Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Molecular Immunology xxx (xxxx) xxx

Contents lists available at ScienceDirect

Molecular Immunology
journal homepage: www.elsevier.com/locate/molimm

Mycobacterium tuberculosis EsxL induces TNF-α secretion through activation


of TLR2 dependent MAPK and NF-κB pathways
Kali Prasad Pattanaik a, Geetanjali Ganguli a, Sumanta Kumar Naik a, Avinash Sonawane b, *
a
School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
b
Discipline of Biosciences and Biomedical Engineering, IIT Indore, Madhya Pradesh, India

A R T I C L E I N F O A B S T R A C T

Keywords: Mycobacterium tuberculosis (Mtb) employs distinct strategies to circumvent host immune responses during the
Mycobacterium tuberculosis infection process. Various Mtb cell-wall associated and secretory proteins are known to play a critical role in the
Tumour necrosis factor α orchestration of host innate immune responses through modulation of signaling pathways. Mtb genome encodes
Toll-like receptor 2
for 23 (EsxA-EsxW) proteins belonging to the ESAT-6 like family; however, most of them are functionally un-
Mitogen-activated protein kinase
Nuclear factor-κB
known. Here, we show that Mtb EsxL induces tumor necrosis factor-alpha (TNF-α) production by activating
nuclear translocation of nuclear factor-κB (NF-κB) via interaction with Toll-like Receptor 2 (TLR2). Blocking or
silencing of TLR2 abrogated nuclear translocation of NF-kB and TNF-α production. Treatment with recombinant
purified EsxL (rEsxL) activated mitogen-activated protein kinase (MAPK) pathway by inducing the phosphory-
lation of extracellular signal-regulated kinase (ERK) and p38 kinase (p38) pathways. At the same time, inhibition
of ERK and p38 down-regulated the expression of TNF-α in rEsxL exposed murine macrophages. Besides TNF-α,
EsxL also induced the production of IL-6 proinflammatory cytokine. Taken together, these results suggest that
EsxL is able to induce TNF-α secretion via TLR2 through activation of NF-κB and MAPK signaling. This study will
help in deducing therapeutic strategies for better control of the disease.

1. Introduction deficient mice were found to be more susceptible to Mtb infection


(Drennan et al., 2004). Upon infection, the activation of MAPKs such as
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is still p38, ERK and JNK induce the cascades of phosphorylation reactions at
considered as one of the deadliest infectious diseases worldwide. It has Th-XXX-Tyr motifs to control pro-inflammatory responses; direct or in-
been reported that approximately 10 million people are infected with direct cross-talk through nuclear translocation of NF-κB to regulate the
TB, out of which 1.5 million people die annually (Global Tuberculosis expression of different genes involved in cell differentiation, prolifera-
Report, 2017). Mtb is an intracellular pathogen that primarily resides tion and death processes (Hayden and Ghosh, 2014).
inside the alveolar macrophages for a longer period by manipulating the It has been shown that various mycobacterial antigenic molecules
immune effector functions of macrophages (Satoh and Akira, 2016; such as lipomannan, MPT83 and Rv1808 trigger the production of
Lugo-Villarino et al., 2011). In the context of Mtb infection, various proinflammatory cytokines such as TNF-α, IL-6, IL-12 and IL-1β through
Toll-like receptors such as TLR1, TLR2, TLR4, TLR6, and TLR9 play an activation of p38, ERK, and NF-kB pathways. Several studies have shown
essential role in the recognition of a pathogen via interaction with that TNF-α is essential for the induction of pro-inflammatory responses
different mycobacterial ligands to initiate the downstream innate im- to activate macrophages during Mtb infection and also in the develop-
mune responses (Mogensen, 2009). Previously, several mycobacterial ment of granuloma during the progression of the disease. TNF-α proin-
proteins such as lipomannan (Quesniaux et al., 2004), lip- flammatory cytokine was found to be increased in pulmonary TB cases
oarabinomannan (Jones et al., 2001), LpqH (19-kDa/Rv3763) (Noss that helps in recruitment of immune cells such as macrophage and
et al., 2001; Pai et al., 2003), LprG (Gehring et al., 2004), LprA (Pecora lymphocytes to form granuloma and covers the infected foci (Wickre-
et al., 2006), MPT83 (Chen et al., 2012), MPB83 (Chambers et al., 2010) masinghe et al., 1999; Roach et al., 1993). Mice deficient in the TNF-α
and LprE (Padhi et al., 2019) have been shown to modulate macrophage receptor or neutralization of TNF-α showed high bacterial burden and
functions through TLR2 dependent MAPK pathways and that TLR2 were more susceptible to Mtb infection (Flynn et al., 1995). TNF-α also

* Corresponding author at: Discipline of Biosciences and Biomedical Engineering, IIT Indore, Khandwa Road, Simrol, Madhya Pradesh, 453552, India.
E-mail address: asonawane@iiti.ac.in (A. Sonawane).

https://doi.org/10.1016/j.molimm.2020.11.020
Received 26 March 2020; Received in revised form 6 November 2020; Accepted 23 November 2020
0161-5890/© 2020 Published by Elsevier Ltd.

Please cite this article as: Kali Prasad Pattanaik, Molecular Immunology, https://doi.org/10.1016/j.molimm.2020.11.020
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

serves as a chemotactic factor to draw inflammatory cells such as 2.2. siRNA transfection
T-lymphocyte into the lower respiratory tract and leads to macrophage
activation and mycobacterial growth inhibition by working synergisti- RAW264.7 murine macrophages (2.5 × 105 cells/well) in DMEM
cally with IFN-γ (Sharma and Bose, 2001).TNF-α is known to inhibit media without antibiotic were seeded in 6-well plates. Cells were then
mycobacterial intracellular replication by activating both inducible ni- incubated at 37 ◦ C and 5% CO2 till confluency reached to 60 %. Cells
tric oxide synthase (iNOS) dependent and independent pathways (Bek- were transfected with 40 pmols of TLR2 siRNA (Santa Cruz Biotech, sc-
ker et al., 2001), facilitate phago-lysosome fusion and enhance the 40257) as per manufacturer’s instructions. Scrambled siRNA was used
apoptosis of Mtb infected cells. However, Mtb is known to as a control. Cells were harvested 24 h post-treatment, and the silencing
down-regulate TNF-α production in order to evade the host efficiency was determined by quantitative real-time PCR (qRT-PCR)
anti-tuberculosis immune mechanisms (Olsen et al., 2016). using gene-specific primers (Table 1).
In addition to the cytokines, chemokines were also found to be
essential to attract immune cells to Mtb infected regions. Several che- 2.3. Western blot analysis
mokines such as CCL2 (MCP1), CCL3 (MIP-1 α), CCL4 (MIP1β) and CLL5
(RANTEs) play a vital role during Mtb infection (Domingo-Gonzalez RAW264.7 cells were treated with different concentrations of rEsxL
et al., 2016; Qiu et al., 2008; Sadek et al., 1998). MIP-1α, MIP-1β, and for different time points and lysed with cell lysis buffer (5 mM EDTA, 5
RANTES induce activation and proliferation of T cells and macrophages mM EGTA, 1 mM PMSF, protease inhibitor cocktail, 10 mM Sodium
to suppress the growth of Mtb within macrophages (Taub et al., 1996; fluoride, 1 mM DTT and 1 mM sodium orthovanadate). The cells were
Chensue et al., 1999). MIP-1α induces production of TNF-α and IL-6 in centrifuged at 13,000 r.p.m. for 30 min at 4 ◦ C. Protein concentration
macrophages, while MIP-1β can modulate MIP-1α synthesis to induce was measured by Bradford assay. To study NF-kβ expression, a nuclear
TNF-α production. Other studies demonstrated that MIP-1α, MIP-1β, and protein extraction kit (ab113474, Abcam) was used for nuclear protein
RANTES could increase phagocytosis and killing of Trypanosoma cruzi by extraction. Nuclear and cytoplasmic fractions were separated as per the
inducing nitric oxide production (Chensue et al., 1999; Villalta et al., manufactures instructions. Proteins were electrophoresed in 12 % SDS-
1998). PAGE gels and transferred to a polyvinylidenedifluoride (PVDF) mem-
Mtb EsxL (Rv1198) belongs to the mycobacterial early secreted brane (GE Healthcare, USA). The membranes were blocked with 5%
antigenic targets of 6-kDa (ESAT6) family proteins. The type VII secre- nonfat milk for 2 h at room temperature (RT) followed by wash with PBS
tion system (T7SS) of Mtb contains five ESX secretion system (ESX1- containing 0.05 % tween-20 (v/v) (PBST, pH 7.4) and incubated over-
ESX5) (Målen et al., 2007). ESAT6 is an extraordinary Mtb virulence as night at 4 ◦ C with primary antibodies such as rabbit anti-ERK2, rabbit
well as antigenic determinant abundantly secreted through ESX secre- anti-p38 (Santa Cruz Biotechnology), mouse anti–phospho-ERK1/2,
tion system (Lewis et al., 2003). ESAT-6 family proteins are known to rabbit anti–phospho-p38, MHCII and MyD88. After a wash with PBST,
induce apoptosis and pro-inflammatory cytokine response via TLR the membranes were incubated for 1–2 h at 37 ◦ C with the appropriate
mediated signaling pathways (Shi et al., 2014; Lin et al., 2019). In this HRP-conjugated anti-mouse IgG or anti-rabbit IgG secondary antibodies
study, we elucidated the role of Mtb EsxL (Rv1198) in the modulation of (1:5000) (Genei). The peroxidase-positive bands were detected using an
macrophage innate immune responses. We have shown that Mtb EsxL is ECL reaction solution (Merck) and visualised by exposure to x-ray film
responsible for the activation of macrophage pro-inflammatory response (XAR5; Kodak, Rochester, NY).
by inducing the secretion of TNF-α through activation of TLR2 depen-
dent MAPK and NF-κB signaling pathways. 2.4. qRT-PCR analysis

2. Material and methods To check the relative expression of different cytokines, RAW264.7
cells (3 × 105 cells/well) were seeded on 6-well plates followed by
2.1. Purification of recombinant EsxL protein treatment with different concentrations of rEsxL. Total RNA was isolated
using TRIzol reagent and cDNA was synthesized using cDNA synthesis
Full-length Rv1198 gene was amplified from Mtb H37Rv genome. kit (Thermo Scientific, AB1453A) as per the manufacturer’s instructions.
Then the amplified product was digested with Nde1 and HindIII and qRT-PCR amplification was carried out using gene-specific primers and
cloned into pET21b vector. The recombinant plasmid was transformed the synthesized cDNA as a template. All reactions were performed in a
into E. coli DH5α. The positive clones were confirmed by colony PCR and total reaction volume of 10 μl using SYBR® Green PCR master mix (Cat.
sequencing using gene-specific primers (Table 1). Finally, the recombi- No. A25742, Applied Biosystems, USA), and carried out in InstaQ96
nant EsxL protein (rEsxL) was expressed in E. coli BL21 (DE3). Protein Real-time PCR (Himedia) with initial denaturation at 95 ◦ C for 2 min,
expression was induced with 0.5 mM IPTG (isopropyl-b-D-thio- final denaturation at 95 ◦ C for 30 s, annealing at 55 ◦ C for 15 s and
galactopyranoside, Sigma) for overnight at 16 ◦ C. Bacterial cells were extension at 72 ◦ C for 30 s. The primers used for the determination of
harvested and stored at −80 ◦ C. Next day cells were lysed using lysis different gene expression are listed in Table 1. The mRNA levels were
buffer (20 mM Tris/HCl, pH 8.0, 5 % glycerol, 300 mM NaCl, 10 mM normalized to the transcript levels of 18 s rRNA and GAPDH and the
imidazole and lysozyme 1 mg/mL) containing protease inhibitor cock- relative fold changes were calculated.
tail (Roche). The harvested cells were then sonicated, centrifuged at
13,000 r.p.m. for 40 min. rEsxL His-tagged protein was purified by af-
finity chromatography using Ni-NTA resin (Qiagen).

Table 1
Primers used in this study.
Gene name Forward primer Reverse Primer

18 srRNA GTAACCCGTTGAACCCCATT GTAACCCGTTGAACCCCATT


GAPDH GAGAGGCCCTATCCCAACTC TTCACCTCCCCATACACACC
TNF-α CACACACACCCTCCTGATTG CTCATTCAACCCTCGAAAA
IL-12 CACACTGGACCAAAGGGACT CTTTGCTTCCCTAGCACCTG
IL-6 GCCTTCTTGGGACTGATGCT TGCCATTGCACAACTCTTTTCT
EsxL AAGGATCCATATGACCATCAACTATCAATTCGG ATTAAGCTTGGCCCAGCTGGAGCCGAC

2
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

2.5. Subcellular localization of NF-kB cytotoxicity on macrophages up to 50 μg/mL concentration (Supple-


mentary Figure S2).
RAW264.7 cells were seeded in 24-well culture plates with glass
coverslips; treated with rEsxL (5 mg/mL) for 0, 15, or 30 min; and 3.2. rEsxL induces production of proinflammatory cytokines and
washed twice with PBS. Then cells were fixed for 20 min using pre- chemokines in murine macrophages
chilled methanol and acetone (1:1). To reduce the background stain-
ing, cells were incubated in PBS containing 5% BSA for 30 min and then ESAT-6 family proteins and ESX secretory proteins are known to
incubated with PhospoNF-kB(cellsignaling-3033) antibody. After incu- modulate the cytokine production, and in particularly enhance IL-6 and
bation for 12 h at 4 ◦ C, the cells were washed with phosphate buffer TNF-α secretion upon interaction with the macrophages (Yang et al.,
saline (PBS) and incubated with FITC conjugated goat anti-rabbit IgG 2015).To examine the role of EsxL in the modulation of host immune
Alexfluor 488(Thermo Scientific cat A32790) for 1 h at RT. Following, responses, macrophages were treated with rEsxL and studied for the
cells were washed and mounted with DAPI-containing ProLong Gold production of a panel of cytokines and chemokines. We found that
Antifade mounting solution (catalog number P-36,931; Invitrogen). NF- treatment with the 2−4 μg concentration of rEsxL significantly induced
kB localization was observed using a fluorescent microscope (Olympus). the production of TNF-α cytokine in macrophages compared to un-
treated and buffer control cells at 24 h post infection (Fig. 1A and B).
2.6. Cytokine profiling Treatment with proteinase K (PK) abrogated the rEsxL mediated in-
duction of TNF-α production at the transcriptional level (Fig. 1A). To
RAW 264.7 cells (2 × 105) in DMEM medium were seeded onto 24- exclude the effect of endotoxin contamination, RAW264.7 cells were
well tissue culture plate and treated with rEsxL (4 μg/mL) for 24 h. Then incubated with LPS (100 ng/mL) or rEsxL (5 μg/mL) with or without
supernatants were collected and cytokine levels were estimated using pretreatment with polymyxin B (PMB) and the TNF-α level was checked
Bioplex kit assay (Bio-Rad). after 24 h (Fig. 1C). As shown, no difference in TNF-α secretion was
observed in with and without PMB treated cells. Since maximum TNF-α
2.7. Pull-down assay secretion was observed at 4 μg rEsxL concentration after 24 h, we
selected these two parameters for the analysis of different cytokines and
Whole-cell protein lysates were isolated from RAW 264.7 cells and chemokines production in macrophages using Bioplex method. We
purified rEsxL-His proteins were added to Ni-NTA beads either alone or found that treatment with rEsxL significantly increased the production
in combination for 2 h on a rocker at 4 ◦ C. The flow-through (FT) of various cytokines and chemokines such as TNF-α, IL-6, MIP1α, MIP1β,
samples were first collected and the beads were then washed (W) with MCP1α and RANTES in macrophages after 24 h (Fig. 1 D–I). However,
ice-cold equilibration buffer (20 mM Tris, pH 8.0, 5 % glycerol 150 mM no significant differences in the production of IL-10 and IL-12p40 cy-
NaCl) containing 20 mM imidazole. Finally bound proteins were eluted tokines were observed in comparison to untreated cells (Figs. 1 J and K).
(E1-E2) in buffer containing 300 mM imidazole. Fractions were elec- These results indicate that rEsxL elicits the production of some proin-
trophoresed on 15 % SDS-PAGE. Western blotting was done with anti- flammatory cytokines and chemokines in macrophages.
TLR2 and anti-rEsxL antibodies.
3.3. rEsxL induces nuclear translocation of NF-kBp65 in murine
2.8. Statistical analysis macrophages

All experiments were performed at least three times. Statistically Previous studies have shown that MtbESAT-6 family proteins TB10.4
significant differences between groups were determined using one-way, and TB9.8 induces TNF-α production through activation and nuclear
two-way ANOVA (Sidak’s & Turkey’s multiple comparison test) and translocation of NF-kB (Liu et al., 2014; Liu et al., 2018). We speculated
Mann-Whitney U_test. Significance were referred as ‘ns’ for non- that Mtb EsxL may also phosphorylate and induce nuclear translocation
significant, * for P < 0.05, ** for P < 0.01 and *** for P ≤ 0.001 **** of NF-kB to enhance proinflammatory cytokine production in macro-
for P ≤ 0.0001. phages. To enumerate this hypothesis, we treated the macrophages with
4 μg/mL of rEsxL and studied the NF-kB level in the cytoplasmic and
3. Results nuclear fractions. We observed a significant time-dependent increase in
nuclear translocation and a concomitant decrease in cytoplasmic
3.1. Purification of rEsxL and generation of polyclonal anti-EsxL NF-kBp65 levels in rEsxL treated macrophages than the untreated cell
antibody (Fig. 2A&B). Immunofluorescence analysis also showed more NF-kB
co-localization (cyan) with nuclei (blue), whereas treatment with pro-
Mycobacterium tuberculosis EsxL (Rv1198), a 94 amino acid residue teinase K showed significant reduction in NF-kB nuclear translocation
protein, is present in the esxKL operon. EsxL belongs to ESX-5 system (Fig. 2C&D). These results demonstrate that rEsxL is indeed involved in
and is reported to be present in the culture filtrates of Mtb H37Rv along NF-kB nuclear translocation.
with EsxK (Rv1197) (Pandey et al., 2017). Previous study has shown
that ESX-5 plays a critical role in the modulation of macrophage immune 3.4. rEsxL triggers TNF-α expression through MAPK pathway
responses and also in cell to cell spread of mycobacteria (Abdallah et al.,
2008; Abdallah et al., 2011). EsxL was amplified from Mtb H37Rv Both P38 and ERK MAPK pathways have been shown to enhance the
genomic DNA, cloned in pET21b vector and expressed in E.coli BL21DE3 synthesis of pro-inflammatory cytokines such as TNF-α. Moreover,
(clone map; Supplementary Figure S1A). rEsxL was purified using MAPK and NF-kB cross-talk were also found to be necessary for the in-
Ni-NTA and the protein purity was confirmed on SDS-PAGE followed by duction of TNF-α expression. We also found that treatment with rEsxL
silver staining (Supplementary Figure S1B). Anti-EsxL polyclonal anti- resulted in phosphorylation of MAPK proteins, p38 and ERK at early
body was generated in rabbit. As shown, the antibody can detect rEsxL time points (Fig. 3A&B), while no significant differences were observed
(Supplementary Figure S1C). Further confirmative analysis was per- in pJNK levels (data not shown). To confirm the involvement of MAPK in
formed by identification of the purified EsxL protein by mass spec- cytokine modulation, we next studied the expression of TNF-α after
trometry analysis (Supplementary Figure S1D). To determine whether chemical inhibition of p38 and ERK by SB203580 (10μM) and U0126
rEsxL exhibit any cytotoxic effect on macrophages, murine macrophages (10 μM), respectively in macrophages. We found that inhibition of p38
RAW264.7 were treated with different concentrations of rEsxL and cell and ERK significantly reduced the induction of TNF-α transcripts by
cytotoxicity was determined by MTT assay. rEsxL did not show any rEsxL as compared to uninhibited cells (Fig. 3E). Similarly, inhibition of

3
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

Fig. 1. RAW264.7 cells were incubated with (A) 2 and 4 (μg/mL) of rEsxL, buffer control (B.C.) and proteinase K (PK) (50 μg/mL) along with rEsxL for 24 h, and total
RNA was isolated from rEsxL treated and control cells, cDNA was synthesized and subsequently TNF-α expression was checked at the transcriptional levels. The fold
change was normalized against 18 s rRNA. (B) 4 μg/mL rEsxL and TNF-α cytokine level was measured in comparison to respective control at different time points. (C)
LPS (100 ng/mL) and rEsxL (4 μg/mL) with and without polymyxin-B (PMB) (10 μg/mL), Pam3CSK4; untreated (control) cells were used as control for rEsxL.The cell
supernatants were collected and TNF-α level was measured by using cytokine assay kit. (D-K) with and without rEsxL (4 μg/mL); Cell supernatant from untreated
(control) and rEsxL treated cells were collected after 24 h post treatment and the released cytokines and chemokines were studied by using bioplex assay. Data are
expressed as the mean + SD.**, P < 0.001;***, P < 0.0001; ns, non significant. The experiment was performed in duplicates.

4
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

Fig. 2. rEsxL induce nuclear translocation of NF-kBp65. (A) Western blot analysis of NF-kBp65 using cytoplasmic and nuclear fraction of the protein samples isolated
from rEsxL treated RAW264.7 macrophages at different time points. (B) Quantitative densitometry of western blot data was performed using ImageJ analysis
software. Graph represent densitometric ratio of cytoplasmic and nuclear NF-kBp65 ratio with respect to respective constitutive control at different time intervals. (C)
Nuclear localization of NF-kBp65 post rEsxL treatment in mouse macrophages was determined using fluorescence microscopy and compared with untreated and
protein kinase treated rEsxL. (D) Fluorescence intensity was quantified using Image J software. The graph represents nuclear NF-kBp65 fluorescence intensity of
control and rEsxL treated variants from three biological replicates; Mean ± SD; *** for P < 0.0001,** for P < 0.001 * for P < 0.05; ns, non-significant.

p38 and ERK also decreased nuclear translocation of NF-kB in rEsxL of NF-kB after blocking the TLR2 receptor by using anti-TLR2 antibody.
treated macrophages in comparison to rEsxL treated cells (Fig. 3C&D). TLR-2 blocking significantly reduced NF-kB translocation to the nucleus
These results demonstrate that rEsxL induces NF-kB nuclear trans- (Fig. 4B&C). On the other hand no significant differences in NF-kBp65
location via p38 and ERK MAPK dependent pathways. translocation and TNF-α expression were observed after treatment
with a TLR4 blocker(TAK242, Sigma) (Supplementary Fig. 4A).
Above results suggested that EsxL regulates TNF-α secretion through
3.5. Induction of TNF-α production and NF-kB translocation by rEsxL is TLR-2 dependent pathway. To further confirm that EsxL interacts with
dependent on TLR2 TLR2, we performed immune pull-down assay with rEsxL. Immunoflu-
orescence study showed TLR2 and EsxL co-localization (Fig. 4D). For
Toll-like receptors play an essential role in the activation of macro- pull-down assay, murine macrophage cell lysates were incubated with
phage immune responses. ESAT-6 proteins such as TB10.4 and TB9.8 are rEsxL protein and passed through Ni-NTA column followed by elution
known to increase TNF-α production through TLR2 pathway (Liu et al., with an elution buffer. As shown, both rEsxL and TLR2 were eluted in
2014; Liu et al., 2018). As shown above, rEsxL treatment up-regulated the same fraction indicating that TLR2-EsxL interacts with each other
TNF-α synthesis (Fig. 1A-D). Next, we investigated the role of TLRs in (Fig. 4E).
the induction of TNF-α by EsxL. For this, TLR2 and TLR4 were either
silenced or chemically inhibited using siRNA or treatment with TLR 4. Discussion
blocker and then studied TNF-α expression in rEsxL treated macro-
phages. Interestingly, TLR2 silenced cells failed to induce TNF-α Various secretory as well as cell-wall associated virulence factors
expression by rEsxL when compared to non-silenced cells (Fig. 4A and have been regarded as the key mediators for the Mtb pathogenicity.
Supplementary Fig. 3A and B). We also studied the nuclear translocation

5
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

Fig. 3. MAPK activation by rEsxL triggers NF-kBp65 nuclear translocation and TNF-α induction. (A)Western blot analysis was performed to determine the phos-
phorylation of p38 and pERK in rEsxL treated RAW 264.7 macrophages at different time points. (B) Quantitative densitometry of western blots was performed using
Image J analysis. The expression was normalized to total p38 and total ERK as loading control. Experiments were performed in biological triplicates. (C) Fluorescence
microscopy analysis of NF-kBp65 nuclear translocation in rEsxL treated RAW264.7 cells in the presence of p38 inhibitor (SB203580,10μM) and ERK inhibitor
(U0126,10μM). (D) The bar graph shows non-significant nuclear translocation of NF-kBp65 at p38 and ERK inhibitor conditions with and without rEsxL of three
biological replicates. (E) Relative expression of TNF-α was studied by using P38 chemical inhibitor (SB203580, 10μM) and ERK inhibitor (U0126, 10μM).The fold
change was normalized against 18 s rRNA. Data are expressed as the mean + SD from).*** for P < 0.005;**** for P < 0.0001; ns, non-significant. The experiment was
performed in triplicates.

Studies showed that mycobacteria have evolved with different secretion (Abdallah et al., 2008; Abdallah et al., 2011). Mtb EsxL, encoded by
mechanisms to transport extracellular proteins across their hydrophobic Rv1198, is a putative interacting ESAT-6 family protein that belongs to
and extremely impermeable cell walls (Stanley et al., 2003; Brodin et al., the ESX-5 group and is encoded by the esxKL operon. EsxL is stated to be
2005). It has been reported that pathogenic mycobacteria strains consist present in the culture filtrates of Mtb H37Rv (Målen et al., 2007). The
up to five T7S secretory systems designated as ESX-1,2,3,4 and 5. patterns of the RNA expression in sputum from diagnosed TB patients
Phylogenetic and comparative genomic analyses have shown that these also displayed a prominent presence of Rv1198 transcripts, and that EsxL
ESX systems are likely to be evolved by gene duplication in the order is highly expressed during active human lung TB infection (Bukka et al.,
ESX-4, ESX-1, ESX-3, ESX-2, and ESX-5 (Gey Van Pittius et al., 2001). A 2012; Pandey et al., 2017). Mtb genome encodes numerous putative
T7S system that has attracted considerable attention in recent times is protein and non-protein antigenic molecules, some of which are enco-
ESX-5, which is limited to pathogenic mycobacterial species like M. ded by ESX-5 system, are capable of eliciting potent innate and adaptive
tubercule, M. leprae and, M. marinum (Gey van Pittius et al., 2006). This immune responses. Therefore, functional characterization of these
ESX-5 system was found to be essential for the formation of granulomas, mycobacterial ESAT-6 family proteins is essential for better under-
cell to cell spread of mycobacteria, and its substrates were shown to standing of the host-pathogen interactions and the design of prospective
actively modulate the immune responses of human macrophages vaccine candidates.

6
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

Fig. 4. rEsxL interacts with TLR2 and triggers TNF-α expression. (A) RAW264.7 mouse macrophages were treated with rEsxL, TLR2 siRNA, scrambled siRNA, and
TLR2 siRNA along with rEsxL for 24 h. Relative fold-expression of TNF-α was studied by normalizing data against 18 s rRNA. (B) NF-kBp65 nuclear translocation was
studied with fluorescence microscopy by blocking TLR2 using anti-TLR2 antibody prior to rEsxL treatment (blue-Nucleus, green-NF-kB). (C) Fluorescence intensity of
nuclear p-65NFkB of RAW264.7 cells blocked with TLR2 antibody and with or without rEsxL treatment were quantified using imageJ analysis software. (D)
Interaction between TLR2 and rEsxL was studied by using immune staining method (blue-nucleus, Red- TLR2 and green rEsxL). (E) Pull down assay was performed to
confirm rEsxL interaction with TLR2. Data are expressed as the mean + SD from).**, P < 0.001;***, P < 0.0001; ns, not significant. The experiment was performed in
triplicates (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article).

Cross-talks between TLRs and mycobacterial ESAT-6 family antigens interacts with the TLR2 receptor to trigger TNF-α secretion in macro-
have been extensively studied over the decades. Among different toll- phages. Previous studies also demonstrated that several mycobacterial
like receptors, TLR2 is considered as an essential link between innate proteins such as MPT83 and Mtb9.8 interacted with TLR2 (Chen et al.,
and adaptive immune responses against invasive mycobacteria and 2012; Liu et al., 2018). The same report also suggested that interaction
other microbial pathogens (Hmama et al., 2015; Pasare and Medzhitov, of mycobacterial MPT83 and Mtb9.8 with TLR2 resulted in subsequent
2005; Takeda and Akira, 2005). However, despite comprehensive activation of the pro-inflammatory TNF-α cytokine. We also found that
studies carried out over the years, the precise molecular mechanism(s) exposure to Mtb EsxL induced TNF-α secretion, along with a panel of
by which mycobacterial ESAT-6 like proteins help in exerting patho- other cytokines and chemokines; while TLR silencing abrogated EsxL
genesis remains poorly understood. Here, we successfully purified induced TNF-α secretion. TNF-α act as a critical regulator of host im-
His-tagged recombinant protein (rEsxL) and also generated anti-EsxL mune responses to Mtb infection, macrophage activation to kill engulfed
antibody. From the pull-down assay, we confirmed that Mtb EsxL bacteria efficiently, and induction of chemokines, cytokines and

7
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

apoptosis. These NF-κB dependent signaling pathway activities have Abdallah, A.M., Bestebroer, J., Savage, N.D.L., De Punder, K., Van Zon, M., Wilson, L.,
Korbee, C.J., Van Der Sar, A.M., Ottenhoff, T.H.M., Van Der Wel, N.N., 2011.
made TNF-α as a key factor for inhibiting mycobacterial growth in
Mycobacterial secretion systems ESX-1 and ESX-5 play distinct roles in host cell
granulomas(Gutierrez et al., 2008; Harris et al., 2008). death and inflammasome activation. J. Immunol. 187, 4744–4753.
Earlier reports demonstrated that Mtb proteins such as PPE26 and Adler, H.S., Kubsch, S., Graulich, E., Ludwig, S., Knop, J., Steinbrink, K., 2007. Activation
MPT83 interact with TLR2 to induce secretion of pro-inflammatory cy- of MAP kinase p38 is critical for the cell-cycle-controlled suppressor function of
regulatory T cells. Blood 109, 4351–4359. https://doi.org/10.1182/blood-2006-09-
tokines through nuclear translocation of NF-kB (Chen et al., 2012; Su 047563.
et al., 2015). Reports suggest that NF-κB mediated responses signifi- Bekker, L.G., Freeman, S., Murray, P.J., Ryffel, B., Kaplan, G., 2001. TNF-alpha controls
cantly control mycobacterial load in TB granulomas, inflammation level intracellular mycobacterial growth by both inducible nitric oxide synthase-
dependent and inducible nitric oxide synthase-independent pathways. J. Immunol.
in lung tissue, and granuloma size (Fallahi-Sichani et al., 2012). Further 166, 6728–6734. https://doi.org/10.4049/jimmunol.166.11.6728.
upon stimulation, NF-kB also induces the transcription of the TNF family Brodin, P., de Jonge, M.I., Majlessi, L., Leclerc, C., Nilges, M., Cole, S.T., Brosch, R., 2005.
of cytokines. Indeed we also found that treatment with rEsxL induced Functional analysis of early secreted antigenic target-6, the dominant T-cell antigen
of Mycobacterium tuberculosis, reveals key residues involved in secretion, complex
NF-kB activation and subsequently TNF-α secretion in macrophages. formation, virulence, and immunogenicity. J. Biol. Chem. 280, 33953–33959.
TNF-α binds to the TNF-receptor (Tnfr)to activate the MAPK https://doi.org/10.1074/jbc.M503515200.
pathway as a primary response. Then secondary response generated Bukka, A., Price, C.T.D., Kernodle, D.S., Graham, J.E., 2012. Mycobacterium tuberculosis
RNA expression patterns in sputum Bacteria Indicate secreted esx factors
from MAPK molecules is known to augment the TNF-α expression. The contributing to growth are highly expressed in active disease. Front. Microbiol. 2,
secondary response is a result of cross-talk between MAPkinases (p38 266. https://doi.org/10.3389/fmicb.2011.00266.
and ERK) and NF-kB, thereby inducing the production of TNF-α (Sabio Chambers, M.A., Whelan, A.O., Spallek, R., Singh, M., Coddeville, B., Guerardel, Y.,
Elass, E., 2010. Non-acylated Mycobacterium bovis glycoprotein MPB83 binds to
and Davis, 2014). In the context of Mtb infection, MAPKs play crucial
TLR1/2 and stimulates production of matrix metalloproteinase 9. Biochem. Biophys.
roles in the activation of innate immunity, T-cell activation, prolifera- Res. Commun. 400, 403–408. https://doi.org/10.1016/j.bbrc.2010.08.085.
tion and differentiation into T-helper cells (Dong et al., 2002; Adler Chen, S.-T., Li, J.-Y., Zhang, Y., Gao, X., Cai, H., 2012. Recombinant MPT83 derived from
et al., 2007). We also found that rEsxL significantly upregulated the Mycobacterium tuberculosis induces cytokine production and upregulates the
function of mouse macrophages through TLR2. J. Immunol. 188, 668–677. https://
phosphorylation of MAPK molecules namely p38 and ERK. Previous doi.org/10.4049/jimmunol.1102177.
studies also demonstrated that Mtb protein such as Rv3402C and MPT83 Chensue, S.W., Warmington, K.S., Allenspach, E.J., Lu, B., Gerard, C., Kunkel, S.L.,
trigger TNF-α production through activation of p38 and ERK MAPK Lukacs, N.W., 1999. Differential expression and cross-regulatory function of RANTES
during mycobacterial (type 1) and schistosomal (type 2) antigen-elicited
signaling pathways (Li et al., 2014; Chen et al., 2012). On the contrary, granulomatous inflammation. J. Immunol. 163, 165–173.
chemical inhibition of p38 and ERK, but not JNK, significantly reduced Domingo-Gonzalez, R., Prince, O., Cooper, A., Khader, S.A., 2016. Cytokines and
NF-kB nuclear translocation and subsequent reduction in the production chemokines in Mycobacterium tuberculosis infection. Microbiol. Spectr. 4 (10)
https://doi.org/10.1128/microbiolspec.TBTB2-0018-2016, 1128/microbiolspec.
of TNF-α cytokine. These results suggested that p38 and ERK MAPK TBTB2-0018–2016.
pathways are essential for the production of TNF-α by Mtb EsxL. Dong, C., Davis, R.J., Flavell, R.A., 2002. MAP kinases in the immune response. Annu.
Rev. Immunol. 20, 55–72. https://doi.org/10.1146/annurev.
immunol.20.091301.131133.
5. Conclusions Drennan, M.B., Nicolle, D., Quesniaux, V.J.F., Jacobs, M., Allie, N., Mpagi, J.,
Frémond, C., Wagner, H., Kirschning, C., Ryffel, B., 2004. Toll-like receptor 2-defi-
In summary, MtbEsxL interacts with TLR2 to activate the production cient mice succumb to Mycobacterium tuberculosis infection. Am. J. Pathol. 164,
49–57. https://doi.org/10.1016/S0002-9440(10)63095-7.
of pro-inflammatory cytokines (specifically TNF-α) through MAPK
Fallahi-Sichani, M., Kirschner, D.E., Linderman, J.J., 2012. NF-κB signaling dynamics
phosphorylation and nuclear translocation of NF-kB. play a key role in infection control in tuberculosis. Front. Physiol. 3, 170. https://
doi.org/10.3389/fphys.2012.00170.
Author’s contribution Flynn, J.L., Goldstein, M.M., Chan, J., Triebold, K.J., Pfeffer, K., Lowenstein, C.J.,
Schreiber, R., Mak, T.W., Bloom, B.R., 1995. Tumor necrosis factor-alpha is required
in the protective immune response against Mycobacterium tuberculosis in mice.
KP designed, performed, analyzed the experiments and wrote the Immunity 2, 561–572. https://doi.org/10.1016/1074-7613(95)90001-2.
manuscript; GG performed the experiments and analyzed the data and Gehring, A.J., Dobos, K.M., Belisle, J.T., Harding, C.V., Boom, W.H., 2004.
Mycobacterium tuberculosis LprG (Rv1411c): a novel TLR-2 ligand that inhibits
contributed to writing up the manuscript; SKN analyzed the data and human macrophage class II MHC antigen processing. J. Immunol. 173, 2660–2668.
contributed to writing up the manuscript; AS designed experiments, https://doi.org/10.4049/jimmunol.173.4.2660.
analyzed the data, obtained funding and wrote the manuscript. Gey van Pittius, N.C., Sampson, S.L., Lee, H., Kim, Y., van Helden, P.D., Warren, R.M.,
2006. Evolution and expansion of the Mycobacterium tuberculosis PE and PPE
multigene families and their association with the duplication of the ESAT-6 (esx)
Declaration of Competing Interest gene cluster regions. BMC Evol. Biol. 6, 95. https://doi.org/10.1186/1471-2148-6-
95.
Gey Van Pittius, N.C., Gamieldien, J., Hide, W., Brown, G.D., Siezen, R.J., Beyers, A.D.,
The authors have no conflict of interest. 2001. The ESAT-6 gene cluster of Mycobacterium tuberculosis and other high G+C
Gram-positive bacteria. Genome Biol. 2 https://doi.org/10.1186/gb-2001-2-10-
Acknowledgement research0044. RESEARCH0044.
World Health Organization. Global tuberculosis report 2017. Geneva; 2017. Report No.
WHO/HTM/TB/2017.23 Contract No.: WHO/HTM/TB; 2017. https://apps.who.int/
This work was supported by grant (BT/PR23317/MED/29/1186/ iris/handle/10665/259366.
2017) from Department of Biotechnology, Government of India and Gutierrez, M.G., Mishra, B.B., Jordao, L., Elliott, E., Anes, E., Griffiths, G., 2008. NF-
kappa B activation controls phagolysosome fusion-mediated killing of mycobacteria
Alexander von-Humboldt Stiftung, Germany (Ref 3.3-IND-1152176-
by macrophages. J. Immunol. 181, 2651–2663. https://doi.org/10.4049/
HFST-E) to AS. KP would like to acknowledge the Council of Science and jimmunol.181.4.2651.
Industrial Research, Government of India, for the research scholarship. Harris, J., Hope, J.C., Keane, J., 2008. Tumor necrosis factor blockers influence
macrophage responses to Mycobacterium tuberculosis. J. Infect. Dis. 198,
1842–1850. https://doi.org/10.1086/593174.
Appendix A. Supplementary data Hayden, M.S., Ghosh, S., 2014. Regulation of NF-κB by TNF family cytokines. Seminars
in Immunology. Elsevier, pp. 253–266.
Supplementary material related to this article can be found, in the Hmama, Z., Peña-Díaz, S., Joseph, S., Av-Gay, Y., 2015. Immunoevasion and
immunosuppression of the macrophage by Mycobacterium tuberculosis. Immunol.
online version, at doi:https://doi.org/10.1016/j.molimm.2020.11.020. Rev. 264, 220–232. https://doi.org/10.1111/imr.12268.
Jones, B.W., Means, T.K., Heldwein, K.A., Keen, M.A., Hill, P.J., Belisle, J.T., Fenton, M.
References J., 2001. Different Toll-like receptor agonists induce distinct macrophage responses.
J. Leukoc. Biol. 69, 1036–1044.
Lewis, K.N., Liao, R., Guinn, K.M., Hickey, M.J., Smith, S., Behr, M.A., Sherman, D.R.,
Abdallah, A.M., Savage, N.D.L., van Zon, M., Wilson, L., Vandenbroucke-Grauls, C.M.J.
2003. Deletion of RD1 from Mycobacterium tuberculosis mimics bacille Calmette-
E., van der Wel, N.N., Ottenhoff, T.H.M., Bitter, W., 2008. The ESX-5 secretion
Guérin attenuation. J. Infect. Dis. 187, 117–123. https://doi.org/10.1086/345862.
system of Mycobacterium marinum modulates the macrophage response.
J. Immunol. 181, 7166–7175.

8
K.P. Pattanaik et al. Molecular Immunology xxx (xxxx) xxx

Li, W., Zhao, Q., Deng, W., Chen, T., Liu, M., Xie, J., 2014. Mycobacterium tuberculosis Qiu, L., Huang, D., Chen, C.Y., Wang, R., Shen, L., Shen, Y., Hunt, R., Estep, J., Haynes, B.
Rv3402c enhances mycobacterial survival within macrophages and modulates the F., Jacobs Jr, W.R., Letvin, N., Du, G., Chen, Z.W., 2008. Severe tuberculosis induces
host pro-inflammatory cytokines production via NF-kappa B/ERK/p38 signaling. unbalanced up-regulation of gene networks and overexpression of IL-22, MIP-
PLoS One 9. https://doi.org/10.1371/journal.pone.0094418 e94418–e94418. 1alpha, CCL27, IP-10, CCR4, CCR5, CXCR3, PD1, PDL2, IL-3, IFN-beta, TIM1, and
Lin, J., Chang, Q., Dai, X., Liu, D., Jiang, Y., Dai, Y., 2019. Early secreted antigenic target TLR2 but low antigen-specific cellular responses. J. Infect. Dis. 198, 1514–1519.
of 6-kDa of Mycobacterium tuberculosis promotes caspase-9/caspase-3-mediated https://doi.org/10.1086/592448.
apoptosis in macrophages. Mol. Cell. Biochem. 457, 179–189. https://doi.org/ Quesniaux, V.J., Nicolle, D.M., Torres, D., Kremer, L., Guérardel, Y., Nigou, J., Puzo, G.,
10.1007/s11010-019-03522-x. Erard, F., Ryffel, B., 2004. Toll-like receptor 2 (TLR2)-dependent-positive and TLR2-
Liu, S., Jia, H., Hou, S., Zhang, G., Xin, T., Li, H., Yuan, W., Guo, X., Gao, X., Li, M., 2014. independent-negative regulation of proinflammatory cytokines by mycobacterial
Recombinant TB10. 4 of Mycobacterium bovis induces cytokine production in lipomannans. J. Immunol. 172, 4425–4434. https://doi.org/10.4049/
RAW264. 7 macrophages through activation of the MAPK and NF-κB pathways via jimmunol.172.7.4425.
TLR2. Mol. Immunol. 62, 227–234. Roach, T.I., Barton, C.H., Chatterjee, D., Blackwell, J.M., 1993. Macrophage activation:
Liu, S., Jia, H., Hou, S., Xin, T., Guo, X., Zhang, G., Gao, X., Li, M., Zhu, W., Zhu, H., lipoarabinomannan from avirulent and virulent strains of Mycobacterium
2018. Recombinant Mtb9.8 of Mycobacterium bovis stimulates TNF-α and IL-1β tuberculosis differentially induces the early genes c-fos, KC, JE, and tumor necrosis
secretion by RAW264. 7 macrophages through activation of NF-κB pathway via factor-alpha. J. Immunol. 150, 1886–1896.
TLR2. Sci. Rep. 8, 1–11. Sabio, G., Davis, R.J., 2014. TNF and MAP kinase signalling pathways. Semin. Immunol.
Lugo-Villarino, G., Hudrisier, D., Tanne, A., Neyrolles, O., 2011. C-type lectins with a 26, 237–245. https://doi.org/10.1016/j.smim.2014.02.009.
sweet spot for Mycobacterium tuberculosis. Eur. J. Microbiol. Immunol. (Bp). 1, Sadek, M.I., Sada, E., Toossi, Z., Schwander, S.K., Rich, E.A., 1998. Chemokines induced
25–40. https://doi.org/10.1556/EuJMI.1.2011.1.6. by infection of mononuclear phagocytes with mycobacteria and present in lung
Målen, H., Berven, F.S., Fladmark, K.E., Wiker, H.G., 2007. Comprehensive analysis of alveoli during active pulmonary tuberculosis. Am. J. Respir. Cell Mol. Biol. 19,
exported proteins from Mycobacterium tuberculosis H37Rv. Proteomics 7, 513–521. https://doi.org/10.1165/ajrcmb.19.3.2815.
1702–1718. https://doi.org/10.1002/pmic.200600853. Satoh, T., Akira, S., 2016. Toll-like receptor signaling and its inducible proteins.
Mogensen, T.H., 2009. Pathogen recognition and inflammatory signaling in innate Microbiol. Spectr. 4 https://doi.org/10.1128/microbiolspec.MCHD-0040-2016,
immune defenses. Clin. Microbiol. Rev. 22, 240–273. https://doi.org/10.1128/ 10.1128/microbiolspec.MCHD-0040–2016.
CMR.00046-08. Sharma, S., Bose, M., 2001. Role of cytokines in immune response to pulmonary
Noss, E.H., Pai, R.K., Sellati, T.J., Radolf, J.D., Belisle, J., Golenbock, D.T., Boom, W.H., tuberculosis. Asian Pacific J. allergy Immunol. 19, 213–219.
Harding, C.V., 2001. Toll-like receptor 2-dependent inhibition of macrophage class II Shi, J., Zhang, H., Fang, L., Xi, Y., Zhou, Y., Luo, R., Wang, D., Xiao, S., Chen, H., 2014.
MHC expression and antigen processing by 19-kDa lipoprotein of Mycobacterium Biochemical and Biophysical Research Communications A novel firefly luciferase
tuberculosis. J. Immunol. 167, 910–918. https://doi.org/10.4049/ biosensor enhances the detection of apoptosis induced by ESAT-6 family proteins of
jimmunol.167.2.910. Mycobacterium tuberculosis. Biochem. Biophys. Res. Commun. 452, 1046–1053.
Olsen, A., Chen, Y., Ji, Q., Zhu, G., De Silva, A.D., Vilchèze, C., Weisbrod, T., Li, W., https://doi.org/10.1016/j.bbrc.2014.09.047.
Xu, J., Larsen, M., Zhang, J., Porcelli, S.A., Jacobs, W.R.C., 2016. Tumor necrosis Stanley, S.A., Raghavan, S., Hwang, W.W., Cox, J.S., 2003. Acute infection and
factor alpha-downregulating genes for the development of antituberculous vaccines. macrophage subversion by Mycobacterium tuberculosis require a specialized
MBio 7, e01023–15. https://doi.org/10.1128/mBio.01023-15. secretion system. Proc. Natl. Acad. Sci. U. S. A. 100, 13001–13006. https://doi.org/
Padhi, A., Pattnaik, K., Biswas, M., Jagadeb, M., Behera, A., Sonawane, A., 2019. 10.1073/pnas.2235593100.
Mycobacterium tuberculosis LprE suppresses TLR2-Dependent cathelicidin and Su, H., Kong, C., Zhu, L., Huang, Q., Luo, L., Wang, H., Xu, Y., 2015. PPE26 induces
autophagy expression to enhance bacterial survival in macrophages. J. Immunol. TLR2-dependent activation of macrophages and drives Th1-type T-cell immunity by
203, 2665–2678. https://doi.org/10.4049/jimmunol.1801301. triggering the cross-talk of multiple pathways involved in the host response.
Pai, R.K., Convery, M., Hamilton, T.A., Boom, W.H., Harding, C.V., 2003. Inhibition of Oncotarget 6, 38517–38537. https://doi.org/10.18632/oncotarget.5956.
IFN-γ-Induced class II transactivator expression by a 19-kDa lipoprotein from Takeda, K., Akira, S., 2005. Toll-like receptors in innate immunity. Int. Immunol. 17,
Mycobacterium tuberculosis : a potential mechanism for immune evasion. 1–14. https://doi.org/10.1093/intimm/dxh186.
J. Immunol. 171, 175–184. https://doi.org/10.4049/jimmunol.171.1.175. Taub, D.D., Turcovski-Corrales, S.M., Key, M.L., Longo, D.L., Murphy, W.J., 1996.
Pandey, H., Tripathi, S., Srivastava, K., Tripathi, D.K., Srivastava, M., Kant, S., Chemokines and T lymphocyte activation: I. Beta chemokines costimulate human T
Srivastava, K.K., Arora, A., 2017. Characterization of culture filtrate proteins Rv1197 lymphocyte activation in vitro. J. Immunol. 156, 2095–2103.
and Rv1198 of ESAT-6 family from Mycobacterium tuberculosis H37Rv. Biochim. Villalta, F., Zhang, Y., Bibb, K.E., Kappes, J.C., Lima, M.F., 1998. The cysteine-Cysteine
Biophys. Acta - Gen. Subj. 1861, 396–408. https://doi.org/10.1016/j. family of chemokines RANTES, MIP-1α, and MIP-1β induce trypanocidal activity in
bbagen.2016.10.013. human macrophages via nitric oxide. Infect. Immun. 66, 4690. LP – 4695.
Pasare, C., Medzhitov, R., 2005. Toll-like receptors: linking innate and adaptive Wickremasinghe, M.I., Thomas, L.H., Friedland, J.S., 1999. Pulmonary epithelial cells
immunity. Adv. Exp. Med. Biol. 560, 11–18. https://doi.org/10.1007/0-387-24180- are a source of IL-8 in the response to Mycobacterium tuberculosis: essential role of
9_2. IL-1 from infected monocytes in a NF-kappa B-dependent network. J. Immunol. 163,
Pecora, N.D., Gehring, A.J., Canaday, D.H., Boom, W.H., Harding, C.V., 2006. 3936–3947.
Mycobacterium tuberculosis LprA is a lipoprotein agonist of TLR2 that regulates Yang, S., Li, F., Jia, S., Zhang, K., Jiang, W., Shang, Y., Chang, K., Deng, S., Chen, M.,
innate immunity and APC function. J. Immunol. 177, 422–429. https://doi.org/ 2015. Early secreted antigen ESAT-6 of mycobacterium tuberculosis promotes
10.4049/jimmunol.177.1.422. apoptosis of macrophages via targeting the microrna155-SOCS1 interaction. Cell.
Physiol. Biochem. 35, 1276–1288. https://doi.org/10.1159/000373950.

You might also like