Вы находитесь на странице: 1из 11

BBAMCB-57531; No.

of pages: 11; 4C: 3, 9


Biochimica et Biophysica Acta xxx (2013) xxxxxx

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta


journal homepage: www.elsevier.com/locate/bbalip

4
5
6
7
8
9
10

3Q1

Patrcia E. Almeida a,c, Natlia R. Roque a, Kelly G. Magalhes a, Katherine A. Mattos b, Livia Teixeira a,
Clarissa Maya-Monteiro a, Ceclia J. Almeida a, Hugo C. Castro-Faria-Neto a, Bernhard Ryffel d,e,
Valrie F.J. Quesniaux d,e, Patrcia T. Bozza a,

Differential TLR2 downstream signaling regulates lipid metabolism and


cytokine production triggered by Mycobacterium bovis BCG infection

Laboratrio de Imunofarmacologia, Instituto Oswaldo Cruz, Fundao Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
Laboratrio de Microbiologia Celular, Instituto Oswaldo Cruz, Fundao Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
c
Laboratrio de Biologia Celular, Universidade Federal de Juiz de Fora, Juiz de Fora, MG, Brazil
d
National Center for Scientic Research (CNRS), UMR7355, Orleans, France
e
Experimental and Molecular Immunology and Neurogenetics, University of Orleans, France
b

i n f o

a b s t r a c t

a r t i c l e

The nuclear receptor PPAR acts as a key modulator of lipid metabolism, inammation and pathogenesis in BCGinfected macrophages. However, the molecular mechanisms involved in PPAR expression and functions during
infection are not completely understood. Here, we investigate signaling pathways triggered by TLR2, the involvement of co-receptors and lipid rafts in the mechanism of PPAR expression, lipid body formation and cytokine
synthesis in macrophages during BCG infection. BCG induces NF-B activation and increased PPAR expression
in a TLR2-dependent manner. Furthermore, BCG-triggered increase of lipid body biogenesis was inhibited by
the PPAR antagonist GW9662, but not by the NF-B inhibitor JSH-23. In contrast, KC/CXCL1 production was
largely dependent on NF-B but not on PPAR. BCG infection induced increased expression of CD36 in
macrophages in vitro. Moreover, CD36 co-immunoprecipitates with TLR2 in BCG-infected macrophages, suggesting its interaction with TLR2 in BCG signaling. Pretreatment with CD36 neutralizing antibodies signicantly
inhibited PPAR expression, lipid body formation and PGE2 production induced by BCG. Involvement of CD36 in
lipid body formation was further conrmed by decreased BCG-induced lipid body formation in CD36 decient
macrophages. Similarly, CD14 and CD11b/CD18 blockage also inhibited BCG-induced lipid body formation,
whereas TNF- synthesis was not affected. Disruption of rafts recapitulates the latter result, inhibiting lipid
body formation, but not TNF- synthesis in BCG-infected macrophages. In conclusion, our results suggest that
CD36-TLR2 cooperation and signaling compartmentalization within rafts, divert host response signaling through
PPAR-dependent and NF-B-independent pathways, leading to increased macrophage lipid accumulation and
down-modulation of macrophage response.
2013 Published by Elsevier B.V.

Article history:
Received 29 April 2013
Received in revised form 4 September 2013
Accepted 1 October 2013
Available online xxxx

Keywords:
Nuclear receptor
TLR
Lipid droplet
CD36
PPARgamma

49

N
C
O

12
13
14
15
16
17
19
18
20
21
22
23
24
25
26

11

R
O

1. Introduction

51
52

Mycobacterium tuberculosis is a global health threat, infecting about


one third of the human population and alone is responsible for almost

48
50

Abbreviations: ADRP, adipose differentiation related protein; BCG, Bacillus Calmette


Gurin; IL, interleukin; NF-B, nuclear factor-B; M-CD, methyl--cyclodextrin; PAMP,
pathogen-associated molecular patterns; PGE2, prostaglandin E2; PPAR, peroxisome
proliferator-activated receptor gamma; TLR, Toll-like receptor
This work was funded by Conselho Nacional de Desenvolvimento Cientco e
Tecnolgico (CNPq/Brazil), PAPES-FIOCRUZ, PRONEX (MCT, Brazil), Fundao de Amparo
Pesquisa do Estado do Rio de Janeiro (FAPERJ, Brazil), French National Center for
Scientic Research (CNRS), CNRS/FIOCRUZ 2009 project No. 24201, and Guggenheim
Foundation.
Corresponding author at: Laboratorio de Imunofarmacologia, Instituto Oswaldo Cruz,
Fundao Oswaldo Cruz, Av. Brasil 4365, Manguinhos, 21045-900 Rio de Janeiro, RJ, Brazil.
E-mail address: pbozza@ioc.ocruz.br (P.T. Bozza).

27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
47
46

2 million deaths annually worldwide [1]. The ability of pathogenic


mycobacteria to adapt to the hostile environment of macrophage has
been instrumental in its success as a pathogen. Mycobacteria interfere
with host trafcking pathways by modulating events in endosomal/
phagosomal maturation pathway to create a protected niche for itself,
the mycobacterial phagosome [24]. Moreover, pathogenic mycobacteria may subvert host signaling response, through upregulation and
highjack of specic metabolic pathways, thus regulating host immune
response and enabling access to nutrients, which ultimately favor
infection establishment [510]. The foam-like macrophages are a cell
type found within granulomatous structures in both experimental
animal models and human disease [1113]. As observed for other
pathological conditions, the foam aspect of macrophages during mycobacteria infection is a reection of intracellular lipid accumulation.
Accumulated evidence demonstrated that newly formed lipid bodies
are structurally distinct cytoplasmic organelles involved in lipid mediator
synthesis with immunomodulatory functions during Mycobacterium

1388-1981/$ see front matter 2013 Published by Elsevier B.V.


http://dx.doi.org/10.1016/j.bbalip.2013.10.008

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

53
54
55
56
57
58
59
60
61
62
63
64
65
66
67
68
69
Q2

93
94
95
96
97
98
99
100
101
102
103
104
105
106
107
108
109
110
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
126
Q3
127
128
129
130
131
132
133
134
135

2.1. Animals

141

Mice were housed with free access to food and water at 22 to 24 C


and a 12 h light/dark cycle until used. Animals weighing 20 to 25 g
from both sexes were used. CD36 [29] and CD14 [39], provided by the
Transgenose Institute, CNRS, Orlans, France, and TLR2 [40], provided
by CECAL, FIOCRUZ, genetically decient mice in a homogeneous
C57BL/6 background were used. All protocols were approved by the
Institutional animal welfare committees.

142
143

2.2. M. bovis BCG and M. smegmatis

149

140

R
O

91
92

2. Materials and methods

138
139

144
145
146
147
148

M. bovis BCG (Moreau strain) dried vaccine (40 mg) was stored at
4 C and M. smegmatis (mc2155) were stored at 20 C. Both
mycobacteria strains were resuspended to a nal concentration of
1 106 colony-forming units (CFU)/mL in RPMI and sonicated just
before use.

150
151

2.3. Macrophage culture and in vitro infection

155

Peritoneal cells from nave C57/BL6 or TLR2/ mice were harvested


with sterile RPMI-1640 cell-culture medium. Peritoneal cells were
allowed to adhere for 2 h at 37 C in a 5% CO2 atmosphere, and washed
twice vigorously with phosphate buffer (PBS) to remove non-adherent
cells. Macrophages (1 106 cells/mL) were adhered in cover slides
within culture plates (24 wells) overnight with RPMI-1640 cellculture medium containing 2% FCS. Macrophages were infected with
BCG (MOI, 1:1) or M. smegmatis (MOI, 1:1) for 24 h at 37 C in 5% CO2
atmosphere. In inhibitory studies, macrophages were treated 30 min
before infection with: neutralizing CD36 monoclonal antibody (Cayman
Chemical Co.) or IgA isotype control (eBiosciences) (2 g/mL), antiCD14 (Cayman Chemical Co.), anti-CD11b/CD18 (eBioscience) or IgG
isotype control (10 g/mL), or the drugs JSH (Sigma) (30 M), lipin or
methyl--cyclodextrin (Sigma) (10 M) at 37 C. Then, macrophages
were infected with BCG for 1 h, and washed with PBS (three times)
to remove non-internalized BCG. Vehicle (DMSO 0.01%) was used
as control. The cell-free supernatants were recovered and stored at
20 C. Cell viability was assessed by trypan blue exclusion at the
end of each experiment and was always greater than 90%.

156

2.4. Pleurisy induced by M. bovis BCG

175

Mice were intrathoracically injected with M. bovis BCG (5 105


bacilli/cavity or 5 106 bacilli/cavity) in 100 L sterile saline. Control
animals received an equal volume (100 L) of sterile saline only. After
24h the animals were killed by CO2 inhalation and their thoracic cavities
were washed with 1 mL of the heparinized PBS (10 UI/mL).

176
177

2.5. Lipid body staining and counting

181

Macrophages were xed in 3.7% formaldehyde in PBS (pH7.4), rinsed


in 0.1 M cacodylate buffer (pH 7.4), stained in 1.5% osmium tetroxide
(30 min), rinsed in water, immersed in 1.0% thiocarbohydrazide
(5 min), rinsed in water, rinsed in 0.1 M cacodylate buffer, reincubated
in 1.5% osmium tetroxide (3 min), rinsed in distilled water, dried, and
mounted for further analysis. The morphology of xed cells was
observed, and lipid bodies were enumerated by light microscopy with
a 100 objective lens in 50 consecutive leukocytes in each slide. To
avoid human interference on this counting, we always ensured that

182
183

89
90

136
137

87
88

infections. We demonstrate here that, indeed, lipid body formation, PGE2


production and PPAR expression during M. bovis BCG infection require
the presence of CD36 together with TLR2, suggesting that cellular
activation is a result of synergic interactions among these receptors.

85
86

83
84

81
82

79
80

77
78

76

74
75

72
73

bovis BCG infection [5,14,15]. In addition, mycobacteria induction and


targeting to lipid body may provide an escape mechanism during
infection due to down-modulation of the macrophage response and/or
acquisition of nutrients, leading to enhanced survival and replication in
host cells [5,13,16,17]. However, the signaling pathway and molecular
mechanisms that regulate lipid body biogenesis during mycobacterial
infection and their contribution to the pathophysiology of tuberculosis
are not clear.
Toll-like receptors are germ-lined encoded receptors of the innate
immune system responsible for recognizing a range of pathogenassociated molecular patterns (PAMPs). Mycobacterial components
are recognized by different members of the TLR family including TLR2
in association with TLR1/TLR6, by TLR4 and by TLR9, and have been
implicated to mediate intracellular signaling in the host response to
bacterial infection [18,19]. TLR2 seems to be very promiscuous and
able to recognize the most diverse set of pathogen-associated motifs
[2022]. Its promiscuity has been attributed to its unique ability to
heterodimerize with TLRs 1 and 6, which direct ligand specicity
towards triacylated versus diacylated lipopeptides, respectively [23]. It
was thus suggested that combinatorial signaling among the TLRs
might account for the breadth of their repertoire [24]. However,
identication of co-receptors that enhances TLR functions suggests
that TLRs act together with other cell surface molecules to link PAMP
recognition to the initiation of inammatory responses. Indeed, recognition of TLR2 agonists is inuenced by several accessory receptors,
including CD14, CD36 [25] and CD11b/CD18 integrin [26]. The study
by Hoebe et al. [27] has shed light into TLR2 heterotypic associations
by demonstrating using forward-genetic mutagenesis that TLR2/TLR6
heterodimers also require CD36. In addition, heterotypic associations
of TLR2/6 with CD36 are not pre-formed and are ligand-induced [28].
CD36 is a transmembrane glycoprotein involved in many biological
processes, such as platelet biology, angiogenesis and in the pathophysiology of atherosclerosis and cardiovascular diseases dening it as
a multi-ligand scavenger receptor. CD36 performs homeostatic functions,
including recognition and phagocytosis of apoptotic cells, senescent cells,
cellular debris, and oxidized LDL [29,30]. Monocyte/macrophage CD36
has been shown to play a critical role in the development of atherosclerotic lesions by its capacity to bind and promote endocytosis of
oxidized low density lipoproteins (oxLDL) and to participate in the
formation of foam cells. In atherosclerosis and Alzheimer's disease,
deposition of the altered self-components oxLDL and amyloid-beta
trigger inammatory response through CD36-induced TLR4TLR6 heterodimer activation, suggesting a new model of TLR heterodimerization
triggered by co-receptor signaling events [31].
Recent studies have demonstrated that mycobacterial components
may regulate expression and function of nuclear receptors [8,3234].
PPAR is a member of the lipid activated nuclear receptor family and
has been demonstrated to function as a key transcriptional regulator
of cell differentiation, inammation and lipid metabolism in macrophages and dendritic cells [35]. Indeed, PPAR is highly expressed in
macrophage-derived foam cells within atherosclerotic lesions where it
plays an important role in lipid homeostasis and metabolism [3638].
Our group demonstrated that BCG-induced PPAR expression, lipid
body formation and TNF- generation were drastically inhibited
in TLR2 decient mice, suggesting a requisite role for TLR2 in BCG
mediated macrophage up-regulation of PPAR protein content [8].
Interestingly, activation of macrophages in vitro with the Mycobacterium
smegmatis failed to induce PPAR expression and lipid body formation,
while inducing TLR2-dependent TNF- production. Thus TLR2 activation, although essential for mycobacterial-induced lipid body formation,
is not sufcient to trigger pathways of lipid body formation and other coreceptors might be involved [8,34].
Since TLR activation seems to be associated with lipid-raft-resident
molecules, such as CD14 and CD36, we hypothesized that CD36 and
other co-receptors such as CD14 and CD11b/CD18 might cooperate
with TLRs, including TLR2, to activate macrophages during mycobacterial

70
71

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

152
153
154

157
158
159
160
161
162
163
164
165
166
Q4
167
168
169
170
171
172
173
174

178
179
180

184
185
186
187
188
189
190

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

2.6. NF-B and PPAR immunolocalization

194

The slides containing peritoneal macrophages were xed in 3%


formaldehyde at room temperature for 10 min, permeabilized with
0.2% Triton, and then blocked with 1% normal donkey serum. After
washing, slides were incubated overnight at 4 C with anti-NF-Bp65
polyclonal antibody or anti-PPAR polyclonal antibody (Santa Cruz)
diluted in 1% normal donkey serum/PBS solution. Non-immune rabbit
serum was used as control. After 3 washes of 5min, slides were incubated
for 1 h at room temperature with the secondary Alexa 546-labeled goat
anti-rabbit immunoglobulin G antibody (Molecular Probes). Incubation
with DAPI (Sigma) for 5 min was used for nuclei counterstaining.

211

209
210

Supernatants from in vitro M. bovis BCG-infected macrophages 212


after 24 h of infection were collected and stored at 20 C until the 213
day of analysis. Cytokines were analyzed simultaneously using Luminex 214

P
D
E
T
C
E

203

2.8. Cytokine analysis

201
202

207
208

199
200

PGE2 levels were measured directly in the supernatant of cultured


cells obtained 24 h after BCG infection. PGE2 was assayed in the cellfree supernatant by enzyme-linked immunoassay (EIA), according to
the manufacturer's instructions (Cayman Chemical Co.).

N
C
O

197
198

206

195
196

2.7. Prostaglandin E2 measurement

193

Immunostained samples were analyzed with a Confocal Laser Scanning 204


Microscope (CLSM 510; Zeiss).
205

the person responsible for counting was blinded to the codes for each
slide.

R
O

191
192

Fig. 1. M. bovis BCG and M. smegmatis induce differential response in macrophages. Mouse peritoneal macrophages obtained from C57BL/6 or TLR2 knockout mice (TLR2/) were infected
in vitro with BCG or M. smegmatis (MOI 1:1). Analysis of lipid body formation, KC synthesis, as well as, NF-B and PPAR expression and localization were performed at 24 h of infection.
(A) Lipid body counting after osmium staining. (B) KC synthesis was analyzed using Luminex technology. Each bar represents the mean SEM from 3 pools of 10 animals. Differences
between control and infected groups are indicated by asterisks (p b 0.05); + represents differences between TLR2+/+ and TLR2/ macrophages infected by BCG. (C) NF-B and
PPAR immunouorescence staining of TLR2+/+ or TLR2/ macrophages unstimulated or stimulated with LPS and non-infected or infected with BCG. DAPI (blue uorescence) was
used in all cell identication of the nuclei. The same magnication was used in all images detection with a 100 objective lens.

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

2.10. Immunoprecipitation

232
233

Briey, macrophages infected by M. bovis BCG 24 h were washed


twice with ice-cold PBS and scraped into 800 L of lysis buffer (10 mM
Tris, pH 7.5, 50 mM NaCl, 1% Triton X-100, 60 mM octylglucoside),
containing protease inhibitors. For pre-clearing, lysates were incubated
with pre-bound to Protein A Sepharose CL 48 (Amersham Bioscience).
After incubation for 4 h at 4 C, the beads were extensively washed
with TNET buffer (50 mM Tris pH 8.0, 150 mM, NaCl, 5 mM EDTA, 1%
Triton X-100) (six times). After that, pre-cleared lysates were incubated

242
243
244

248
249
250
251
252
253
254
255
256
257
258
259

260

The results were expressed as mean SEM and were analyzed 261
statistically by means of variance followed by NewmanKeulsStudent's 262
test or Student's t test with the level of signicance set at p b 0.05.
263

C
E
R
R

238
239

2.12. Statistical analysis

236
237

234
235

227
228

225
226

246
247

231

223
224

Cell lysates were prepared in reducing and denaturing conditions


and subjected to SDS-PAGE. Samples were submitted to electrophoresis
in 10% acrylamide gradient SDS-PAGE gels. After transfer onto nitrocellulose membranes, non-specic binding sites were blocked with 5%
non-fat milk in Tris buffered saline-Tween (TBST; 50 mM TrisHCl,
pH 7.4, 150 mM NaCl, 0.05% Tween 20). Membranes were probed
with the polyclonal antibodies, anti-CD36 and anti-PPAR (Cayman
Chemical Co.), anti-TLR2 (eBioscience Cd282) or anti--actin mAb
(BD Transduction Laboratories) in TBST with 1% non-fat dry milk.
Proteins of interest were then identied by incubating the membrane
with HRP-conjugated secondary antibodies in TBST, followed by detection of antigenantibody complexes by Supersignal Chemiluminescence
(Pierce). For densitometry analysis, images were analyzed in the
software Scion Image (International University Bremen).

229
230

Murine macrophages were detached with ice-cold 6 mM PBS-EDTA


and gentle scraping. Cells were washed in PBS in blocking buffer (2%
FBS in PBS). FITC-conjugated anti-CD36 antibody (Cayman Chemical
Co.) 0.5 mg/mL or with an irrelevant FITC-conjugated isotype (Santa
Cruz) for 1h at 4C. The cells were xed with 4% (w/v) paraformaldehyde,
and then uorescence measured at FL1 channel was detected by a
FACSCalibur ow cytometer (BD Biosciences, Heidelberg, Germany),
and at least 10,000 cells were analyzed/sample. Quantitative data
analysis was performed using WinMDI analysis software and expressed
as percent of CD36 positive cells.

245

R
O

221
222

2.11. Western-blot analysis

2.9. Flow cytometry analysis for CD36 expression

240
241

220

217
218

overnight at 4C with anti-CD36 polyclonal antibody (Cayman Chemical


Co.) or IgG rabbit control (R&DSystems) 2.5 g/mL. Finally, the beads
were resuspended in 3 sample buffer, separated by SDS-PAGE (10%)
and processed for immunoblotting. Each experiment was repeated at
least three times independently, with virtually identical results.

219

technology on the Bio-Plex system (Bio-Rad, Hercules, CA). Fifty L of


sample was analyzed using a mouse multiplex cytokine kit obtained
and assayed according to the manufacturer's instructions (Upstate,
Waltham, MA). Data analyses were performed with the Bio-Plex
Manager software.

215
216

Fig. 2. Inhibition of NF-B does not interfere with lipid body formation, although it is able to inhibit cytokine synthesis after infection by M. bovis BCG. Peritoneal macrophages were treated
with vehicle, JSH-23 (10 M or 30 M) or GW9662 (1 M) 30 min before infection with BCG (MOI 1:1). (A and B) Lipid body counting after osmium staining in peritoneal macrophages
treated with vehicle or JSH-23 in non-infected or BCG-infected during 3 h (A) or 24 h (B), (C) parallel enhancement of KC synthesis at 3 h was analyzed using Luminex technology, (D and E)
lipid body counting after osmium staining in peritoneal macrophages treated with vehicle or GW9662 non-infected or infected with BCG during 3 h (D) and 24 h (E); (F) TNF- synthesis
analysis by Luminex technology. Each bar represents the mean SEM from 3 pools of 10 animals. Differences between control and infected macrophages are indicated by asterisks
(p b 0.05). + represents differences between BCG and BCG in the presence of JSH-23 or GW 9662.

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

Lipid body formation in leukocytes is a highly regulated event that


depends on the interaction of cellular receptors with their ligands, and
newly formed lipid bodies have been associated with intracellular
pathogen survival advantages in HCV [41,42], dengue [43], Trypanosoma
cruzi [44] and mycobacterial infections [5,8,10,15,45]. The TLR-mediated
pathogen recognition and activation in the mechanism of lipid body
formation have been documented [5,46]. We recently demonstrated
that TLR2 activation, although essential, is not sufcient to trigger lipid
body formation during M. bovis BCG infection [8], suggesting the
involvement of cooperative signaling pathways. Here, we rst investigated the requirement of TLR2 and the involvement of NF-B and
PPAR activation in M. bovis BCG or M. smegmatis induced macrophage
activation. As shown in Fig. 1AB, M. bovis BCG infection triggers
both lipid body formation and CXCL1/KC production in macrophages
in a mechanism highly dependent on TLR2 activation. In contrast,
M. smegmatis was unable to induce lipid body biogenesis (Fig. 1A),
while effectively inducing CXCL1/KC in a TLR2-dependent way (Fig. 1B).
As seen in Fig. 1C, M. bovis BCG infection induced both NF-B nuclear
translocation and PPAR expression in wild-type macrophages, a
phenomenon that was drastically inhibited in TLR2/ macrophages.
Thus, differential signaling pathways are triggered by TLR2 in response
to mycobacteria.

285
286
287
288

283
284

281
282

279
280

277
278

275
276

273
274

N
C
O

271
Q5
272

269
270

267

A number of questions remain about how TLR2 and its coreceptors recognize the diverse set of physiological ligands with
TLR2 agonist activity and how they trigger differential intracellular
signaling pathways. We assessed the relative contribution of the
NF-B and the PPAR pathways in cytokine expression versus lipid
body formation by M. bovis BCG activated macrophages.
NF-B is a transcription factor with a central role in TLR-triggered
signaling pathways. It is composed of hetero- or homodimers of members of the Rel family of proteins, such as RelA (p65), RelB, cRel, p50
and p52 [47]. TLR ligands activate downstream signaling molecules
that include MyD88, IL-1 receptor-associated kinase, and tumor necrosis
factor (TNF) receptor-associated factor 6, which activates IB kinase
(IKK) complex [48,49]. IB degradation unmask the nuclear localization
signal of NF-B, allowing the transcription factor to translocate to the
nucleus, and then NF-B binds to the promoter region of immune and
inammatory genes for transcriptional regulation [50]. The aromatic
diamine 4-methyl-N1-(3-phenyl-propyl)-benzene-1,2-diamine (JSH23) compound inhibits nuclear translocation of NF-B p65 without
affecting IB degradation, [51]. It has been demonstrated that JSH-23
causes down-regulation of LPS-inducible cytokines and enzymes, and
inhibits LPS-induced apoptosis of the RAW 264.7 cells [51]. Here, we
used JSH-23 to determine whether inhibition of NF-B activation
would affect cytokine production and lipid body formation in BCGstimulated peritoneal macrophages. As shown in Fig. 2, although the

3.1. M. bovis BCG and M. smegmatis induce differential responses


in macrophages

R
O

265
266

268

3.2. Distinct transcriptionally regulated pathways are required for cytokine 289
and lipid metabolism control during infection by M. bovis BCG
290

3. Results

264

Fig. 3. M. bovis BCG induces CD36 expression and TLR2 heterodimerization. Murine peritoneal macrophages were cultured in the absence or presence of M. bovis BCG or M. smegmatis
(MOI 1:1) for 24 h. CD36 expression was determined by medium uorescence intensity of FITC (FL1) by ow cytometric analysis, (A) histogram representing macrophage
autouorescence control of non-labeled cells (white) or CD36 labeled cells as follows: non-stimulated cells (black), M. smegmatis-infected cells (light gray), and M. bovis BCG-infected
cells (dark gray). (B) Graph represents percentage of CD36 positivity in macrophages infected or not with M. smegmatis or M. bovis BCG. (C) TLR2 was detected by Western blot with
anti-TLR2 antibody after immunoprecipitation with anti-CD36 antibody as described under Materials and methods section. (D) Murine peritoneal macrophages were pre-treated with
vehicle or anti-CD36 antibody (2 g/mL) for 30 min. After pre-treatment, macrophages were infected or not with M. bovis BCG for 24 h. Total macrophage cell lysates were separated
by SDS-PAGE (10%) and submitted to Western blotting for anti-PPAR. The graph represents the densitometric analysis (arbitrary units (A.U.)) of the Western blotting bands. The
image is representative of at least two different blots.

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

291
292
293
294
295
296
297
298
299
300
301
302
303
304
305
306
307
308
309
310
311
312
313
314

324
325
326
327
328
329
330

3.3. CD36 expression interacts with TLR2 and enhances PPAR expression

332

Various molecules have been described as co-receptors acting in


conjunction with TLR2, such as CD36, CD14, TLR1 and TLR6. Considering
the role of CD36 in lipid metabolism, macrophage differentiation and
pathogen recognition, we investigated the role of CD36 in lipid body
formation in mouse peritoneal macrophages infected by BCG. As shown
in Fig. 3A and B, M. bovis BCG, but not M. smegmatis infection induces
an increase in CD36 protein expression in macrophages. Increased
CD36 protein content was noted within 90min, and was maximal within
3h, occurring in parallel but preceding the maximal lipid body formation
that is noted within 24 h (data not shown). Several studies in the
literature show that TLR2 and CD36 dimerize upon various stimuli,
such as lipoproteins, lipotechoic acid or diacylglycerides [27,54,55].
Here we demonstrated that CD36 and TLR2 co-immunoprecipitate in

349
350
351
352

355
356
357
358
359
360
361
362
363
364
365
366
367
368
369

In addition to CD36, recognition of TLR2 agonists is inuenced by 372


several accessory receptors, including CD14 and CD11b/CD18 integrin, 373

C
E
R

343
344

347
348

3.5. CD14 and CD11b/CD18 contribute to lipid body formation, but not to 370
TNF- expression in response to M. bovis BCG infection
371

341
342

339
340

337
338

335
336

333
334

To investigate the functional role of CD36 activation during M. bovis


BCG infection, we used a neutralizing antibody for this receptor. As
shown in Fig. 4, addition of anti-CD36 antibody signicantly inhibited
lipid body formation during BCG infection (Fig. 4A), while the isotype
control IgA had no effect. The role of CD36 in lipid metabolism regulation during BCG infection was conrmed in CD36 genetically
decient animals. As shown in Fig. 4, BCG-induced lipid body formation
was signicantly decreased when cells from CD36 decient animals
were compared to cells from wild-type animals, both in vitro (Fig. 4B)
and in vivo (Fig. 4C). PGE2 production was also inhibited by pretreatment with the neutralizing antibody anti-CD36 (Fig. 4D), although
TNF- and IL-1 synthesis remained unaltered (Fig. 4E and F), during
BCG infection at 24 h in vitro. These results indicate that CD36 has a
crucial role in signaling lipid body biogenesis and prostanoid production
by M. bovis BCG infection.

331

345
346

3.4. CD36 is required for lipid body formation and PGE2 production, but not 353
cytokine synthesis during BCG infection
354

322
323

321

R
O

319
320

macrophages 24 h after infection by M. bovis BCG (Fig. 3C lane 4),


which is not observed in non-infected cells (Fig. 3C lane 1).
Next, we evaluated whether CD36 blockade affects PPAR expression.
As shown in Fig. 3D, PPAR expression was partially inhibited after CD36
neutralization (Fig. 3D lane 4). In summary, these results suggest that
CD36 activation favors PPAR expression, while CD36 expression may
be amplied by PPAR in a positive feedback loop during M. bovis BCG
infection.

317
318

pretreatment with JSH-23 (30 M) signicantly inhibited the production of CXCL1/KC (Fig. 2C), it failed to inhibit lipid body formation
(Fig. 2AB) in peritoneal macrophages at 3 and 24 h after infection.
Recent studies have shown that PPAR activation inhibits NF-B
and MAP kinase pathways, two major signaling pathways that regulate
pro-inammatory responses triggered by TLR activation [52,53]. We
next investigated the role of PPAR on M. bovis BCG-induced lipid
body formation and TNF- production in macrophages. As shown in
Fig. 2DF, M. bovis BCG infection signicantly increased lipid body
formation and synthesis of TNF-. Pretreatment with PPAR antagonist
GW9662 was able to inhibit lipid body formation at 3h and 24h (Fig. 2D
and E, respectively), although it failed to modify BCG-induced TNF-
production by macrophages (Fig. 2F). These results suggest that TLR2
activation by M. bovis BCG triggers differential transcriptional regulated
NF-B and the PPAR pathways, leading to cytokine production and
lipid body formation, respectively.

315
316

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

Fig. 4. Lipid body formation, PGE2 production, but not cytokine synthesis is dependent of CD36 expression during M. bovis BCG infection. Peritoneal macrophages were treated with vehicle
or anti-CD36 antibody (2 g/mL) for 30 min before infection with BCG (MOI 1:1) during 24 h (A, D, E and F). (A, B and C) Lipid body counting was performed after osmium staining (A) in
peritoneal macrophages infected or not with BCG and treated or not with anti-CD36, (B) in peritoneal macrophages from CD36+/+ or CD36/ mice in vitro, and (C) in total cells from the
intrathoracic cavity of CD36+/+ or CD36/ mice in vivo. (D) PGE2 production was measured in medium of macrophage cultures by an enzyme immunoassay. (E) TNF- and (F) IL-1
cytokine in medium of macrophage cultures were analyzed using Luminex technology. Each bar represents the mean SEM from 3 pools of cells from 10 animals each. Differences
between control and infected groups are indicated by asterisks (p b 0.05). + represents differences between BCG and BCG in the presence of anti-CD36. # represents differences between
CD36+/+ and CD36/ infected by BCG.

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

384
385
386
387

3.6. Raft disruption inhibits both lipid body formation and PGE2, but not
TNF- release induced by M. bovis BCG

399

4. Discussion

400

Mycobacteria-induced manipulation of host cell lipid metabolism is


now recognized as an important event for persistence and successful
establishment of infection in tuberculosis and leprosy. We and others,
have recently established a key role for PPAR in the regulation of
lipid metabolism, lipid body formation and immune response in mycobacterium infection [8,3234]. However, the mechanisms that regulate
PPAR expression and activation are not well understood. Our study
provides novel evidence that CD36-TLR2 cooperation, association with
CD11b and CD14, and signaling compartmentalization within rafts,
divert host response signaling with increased expression and activation
of PPAR through NF-B-independent pathways, leading to increased
macrophage lipid accumulation and down-modulation of macrophage
response.
The signaling cascade initiated by activation of the TLRs results in
translocation of the transcription factor NF-B, which subsequently
induces the expression of cytokines. TLR2 signaling was also demonstrated as an essential step for PPAR expression and macrophage
lipid accumulation following M. bovis BCG infection [8]. To clarify the
downstream mechanism by which TLR2 triggers increased PPARdependent lipid metabolism alterations, we evaluated the role of
NF-B activation in this phenomenon. Notably, JSH-23, a selective
NF-B transcriptional inhibitor, at doses that signicantly inhibited
KC/CXCL-1 production failed to inhibit BCG-induced lipid body formation. Indeed, NF-B inhibition by JSH-23 within 3 h leads to enhanced
lipid body formation induced by BCG, suggesting NF-B as a negative
regulator of lipid body formation. The mechanism and pathways involved in this negative regulation deserve further investigation.
In contrast the PPAR antagonist GW 9662 was able to inhibit BCGinduced lipid body formation, but not TNF- release. Together, these
results suggest that distinct signaling pathways downstream of TLR2
are involved in cytokine expression and lipid metabolism regulation.
Accordingly, macrophage activation in vitro with M. smegmatis, or
Pam3CSK4 (not shown), both potent TLR2 ligands, at doses that signicantly induced TLR2-dependent CXCL1 production, were unable to
induce lipid body biogenesis in macrophages, suggesting that TLR2

403
404
405
406
407
408
409
410
411
412
413
414
415
416
417
418
419
420
421
422
423
424
425
426
427
428
429
430
431
432
433
434

401
402

395
396

394

392
393

N
C
O

390
391

388
389

397
398

Localization of TLR2 and co-receptors may be important for function. It has been demonstrated in several model systems that CD36,
CD14 and CD11b/CD18 may compartmentalize with TLR2 in rafts.
Thus, we next evaluated the involvement of rafts in BCG infection.
As shown in Fig. 6, drugs that disrupt rafts, including methyl-cyclodextrin and lipin, signicantly inhibited lipid body formation
(Fig. 6A and B). Treatment with methyl--cyclodextrin also inhibited
BCG-induced PGE2 production (Fig. 6C), although TNF- release remained unaltered (Fig. 6D).
In summary, these results indicate that TLR2 cooperates with CD36,
CD14 and CD11b/CD18 to activate lipid body production, prostanoid
and cytokine synthesis, engaging distinct signaling pathways, which
involve NF-B and PPAR.

383

381
382

R
O

380

378
379

376
377

alone is not sufcient and that TLR2 associated co-receptors involved 435
in bacterial recognition may account for the recruitment of signaling 436
molecules important for PPAR expression and lipid body formation [8]. 437

which act in concert to modulate TLR activity [18,21]. Thus, we studied


the participation of CD14 and integrins, as putative co-receptors of TLR2
in cell signaling triggered by mycobacterial infection.
As shown in Fig. 5A, lipid body formation in macrophages from CD14
genetically decient animals is signicantly diminished compared to
macrophages from wild type mice. Accordingly, the neutralization of
CD11b/CD18 or CD14 inhibits lipid body formation (Fig. 5B) and PGE2
production (data not shown), but not TNF- synthesis (Fig. 5C).
Therefore, CD14 and CD11b/CD18 are also engaged in BCG-infected
macrophages leading to lipid body formation and PGE2 synthesis.

374
375

Fig. 5. Involvement of CD14 and CD11b/CD18 in lipid body formation, but not TNF-
synthesis during M. bovis BCG infection. (A) Lipid body counting of peritoneal macrophage
obtained from wild type (CD14+/+) or CD14 decient (CD14/) were treated in vitro
with vehicle or BCG for 24 h. (BC) Peritoneal macrophages from wild type mice were treated
with vehicle, anti-CD14 (10 M) or anti-CD11b/CD18 (10 M) for 30 min before infection
with BCG for 24 h. (B) Lipid body counting was performed after osmium staining, (C) TNF in the media of macrophage cultures was analyzed using Luminex technology. Each bar
represents the mean SEM from 3 independent pools of 10 animals each. Differences
between control and infected groups are indicated by asterisks (p b 0.05). + represent
differences between CD14+/+ and CD14/ macrophages infected with BCG. # represents
differences between BCG and BCG in the presence of anti-CD14 or anti-CD11b/CD18.

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

R
O

448
449
450
451
452
453
454
455
456
457
458
459
460
461
462
463
464
465
466
467
468

446
447

444
445

442
443

440
441

It is increasingly recognized that TLR2-dependent signaling is modulated by the concomitant interaction with co-receptors and depending
on the co-receptor usage, distinct downstream signaling pathways
might be recruited, leading to differential signaling compartmentalization and responses [28]. Several co-receptors for TLR2 have been
characterized, which may contribute to the diversity of ligands able to
trigger TLR2-dependent responses. Heterodimerization of TLR2 with
other members of the TLR family forming the TLR2-TLR1 and TLR2TLR6 heterodimers have been well characterized to regulate signaling,
including cytokine production [25]. TLR6 was demonstrated to be
dispensable for regulating macrophage lipid metabolism upon BCG
infection [8]. However, TLR6 participates in the metabolic response
towards M. leprae in Schwann cells [46], suggesting that different coreceptors involved in lipid metabolism signaling may be recruited
according to the pathogen and cell system.
Next, we addressed the involvement of CD36, in cooperation with
TLR2, to modulate the BCG effects on host lipid metabolism. Monocyte/
macrophage CD36 has been shown to play a critical role in the
development of atherosclerotic lesions by its capacity to bind and
promote endocytosis of oxLDL [56]. It has also been implicated in the
formation of foam cells [57]. CD36 was demonstrated to cooperate
with TLR2 in sensing bacteria and bacterial ligands, acting as a coreceptor for the induction of mycobacterial uptake and cytokine
generation [28]. We showed that BCG triggered increased expression
of CD36. BCG-induced PPAR expression, lipid body formation and
PGE2 formation, but not cytokine production, were signicantly inhibited by CD36 neutralizing antibodies. The involvement of CD36 was
further conrmed by the impairment of BCG-induced lipid body
formation in vitro and in vivo in macrophages from CD36 decient
animals, suggesting that host cell lipid metabolism alteration induced
by infection is a result of synergic interactions between CD36 and

438
439

Fig. 6. Raft disruption inhibits both lipid body formation and PGE2 production, but not TNF- synthesis induced by M. bovis BCG. Peritoneal macrophages were treated with vehicle, lipin
(10 M) (A) or methyl--cyclodextrin (Sigma) (10 M) (B) for 30 min, and then infected with BCG (MOI 1:1) for 24 h. Lipid body counting in peritoneal macrophages with different
treatments after osmium staining (A and B). (C) PGE2 production and (D) TNF- synthesis in peritoneal macrophages infected or not with BCG and treated with vehicle or methyl-cyclodextrin a. Each bar represents the mean SEM from 3 independent pools of 10 animals each. Differences between control and infected groups are indicated by asterisks
(p b 0.05). + represents differences between BCG and BCG in the presence of lipin, # represents differences between BCG and BCG in the presence of methyl--cyclodextrin.

TLR2. Indeed, impairment of either TLR2 or CD36 inhibited BCGinduced PPAR expression and lipid droplet formation in macrophages.
Accordingly, the co-immunoprecipitation of CD36 and TLR2 in cells
activated with M. bovis BCG infection in vitro suggests that heterodimerization occurred between TLR2 and CD36 and signaling downstream of TLR2/CD36 trigger pathways of increased PPAR expression
and lipid metabolism regulation in macrophages during mycobacterial
infection. Corroborating with the observations that increased PPAR
expression and lipid body biogenesis is associated with detrimental
ability of host cells to control mycobacteria growth acting as an escape
mechanism of intracellular pathogens, Hawkes et al, [58] demonstrated
that CD36 deciency confers resistance to mycobacterial infection.
These authors demonstrated that reduced intracellular survival of mycobacteria in the CD36/ macrophage, as well as, a role of CD36 in the
cellular events involved in granuloma formation that promote early
bacterial expansion and dissemination.
BCG-induced lipid formation was only halved in CD36-decient
cells, indicating that some other co-receptors might be involved and
that compensatory gene expression might occur in genetically decient
mice. Indeed, partial redundancy of the different pattern recognition
receptors was documented and a double deciency in scavenger
receptors CD36 plus SR-A had no marked effect on the control of
M. tuberculosis infection [59].
The mechanisms by which CD14 contributes as a co-receptor to
promote TLR2 signaling are poorly understood. TLR2 is activated by a
variety of bacterial products, including some that depend on CD14 for
sensitive responses. CD14 may participate in trafcking and/or increasing bioavailability of TLR2 ligands [55,60]. Of note, this receptor
was shown to be highly concentrated in lipid raft microdomains of
monocyte lineage [61]. In addition, Nielsen et al. (2008) [54] demonstrated that signaling in response to lipoteichoic acid, preferentially

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

469
470
471
472
473
474
475
476
477
478
479
480
481
482
483
484
485
486
487
488
489
490
491
492
493
494
495
496
497
498
499

R
O

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

505
506
507
508
509
510
511
512
513
514
515
516
517
518
519
520
521
522
523
524
525
526
527

503
504

occurs at the plasma membrane and requires CD36 and CD14 as coreceptors for TLR2. Corroborating with these data, Hajishengallis et al.
have shown in an elegant experiment the induction of a multireceptor
complex in lipid rafts, comprising TLR2, CD14 and CD11b/CD18 upon
stimulation with LPS from Porphyromonas gingivalis and mbriae [62].
Whether similar multireceptor complex in lipid rafts are formed after
mycobacterial infection needs further investigation.
Here, we demonstrate that, beyond TLR2 and CD36, blockade of CD14
and CD11b/CD18 also inhibited M. bovis BCG-induced lipid droplet
formation and PGE2 production, suggesting that in addition to CD36
other TLR2 co-receptors may regulate mycobacterial infection-triggered
alterations in host lipid metabolism. CD36, CD11b/CD18, and CD14 are
all lipid-raft-resident molecules [28], and raft signaling compartmentalization may modulate TLR mechanism of activation and downstream
signaling. Membrane lipid rafts are enriched in cholesterol and play an
important role as signaling platforms. Spatial concentration of receptors
on the membrane can allow rapid and efcient coupling with ligands
and effectors of the intracellular signaling system [63]. Lipid rafts do
not only facilitate receptor interactions by compartmentalization of
molecules on the plasma membrane, but also seem to play a role in the
intracellular activation pathways during mycobacterial infections. Here,
we show that during M. bovis BCG-infection of macrophages, stability
of rafts is important for lipid body formation and PGE2 production,
suggesting that TLR2 and co-receptors interact in these membrane
domains to modulate host cell lipid metabolism.
In conclusion, we uncovered novel connections between TLR2/CD36
activation, in association with CD11b/CD18 and CD14, and PPAR
expression and activation. Moreover, our results suggest that TLR2

N
C
O

501
502

500

Fig. 7. TLR2/CD36 heterodimers divert intracellular signaling toward increased PPAR expression and lipid accumulation during M. bovis BCG infection. Mycobacterium bovis BCG infection
triggers different TLR2-dependent intracellular signaling cascades with PPAR and NF-B activation. The CD36-TLR2 cooperation and signaling compartmentalization within rafts, with
participation of both CD14 and CD11b/CD18, diverts host response signaling with increased expression and activation of PPAR through NF-B-independent pathways, leading to
increased macrophage lipid accumulation and down-modulation of macrophage response.

signaling compartmentalization within rafts is essential for myco- 528


bacterial induced regulation of host lipid metabolism but not for pro- 529
inammatory cytokine production (Fig. 7).
530

References
[1] W.H. Organization, Global tuberculosis control, WHO/HTM/TB/2011.16 Geneva:
WHO Report, 2011, p. 258.
[2] J.A. Armstrong, P.D. Hart, Phagosomelysosome interactions in cultured macrophages infected with virulent tubercle bacilli. Reversal of the usual nonfusion
pattern and observations on bacterial survival, J. Exp. Med. 142 (1975) 116.
[3] D.G. Russell, Mycobacterium tuberculosis and the intimate discourse of a chronic
infection, Immunol. Rev. 240 252268.
[4] D.G. Russell, H.C. Mwandumba, E.E. Rhoades, Mycobacterium and the coat of many
lipids, J. Cell Biol. 158 (2002) 421426.
[5] H. D'Avila, R.C. Melo, G.G. Parreira, E. Werneck-Barroso, H.C. Castro Faria Neto, P.T.
Bozza, Mycobacterium bovis BCG induces TLR 2-mediated formation of lipid bodies:
intracellular domains for eicosanoid synthesis in vivo, J. Immunol. 176 (2006)
30873097.
[6] D. Cruz, A.D. Watson, C.S. Miller, D. Montoya, M.T. Ochoa, P.A. Sieling, M.A. Gutierrez,
M. Navab, S.T. Reddy, J.L. Witztum, A.M. Fogelman, T.H. Rea, D. Eisenberg, J. Berliner,
R.L. Modlin, Host-derived oxidized phospholipids and HDL regulate innate
immunity in human leprosy, J. Clin. Invest. 118 (2008) 29172928.
[7] H. D'Avila, N.R. Roque, R.M. Cardoso, H.C. Castro-Faria-Neto, R.C. Melo, P.T. Bozza,
Neutrophils recruited to the site of Mycobacterium bovis BCG infection undergo
apoptosis and modulate lipid body biogenesis and prostaglandin E production by
macrophages, Cell. Microbiol. 10 (2008) 25892604.
[8] P.E. Almeida, A.R. Silva, C.M. Maya-Monteiro, D. Torocsik, H. D'Avila, B. Dezso, K.G.
Magalhaes, H.C. Castro-Faria-Neto, L. Nagy, P.T. Bozza, Mycobacterium bovis bacillus
CalmetteGuerin infection induces TLR2-dependent peroxisome proliferatoractivated receptor gamma expression and activation: functions in inammation,
lipid metabolism, and pathogenesis, J. Immunol. 183 (2009) 13371345.
[9] M.J. Kim, H.C. Wainwright, M. Locketz, L.G. Bekker, G.B. Walther, C. Dittrich, A.
Visser, W. Wang, F.F. Hsu, U. Wiehart, L. Tsenova, G. Kaplan, D.G. Russell, Caseation

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

531
532
533
534
535
536
537
538
Q6
539
540
541
542
543
544
545
546
547
548
549
550
551
552
553
554
555
556
557
558
559

[19]
[20]

[21]

[22]

[23]

[24]

[25]

[26]

[27]

[29]

[30]

[31]

[32]

[33]

[34]

[28]

R
O

[18]

[17]

[16]

[35] I. Szatmari, L. Nagy, Nuclear receptor signalling in dendritic cells connects lipids, the
genome and immune function, EMBO J. 27 (2008) 23532362.
[36] L. Nagy, P. Tontonoz, J.G. Alvarez, H. Chen, R.M. Evans, Oxidized LDL regulates
macrophage gene expression through ligand activation of PPARgamma, Cell 93
(1998) 229240.
[37] L. P.N. Tontonoz, J.G. Alvarez, V.A. Thomazy, R.M. Evans, PPARgamma promotes
monocytes/macrophage differentiation and uptake of oxidized LDL, Cell 93 (1998)
241252.
[38] A. Chawla, Y. Barak, L. Nagy, D. Liao, P. Tontonoz, R.M. Evans, PPAR-gamma
dependent and independent effects on macrophage-gene expression in lipid
metabolism and inammation, Nat. Med. 7 (2001) 4852.
[39] K.J. Moore, L.P. Andersson, R.R. Ingalls, B.G. Monks, R. Li, M.A. Arnaout, D.T.
Golenbock, M.W. Freeman, Divergent response to LPS and bacteria in CD14decient murine macrophages, J. Immunol. 165 (2000) 42724280.
[40] O. Takeuchi, K. Hoshino, T. Kawai, H. Sanjo, H. Takada, T. Ogawa, K. Takeda, S. Akira,
Differential roles of TLR2 and TLR4 in recognition of gram-negative and
gram-positive bacterial cell wall components, Immunity 11 (1999) 443451.
[41] S. Boulant, P. Targett-Adams, J. McLauchlan, Disrupting the association of hepatitis C
virus core protein with lipid droplets correlates with a loss in production of
infectious virus, J. Gen. Virol. 88 (2007) 22042213.
[42] Y. Miyanari, K. Atsuzawa, N. Usuda, K. Watashi, T. Hishiki, M. Zayas, R.
Bartenschlager, T. Wakita, M. Hijikata, K. Shimotohno, The lipid droplet is an
important organelle for hepatitis C virus production, Nat. Cell Biol. 9 (2007)
10891097.
[43] M.M. Samsa, J.A. Mondotte, N.G. Iglesias, I. Assuncao-Miranda, G. Barbosa-Lima, A.T.
Da Poian, P.T. Bozza, A.V. Gamarnik, Dengue virus capsid protein usurps lipid
droplets for viral particle formation, PLoS Pathog. 5 (2009) e1000632.
[44] H. D'Avila, C.G. Freire-de-Lima, N.R. Roque, L. Teixeira, C. Barja-Fidalgo, A.R. Silva,
R.C. Melo, G.A. Dosreis, H.C. Castro-Faria-Neto, P.T. Bozza, Host cell lipid bodies
triggered by Trypanosoma cruzi infection and enhanced by the uptake of apoptotic
cells are associated with prostaglandin E(2) generation and increased parasite
growth, J. Infect. Dis. 204 (2011) 951961.
[45] K.A. Mattos, H. D'Avila, L.S. Rodrigues, V.G. Oliveira, E.N. Sarno, G.C. Atella, G.M.
Pereira, P.T. Bozza, M.C. Pessolani, Lipid droplet formation in leprosy: Toll-like
receptor-regulated organelles involved in eicosanoid formation and Mycobacterium
leprae pathogenesis, J. Leukoc. Biol. 87 (2010) 371384.
[46] K.A. Mattos, V.G. Oliveira, H. D'Avila, L.S. Rodrigues, R.O. Pinheiro, E.N. Sarno, M.C.
Pessolani, P.T. Bozza, TLR6-driven lipid droplets in Mycobacterium leprae-infected
Schwann cells: immunoinammatory platforms associated with bacterial persistence,
J. Immunol. 187 (2011) 25482558.
[47] P.A. Baeuerle, D. Baltimore, NF-kappa B: ten years after, Cell 87 (1996) 1320.
[48] M. Muzio, G. Natoli, S. Saccani, M. Levrero, A. Mantovani, The human Toll signaling
pathway: divergence of nuclear factor kappaB and JNK/SAPK activation upstream
of tumor necrosis factor receptor-associated factor 6 (TRAF6), J. Exp. Med. 187
(1998) 20972101.
[49] H. Wesche, W.J. Henzel, W. Shillinglaw, S. Li, Z. Cao, MyD88: an adapter that recruits
IRAK to the IL-1 receptor complex, Immunity 7 (1997) 837847.
[50] V.J. Palombella, O.J. Rando, A.L. Goldberg, T. Maniatis, The ubiquitinproteasome
pathway is required for processing the NF-kappa B1 precursor protein and the
activation of NF-kappa B, Cell 78 (1994) 773785.
[51] H.M. Shin, M.H. Kim, B.H. Kim, S.H. Jung, Y.S. Kim, H.J. Park, J.T. Hong, K.R. Min, Y. Kim,
Inhibitory action of novel aromatic diamine compound on lipopolysaccharideinduced nuclear translocation of NF-kappaB without affecting IkappaB degradation,
FEBS Lett. 571 (2004) 5054.
[52] P. Desreumaux, L. Dubuquoy, S. Nutten, M. Peuchmaur, W. Englaro, K. Schoonjans, B.
Derijard, B. Desvergne, W. Wahli, P. Chambon, M.D. Leibowitz, J.F. Colombel, J.
Auwerx, Attenuation of colon inammation through activators of the retinoid X
receptor (RXR)/peroxisome proliferator-activated receptor gamma (PPARgamma)
heterodimer. A basis for new therapeutic strategies, J. Exp. Med. 193 (2001)
827838.
[53] S. Ogawa, J. Lozach, C. Benner, G. Pascual, R.K. Tangirala, S. Westin, A. Hoffmann, S.
Subramaniam, M. David, M.G. Rosenfeld, C.K. Glass, Molecular determinants of
crosstalk between nuclear receptors and Toll-like receptors, Cell 122 (2005) 707721.
[54] N.J. Nilsen, S. Deininger, U. Nonstad, F. Skjeldal, H. Husebye, D. Rodionov, S. von
Aulock, T. Hartung, E. Lien, O. Bakke, T. Espevik, Cellular trafcking of lipoteichoic
acid and Toll-like receptor 2 in relation to signaling: role of CD14 and CD36,
J. Leukoc. Biol. 84 (2008) 280291.
[55] M.G. Drage, N.D. Pecora, A.G. Hise, M. Febbraio, R.L. Silverstein, D.T. Golenbock, W.H.
Boom, C.V. Harding, TLR2 and its co-receptors determine responses of macrophages
and dendritic cells to lipoproteins of Mycobacterium tuberculosis, Cell. Immunol. 258
(2009) 2937.
[56] A.C. Nicholson, J. Han, M. Febbraio, R.L. Silversterin, D.P. Hajjar, Role of CD36, the
macrophage class B scavenger receptor, in atherosclerosis, Ann. N. Y. Acad. Sci.
947 (2001) 224228.
[57] S. Collot-Teixeira, J. Martin, C. McDermott-Roe, R. Poston, J.L. McGregor, CD36 and
macrophages in atherosclerosis, Cardiovasc. Res. 75 (2007) 468477.
[58] M. Hawkes, X. Li, M. Crockett, A. Diassiti, C. Finney, G. Min-Oo, W.C. Liles, J. Liu, K.C.
Kain, CD36 deciency attenuates experimental mycobacterial infection, BMC Infect.
Dis. 10 (2010) 299.
[59] N. Court, V. Vasseur, R. Vacher, C. Fremond, Y. Shebzukhov, V.V. Yeremeev, I. Maillet,
S.A. Nedospasov, S. Gordon, P.G. Fallon, H. Suzuki, B. Ryffel, V.F. Quesniaux, Partial
redundancy of the pattern recognition receptors, scavenger receptors, and C-type
lectins for the long-term control of Mycobacterium tuberculosis infection, J. Immunol.
184 (2010) 70577070.
[60] T. Vasselon, P.A. Detmers, D. Charron, A. Haziot, TLR2 recognizes a bacterial
lipopeptide through direct binding, J. Immunol. 173 (2004) 74017405.

[15]

[14]

[13]

[12]

[11]

[10]

of human tuberculosis granulomas correlates with elevated host lipid metabolism,


EMBO Mol. Med. 2 258274.
K.A. de Mattos, E.N. Sarno, M.C. Pessolani, P.T. Bozza, Deciphering the contribution of
lipid droplets in leprosy: multifunctional organelles with roles in Mycobacterium
leprae pathogenesis, Mem. Inst. Oswaldo Cruz 107 (Suppl. 1) (2012) 156166.
J. Gateld, J. Pieters, Essential role for cholesterol in entry of mycobacteria into
macrophages, Science 288 (2000) 16471650.
E. Anes, M.P. Kuhnel, E. Bos, J. Moniz-Pereira, A. Habermann, G. Grifths, Selected
lipids activate phagosome actin assembly and maturation resulting in killing of
pathogenic mycobacteria, Nat. Cell Biol. 5 (2003) 793802.
P. Peyron, J. Vaubourgeix, Y. Poquet, F. Levillain, C. Botanch, F. Bardou, M. Daffe, J.F.
Emile, B. Marchou, P.J. Cardona, C. de Chastellier, F. Altare, Foamy macrophages from
tuberculous patients' granulomas constitute a nutrient-rich reservoir for M. tuberculosis
persistence, PLoS Pathog. 4 (2008) e1000204.
H. D'Avila, P.E. Almeida, N.R. Roque, H.C. Castro-Faria-Neto, P.T. Bozza, Toll-like
receptor-2-mediated C-C chemokine receptor 3 and eotaxin-driven eosinophil
inux induced by Mycobacterium bovis BCG pleurisy, Infect. Immun. 75 (2007)
15071511.
H. D'Avila, C.M. Maya-Monteiro, P.T. Bozza, Lipid bodies in innate immune response
to bacterial and parasite infections, Int. Immunopharmacol. 8 (2008) 13081315.
K.A. Mattos, F.A. Lara, V.G. Oliveira, L.S. Rodrigues, H. D'Avila, R.C. Melo, P.P. Manso,
E.N. Sarno, P.T. Bozza, M.C. Pessolani, Modulation of lipid droplets by Mycobacterium
leprae in Schwann cells: a putative mechanism for host lipid acquisition and
bacterial survival in phagosomes, Cell. Microbiol. 13 (2011) 259273.
O. Neyrolles, R. Hernandez-Pando, F. Pietri-Rouxel, P. Fornes, L. Tailleux, J.A. Barrios
Payan, E. Pivert, Y. Bordat, D. Aguilar, M.C. Prevost, C. Petit, B. Gicquel, Is adipose
tissue a place for Mycobacterium tuberculosis persistence? PLoS ONE 1 (2006) e43.
B. Ryffel, M. Jacobs, S. Parida, T. Botha, D. Togbe, V. Quesniaux, Toll-like receptors and
control of mycobacterial infection in mice, Novartis Found. Symp. 279 (2006)
127139 (discussion 139141, 216129).
J. Kleinnijenhuis, M. Oosting, L.A. Joosten, M.G. Netea, R. Van Crevel, Innate immune
recognition of Mycobacterium tuberculosis, Clin. Dev. Immunol. 2011 (2011) 405310.
A. Yoshimura, E. Lien, R.R. Ingalls, E. Tuomanen, R. Dziarski, D. Golenbock, Cutting
edge: recognition of Gram-positive bacterial cell wall components by the innate
immune system occurs via Toll-like receptor 2, J. Immunol. 163 (1999) 15.
T.H. Flo, O. Halaas, E. Lien, L. Ryan, G. Teti, D.T. Golenbock, A. Sundan, T. Espevik, Human
Toll-like receptor 2 mediates monocyte activation by Listeria monocytogenes, but not
by group B streptococci or lipopolysaccharide, J. Immunol. 164 (2000) 20642069.
T.H. Flo, L. Ryan, E. Latz, O. Takeuchi, B.G. Monks, E. Lien, O. Halaas, S. Akira, G.
Skjak-Braek, D.T. Golenbock, T. Espevik, Involvement of Toll-like receptor (TLR) 2
and TLR4 in cell activation by mannuronic acid polymers, J. Biol. Chem. 277
(2002) 3548935495.
J.Y. Kang, X. Nan, M.S. Jin, S.J. Youn, Y.H. Ryu, S. Mah, S.H. Han, H. Lee, S.G. Paik, J.O.
Lee, Recognition of lipopeptide patterns by Toll-like receptor 2-Toll-like receptor 6
heterodimer, Immunity 31 (2009) 873884.
A. Ozinsky, D.M. Underhill, J.D. Fontenot, A.M. Hajjar, K.D. Smith, C.B. Wilson, L.
Schroeder, A. Aderem, The repertoire for pattern recognition of pathogens by the
innate immune system is dened by cooperation between Toll-like receptors,
Proc. Natl. Acad. Sci. U. S. A. 97 (2000) 1376613771.
M. Triantalou, F.G. Gamper, R.M. Haston, M.A. Mouratis, S. Morath, T. Hartung, K.
Triantalou, Membrane sorting of Toll-like receptor (TLR)-2/6 and TLR2/1
heterodimers at the cell surface determines heterotypic associations with CD36
and intracellular targeting, J. Biol. Chem. 281 (2006) 3100231011.
D. Strapagiel, K. Kasztalska, M. Druszczynska, M. Kowalewicz-Kulbat, A. Vrba, A.
Matusiak, M. Chmiela, W. Rudnicka, Monocyte response receptors in BCG driven
delayed type hypersensitivity to tuberculin, Folia Histochem. Cytobiol. 46 (2008)
353359.
K. Hoebe, P. Georgel, S. Rutschmann, X. Du, S. Mudd, K. Crozat, S. Sovath, L. Shamel,
T. Hartung, U. Zahringer, B. Beutler, CD36 is a sensor of diacylglycerides, Nature 433
(2005) 523527.
M. Triantalou, F.G. Gamper, P.M. Lepper, M.A. Mouratis, C. Schumann, E.
Harokopakis, R.E. Schifferle, G. Hajishengallis, K. Triantalou, Lipopolysaccharides
from atherosclerosis-associated bacteria antagonize TLR4, induce formation of
TLR2/1/CD36 complexes in lipid rafts and trigger TLR2-induced inammatory
responses in human vascular endothelial cells, Cell. Microbiol. 9 (2007) 20302039.
M. Febbraio, D.P. Hajjar, R.L. Silverstein, CD36: a class B scavenger receptor involved
in angiogenesis, atherosclerosis, inammation, and lipid metabolism, J. Clin. Invest.
108 (2001) 785791.
L.M. Stuart, J. Deng, J.M. Silver, K. Takahashi, A.A. Tseng, E.J. Hennessy, R.A.
Ezekowitz, K.J. Moore, Response to Staphylococcus aureus requires CD36-mediated
phagocytosis triggered by the COOH-terminal cytoplasmic domain, J. Cell Biol. 170
(2005) 477485.
C.R. Stewart, L.M. Stuart, K. Wilkinson, J.M. van Gils, J. Deng, A. Halle, K.J. Rayner, L.
Boyer, R. Zhong, W.A. Frazier, A. Lacy-Hulbert, J. El Khoury, D.T. Golenbock, K.J.
Moore, CD36 ligands promote sterile inammation through assembly of a Toll-like
receptor 4 and 6 heterodimer, Nat. Immunol. 11 155161.
M.V. Rajaram, M.N. Brooks, J.D. Morris, J.B. Torrelles, A.K. Azad, L.S. Schlesinger,
Mycobacterium tuberculosis activates human macrophage peroxisome proliferatoractivated receptor gamma linking mannose receptor recognition to regulation of
immune responses, J. Immunol. 185 (2010) 929942.
S. Mahajan, H.K. Dkhar, V. Chandra, S. Dave, R. Nanduri, A.K. Janmeja, J.N. Agrewala,
P. Gupta, Mycobacterium tuberculosis modulates macrophage lipid-sensing nuclear
receptors PPARgamma and TR4 for survival, J. Immunol. 188 (2012) 55935603.
P.E. Almeida, A.B. Carneiro, A.R. Silva, P.T. Bozza, PPARgamma expression and
function in mycobacterial infection: roles in lipid metabolism, immunity, and
bacterial killing, PPAR Res. 2012 (2012) 383829.

560
561
Q7
562
563
564
565
566
567
568
569
570
571
572
573
574
575
576
577
578
579
580
581
582
583
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
604
605
606
607
608
609
610
611
612
613
614
615
616
617
618
619
620
621
622
623
624
625
626
627
628
629
630
631
632
633
634
635
Q8
636
637
638
639
640
641
642
643
644
645

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

10

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

646
647
648
649
650
651
652
653
654
655
656
657
658
659
660
661
662
663
664
665
666
667
668
669
670
671
672
673
674
675
676
677
678
679
680
681
682
683
684
685
686
687
688
689
690
691
692
693
694
695
696
697
698
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
721
722
723
724
725
726
727
728
729
730
731

P.E. Almeida et al. / Biochimica et Biophysica Acta xxx (2013) xxxxxx

732
733
734
735
736
737

[61] A. Pfeiffer, A. Bottcher, E. Orso, M. Kapinsky, P. Nagy, A. Bodnar, I. Spreitzer, G.


Liebisch, W. Drobnik, K. Gempel, M. Horn, S. Holmer, T. Hartung, G. Multhoff, G.
Schutz, H. Schindler, A.J. Ulmer, H. Heine, F. Stelter, C. Schutt, G. Rothe, J. Szollosi,
S. Damjanovich, G. Schmitz, Lipopolysaccharide and ceramide docking to CD14
provokes ligand-specic receptor clustering in rafts, Eur. J. Immunol. 31 (2001)
31533164.

11

[62] G. Hajishengallis, R.I. Tapping, E. Harokopakis, S. Nishiyama, P. Ratti, R.E. Schifferle, E.A.
Lyle, M. Triantalou, K. Triantalou, F. Yoshimura, Differential interactions of mbriae
and lipopolysaccharide from Porphyromonas gingivalis with the Toll-like receptor
2-centred pattern recognition apparatus, Cell. Microbiol. 8 (2006) 15571570.
[63] K. Simons, D. Toomre, Lipid rafts and signal transduction, Nat. Rev. Mol. Cell Biol. 1
(2000) 3139.

N
C
O

R
O

745

Please cite this article as: P.E. Almeida, et al., Differential TLR2 downstream signaling regulates lipid metabolism and cytokine production
triggered by Mycobacterium bovis BCG infection, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbalip.2013.10.008

738
739
740
741
742
743
744

Вам также может понравиться