular signaling that results in cell migration. Recently, the closest relative of ACKR3, GPR182, has been partially deorphanized as a potential novel ACKR. We confirm and extend previous studies by identifying further ligands that classify GPR182 as a broadly scavenging chemokine receptor. We validate the “atypical” nature of the receptor, wherein canonical G-protein-dependent intracellular signaling is not activated following ligand stimulation. However, β-arrestins are required for ligand-independent internalization and chemokine scavenging whereas the C-terminus is in part dispensable. In the absence of GPR182 in vivo , we observed elevated chemokine levels in the serum but also in SLO interstitium. We also reveal that CXCL13 and CCL28, which do not bind any other ACKR, are bound and efficiently scavenged by GPR182. Moreover, we found a cooperative relationship between GPR182 and ACKR3 in regulating serum CXCL12 levels, and between GPR182 and ACKR4 in controlling CCL20 levels. Furthermore, we unveil a new phenotype in GPR182-KO mice, in which we observed a reduced marginal zone (MZ), both in size and in cellularity, and thus in the T-independent antibody response. Taken together, we and others have unveiled a novel, broadly scavenging chemokine receptor, which we propose should be named ACKR5.
... The role of ACKR2 in the inflammatory response has been investigated in several animal models, including chemical-and bacterial-induced inflammation, autoimmune disease and alloimmune disease, but with limited exploration in viral infection (16,26,(29)(30)(31)(32). Contradictory findings have been reported in both experimental autoimmune encephalomyelitis and experimental colitis where opposing disease phenotypes were reported in mice with deletion of ACKR2 (29, 30, 33, 34). ...
Article
Full-text available
Herpes stromal keratitis (HSK) is a blinding corneal disease caused by herpes simplex virus-1 (HSV-1), a common pathogen infecting most of the world’s population. Inflammation in HSK is chemokine-dependent, particularly CXCL10 and less so the CC chemokines. The atypical chemokine receptor-2 (ACKR2) is a decoy receptor predominantly for pro-inflammatory CC chemokines, which regulates the inflammatory response by scavenging inflammatory chemokines thereby modulating leukocyte infiltration. Deletion of ACKR2 exacerbates and delays the resolution of the inflammatory response in most models. ACKR2 also regulates lymphangiogenesis and mammary duct development through the recruitment of tissue-remodeling macrophages. Here, we demonstrate a dose-dependent upregulation of ACKR2 during corneal HSV-1 infection. At an HSV inoculum dose of 5.4 x 10 ⁵ pfu, but not at higher dose, ACKR2 deficient mice showed prolonged clinical signs of HSK, increased infiltration of leukocytes and persistent corneal neovascularization. Viral clearance and T cell activation were similar in ACKR2 -/- and wild type mice, despite a transient diminished expression of CD40 and CD86 in dendritic cells. The data suggest that ACKR2 fine-tunes the inflammatory response and the level of neovascularization in the HSK.
... This concept is supported by observations that different inhibitory receptors also act as vital modulators of TB immune control. Mice lacking the chemokine scavenger D6 or the negative regulator of the IL-1 system, Toll/IL-1 receptor 8 (TIR8), were rapidly killed by low-dose M. tuberculosis challenges accompanied by considerable local and systemic inflammation [174,175]. Interestingly, both studies showed no differences in M. tuberculosis growth kinetics between KO and wild-type groups, suggesting that TB disease was driven by hyperinflammation not bacterial growth. In humans, classical epidemiological surveys showed that a strong tuberculin response in childhood is associated with an increased risk for TB disease later in life, implying disease associates with heightened immune responses [176]. ...
Article
Full-text available
By attenuating T cell activation, immune checkpoints (ICs) limit optimal anti-tumor responses, and immune checkpoint inhibition (ICI), has emerged as a highly effective new therapy for a broad range of cancers. However, boosting T cell immunity in cancer patients by blocking the PD-1/PDL-1 axis can trigger reactivation of latent tuberculosis (TB). This phenomenon appears to contradict the prevailing thought that enhancing T cell immunity to Mycobacterium tuberculosis (Mtb) will improve immune control of this pathogen. In support of this anecdotal human data, several murine studies have shown that PD-1 deficiency leads to severe TB disease and rapid death. These observations warrant a serious reconsideration of what constitutes effective TB immunity and how ICs contribute. Through restraining T cell responses, ICs are critical to preventing excessive tissue damage and maintaining a range of effector functions. Supporting this notion, inhibitory receptors limit pathology in respiratory infections such as influenza, where loss of negative immune regulation resulted in progressive immunopathology. In this review, we analyze the mechanisms of ICs in general and their role in TB in particular. We conclude with a reflection on the emerging paradigm and avenues for future research.
... The expression level of D6 is increased in lymphatic endothelium and intestinal leucocytes of IBD patients, with a much more elevated level in patients with IBD-associated CRC, implicating its significance in lymphatic system functions. Deficiency of D6 is associated with aggravated inflammation, CC chemokines accumulation, and leucocytic infiltration [44,45]. ...
Article
Full-text available
Inflammatory bowel disease (IBD) is characterized by intense immune dysregulation, gut microbiota imbalance, and intestinal epithelium destruction. Among the factors that contribute to the pathogenesis of IBD, lymphatics have received less attention, hence less studied, characterized, and explored. However, in recent years, the role of the lymphatic system in gastrointestinal pathophysiology continues to be highlighted. This paper examines the implications of lymphatic changes in IBD pathogenesis related to immune cells, gut microbiota, intestinal and mesenteric epithelial barrier integrity, and progression to colorectal cancer (CRC). Therapeutic targets of lymphatics in IBD studies are also presented. Available studies indicate that lymph nodes and other secondary lymphatic tissues, provide highly specialized microenvironments for mounting effective immune responses and that lymphatic integrity plays a significant role in small intestine homeostasis, where the lymphatic vasculature effectively controls tissue edema, leukocyte exit, bacterial antigen, and inflammatory chemokine clearance. In IBD, there are functional and morphological alterations in intestinal and mesenteric lymphatic vessels (more profoundly in Crohn's disease [CD] compared to ulcerative colitis [UC]), including lymphangiogenesis, lymphangiectasia, lymphadenopathy, and lymphatic vasculature blockade, affecting not only immunity but gut microbiota and epithelial barrier integrity. While increased lymphangiogenesis is primarily associated with a good prognosis of IBD, increased lymphangiectasia, lymphadenopathy, and lymphatic vessel occlusion correlate with poor prognosis. IBD therapies that target the lymphatic system seek to increase lymphangiogenesis via induction of lymphangiogenic factors and inhibition of its antagonists. The resultant increased lymphatic flow coupled with other anti-inflammatory activities restores gut homeostasis.
... Cell membranes and proteins of some organisms may act as receptors or primers that suppress or amplify AMR [263,264]. However, the existence and nature of such primers and receptors remain poorly understood in the case of AMR in insects, rodents, and pets. ...
Article
Full-text available
The present paper reviews the occurrence of antimicrobial resistance (AMR) in insects, rodents and pets. Insects (e.g., houseflies, cockroaches), rodents (rats, mice), and pets (dogs, cats) act as reservoirs of AMR for first-line and last-resort antimicrobial agents. AMR proliferates in insects, rodents, and pets, and their skin and gut systems. Subsequently, insects, rodents and pets act as vectors that disseminate AMR to humans via direct contact, human food contamination, and horizontal gene transfer. Thus, insects, rodents, and pets may act as sentinels or bioindicators of AMR. Human health risks are discussed, including those unique to low-income countries. Current evidence on human health risks is largely inferential and based on qualitative data, but comprehensive statistics based on quantitative microbial risk assessment (QMRA) are still lacking. Hence, tracing human health risks of AMR to insects, rodents, and pets remains a challenge. To safeguard human health, mitigation measures are proposed based on the one-health approach. Future research should include human health risk analysis using QMRA, and the application of in-silico techniques, genomics, network analysis, and machine learning to understand the role of household insects, rodents and pets in the persistence, circulation and health risks of AMR.
Article
Full-text available
Background/Aim: Atypical chemokine receptor (ACKR) family suppresses chemokine response and keeps the inflammatory state under control. This study investigated ACKR2 serum levels, which are thought to have an effect on the extreme inflammatory state in Crohn's disease (CD). Methods: Active and newly diagnosed Crohn's patients under treatment and a healthy control group were included in this prospective case-control study. Patients under the age of 18 years and those with Crohn's disease in remission were excluded. Clinical, demographic, laboratory parameters and serum ACKR2 levels of the patients were examined. Disease activity was evaluated using the simplified endoscopic score for Crohn’s disease (SES-CD) and Crohn’s disease activity index (CDAI) index. The relationship between disease activity and serum ACKR2 was evaluated using the Spearman correlation analysis. Results: A total of 119 subjects (66 CD patients and 53 healthy controls) were included in the study. Serum ACKR2 level was significantly lower in the CD group (4.80 ng/mL) compared to the control group (11.15 ng/mL) (P<0.001). In the correlation analysis between ACKR2 level and disease activity indicators, there was a weak positive correlation with SES-CD and CDAI (r = 0.350 P=0.004, r = 0.252, P=0.041, respectively). Conclusion: Our data show that the ACKR2 level in active CD is quite low compared to the control group. Despite the increase in disease activity, it is not upregulated at a sufficient level and may have adverse effects on the progression of the disease.
Article
Full-text available
Summary The expressionofprotective immunity toMycobacterium tuberculosisinmice ismediatedby T lymphocytesthatsecretecytokines .Thesemoleculesthenmediateavarietyofroles,including theactivation ofparasitized hostmacrophages,andtherecruitment ofothermononuclearphagocytes tothesiteoftheinfectioninordertoinitiate granulomaformation.Among thesecytokines, interferon y (IFN-y)isbelieved toplayakeyroleistheseevents .Inconfirmation ofthishypothesis, we show inthisstudythatmiceinwhich theIFN-y genehasbeendisruptedwereunableto containor controla normallysublethaldoseof M. tuberculosis,deliveredeitherintravenously oraerogenically .Insuchmice,aprogressive andwidespreadtissue destruction andnecrosis, associated withveryhighnumbersofacid-fast bacilli, wasobserved.Incontrast, despitethelackofprotective immunity,some DTH-like reactivity couldstill beelicited .Thesedata,therefore, indicatethat althoughIFN-y may notbe neededforDTH expression, itplaysapivotaland essential role inprotective cellular immunity totuberculosis infection .
Article
Full-text available
Understanding the immunological mechanisms of protection and pathogenesis in tuberculosis remains problematic. We have examined the extent to which tumor necrosis factor-alpha (TNF alpha) contributes to this disease using murine models in which the action of TNF alpha is inhibited. TNF alpha was neutralized in vivo by monoclonal antibody; in addition, a mouse strain with a disruption in the gene for the 55 kDa TNF receptor was used. The data from both models established that TNF alpha and the 55 kDa TNF receptor are essential for protection against tuberculosis in mice, and for reactive nitrogen production by macrophages early in infection. Granulomas were formed in equal numbers in control and experimental mice, but necrosis was observed only in mice deficient in TNF alpha or TNF receptor. TNF alpha and the 55 kDa TNF receptor are necessary conditions for protection against murine M. tuberculosis infection, but are not solely responsible for the tissue damage observed.
Article
Full-text available
We show here that infection of murine macrophages with various strains of Mycobacterium tuberculosis induces the rapid in vitro expression of genes encoding chemokines macrophage inflammatory protein 1 alpha and macrophage inflammatory protein 2, which recruit neutrophils to sites of infection, and macrophage-recruiting chemokines 10-kDa, interferon-inducible protein (IP-10) and macrophage chemotactic protein 1. Three strains of M. tuberculosis, Erdman and the clinical isolates CSU 22 and CSU 46, induced similar levels of secretion of macrophage chemotactic protein 1 from infected macrophage monolayers; however, the Erdman strain failed to induce levels of secretion of tumor necrosis factor alpha similar to those induced by either CSU 22 or CSU 46. Using a low-dose aerosol infection model, we also found that while the Erdman strain induced negligible increases in chemokine mRNA levels in the lungs, infection with either CSU 22 or CSU 46 resulted in greater levels of mRNA production for all four chemokines tested. The growth of these strains in the lungs was, however, equally well contained by acquired host immunity. These data allow us to hypothesize that the chemokine response in the lungs probably does not control the protective granulomatous response and that perhaps other T-cell- or macrophage-associated cytokines such as tumor necrosis factor alpha or interleukin 12 may be involved in this process.
Article
The expression of protective immunity to Mycobacterium tuberculosis in mice is mediated by T lymphocytes that secrete cytokines. These molecules then mediate a variety of roles, including the activation of parasitized host macrophages, and the recruitment of other mononuclear phagocytes to the site of the infection in order to initiate granuloma formation. Among these cytokines, interferon gamma (IFN-gamma) is believed to play a key role is these events. In confirmation of this hypothesis, we show in this study that mice in which the IFN-gamma gene has been disrupted were unable to contain or control a normally sublethal dose of M. tuberculosis, delivered either intravenously or aerogenically. In such mice, a progressive and widespread tissue destruction and necrosis, associated with very high numbers of acid-fast bacilli, was observed. In contrast, despite the lack of protective immunity, some DTH-like reactivity could still be elicited. These data, therefore, indicate that although IFN-gamma may not be needed for DTH expression, it plays a pivotal and essential role in protective cellular immunity to tuberculosis infection.
Article
The lymphatic vessels (lymphatics) play an important role in channeling fluid and leukocytes from the tissues to the secondary lymphoid organs. In addition to driving leukocyte egress from blood, chemokines have been suggested to contribute to leukocyte recirculation via the lymphatics. Previously, we have demonstrated that binding sites for several pro-inflammatory beta -chemokines are found on the endothelial cells (ECs) of lymphatics in human dermis, sere, using the MIP-1 alpha isoform MIP-1 alphaP, me have extended these studies to further support the contention that the in situ chemokine binding to afferent lymphatics exhibits specificity akin to that observed in vitro with the promiscuous beta -chemokine receptor D6. We have generated monoclonal antibodies to human D6 and showed D6 immunoreactivity on the ECs Lining afferent lymphatics, confirmed as such by staining serial skin sections with antibodies against podoplanin, a known lymphatic EC marker. In parallel, in situ hybridization on skin with antisense D6 probes demonstrated the expression of D6 mRNA by lymphatic ECs. D6-immunoreactive lymphatics were also abundant in mucosa and submucosa of small and large intestine and appendix, but not observed in several other organs tested. In lymph nodes, D6 immunoreactivity was present on the afferent lymphatics and also in subcapsular and medullary sinuses. Tonsilar lymphatic sinuses were also D6-positive. Peripheral blood cells and the ECs of blood vessels and high endothelial venules were consistently nonreactive with anti-D6 antibodies. Additionally, we have demonstrated that D6 immunoreactivity is detectable in some malignant vascular tumors suggesting they may be derived from, or phenotypically similar to, lymphatic ECs. This is the first demonstration of chemokine receptor expression by lymphatic ECs, and suggests that D6 may influence the chemokine-driven recirculation of leukocytes through the lymphatics and modify the putative chemokine effects on the development and growth of vascular tumors.
Article
Granuloma formation in the liver of mice infected with BCG coincides with local TNF synthesis. Injection of rabbit anti-TNF antibody, after 1 or 2 weeks of infection, dramatically interferes with the development of granulomas (both in number and size, large epithelioid cells failing to appear) and subsequent mycobacterial elimination. Furthermore, fully developed BCG granulomas, after 3 weeks of infection, rapidly regress after anti-TNF treatment. Antibody treatment also prevents or suppresses accumulation of TNF mRNA and protein, which resumes after disappearance of the antibody. Peritoneal macrophages exposed to TNF transiently accumulate TNF mRNA, and show an enhanced increase in TNF mRNA in response to gamma interferon. We propose that TNF released from macrophages in the microenvironment of developing granulomas is involved in a process of autoamplification: acting in an autocrine or paracrine way, it enhances its own synthesis and release, thus favoring further macrophage accumulation and differentiation leading to bacterial elimination.
Article
We have constructed transgenic mice in which the mouse mammary tumor virus long terminal repeat controls the expression of murine monocyte chemoattractant protein-1 (MCP-1). Several independently derived lines of transgenic mice constitutively expressed MCP-1 protein in a variety of organs. Protein extracts from these organs had substantial in vitro monocyte chemoattractant activity that was neutralized by an anti-MCP-1 Ab, indicating that transgenic MCP-1 protein is biologically active. However, no transgenic mouse at any age displayed monocyte infiltrates in MCP-1-expressing organs. Two transgenic lines had circulating MCP-1 levels of 13 to 26 ng/ml, which is a concentration sufficient to induce maximal monocyte chemotaxis in vitro. These transgenic lines showed a 1 to 1.5 log greater sensitivity to infection with Listeria monocytogenes and Mycobacterium tuberculosis. A third transgenic line had lower serum levels of MCP-1 and was resistant to L. monocytogenes. The results suggest that this transgenic model is one of monocyte nonresponsiveness to locally produced MCP-1 due to either receptor desensitization or neutralization of a chemoattractant gradient by high systemic concentrations of MCP-1. Regardless of the mechanism, the data indicate that constitutively high levels of MCP-1 expression do not induce monocytic infiltrates, and that MCP-1 is involved in the host response to intracellular pathogens.
Article
Tumor necrosis factor (TNF) plays a pivotal role in inflammatory phenomena that culminate in either pathogenesis or resistance in mycobacterial disease. The regulatory role of TNF in murine tuberculosis was examined by administering a recombinant adenovirus encoding a fusion protein consisting of the human 55-kDa TNF receptor extracellular domain and the mouse IgG heavy chain domain (AdTNFR). During acute infections with Mycobacterium tuberculosis, AdTNFR pretreatment induced elevated mycobacterial burdens of 1 log10 in the tissues of H37Ra-infected mice and 2 log10 (spleen and liver) and 4 log10 (lungs) in H37Rv-infected mice. In mice infected chronically with H37Rv, AdTNFR treatment induced a 3-log10 increase of M. tuberculosis in the lungs, in which a tuberculous bronchopneumonia developed with numerous acid-fast bacilli visible in alveoli and bronchi. Administration ofAdTNFR may serve as a useful model for studying the pathogenesis and chemotherapy of progressive primary tuberculosis.