You are on page 1of 10

DNA AND CELL BIOLOGY

Volume 36, Number 1, 2017


ª Mary Ann Liebert, Inc.
Pp. 58–66
DOI: 10.1089/dna.2016.3418

Wnt5a Deficiency Regulates Inflammatory Cytokine


Secretion, Polarization, and Apoptosis
in Mycobacterium tuberculosis-Infected Macrophages

Deming Chen,1 Guobao Li,2 Xiangdong Fu,2 Pei Li,2 Jie Zhang,2 and Lan Luo2

Tuberculosis, an infectious disease caused by Mycobacterium tuberculosis (MTB), is one of the global public health
catastrophes. Wnt signaling has recently been identified to exert immunoregulatory functions in a variety of
inflammatory and infectious diseases, including tuberculosis. The opposite expression of Wnt5a in human and mice
during MTB infection drives us to explore the roles and biological significances of reduced Wnt5a for MTB-treated
mice. In our study, the reduction of WNT5A in MTB-treated mice lung tissues or MTB-infected mice bone marrow-
derived macrophages (BM-Mø) was in a dose- and time-dependent manner. Then, WNT5A-silenced mice, secreted
frizzled-related protein 1 (SFRP1)-overexpressed or -silenced mice BM-Mø, were constructed to regulate Wnt5a
levels. When Wnt5a is deficient, MTB-induced increases of pro-inflammatory cytokines (TNF-a, IL-1b, IL-12, and
IL-6) can be markedly attenuated in mice lung tissues or BM-Mø. Besides, external disturbance triggered that Wnt5a
lower expression can induce Mø to be M2 phenotype and enhance cell apoptosis of MTB-infected mice BM-Mø.
Hence, the reduction of Wnt5a is a tactful strategy adopted by Mø to resistant MTB-induced immune responses and
to enhance MTB-induced Mø apoptosis in mice. Our study revealed a new style for Mø to manipulate themselves
against MTB infection. Our research identifies that Wnt5a deficiency can regulate inflammatory cytokine secretion,
polarization, and apoptosis in MTB-infected Mø.

Keywords: Sfrp1, Wnt5a, polarization, inflammatory cytokine, Mycobacterium tuberculosis, macrophage

Introduction survival strategy for virulent MTB (Kremer et al., 1997). The
immune response of Mø to MTB infections has been suffi-

T uberculosis is one of the global public health catas-


trophes leading to more than 8 million cases of illness
and 2 million deaths annually (Dheda et al., 2005). The
ciently studied in great detail. On the contrary, still little is
known about how Mø manipulate themselves to resistant MTB-
induced immune responses and apoptosis.
pathogenesis of tuberculosis is characterized by the granu- Wnt signaling pathways are well known as a group of
lomatous lesion formation in the lung due to Mycobacterium highly conserved principles among metazoan species, which
tuberculosis (MTB) infection (Russell et al., 2009). The first are responsible for essential biological processes in em-
cell type encountered by invaded MTB is the alveolar bryogenesis and tissue homeostasis (Angers and Moon,
macrophages (Mø) (Ernst, 1998; Heldwein and Fenton, 2009; van Amerongen and Nusse, 2009). Wnt family pro-
2002), which are the primary host cells for MTB, and fre- teins regulate multiple cellular functions, including prolif-
quently aggregate to form the early granuloma core medi- eration, apoptosis, migration, survival, differentiation, and
ated by the binding of hyaluronic acid to CD44 on Mø polarity, by activating various intracellular signaling cas-
surface (Love et al., 1979; Green et al., 1988). cades (Kikuchi et al., 2009). In essence, Wnts have also
It is well demonstrated that Mø play a pivotal role in in- been identified to exert immunoregulatory functions in a
flammation by releasing of and responding to numerous in- variety of inflammatory and infectious disease, including
flammatory cytokines and chemokines (Pereira et al., 2008). tuberculosis (Schaale et al., 2013).
Within Mø, MTB is able to stably multiply and can efficiently Currently, there are at least 19 Wnt members that have been
induce Mø apoptosis, which is associated with killing of intra- revealed to be present in human and mice (Kikuchi et al.,
cellular MTB (Molloy et al., 1994; Oddo et al., 1998). Hence, it 2009). Among which, Wnt5a, negatively regulated by secreted
seems that inhibiting apoptosis of Mø might be an effective frizzled-related protein 1 (Sfrp1) (Surinkaew et al., 2015), was

1
ICU, Shenzhen Third People’s Hospital, Shenzhen, China.
2
Department of Pulmonary Diseases, Shenzhen Third People’s Hospital, Shenzhen, China.

58
WNT5A DEFICIENCY REGULATES MTB-INFECTED MØ 59

reported to be the first member of Wnt family (Schaale et al., lung tissues of these mice were removed to prove the suc-
2013). Generally, Wnt5a was induced in multiple inflamma- cess of infection by colony enumeration assay (Hölscher
tory diseases, such as sepsis (Pereira et al., 2008), atheroscle- et al., 2001). After infection for indicated days (0, 7, 14, or
rosis (Christman et al., 2008), and tuberculosis (Blumenthal 21 days, n = 3 for each group), lung tissues of these mice
et al., 2006), and commonly exert pro-inflammatory effects on were removed for use in following experiments. Experi-
MTB-infected human Mø (Blumenthal et al., 2006; Schaale ments were performed under BSL3 conditions.
et al., 2011). While in mice, Reiling and colleagues discovered
that Wnt5a was markedly reduced in the lung tissue of MTB- Construction of Wnt5a siRNA recombinant adenovirus
treated mice, as well as in MTB-infected mice bone marrow-
derived macrophages (BM-Mø) (Schaale et al., 2013), which Wnt5a special and nonspecial (normal-control, NC) siR-
aroused our interest in the roles and biological significances of NA sequences were designed and synthesized by Gene-
reduced Wnt5a in MTB-infected mice BM-Mø. Pharma Co., Ltd. (Shanghai, China). Sequences were listed
In the present study, the reducing of WNT5A in MTB- as follows: Special: 5¢-GAA GCC CAU UGG AAU AUU
treated mice or MTB-infected mice BM-Mø was verified ATT-3¢ (sense) and 5¢-UAA UAU UCC AAU GGG CUU
first. Through silencing or inhibiting of WNT5A gene facti- CTT-3¢ (antisense); Nonspecial: 5¢-UUC UCC GAA
tiously, we found that MTB infection, which induced in- CGU GUC ACG UUU-3¢ (sense) and 5¢-ACG UGA CAC
creases of inflammatory cytokine secretion, can be attenuated GUU CGG AGA AUU-3¢ (antisense). The Wnt5a siRNA
in Wnt5a lacked mice or mice BM-Mø and enhanced in recombinant adenovirus (Ad-Wnt5a-siRNA) was con-
Wnt5a overexpressed mice BM-Mø. Besides, when Wnt5a structed as previously described (Yang et al., 2014). Briefly,
was insufficient, Mø phenotype trended to be M2 and cell the synthesized siRNA sequences were amplified and sub-
apoptosis was significantly enhanced in MTB-infected BM- cloned into pAdTrack-cytomegalovirus (CMV) plasmids
Mø. These findings hinted us that, in mice, Wnt5a deficiency (Stratagene-Agilent Technologies, Waldbronn, Germany).
can lead to a resistance of Mø to MTB-induced immune The obtained pAdTrack-CMV-Wnt5a-siRNA or pAdTrack-
responses and reinforcement of MTB-induced Mø apoptosis. CMV-NC plasmids were homologously recombined with
pAdEasy-1 (Stratagene) in Escherichia coli strain BJ5183
(Stratagene). Then, the secondarily obtained recombinant
Materials and Methods
plasmids were transfected into HEK-293 cells (ATCC). After
Mycobacteria preparation amplification and purification, virus titers were measured by
the p24 ELISA Kit (Cell Biolabs, Inc., San Diego, CA).
M.tuberculosis H37Rv was purchased from American
Type Culture Collection (ATCC, Manassas, VA). Experi-
Treatment of mice with recombinant Ad-Wnt5a-siRNA
ments were performed under biosafety level 3 (BSL3)
conditions. H37Rv was cultured in Middlebrook 7H9 broth Through caudal vein injection, mice received 200 mL phos-
(Difco, Detroit, MI) containing 10% oleic acid albumin- phate buffer saline (PBS), which were used as Mock group
dextrose-catalase (OADC; Difco), 0.05% Tween 80 (Sigma- (n = 3), 200 mL recombinant Ad-NC (1 · 1010 plaque-forming
Aldrich, St. Louis, MO), and 0.2% glycerol (Sigma) at 37C units, PFUs) as rAd-NC group (n = 3), and 200 mL recombinant
in a shaking incubator until log phase (OD600*0.6) (Ven- Ad-Wnt5a-siRNA (1 · 1010 PFUs) as rAd-Wnt5a-siRNA group
ketaraman et al., 2005; Chandolia et al., 2014). Before use, (n = 9). Mice without any treatment were used as Control group
H37Rv was dispersed by aspiration through a 29-gauge (n = 3). After 14 days, a secondary injection (200 mL PBS to
needle (BD Biosciences, Franklin Lakes, NJ), vortexed, and mice in Mock group, 200 mL recombinant Ad-NC to mice in
then sonicated for 15 s in a bath sonicator (100 Watt, KQ- rAd-NC group, and 200 mL recombinant Ad-Wnt5a-siRNA to
100DB; Kunshan Ultrasound Co., Ltd., China). After soni- mice in rAd-Wnt5a-siRNA group) was performed. After an-
cation, suspensions were allowed to settle for 10 min, and the other 14 days, three mice in each group were sacrificed to
upper 500 mL were removed and stored for use in following analyze Wnt5a levels in lung tissues. The rest mice in rAd-
experiments (Schaale et al., 2013). For the determination of Wnt5a-siRNA group (n = 9–3) continuously received H37Rv
the number of colony-forming units (CFUs), 10-fold serial infection or not (1000 CFU for 21 days) as described above.
dilutions of the prepared H37Rv strain were plated in tripli- Meanwhile, normal mice treated with or without H37Rv (1000
cate onto Middlebrook 7H11 agar plates (Difco) containing CFU for 21 days) were used as appropriate controls, respectively.
10% OADC and incubated at 37C. After incubating for
21 days, CFU was calculated (Rahman et al., 2014). Preparation of murine BM-Mø
Murine BM-Mø were generated as previously described
Treatment of mice with H37Rv by aerosol infection
(Reiling et al., 2001). First, the femurs of three mice were
C57BL/6 mice (8 weeks, 24 – 0.7 g) were purchased from separated and flushed with HBSS (Sigma). To remove fi-
Shanghai Laboratory Animal Center, Chinese Academy of broblasts, the obtained cells were cultured at 37C overnight
Sciences (Shanghai, China) and housed under specific in DMEM (Sigma) containing 10% heat-inactivated FBS
pathogen-free conditions in accordance with institutional (Sigma), 10 mM HEPES (Sigma), 10 mM glutamine (Sigma),
guidelines. All animal experiments were approved by the 1 mM sodium pyruvate (Sigma), and 50 ng/mL macrophage
Animal Care and Use Committee of Shenzhen Third Peo- colony stimulating factor (M-CSF; Sigma). Then, the adherent
ple’s Hospital. For MTB infection, mice were infected with cells were collected and cultured in DMEM containing M-CSF
different doses of H37Rv (CFU: 0, 100, 500, and 1000; n = 3 (50 ng/mL) for 9 days. Cells were collected to identify the
for each group) through the aerosol route as described pre- markers (F4/80 and CD11b) of Mø by Flow cytometry.
viously (Reiling et al., 2002). After infection for 7 days, Fluorescein isothiocyanate (FITC)-labeled rat monoclonal
60 CHEN ET AL.

anti-mouse F4/80 antibody (Abcam, Cambridge, MA) and logical Engineering Technology and Service Corporation Ltd.
allophycocyanin (APC)-labeled rat monoclonal anti-mouse (Shanghai, China) and listed in Table 1. All reactions were
CD11b antibody (Abcam) were used for Mø identification. performed in triplicate. Data were calculated using 2-DDCt
method and normalized to b-actin.
Treatment of BM-Mø with H37Rv
Western blot analysis
In vitro experiments, BM-Mø in 24-well plates were in-
fected with DMEM containing different doses of H37Rv Proteins from mice lung tissues or BM-Mø were isolated
(multiplicities of infection, MOI: 0:1, 3:1, 6:1, or 9:1) for using RIPA lysis buffer (Sigma). Protein concentrations were
indicated times (0, 4, 8, or 12 h). Then, the plates were measured by BCA (Sigma). Then, 40 mg total proteins were
gently washed with HBSS (Sigma). After washing, cells separated by 10% SDS-PAGE gel and transferred to PVDF
were collected, and Wnt5a levels were measured by real- membranes (Sigma). After incubated with 0.5% skimmed milk
time quantitative polymerase chain reaction (RT-qPCR) and power at room temperature for 1 h, target proteins in the
Western blots. For inflammatory cytokine (TNF-a, IL-1b, membranes were probed with primary antibodies overnight at
IL-12, IL-6, Arg-1, MRC-1, and TGF-b) level detection by 4C. After washing, the membranes were incubated with cor-
RT-qPCR, cells were collected at 12 h postinfection with responding horseradish peroxidase (HRP)-conjugated second-
heat-inactivated H37Rv or H37Rv at a MOI of 6:1. ary antibodies at room temperature for 1.5 h. The bands were
visualized using enhanced chemiluminescence (Pierce, Rock-
Construction and transfection of Sfrp1 ford, IL). The levels of b-actin were used as internal reference.
overexpression vector All antibodies (Abs) were purchased from Abcam. Primary
Abs: rabbit polyclonal anti-Wnt5a Ab (ab72583; 1:2000), anti-
The cDNA of Sfrp1 was subcloned from total RNA of
b-actin Ab (ab8227; 1:3000); Secondary Abs: HRP-conjugated
mice BM-Mø with Pyrobest DNA polymerase (Takara,
goat polyclonal anti-rabbit IgG Ab (ab6721; 1:4000).
Kyoto, Japan) and inserted into a pcDNA3.1/V5-HisA vector
(Invitrogen, Carlsbad, CA). The obtained pcDNA3.1/V5-
Cell apoptosis assay
HisA-Sfrp1-vectors or pcDNA3.1/V5-HisA-empty-vectors
were transfected into BM-Mø in 6-well plates using Lipo- Mice BM-Mø prepared above were allowed to receive
fectamine 2000 (Sigma). After transfection for 48 h, cells heat-inactivated H37Rv or H37Rv infection at a MOI of 6: 1
were collected to analyze and verify Wnt5a levels. Mean- for 12 h. Then, cell apoptosis was analyzed by flow cy-
while, pcDNA3.1/V5-HisA-Sfrp1-vector transfected cells were tometry. Cells were stained by reagents in Annexin V-FITC
arranged to receive heat-inactivated H37Rv or H37Rv infection Apoptosis Detection Kits (Sigma) as per the manufacturer’s
with a MOI of 6:1 for 12 h. Then, mRNA levels of inflamma- proposal. In brief, cells were collected postinfection and
tory cytokine (TNF-a, IL-1b, IL-12, IL-6, Arg-1, MRC-1, and washed twice with PBS and then resuspended in 1· binding
TGF-b) in the above cells were analyzed by RT-qPCR. buffer (1 · 106 cells/mL) in a test tube. Next, 5 mL Annexin
V-FITC conjugate and 10 mL propidium iodide solution
Silencing of Sfrp1 expression with siRNA interference were added into the tube sequentially. After being incubated
in the dark for 10 min, cells were analyzed using a FACS
For Sfrp1 gene silence, siRNA transfection was per-
analyzer (BD Biosciences, San Jose, CA).
formed as per the manufacturer’s protocol. In brief, 0.2 nmol
mouse Sfrp1 siRNA (Thermo Fisher Scientific, Waltham,
MA) or corresponding Silencer Select Negative Control Table 1. List of Primers
No. 1 siRNA and 20 mL Lipofectamine 2000 were incubated
in 2 mL DMEM at room temperature for 1.5 h. The above Gene name Sequence from 5¢ to 3¢
mixtures were added to BM-Mø in six-well plates and cultured
at 37C for 48 h. Then, cells were collected to verify Wnt5a Wnt5a F GAATCCCATTTGCAACCCCTCACC
R GCTCCTCGTGTACATTTTCTGCCC
levels. Besides, Sfrp1-silenced cells were continuously in- TNF-a F GTGACAAGCCTGTAGCCCA
fected with heat-inactivated H37Rv or H37Rv with a MOI of R AAAGTAGACCTGCCCGGAC
6:1 for 12 h. Then, cells were collected to analyze inflamma- IL-1b F ATGGCAATCGTTCCTGAACTCAAC
tory cytokine secretion (TNF-a, IL-1b, IL-12, and IL-6). R CAGGACAGGTATAGATTCTTTCCTTT
IL-12p40 F ATCGTTTTGCTGGTGTCTCC
Real-time quantitative PCR R CTTTGTGGCAGGTGTACTGG
IL-6 F AGTAAGTTCCTCTCTGCAAGAGACT
Total RNA of mice lung tissues or BM-Mø was extracted R CACTAGGTTTGCCGAGTAGATCTC
using the High Pure RNA Kit (Roche, Mannheim, Germany). Sfrp1 F TCCTCCATGCGACAACGA
The purity and integrity of the obtained RNA were controlled R TGATTTTCATCCTCAGTGCAAACT
by absorbance at A 260/280. For each sample, 4 mg total RNA Arg-1 F TGACATCAACACTCCCCTGACAAC
was reverse transcribed using a High Capacity cDNA Archive R GCCTTTTCTTCCTTCCCAGCAG
Kit (Applied Biosystems, Foster City, CA) in a reaction vol- MRC-1 F TCTTTTACGAGAAGTTGGGGTCAG
ume of 30 mL. The acquired cDNA was used for amplification R ATCATTCCGTTCACCAGAGGG
using SYBR Green PCR Amplification Reagent Kit (Qiagen, TGF-b F AGACGGAATACAGGGCTTTCGATTCA
R CTTGGGCTTGCGACCCACGTAGTA
Hilden, Germany) as per the manufacturer’s instructions. b-actin F ATCGTGGGGCGCCCCAGGCACC
RT-qPCR was performed on an Applied Biosystems PRISM R CTCCTTAATGTCACGCACGATTTC
7500 Fast Sequence Detection System (Thermo Fisher Sci-
entific). Primers were synthesized by Shanghai Sangon Bio- F, forward; R, reverse.
WNT5A DEFICIENCY REGULATES MTB-INFECTED MØ 61

Statistical analyses suggests that Wnt5a mRNA level is significantly reduced at


All experiments were performed in triplicate in three in- each MOI (*p < 0.05) postinfection for 4 h. Based on the re-
dependent assays. Data were analyzed by Student’s t-test, sults, BM-Mø in 0:1 or 6: 1 MOI group were continuously
expressed as mean – standard deviation, and normalized to cultured for 8 or 12 h. Similarly, Wnt5a mRNA levels were
b-actin. *p < 0.05 or **p < 0.01 indicates a statistically sig- dramatically reduced with time in 6:1 MOI group (*p < 0.05,
nificant difference. All the statistical analyses were per- Fig. 1F). Together, these results reveal that Wnt5a ubiquitously
formed by SPSS 19.0 software. For Western blots, data were reduced temporality and spatiality in H37Rv-treated mice lung
analyzed using Image-Pro 6.0 software. tissues or in H37Rv-infected mice BM-Mø. And also, 1000
CFU for 21 days in vivo and 6: 1 MOI for 12 h in vitro were
Results
chosen for following experiments.

Wnt5a is reduced in MTB-infected mice Wnt5a deficiency attenuates MTB-induced


lung tissues or BM-Mø inflammatory cytokine secretion in mice lung tissues
As shown in Figure 1A, data from colony enumeration as- To explore the biological significance of reduced Wnt5a
says proved that H37Rv aerosol infection procedures were for MTB-treated mice, expression of Wnt5a was further
successful, which can satisfy the following research. Through silenced artificially by siRNA interference in vivo, which
flow cytometry, we confirmed that BM-Mø were successfully was verified by RT-qPCR (Fig. 2A) and Western blot
induced by M-CSF (Fig. 1D). To determine the proper dose (Fig. 2B). Then, mRNA levels of inflammatory cytokines
and time of MTB infection, mice were treated with H37Rv by were detected in mice lung tissues. The results shown in
different CFU, and BM-Mø were infected with H37Rv by Figure 2C suggest that TNF-a mRNA level was signifi-
different MOI as described in methods. In vivo assay, results cantly higher in H37Rv-treated group than that in untreated
shown in Figure 1B indicate that Wnt5a mRNA level is sig- group (**p < 0.01), which is due to multifactor and can be
nificantly reduced only at 1000 CFU (*p < 0.05) posttreatment partly reversed in H37Rv-treated Wnt5a-silenced group
for 7 days with a slight change. Based on the above result, the (*p < 0.05). And also, similar results can be observed for
rest of mice in 0 or 1000 CFU group was continuously raised to IL-1b (Fig. 2D), IL-12 (Fig. 2E), and IL-6 (Fig. 2F) detec-
14 or 21 days. As shown in Figure 1C, we observed that Wnt5a tion. These results indicate that Wnt5a with almost entire
mRNA levels were markedly reduced with infection time in deficiency can partially attenuate MTB-induced inflam-
1000 CFU group (*p < 0.05). In vitro assay, the result (Fig. 1E) matory responses in mice.

FIG. 1. Wnt5a mRNA levels in H37Rv-treated mice lung tissues or H37Rv-infected mice BM-Mø. Mice were allowed to receive
H37Rv treatment by different CFU (0, 100, 500, or 1000) for indicated days (0, 7, 14, or 21). The success of H37Rv aerosol infection
procedure by different CFU for 7 days was verified by colony enumeration assays. (A) CFU in mice lung tissues. RT-qPCR was
performed to measure Wnt5a mRNA levels. (B, C) Relative Wnt5a mRNA level in mice lung tissues. *p < 0.05 versus CFU = 0 or/
and Day = 0 group. (D) The markers (F4/80 and CD11b) for mice BM-Mø were identified by flow cytometry. Mice BM-Mø received
H37Rv infection by different MOI (0:1, 3:1, 6:1, 9:1) for indicated hours (0, 4, 8, 12). (E, F) Relative Wnt5a mRNA level in mice BM-
Mø. *p < 0.05 versus MOI = 0:1 or/and Hour = 0 group. BM-Mø, bone marrow-derived macrophages; CFU, colony-forming unit;
MOI, multiplicities of infection; RT-qPCR, real-time quantitative polymerase chain reaction.
62 CHEN ET AL.

FIG. 2. Wnt5a and inflammatory cytokine levels in mice lung tissues. Mice were allowed to receive H37Rv infection or
not by 1000 CFU for 21 days. Wnt5a levels were verified by RT-qPCR and Western blot. Inflammatory cytokine levels were
detected by RT-qPCR. (A) Relative Wnt5a mRNA level in lung tissues. (B) Expression of Wnt5a in lung tissues. (A, B)
**p < 0.01 versus Control group. Levels of mRNA were measured in lung tissues for TNF-a (C), IL-1b (D), IL-12 (E), and
IL-6 (F). (C–F) For H37Rv-treated group, *p < 0.05 or **p < 0.01 versus untreated group; for H37Rv-treated Wnt5a-
silenced group, *p < 0.05 versus H37Rv-treated group.

Inhibition of Wnt5a by Sfrp1 overexpression (Arg-1), mannose receptor (MRC-1), TGF-b, and TNF-a was
attenuates MTB-induced inflammatory cytokine measured in heat-inactivated H37Rv-infected, H37Rv-infected,
secretion in mice BM-Mø or uninfected cells by RT-qPCR, and Western blot. The results
Continuously, in vitro, we further explored the effects of shown in Figure 4A suggest that Arg-1 level is dramatically
up- or downregulated Wnt5a on inflammatory cytokine se- decreased in H37Rv-infected cells (*p < 0.05), which can be
cretion in mice BM-Mø. First, the up- or downregulation of slightly reversed in H37Rv-infected Sfrp1-overexpressed cells
Wnt5a was fulfilled in Sfrp1-silenced or -overexpressed (*p < 0.05). Similar results can be observed for MRC-1
mice BM-Mø, which were verified by RT-qPCR (Fig. 3A) (Fig. 4B) and TGF-b (Fig. 4C) detection, while opposite results
and Western blot (Fig. 3B). Following, mRNA levels of for TNF-a detection (Fig. 4D). For these molecules, there was
inflammatory cytokines were detected. The results shown in no difference between uninfected and heat-inactivated H37Rv-
Figure 3C suggest that TNF-a mRNA level was markedly infected cells. The results suggest that inhibition of Wnt5a by
higher in H37Rv-infected cells than that in uninfected or heat- Sfrp1 can induce mice BM-Mø trending to M2 phenotype in
inactivated H37Rv-infected cells (**p < 0.01), which can be MTB-infected cells, which hints that the reduced Wnt5a can
attenuated in H37Rv-infected Sfrp1-overexpressed cells also have a regulation effect on BM-Mø phenotype. This
(*p < 0.05), but enhanced in H37Rv-infected Sfrp1-silenced finding reveals a novel way for Mø to manipulate themselves to
cells (*p < 0.05). Similar results can also be observed for IL-1b resistant MTB-induced immune responses.
(Fig. 3D), IL-12 (Fig. 3E), and IL-6 (Fig. 3F) detection. For
these inflammatory cytokines, there was no difference between Inhibition of Wnt5a by Sfrp1 leads to apoptosis
uninfected and heat-inactivated H37Rv-infected cells. These increase of MTB-infected mice BM-Mø
results reveal that inhibition of Wnt5a by Sfrp1 can attenuate
MTB-induced inflammatory cytokine secretion in mice BM- To explore whether the reduced Wnt5a influences the apo-
Mø, which is in accordance with the results in vivo. ptosis of MTB-infected mice BM-Mø, cell apoptosis was de-
tected in heat-inactivated H37Rv- infected, H37Rv-infected,
or uninfected cells (Fig. 5A). As shown in Figure 5B, cell
Inhibition of Wnt5a by Sfrp1 induces mice BM-Mø
apoptosis rate was significantly increased in H37Rv-infected
trending to M2 phenotype in MTB-infected cells
cells (*p < 0.05), which can be further enhanced in H37Rv-
To examine whether the reduced Wnt5a has an influence on infected Sfrp1-overexpressed cells (*p < 0.05). These results
the phenotype of mice BM-Mø, the expression of arginase-1 indicate that inhibition of Wnt5a by Sfrp1 can lead to apoptosis
WNT5A DEFICIENCY REGULATES MTB-INFECTED MØ 63

FIG. 3. The effects of up- or downregulated Wnt5a on inflammatory cytokine secretion in mice BM-Mø. Mice BM-Mø
prepared in methods were allowed to receive heat-inactivated H37Rv infection or H37Rv infection by a MOI of 6:1 for 12 h.
Wnt5a levels were verified by RT-qPCR and Western blot. Inflammatory cytokine levels were detected by RT-qPCR. (A)
Relative Wnt5a mRNA level in BM-Mø. (B) Expression of Wnt5a in BM-Mø. (A, B) *p < 0.05 versus pcDNA3.1/V5-HisA-
empty or siRNA-NC cells. Levels of mRNA were measured in BM-Mø for TNF-a (C), IL-1b (D), IL-12 (E), and IL-6 (F).
(C–F) For H37Rv-infected cells, *p < 0.05 or **p < 0.01 versus uninfected or heat-inactivated H37Rv-infected cells; for
H37Rv-infected Sfrp1-overexpressed or -silenced cells, *p < 0.05 versus H37Rv-infected cells.

increase of MTB-infected mice BM-Mø, which results in the Discussion


destruction of survival environment for MTB. Hence, we
conclude that the reducing of Wnt5a is a tactful strategy Wnt proteins are well known for their role in embryonic
adopted by mice BM-Mø to manipulate theirselves to resistant development and tissue homeostasis (Angers and Moon,
MTB infection. 2009; van Amerongen and Nusse, 2009). Based on recently
64 CHEN ET AL.

FIG. 4. Changes of Arg-1,


MRC-1, TGF-b, and TNF-a
levels in mice BM-Mø. Mice
BM-Mø prepared in methods
were allowed to receive heat-
inactivated H37Rv infection
or H37Rv infection by a MOI
of 6:1 for 12 h. Levels of
mRNA were measured in
BM-Mø by RT-qPCR for
Arg-1 (A), MRC-1 (B),
TGF-b (C), and TNF-a (D).
For H37Rv-infected cells,
*p < 0.05 versus uninfected
or heat-inactivated H37Rv-
infected cells; for H37Rv-
infected Sfrp1-overexpressed
cells, *p < 0.05 versus
H37Rv-infected cells.

accumulated evidences, Wnt signaling was also shown to be induce Mø to trend to M2 phenotype, and increase cell
involved in the regulation of inflammatory processes and apoptosis of MTB-infected mice BM-Mø, which may lead
infectious diseases (Schaale et al., 2011). In this study, we to a prominent protective effect on mice from MTB infec-
discovered that external disturbance triggered Wnt5a lower tion. Our data revealed a novel strategy that was adopted by
expression can attenuate inflammatory cytokine secretion, Mø to manipulate themselves to resistant MTB-induced

FIG. 5. Changes of cell apoptosis in mice BM-Mø. Mice BM-Mø prepared in methods were allowed to receive heat-
inactivated H37Rv infection or H37Rv infection by a MOI of 6:1 for 12 h. (A) Cell apoptosis was measured by Annexin V-
FITC Apoptosis Detection Kits. (B) Quantification of relative apoptotic mice BM-Mø. For H37Rv-infected cells, *p < 0.05
versus uninfected or heat-inactivated H37Rv-infected cells; for H37Rv-infected Sfrp1-overexpressed cells, *p < 0.05 versus
H37Rv-infected cells.
WNT5A DEFICIENCY REGULATES MTB-INFECTED MØ 65

immune responses and enhance MTB-induced Mø apoptosis above, which will be illuminated in our further study. In
in mice, which can increase our understanding of the in- summary, our study revealed that Wnt5a deficiency can lead
teraction between MTB and Mø. to a resistance of Mø to MTB-induced immune responses
Currently, the pro-inflammatory effects of Wnt5a have and reinforce MTB-induced Mø apoptosis in mice.
been found in several inflammatory diseases, such as sepsis
(Chen et al., 2015), atherosclerosis (Bhatt et al., 2012), and Disclosure Statement
rheumatoid arthritis (de Sousa Rabelo et al., 2010). Wnt5a
can act through Ca2+/calmodulin-dependent protein kinase No competing financial interests exist.
(CaMKII) and that this pathway can contribute to the in-
flammatory response of Mø (Pereira et al., 2008), which is References
the first cell type encountered by invaded MTB (Ernst, Angers, S., and Moon, R.T. (2009). Proximal events in Wnt
1998). A Blumenthal et al. (2006) previously found that signal transduction. Nat Rev Mol Cell Biol 10, 468–477.
Wnt5a was induced in MTB-infected human Mø and plays a Benoit, M., Desnues, B., and Mege, J-L. (2008). Macrophage
pro-inflammatory effect. Similarly, our data also identified polarization in bacterial infections. J Immunol 181, 3733–
the pro-inflammatory roles of Wnt5a in MTB-infected mice 3739.
BM-Mø. Differently, reduction of Wnt5a was ubiquitously Bhatt, P.M., Lewis, C.J., House, D.L., Keller, C.M., Kohn, L.D.,
observed in MTB-treated mice lung tissues or MTB-infected Silver, M.J., et al. (2012). Increased Wnt5a mRNA expression in
mice BM-Mø in our study, which was in accordance with advanced atherosclerotic lesions, and oxidized LDL treated hu-
the findings by Reiling and colleagues (Schaale et al., 2013). man monocyte-derived macrophages. Open Circ Vasc J 5, 1–7.
The different response of Wnt5a expression to MTB- Blumenthal, A., Ehlers, S., Lauber, J., Buer, J., Lange, C.,
infection in human and mice Mø may hint different inter- Goldmann, T., et al. (2006). The Wingless homolog WNT5A
action pattern between MTB and Mø. and its receptor Frizzled-5 regulate inflammatory responses of
In our study, Wnt5a was proved to be significantly re- human mononuclear cells induced by microbial stimulation.
duced in MTB-treated mice lung tissues or MTB-infected Blood 108, 965–973.
mice BM-Mø in dose and time-dependent manner. In Wnt5a Chandolia, A., Rathor, N., Sharma, M., Saini, N.K., Sinha, R.,
deficiency mice lung tissues or BM-Mø, MTB-induced in- Malhotra, P., et al. (2014). Functional analysis of mce4A
creases of pro-inflammatory cytokines, including TNF-a, gene of Mycobacterium tuberculosis H37Rv using antisense
IL-1b, IL-12, and IL-6, can be markedly attenuated, which approach. Microbiol Res 169, 780–787.
indirectly reflected the pro-inflammatory effect of Wnt5a Chen, M., Zhong, W., Hu, Y., Liu, J., and Cai, X. (2015).
Wnt5a/FZD5/CaMKII signaling pathway mediates the effect
during MTB infection. Among these cytokines, TNF-a is
of BML-111 on inflammatory reactions in sepsis. Int J Clin
essential for lung granuloma formation of tuberculosis
Exp Med 8, 17824–17829.
patients (Chensue et al., 1994). Chensue, S.W., Warmington, K., Ruth, J., Lincoln, P., Kuo,
For the past few years, the concept of Mø polarization has M-C., and Kunkel, S.L. (1994). Cytokine responses during
been widely developed and further sophisticated (Schaale mycobacterial and schistosomal antigen-induced pulmonary
et al., 2013). At present, Mø are deemed as a spectrum of granuloma formation. Production of Th1 and Th2 cytokines
phenotypic subtypes ranging from classically activated Mø and relative contribution of tumor necrosis factor. Am J Pa-
(M1) to the alternatively activated Mø (M2) based on gene thol 145, 1105–1113.
expression induced in response to pathogen- or cytokine- Christman, M.A., Goetz, D.J., Dickerson, E., McCall, K.D.,
derived stimulation (Benoit et al., 2008; Mosser and Lewis, C.J., Benencia, F., et al. (2008). Wnt5a is expressed in
Edwards, 2008). In this study, meanwhile, we discovered murine and human atherosclerotic lesions. Am J Physiol
that Wnt5a deficiency can contribute to the tendency of Heart Circ Physiol 294, H2864–H2870.
mice Mø to be M2 phenotype, which can be reflected by the de Sousa Rabelo, F., da Mota, L.M.H., Lima, R.A.C., Lima,
slightly reversed expressions of Arg-1, MRC-1, TGF-b, and F.A.C., Barra, G.B., de Carvalho, J.F., et al. (2010). The Wnt
TNF-a in MTB-infected Wnt5a deficiency mice BM-Mø. signaling pathway and rheumatoid arthritis. Autoimmun Rev
Among the above molecules, Arg-1 and MRC-1 are the 9, 207–210.
well-known markers of M2 Mø (Yang et al., 2012). Dheda, K., Booth, H., Huggett, J.F., Johnson, M.A., Zumla, A.,
As always, the interaction between MTB and Mø is and Rook, G.A. (2005). Lung remodeling in pulmonary tu-
complex and difficult to illustrate. When MTB enters into berculosis. J Infect Dis 192, 1201–1210.
the distal airways, patrolling Mø can provide a critical in- Ernst, J.D. (1998). Macrophage receptors for Mycobacterium
tracellular niche that is required for MTB to infect alveolar tuberculosis. Infect Immun 66, 1277–1281.
Green, S.J., Tarone, G., and Underhill, C.B. (1988). Aggregation of
Mø and establish infection in the human host (Lee et al.,
macrophages and fibroblasts is inhibited by a monoclonal anti-
2009). However, induction of Mø apoptosis is associated body to the hyaluronate receptor. Exp Cell Res 178, 224–232.
with the killing of intracellular MTB (Molloy et al., 1994; Heldwein, K.A., and Fenton, M.J. (2002). The role of Toll-like
Oddo et al., 1998). In our study, Mø apoptosis was signifi- receptors in immunity against mycobacterial infection. Mi-
cantly increased in MTB-infected mice BM-Mø and further crobes Infect 4, 937–944.
markedly enhanced in MTB-infected Wnt5a deficiency mice Hölscher, C., Atkinson, R.A., Arendse, B., Brown, N., My-
BM-Mø, which are unfavorable for MTB survival. Hence, burgh, E., Alber, G., et al. (2001). A protective and agonistic
the reduction of Wnt5a in MTB-infected mice BM-Mø is a function of IL-12p40 in mycobacterial infection. J Immunol
tactful strategy adopted by Mø against MTB infection. Be- 167, 6957–6966.
sides, we guess that Wnt5a deficiency induced BM-Mø Kikuchi, A., Yamamoto, H., and Sato, A. (2009). Selective
apoptosis increase may entirely or partially contribute to the activation mechanisms of Wnt signaling pathways. Trends
reduction of inflammatory cytokine secretion mentioned Cell Biol 19, 119–129.
66 CHEN ET AL.

Kremer, L., Estaquier, J., Brandt, E., Ameisen, J.C., and Locht, Schaale, K., Brandenburg, J., Kispert, A., Leitges, M., Ehlers,
C. (1997). Mycobacterium bovis Bacillus Calmette Guerin S., and Reiling, N. (2013). Wnt6 is expressed in granuloma-
infection prevents apoptosis of resting human monocytes. Eur tous lesions of Mycobacterium tuberculosis—infected mice
J Immunol 27, 2450–2456. and is involved in macrophage differentiation and prolifera-
Lee, J., Hartman, M., and Kornfeld, H. (2009). Macrophage tion. J Immunol 191, 5182–5195.
apoptosis in tuberculosis. Yonsei Med J 50, 1–11. Schaale, K., Neumann, J., Schneider, D., Ehlers, S., and Reiling,
Love, S., Shannon, B., Myrvik, Q., and Lynn, W. (1979). Char- N. (2011). Wnt signaling in macrophages: augmenting and
acterization of macrophage agglutinating factor as a hyaluronic inhibiting mycobacteria-induced inflammatory responses. Eur
acid-protein complex. J Reticuloendothel Soc 25, 269–282. J Cell Biol 90, 553–559.
Molloy, A., Laochumroonvorapong, P., and Kaplan, G. (1994). Surinkaew, S., Dawson, K., Qi, X.-Y., Sun, Y., Huang, H.,
Apoptosis, but not necrosis, of infected monocytes is coupled Chen, Y., et al. (2015). Role of Wnts and secreted frizzled-
with killing of intracellular bacillus Calmette-Guerin. J Exp related protein-1 in atrial fibroblast pathophysiology during
Med 180, 1499–1509. atrial fibrotic remodeling. Circulation 132, A11625-A.
Mosser, D.M., and Edwards, J.P. (2008). Exploring the full spec- van Amerongen, R., and Nusse, R. (2009). Towards an inte-
trum of macrophage activation. Nat Rev Immunol 8, 958–969. grated view of Wnt signaling in development. Development
Oddo, M., Renno, T., Attinger, A., Bakker, T., MacDonald, 136, 3205–3214.
H.R., and Meylan, P.R. (1998). Fas ligand-induced apoptosis Venketaraman, V., Dayaram, Y.K., Talaue, M.T., and Connell,
of infected human macrophages reduces the viability of in- N.D. (2005). Glutathione and nitrosoglutathione in macro-
tracellular Mycobacterium tuberculosis. J Immunol 160, phage defense against Mycobacterium tuberculosis. Infect
5448–5454. Immun 73, 1886–1889.
Pereira, C., Schaer, D.J., Bachli, E.B., Kurrer, M.O., and Yang, L., Chu, Y., Wang, Y., Zhao, X., Xu, W., Zhang, P., et al.
Schoedon, G. (2008). Wnt5A/CaMKII signaling contributes (2014). siRNA-mediated silencing of Wnt5a regulates in-
to the inflammatory response of macrophages and is a target flammatory responses in atherosclerosis through the MAPK/
for the antiinflammatory action of activated protein C and NF-kB pathways. Int J Mol Med 34, 1147–1152.
interleukin-10. Arterioscler Thromb Vasc Biol 28, 504–510. Yang, Q., Shi, Y., He, J., and Chen, Z. (2012). The evolving
Rahman, M.A., Sobia, P., Gupta, N., Van Kaer, L., and Das, G. story of macrophages in acute liver failure. Immunol Lett
(2014). Mycobacterium tuberculosis subverts the TLR-2- 147, 1–9.
MyD88 pathway to facilitate its translocation into the cytosol.
PLoS One 9, e86886.
Reiling, N., Klug, K., Krallmann-Wenzel, U., Laves, R., Goyert, Address correspondence to:
S., Taylor, M.E., et al. (2001). Complex encounters at the Guobao Li, MS
macrophage-mycobacterium interface: studies on the role of the Department of Pulmonary Diseases
mannose receptor and CD14 in experimental infection models Shenzhen Third People’s Hospital
with Mycobacterium avium. Immunobiology 204, 558–571. No.29, Bulan Road
Reiling, N., Hölscher, C., Fehrenbach, A., Kröger, S., Longgang District
Kirschning, C.J., Goyert, S., et al. (2002). Cutting edge: toll- Shenzhen 518112
like receptor (TLR) 2-and TLR4-mediated pathogen recog- China
nition in resistance to airborne infection with Mycobacterium
tuberculosis. J Immunol 169, 3480–3484. E-mail: guobaoligbl@163.com
Russell, D.G., Cardona, P-J., Kim, M-J., Allain, S., and Altare,
F. (2009). Foamy macrophages and the progression of the Received for publication June 16, 2016; received in revised
human tuberculosis granuloma. Nat Immunol 10, 943–948. form September 26, 2016; accepted September 26, 2016.
Copyright of DNA & Cell Biology is the property of Mary Ann Liebert, Inc. and its content
may not be copied or emailed to multiple sites or posted to a listserv without the copyright
holder's express written permission. However, users may print, download, or email articles for
individual use.

You might also like