2020 Rodrigues TS JLB

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

DOI: 10.1002/JLB.

3MA0520-113R

ARTICLE

Mycobacterium tuberculosis-infected alveolar epithelial


cells modulate dendritic cell function through the
HIF-1𝜶-NOS2 axis

Tamara Silva Rodrigues1 * Annie Rocio Piñeros Alvarez2 * Ana Flávia Gembre1
Maria Fernanda Pereira de Araújo Demonte Forni4 Bruno Marcel Silva de Melo3 *
José Carlos Farias Alves Filho3 * Niels Olsen Saraiva Câmara4 *
Vânia Luiza Deperon Bonato1 *

1 Department of Biochemistry and Immunology,

Ribeirao Preto Medical School, University of Sao Abstract


Paulo, Ribeirao Preto, Brazil Tuberculosis kills more than 1 million people every year, and its control depends on the effec-
2 Indiana University – Purdue University
tive mechanisms of innate immunity, with or without induction of adaptive immune response. We
Indianapolis, Indianapolis, Indiana, USA investigated the interaction of type II alveolar epithelial cells (AEC-II) infected by Mycobacterium
3 Department of Pharmacology, Ribeirao Preto
tuberculosis with dendritic cells (DCs). We hypothesized that the microenvironment generated by
Medical School, University of Sao Paulo, Ribeirao
Preto, Brazil
this interaction is critical for the early innate response against mycobacteria. We found that AEC-
4 Transplantation Immunology Laboratory, II infected by M. tuberculosis induced DC maturation, which was negatively regulated by HIF-1𝛼-
Department of Immunology, Institute of inducible NOS2 axis, and switched DC metabolism from an early and short peak of glycolysis to a
Biomedical Sciences, University of Sao Paulo, low energetic status. However, the infection of DCs by M. tuberculosis up-regulated NOS2 expres-
Sao Paulo, Brazil
sion and inhibited AEC-II-induced DC maturation. Our study demonstrated, for the first time, that
Correspondence
HIF-1𝛼-NOS2 axis plays a negative role in the maturation of DCs during M. tuberculosis infection.
Vânia Luiza Deperon Bonato, Department of
Biochemistry and Immunology, University of Sao Such modulation might be useful for the exploitation of molecular targets to develop new thera-
Paulo, Ribeirao Preto Medical School, FMRP- peutic strategies against tuberculosis.
USP. Av. Bandeirantes 3900, Ribeirao Preto, SP
14049–900, Brazil.
KEYWORDS
Email: vlbonato@fmrp.usp.br
* Basic and Applied Immunology Program – epithelial cells, dendritic cells, mycobacterium tuberculosis, inducible nitric oxide synthase, hypoxia
FMRP-USP. Inducible factor alpha

1 INTRODUCTION to Mycobacterium tuberculosis are latently infected, innate immunity,


with or without induction of memory/adaptive immune response, must
The World Health Organization (WHO) estimates that there are play a critical role in the balance of progression of infection and
around 10 million people feeling ill with tuberculosis (TB), and approx- immunopathology.2 In this context, it is imperative to understand the
imately 7 million new cases were notified in 2018. The disease crosstalk among the first cells infected by M. tuberculosis, as these cells
accounted for 1.5 million of deaths in the same year, making it 1 of the drive the molecular mechanisms essential for controlling the dissemi-
10 leading causes of death by infectious diseases in the world (World nation of mycobacteria.
Health Organization).1 Because the great majority of subjects exposed The alveolar space consists mainly of 2 epithelial cells (ECs): type I
and type II alveolar epithelial cells (AEC-I and AEC-II). AEC-I are squa-
mous cells that constitute ∼95% of alveolar space, being the majority of
Abbreviations: AEC, alveolar epithelial cells; BCG, Bacillus Calmette Guerin; BMDC, bone
marrow-derived dendritic cell; BMDM, bone marrow-derived macrophages; ECAR,
ECs in the alveolar space. They are important for gas exchange due to
extracellular acidification rate; HIF-1𝛼, hypoxia inducible factor alpha; MMP1, their large surface area very close to capillaries.3–5 AEC-II are cuboidal
metalloproteinase 1; NIC, noninfected MLE-15 conditioned medium; OCR, oxygen
cells and comprises ∼5% of the alveolar space. They protect the lungs
consumption rate; OXPHOS, oxidative phosphorylation; TB, tuberculosis.

Received: 12 February 2020 Revised: 8 May 2020 Accepted: 17 May 2020


J Leukoc Biol. 2020;1–14. www.jleukbio.org ○
c 2020 Society for Leukocyte Biology 1
2 RODRIGUES ET AL .

from collapse, act on tissue repair after injury, and contribute to the 2 RESULTS
protection of the alveolar space through the secretion of surfactant
proteins.3,6,7 2.1 AECs are permissive to M. tuberculosis infection,
Human AEC-II (A549 cell line) take up M. tuberculosis through glyco- and they produce proinflammatory mediators
proteins described as syndecans8 and they are permissive to M. tuber-
culosis infection.9 AEC-II infected with M. tuberculosis secrete antimi- First, we determined if MLE-15, a mouse lung EC line, would be

crobial peptides,10,11,62 IFN-𝛾,12 CXCL8,13–15 CXCL5,16 NO,17,18 permissive to M. tuberculosis growth, as previously described for A549

CCL2,13 and metalloproteinase 1 (MMP1).19 However, EC may also cells, an adenocarcinomic human alveolar basal EC line.9 MLE-15

play an anti-inflammatory role as bronchial EC infected with Bacillus cells were also permissive to M. tuberculosis infection and allowed a

Calmette Guerin (BCG) secreted IL-10 and IL-22.20 progressive bacillus growth over 72 h of infection (Figs. 1A and 1B),

The interplay of AEC-II and macrophages has been investigated indicating that MLE-15 cells are unable to control bacterial growth.

in M. tuberculosis infection.21,22 AEC-II stimulated with keratinocyte Next, we investigated if M. tuberculosis-infected MLE-15 cells would

growth factor secreted GM-CSF which activates the microbicidal abil- produce inflammatory mediators. An increased production of nitrite

ity of bone marrow-derived macrophages (BMDM) by mechanisms (NO2- ), IL-6, CCL5, IFN-𝛾, and S100A9 was observed in the super-

dependent on NO production and phagolysosome fusion.21 In a differ- natant of infected cells (Figs. 1C–1G), whereas IL-1𝛽 and IL-10 were

ent approach, supernatants from THP-1 cells infected with M. tuber- not detected (data not shown).

culosis induced the secretion of S100 proteins by human primary


bronchial ECs.23 Recently, it was shown that the components of the
2.2 AECs infected by M. tuberculosis positively
alveolar lining fluid influence not only M. tuberculosis growth rate inside
AEC-II, but also the ability of those cells to modulate macrophage
modulate maturation of DCs
inflammatory response.24 Although AEC-II stimulated with keratinocyte growth factor increased
Besides AEC and macrophages, lung dendritic cells (DCs) can also the ability of BMDM to kill M. tuberculosis,21 it remains unclear if AEC-
be infected by M. tuberculosis.25,26 M. tuberculosis hampers DC matu- II infected by M. tuberculosis affect DC activation. To address this issue,
ration and migration. For instance, the expression of CD80 and CD86 we used 2 approaches to investigate the direct or indirect effect of
was only significantly enhanced 21 days post infection in the lymph M. tuberculosis-infected AEC-II on BMDC activation. The coculture of
nodes of infected BALB/c mice.27 It has been shown that M. tubercu- M. tuberculosis-infected MLE-15 and BMDC (Supplemental Fig. 1A)
losis can also prevent DC-mediated antigen presentation by a mech- increased the expression of CD80 (Supplemental Figs. 1B and 1C) and
anism independent of MHC-II regulation.28 DC activation also relies CD86 (Supplemental Figs. 1D and 1E), as well as increased the produc-
on metabolic changes. The activation of immature DCs requires switch tion of IL-1𝛽 (Supplemental Fig. 1F), IL-6 (Supplemental Fig. 1G), and
from oxidative phosphorylation (OXPHOS) and oxidation of fatty IL-10 (Supplemental Fig. 1H).
acids to higher glycolysis rate.29 Mammalian Target of Rapamycin and To assess the indirect effect of MLE-15, BMDCs were cul-
hypoxia inducible factor alpha (HIF-1𝛼) transcription factors have been tured/stimulated with supernatants obtained from M. tuberculosis-
shown to be critical for the switch to glycolysis during DC activation.29 infected MLE-15, which were described as M. tuberculosis-infected
However, it has also been shown that HIF-1𝛼 accumulation could MLE-15 conditioned medium (IC). Noninfected MLE-15 conditioned
induce prolonged down-regulation of CD80 and CD86 expression by medium (NIC) was used as a control (Fig. 2A). In addition, BMDCs
a mechanism dependent on increased inducible NOS2 expression, an were left unstimulated as a negative control or were stimulated
important HIF-1𝛼 target gene.30 with LPS as a positive control for BMDC activation. BMDCs were
The role of HIF-1𝛼–NOS2 axis and the associated metabolic sta- characterized as F4/80− CD11c+ CD11b+ MHC-II+ cells (data not
tus of DC activation in TB have not been explored. Besides, in the shown). IC increased the frequency of MHC-II-expressing DC (Fig. 2B),
context of TB, it has not been determined if the interference of CCR7 (Fig. 2C), CD80 (Fig. 2D), and CD86 (Fig. 2E) in comparison with
the microenvironment in the alveolar space, such as the infected NIC and unstimulated BMDC. As a positive control, LPS increased
ECs, could interfere with DC activation. To address this, we cul- the frequency of MHC-II-expressing DC (Fig. 2B), CCR7 (Fig. 2C),
tured CD11c+ CD11b+ F4/80− bone marrow-derived dendritic cells CD80 (Fig. 2D), and CD86 (Fig. 2E) in comparison with unstimulated
(BMDCs) with conditioned media obtained from the supernatants of BMDC. Besides, IC induced the production of both proinflammatory
M. tuberculosis-infected AEC-II and investigated the maturation and and anti-inflammatory cytokines by BMDC compared with NIC. A
activation status of DCs. We found that M. tuberculosis-infected AEC- significant increase in IL-1𝛽 (Fig. 2F), IL-10 (Fig. 2G), and IL-6 (Fig. 2H)
II induced DC maturation through a pathway partially dependent was observed when BMDCs were cultured with IC. Dotted line in the
on low levels of HIF-1𝛼-NOS2 expression. However, in vivo assays Fig. 2H represents the levels of IL-6 quantified in the supernatants of
showed that there was a higher expression of HIF-1𝛼 gene and higher AEC-II infected by M. tuberculosis (CM-IC). IL-1𝛽 and IL-10 were not
frequency of NOS2+ CD11b+ DCs in the lungs of M. tuberculosis- detected in the cultures of infected or uninfected AEC-II. These find-
infected mice. Therefore, in DCs infected with M. tuberculosis, infection ings show that indirect contact with AEC-II infected by M. tuberculosis
increases NOS2 expression such that AEC-II loses their ability to mod- induced the maturation of DCs and a mixed profile of proinflamma-
ulate DC maturation. tory and anti-inflammatory cytokines. Next, we determined if the
RODRIGUES ET AL . 3

F I G U R E 1 Alveolar epithelial cells are permissive to M. tuberculosis infection and exhibit a proinflammatory profile. Immortalized murine
lung epithelial cells (MLE-15) (1 × 106 cells/well) were infected with M. tuberculosis H37Rv (MOI10). (A) At 4 h after infection, M. tuberculosis was
observed by electronic microscopy. Full black arrows show the bacilli inside the cells. Dotted arrows show the vacuoles formed upon infection.
(B) CFU number was counted at 24, 48, and 72 h of infection. (C) NO2- production, and (D–G) IL-6, CCL5, IFN-𝛾, S100A9 secretion at 48 h of
infection by Griess assay and ELISA, respectively. The results are representative of 3 independent experiments expressed as the mean ± SEM
(n = 4/experimental condition). *P ≤ 0.05. CFU, colony forming units

maturation and activation of BMDC by IC would induce the prolifera- OXPHOS, measured as oxygen consumption rate (OCR) (Figs. 3A and
tion of naive CD4+ T cells. To do this, BMDCs were exposed to NIC or 3C). BMDC activation was characterized by metabolic shift toward
IC, and subsequently cocultured with CD4+ CD62L+ cells stimulated glycolysis, characterized by elevated levels of ECAR and decreased
with mAb anti-CD3. Figure 2I shows that BMDC previously stimulated respiration (Figs. 3E and 3F). BMDC exposed to NIC exhibited the
with IC significantly increased the proliferation of naive CD4+ T cells same pattern described for the cells cultured in media only, with
compared with CD4+ T cells stimulated with NIC. higher oxidative metabolism (Fig. 3B). However, cells exposed to IC
To confirm that the activation of BMDC was indeed associated with exhibited decreased ATP generation-dependent oxygen consumption
mediators secreted by infected AEC-II and not compounds from the (Fig. 3B), suggesting an impairment in the oxidative metabolism, and
mycobacterium, BMDCs were stimulated for 48 h with filtered media a prominent metabolic shift toward glycolysis as evident by more
from M. tuberculosis culture, which consisted only of mycobacterium than 4-fold increase in the levels of extracellular acidification rate
released products. M. tuberculosis-secreted products (Mtb-media) (ECAR) (Fig. 3E). After 24 h, the pattern of LPS stimulation remained
significantly reduced the expression of MHC-II (Supplemental Figs. consistent with that in the literature, with a significant glycolytic shift
2A and 2B), CCR7 (Supplemental Figs. 2C and 2D), and CD86 (Sup- (Figs 3C and 3F). BMDC exposed to NIC presented an even clearer
plemental Figs. 2E and 2F), and had no effect on NOS2 expression pattern of solid and strong oxidative metabolism, with increased levels
(Supplemental Figs. 2G and 2H) in BMDC. These findings show that of basal, maximal, ATP-linked, and spare capacity respiration when
mediators secreted by infected MLE-15 cells, and not compounds compared with the 2 h stimulation (Figs. 3B and 3D). In addition,
from the mycobacterium, induce the maturation and activation they showed lower levels of ECAR than those stimulated with LPS
of BMDC. (Figs. 3E and 3F). Although ECAR levels were elevated in BMDC
stimulated with IC for 2 h, the levels were consistently lower at 24 h of
IC stimulation compared with LPS and NIC stimulation (Fig. 3F). While
2.3 AECs infected by M. tuberculosis modulate
the cells were still able to respond to all the injected drugs, the overall
metabolism of DCs and HIF-1𝜶 accumulation
levels of oxygen consumption were greatly decreased (Fig. 3D), and
A metabolic switch to glycolysis has been shown to be involved in DC they displayed the lowest levels of ECAR after 24 h of IC stimulation
activation.29,31 Next, we investigated if the activation induced by MLE- compared with other culture conditions (Fig. 3F).
15 cells infected by M. tuberculosis would induce the same metabolic Next, we assessed the expression of HIF-1𝛼 because of its role in
pattern in BMDC. BMDCs left unstimulated (media) or stimulated with the maintenance of high glycolytic rates.32–35 HIF-1𝛼 gene and protein
LPS as negative and positive controls, respectively, confirmed that the expression had an early significant increase at 12 h and a reduction at
basal activation of BMDC was characterized by increased levels of 24 h (Figs. 3G and 3H) after IC stimulation. These findings support the
4 RODRIGUES ET AL .

F I G U R E 2 Alveolar epithelial cells infected by M. tuberculosis positively modulate maturation of DCs. (A) BMDC (1 × 106 cells/well) were
cultured for 24 h in different conditions: media (-), LPS 100 ng/ml (LPS), supernatants from noninfected MLE-15 cells (NIC), and M. tuberculosis-free
supernatants from infected MLE-15 cells (IC). (B) MHC-II, (C) CCR7, (D) CD80, (E) CD86 expression on CD11c+ CD11b+ F4/80− cells were analyzed
by flow cytometry. (F) IL-1𝛽, (G) IL-10, and (H) IL-6 secretion were measured at 24 h by ELISA. (I) Flow cytometry analysis of Ki67, a marker of
proliferation of naive CD4+ CD62L+ cells stimulated with mAb anti-CD3 and cocultured for 96 h with nonstimulated BMDC, or BMDC previously
stimulated with NIC or IC for 24 h. The results are representative of 3 independent experiments expressed as the mean ± SEM (n = 4/experimental
condition). *P ≤ 0.05; & P ≤ 0.05 compared with control (-)

early glycolytic peak followed by a lower rate of glycolysis as indicated 2.4 HIF-1𝜶-NOS2 axis plays a negative role in
by the low ECAR at 24 h of stimulation with IC (Fig. 3F). In addition, maturation of DCs
we found an early increase in the gene expression of the glucose trans-
Considering the early peak of glycolysis and the initial accumulation
porter, glut1 (Fig. 3I), and hk2 (hexokinase 2) (Fig. 3J) in IC-stimulated
of HIF-1𝛼, we hypothesized that HIF-1𝛼 would play a role in the
BMDC when compared with NIC-stimulated BMDC. These findings
increased maturation of DCs induced by infected MLE-15 cells. A
indicate an early and nonpersistent increase in both glycolysis and HIF-
prolyl-hydroxylase inhibitor (DMOG) was used to maintain the expres-
1𝛼 expression. To confirm our findings, the expression of NOS2, a HIF-
sion of HIF-1𝛼. DCs treated concomitantly with IC and DMOG lost the
1𝛼 target gene, was also measured by flow cytometry. BMDC exposed
ability to regulate MHC-II (Fig. 4A), CCR7 (Fig. 4B), CD80 (Fig. 4C), and
to IC exhibited no change in NOS2 expression (Figs. 3K and 3L) in com-
CD86 (Fig. 4D), when compared with those stimulated with only IC. As
parison with unstimulated BMDC, BMDC exposed to NIC, and those
expected, the addition of DMOG in BMDC cultures stimulated with IC
stimulated with LPS. NO2- detection in the supernatants of BMDC
induced the expression NOS2 in the CD11b+ DCs population (Fig. 4E).
stimulated with IC are NO2- levels secreted by the single infected MLE-
Interestingly, the majority of NOS2+ cells did not express costimula-
15 cells (CM-IC, dotted line), indicating that the BMDC stimulated with
tory molecules while cells expressing costimulatory molecules barely
IC did not produce extra levels of NO2- (Fig. 3M).
marked positive for NOS2 as shown by the representative analysis
RODRIGUES ET AL . 5

F I G U R E 3 Alveolar epithelial cells infected by M. tuberculosis modulate metabolism of DCs and HIF-1𝜶 accumulation. BMDC (1 × 106
cells/well) were cultured in different conditions: media (-), LPS 100 ng/ml (LPS), supernatants from noninfected MLE-15 cells (NIC) and M.
tuberculosis-free supernatants from infected MLE-15 cells (IC). Real time whole-cell respiration was assessed in (A and B) 2 and (C and D) 24 h.
Oligomycin, CCCP and rotenone plus antimycin were added where indicated. Levels of extracellular acidification rate were measured as a proxy for
lactate production during glycolysis at (E) 2 h and (F) 24 h of induction. (G and H) Gene and protein expression of hif-1𝛼/HIF-1𝛼, gene expression of
(I) glut1 and (J) hk2 (hexokinase 2) were evaluated at indicated times. (K) Frequency of CD11c+ CD11b+ MHC-II+ F4/80− NOS2+ and (L) NOS2 MFI
at 24 h of culture. (M) Levels of NO2- . The results are representative of 3 independent experiments expressed as the mean ± SEM (n = 4/experi-
mental condition). *P ≤ 0.05; & P ≤ 0.05 compared with control (-)
6 RODRIGUES ET AL .

F I G U R E 4 HIF-1𝜶-NOS2 axis plays a negative role in maturation of DCs. (A–F) BMDC (1 × 106 cells/well) from WT mice were cultured for 24 h
in different conditions: media (-), LPS 100 ng/ml (LPS), supernatants from noninfected MLE-15 cells (NIC) and supernatants from infected MLE-15
cells (IC), and IC + DMOG (200 𝜇M). (A) MHC-II, (B) CCR7, (C) CD80, (D) CD86, and (F) NOS2 expression were evaluated by flow cytometry. (F)
Representative analysis of costimulatory molecules and NOS2 expression by flow cytometry. (G–J) BMDC (1 × 106 cells/well) from WT and nos2-/-
mice were cultured for 24 h in different conditions: supernatants from noninfected MLE-15 cells (NIC) and supernatants from infected MLE-15
cells (IC), and IC + DMOG (200 𝜇M). (G and H) MHC-II, and (I and J) CD86 were evaluated by flow cytometry. The results are representative of 3
independent experiments expressed as the mean ± SEM (n = 4/experimental condition). *P ≤ 0.05; and P ≤ 0.05 compared with control (-), ns, not
significant

of MHC-II-, CCR7-, CD80-, CD86-, and NOS2-expressing DCs in the Next, we evaluated the expression of MHC-II, CD80, CD86, and
Fig. 4F. CCR7 on BMDC generated from WT or nos2−/− mice during the con-
DCs treated with IC plus DMOG produced higher levels of IL-1𝛽 comitant treatment with IC and DMOG. As previously observed, IC
(Supplemental Fig. 3A), IL-6 (Supplemental Fig. 3B), and IL-10 (Supple- stimulation positively regulated MHC-II (Figs 4G and 4H) and CD86
mental Fig. 3C) compared with DCs stimulated with IC. As expected, (Figs. 4I and 4J), whereas the addition of DMOG to IC media reduced
NO2- production was dependent on HIF-1𝛼. Dotted line represents the expression of both molecules. The deficiency of NOS2 in DCs
NO2- production by M. tuberculosis-infected MLE-15 cells (Supplemen- had no effect on the expression of MHC-II (Figs. 4G–4I) and CD86
tal Fig. 3D). (Figs. 4G–4I).
RODRIGUES ET AL . 7

The collected findings of in vitro experiments addressing the inter- CCR7 expression, along with the secretion of IL-1𝛽, IL-6, and IL-10.
action of AEC-II and DCs show that the maturation of DCs induced The maturation of DCs is dependent on soluble mediators, secreted
by products secreted after the infection of MLE-15 by M. tuberculo- by infected AEC-II, which switch the metabolism of DCs from an early
sis is associated with a mixed metabolic pattern (low OCR and ECAR), and short glycolysis induction to a low as well as a mixed OXPHOS
reduced expression of HIF-1𝛼 and NOS2, and absence or low produc- and glycolysis pattern. Moreover, the outcome of DC activation is
tion of NO2- . DC maturation, characterized by positive regulation of the absence of HIF-1𝛼-induced NOS2 expression, and consequently, a
MHC-II, CD80, CD86, and CCR7 expression, is dependent on reduced basal production of NO. However, the maturation of DCs induced by M.
expression of HIF-1𝛼 and NOS2. tuberculosis-infected AEC-II is dependent on whether DCs have been
previously infected by the mycobacterium or not. This is a critical issue
to be addressed because the interaction of infected AEC-II might take
2.5 M. tuberculosis hampers AEC-II ability to induce
place with either noninfected or infected DCs in the airways. Our study
maturation of DCs through HIF-1𝜶–NOS2 axis
highlights that the interaction of infected AEC-II with noninfected DCs
We transferred our in vitro findings to an in vivo microenvironment and their maturation determine the induction of adaptive immune
by evaluating HIF-1𝛼 gene expression in the lungs of animals infected response, and the subversion of DC maturation by M. tuberculosis infec-
with M. tuberculosis (Fig. 5A). We found an increased gene expression of tion cannot be restored by contact with infected AEC-II. In the con-
HIF-1𝛼 in the lungs of 30 day-infected animals (Fig. 5B). In addition, we text of leishmaniasis, noninfected DCs activated by soluble molecules
verified the presence of CD11b+ and NOS2+ in lung CD11c+ MHC-II+ of the parasite induces the production of TNF and the activation of T
myeloid cells from infected animals, whereas the expression of NOS2 cells.36 Similarly, in TB, we suggest that optimal migration and activa-
was absent in the cells from noninfected animals (Figs. 5C–5E). Next, tion may also occur through noninfected DCs, which might induce T
we investigated the interaction of AEC-II with M. tuberculosis-infected cell priming. The activation of noninfected DCs could be dependent
BMDC (Fig. 5F). Corroborating to in vivo results, M. tuberculosis is able on the microenvironment such as soluble mediators secreted by M.
to increase NOS2 expression in infected DCs (Figs. 5G and 5H). Fur- tuberculosis-infected AEC-II, and on the uptake of the extracellular vesi-
thermore, there was no significant difference in the NOS2 expres- cles, exosomes, apoptotic bodies, and nonvesicular antigens released
sion of infected BMDC and that of infected BMDC stimulated with IC by infected cells.37,38
(Figs. 5G and 5H). Clearly, Mtb-infected cells had a decrease in NOS2 The in vitro infection of DCs with mycobacteria, BCG,39 or M.
expression when treated with HIF-1𝛼 inhibitor (Figs. 5G and 5H). Fig- tuberculosis40 increased CD80 and CD86 expression. However, in vivo
ures 5I–5L show that infected DCs stimulated with IC were not capa- M. tuberculosis infection down-regulated CD80 and CD86 expression
ble of up-regulating MHC-II or CD86, as previously observed for non- in lung myeloid cells.27 It has been reported that M. tuberculosis is effi-
infected BMDC stimulated with IC. IC plus DMOG were not involved cient in reducing the expression of MHC-II, and consequently the func-
in the down-regulation of MHC-II and CD86, as previously observed tion of DCs.41 Therefore, the down-regulation of MHC and costimu-
for noninfected BMDC stimulated with IC plus DMOG. The inability of latory molecules is 1 of the many strategies used by M. tuberculosis to
infected DCs to up-regulate MHC-II and CD86 was directly dependent disrupt the activation of immune response. Besides, in vivo, we found a
on NOS2 because M. tuberculosis-infected DCs from nos2-deficient higher gene expression of HIF-1𝛼 in the lungs of mice infected with M.
mice exhibited a significant increase in the expression of MHC-II and tuberculosis. A previous study reported that HIF-1𝛼 accumulated in the
CD86. These data suggest that M. tuberculosis infection induces the lungs of TB patients with chronic infection, and it was associated with
production of NO by DCs. Although M. tuberculosis infection inhibited MMP1 expression and lung cavitation.42 In addition, the accumulation
the infected AEC-II-induced maturation of DC, it induced the secretion of HIF-1𝛼 in the lungs of infected BALB/c mice indicate that this tran-
of high levels of IL-1 𝛽 and IL-6 (Figs. 5M and 5N). Furthermore, soluble scription factor is a negative regulator of apoptosis and an indicator of
mediators secreted by infected AEC-II had no additional influence on the chronic phase of infection.43
the production of these cytokines. Similarly to what was reported by previous studies with A549
Figure 6 outlines our findings. AECs infected by M. tuberculo- human cell line,12–14,44–46 we demonstrated that murine M.
sis induced DC maturation, which was negatively regulated by HIF- tuberculosis-infected AEC-II also secrete proinflammatory molecules.
1𝛼-NOS2 axis. However, such modulation was not observed in M. We also showed that MLE-15 cells are permissive to M. tuberculosis
tuberculosis-infected DCs, which exhibited a significant increase of infection, and they are unable to control the bacillus growth. An
NOS2 expression that was not affected by supernatants of infected extensive characterization of soluble mediators secreted by AEC-II
AEC-II cells (IC). infected by M. tuberculosis is important to seek potential targets that
induce the activation of DCs. In addition to IL-6, IFN-𝛾, S100A9, and
CCL5, infected MLE-15 produce high levels of NO, and we speculate
3 DISCUSSION that some of these molecules modulates the metabolism of DCs, and
consequently their maturation and activation.
In the present study, we show, for the first time, that murine AEC-II Although the interaction of AEC-II and DCs has not been investi-
(MLE-15 cell line) infected by M. tuberculosis induce the maturation of gated in TB, the interaction of M. tuberculosis-infected macrophages
DCs, characterized by the up-regulation of MHC-II, CD80, CD86, and with noninfected AEC-II has been investigated. It was recently
8 RODRIGUES ET AL .

F I G U R E 5 M. tuberculosis hampers AEC-II ability of inducing maturation of DCs through HIF-1𝜶-NOS2 axis. (A) C57BL/6 mice were infected
with 1 × 105 M. tuberculosis by intratracheal route. (B) HIF-1𝛼 gene expression and (C–E) CD11c+ MHC-II+ CD11b+ NOS2+ cells were evaluated
in the lungs at 30 days of infection. (F) BMDC from WT and/ or nos2−/− mice were infected with M. tuberculosis (MOI2) and cultured in different
in different conditions: media (-), HIF-1𝛼 inhibitor (HIF-1𝛼 i, 100 𝜇M), supernatants from noninfected MLE-15 cells (NIC) and supernatants from
infected MLE-15 cells (IC). (G and H) NOS2, (I and J) MHC, (K and L) CD86, (M) IL-1𝛽, and (N) IL-6 were evaluated at 24 h of infection. The results
are representative of 3 independent experiments expressed as the mean ± SEM (n = 4/experimental condition). *P ≤ 0.05; & P ≤ 0.05 compared with
control (-)
RODRIGUES ET AL . 9

F I G U R E 6 Interaction of type II alveolar epithelial cells with DCs in the M. tuberculosis infection. Alveolar epithelial cells infected by M. tuber-
culosis secrete mediators that induce the maturation of noninfected DCs by a mechanism dependent on negative modulation of HIF1𝛼-NOS2 axis.
On the contrary, M. tuberculosis infection impairs that maturation of DCs, and alveolar epithelial cells have no capacity to overcome that

reported that macrophages infected by M. tuberculosis positively colytic enzymes, even in the presence of oxygen.51 The energy source
modulated MMP1 production by A549 human cell line.42 In addi- of M. tuberculosis-infected macrophages is dependent on aerobic
tion, noninfected MLE-15 treated with keratinocyte growth factor glycolysis.52 IFN-𝛾 activation induced the accumulation of HIF-1𝛼 in
secreted GM-CSF and enhanced the death of M. tuberculosis-infected infected macrophages, and positively regulated the secretion of IL-1𝛽,
macrophages.21 Therefore, it is worth noting that the interaction of IL-6, NO, and eicosanoids.53 In addition, the deficiency of HIF-1𝛼 in
infected macrophages with noninfected AEC-II has been investigated, macrophages impaired mycobacteria death, which inhibits the ability
but neither the interaction of the contrary nor that of AEC-II with DCs of IFN-𝛾 to control the pathogen’s growth54 and indicates that IFN-𝛾
has been studied. downstream signaling needs HIF-1𝛼-dependent pathways. Similarly,
Although several studies reported that M. tuberculosis or M. tuber- in zebra fish model, HIF-1𝛼 was important in the development of
culosis antigens increase or decrease the response of DCs,25,27,28,47 it resistance against M. marinum infection through the up-regulation
has not been reported that other cells in the microenvironment are of NOS2 expression.55 However, the role of HIF-1𝛼 and cellular
involved in this process. In the present study, M. tuberculosis-infected metabolism in DCs during M. tuberculosis infection has not yet been
MLE-15 cells secreted soluble mediators that induced the maturation explored. Our findings show that at the late stage of DC activation by
of BMDC. Therefore, M. tuberculosis-infected AEC-II indirectly acti- AEC-II, there was no stabilization of HIF-1𝛼 protein and DCs remained
vated the maturation of DCs. in a metabolic state of adaptation with low rate of glycolysis and
Cell metabolism plays an important role in immune activation.48 OXPHOS. Besides, there was a basal expression of NOS2 after 24 h
It has been reported that DC activation is associated with shift in of stimulation, corroborating the associated lower accumulation of
metabolism from OXPHOS to glycolysis.49 The early increase in the HIF-1𝛼 at this time-point. DMOG treatment, which stabilizes HIF-1𝛼
expression of proteins associated with the glycolytic pathway such expression, induced a significant reduction in the expression of MHC-
as GLUT1 and HK2, as detected by extracellular acidification on the II, CD80, and CD86, indicating that prolonged accumulation of HIF-1𝛼
Seahorse equipment, suggested the participation of HIF-1𝛼, which was plays a negative role in the maturation of DCs. Our findings confirm
also accumulated at the early stage of cell culture. The involvement of the negative role of HIF-1𝛼 in DC maturation and consequently, its
HIF-1𝛼 in the metabolic reprogramming of cells for aerobic glycolysis is involvement in impaired DC-induced activation of naive T cells.30 HIF-
well established, even under normal oxygen conditions, a phenomenon 1𝛼, via NOS2-NO axis, induced mitochondrial dysfunction, inhibiting
known as Warburg Effect.32,50 This phenomenon is primarily found OXPHOS and switching cell metabolism to aerobic glycolysis as the
in cancer cells, as they have increased expression of HIF-1𝛼 and gly- only energy source.30 In the present study, we expected a high rate of
10 RODRIGUES ET AL .

both glycolysis and OXPHOS, but instead, DCs used both glycolysis USA) supplemented with 2% FBS (Thermo Fisher Scientific, Waltham,
and OXPHOS at reduced rates. To address the role of HIF-1𝛼-NOS2 MA, USA), 5 mg/L insulin, 10 𝜇g/ml transferrin, 3 ng/ml selenite,
axis in the maturation of DCs induced by M. tuberculosis-infected sodium, 10 nM hydrocortisone, 10 nM 𝛽-estradiol, and 1% peni-
AEC-II, we differentiated BMDC from NOS2-deficient mice, and cillin/streptomycin (complete RPMI).
confirmed that HIF-1𝛼 signaling depends on NOS2 expression and NO Dendritic cells were differentiated from bone marrow cells from
production for the down-regulation of DC maturation. C57BL/6 mice. After removing the femurs and performing the bone
Prolonged HIF-1𝛼 stabilization also had a negative role in the marrow flush, cells were incubated in RPMI media supplemented
expression of CCR7. A recent study demonstrated that early increase with 20 ng/ml GM-CSF, 10% FBS, 1% glutamine, and 0.1% 𝛽-
in the rate of glycolysis is essential for the oligomerization of CCR7.56 mercaptoethanol. In the third day, GM-CSF was added maintaining the
Therefore, the initial increase in the rate of glycolysis might also be 20 ng/ml concentration. After 7 days of culture, the cells are essentially
important for the up-regulation of CCR7. However, energy balance CD11c+ CD11b+ DCs. Atmosphere for both cells: 95% air; 5% CO2 .
via the 2 pathways, glycolysis and OXPHOS, may be essential for Temperature: 37◦ C.
the maintenance of the phenotype found in the present study. Low
rates of glycolysis and OXPHOS following indirect contact with M.
4.4 In vitro infection
tuberculosis-infected AEC-II and the effect of HIF-1𝛼-NOS2 axis in the
maturation of DC is the novelty of this study. In mouse model, NOS2 MLE-15 murine immortalized cells (1 × 106 cells) or BMDCs (previ-
played a protective role against M. tuberculosis infection,57,58 and the ously differentiated from bone marrow cells in RPMI media supple-
HIF-1𝛼-NOS2 axis has been studied in the context of macrophage mented with 20 ng/ml GM-CSF, 10% FBS 1% glutamine, and 0,1%𝛽-
killing.53,54 However, we show for the first time that HIF-1𝛼-NOS2 mercaptoethanol for 7 days) were placed in 24 wells plaques (Corn-
axis plays a negative role in the maturation of DCs. ing Incorporated, New York, USA). After 24 h, MLE-15 or BMDCs were
Finally, the crosstalk between nonhematopoietic cells (AEC-II) and infected with M. tuberculosis H37Rv for 4 h. MLE-15 was infected with
hematopoietic cells (DCs) during TB opens a new perception on the MOI10 and BMDC with MOI2. Bacilli growth by CFU (Counting Form-
function of AEC-II in the modulation of a microenvironment that facil- ing Units) counts was evaluated at times 4, 24, 48, and 72 h post infec-
itates the control of infection. The interaction of AEC-II infected by tion. After washing, cells were lysed with 0.05% saponin. Serial dilu-
M. tuberculosis and DCs generates a microenvironment suitable for tions were plated on 7H11 media (Difco Laboratories, Detroit, MI,
the activation of adaptive immune response. This might represent the EUA), and number of CFU was counted after incubation for 28–30 days
mechanism associated with latent infection, as well as the opportunity at 37◦ C and 5% CO2 . In order to inhibit HIF-1𝛼, a HIF-1𝛼 inhibitor (Cal-
to exploit molecular targets to develop new therapeutic tools. biochem, San Diego, CA, USA) 100 𝜇M was used in infected cells.

4.5 In vivo infection


4 METHODS C57BL/6 mice were randomly divided in noninfected (control) and
infected groups. Infection was performed by intratracheal administra-
4.1 Animals tion of 1 × 105 bacilli.60 The lower and middle right lobes of the lungs
C57BL/6 mice and nos2−/− mice (6–8 weeks) were obtained from the were collected and processed as previously reported.61 Part of the
breeding facility of the Ribeirao Preto Medical School at University cells (1 × 106 were used for DC characterization by flow cytometry
Sao Paulo (FMRP-USP). All animals were housed in Animal Biological analysis. For the CFU assay, serial dilutions (100, 1000, 10,000, and
Safety Level 3 (ABSL3) with sterile water and food. Mice experiments 100,000) of digested lungs were plated on supplemented 7H11 agar
were conducted in accordance with the ethical guide on the use of ani- medium. CFU number was counted after 28 days of incubation at 37◦ C,
mals of FMRP-USP, and were approved by the Ethics Committee on and the results were expressed as the log10 of CFU/lung. Lung cells
Animal Use of FMRP-USP (protocol number 198/2017). were counted in a CountessTM automated cell counter (Thermo Fisher
Scientific, Waltham, MA, USA). No blinding was used in animal experi-
ments.
4.2 Bacteria cultivation
M. tuberculosis H37Rv strain (American Type Culture Collection 27294) 4.6 In vitro experimental models
was grown in 7H9 Middlebrook Broth (DIFCO Laboratories, Detroit,
MI, EUA) for 10–11 days at 37◦ C. The culture was collected as previ- 4.6.1 DC and MLE-15 indirect contact assay
ously described.59 1 × 106 immortalized mouse lung epithelial cells (MLE-15) were plated
in 24 wells plate overnight for adhesion. Next, the cells were incubated
for 4 h with M. tuberculosis H37Rv (MOI 10). The supernatants or
4.3 Epithelial and DCs
conditioned medium (CM) of noninfected (NIC) or infected (IC) cells
Immortalized murine lung epithelial cells (MLE-15) were used. MLE- was collected after 48 h of incubation. CM was filtered with 0.22 𝜇M
15 cells were cultured in RPMI medium (Sigma–Aldrich, St Louis, MO, filter (Corning Incorporated, New York, USA) to confirm the absence
RODRIGUES ET AL . 11

of bacilli. Concurrently, BMDCs, predominantly immature CD11c+ of 95◦ C for 15 s, an annealing phase at 58◦ C for 30 s and an exten-
CD11b+ , were incubated for 24 h with IC or NIC as well as media (neg- sion phase at 72◦ C for 30 s. The threshold cycle (Ct) was used to ana-
ative control), LPS 100 ng/ml (positive control). In order to stabilize lyze samples. Gene expression was calculated as 2−(ΔΔCt), where ΔΔ
HIF-1𝛼, a prolylhydroxylase inhibitor, DMOG, dimetiloxaloilglicine, Ct = Δ Ct (sample) − Δ Ct (calibrator) and where Δ Ct (sample) = Ct
(Calbiochem, San Diego, CA, USA) 200 𝜇M was used in addition to IC. (target gene) − Ct (normalizer = 𝛽-Actin). The primer sequences used
Cells were analyzed for expression of surface markers, cytokines, HIF- for detection are the following: 𝛽-Actin (forward, 5′ -CCC TAG GCA
1𝛼, and target genes. To exclude the role of M. tuberculosis products in CCA GGG TGT GA-3′ ; reverse, 5′ -GCC ATG TTC AAT GGG GTA CTT
our results from IC group, we used as a control, M. tuberculosis-media, C-3′ ), hif-1𝛼 (forward: 5′ -ACC TTC ATC GGA AAC TCC AAA G-3′ ;
which consisted in RPMI media incubated with M. tuberculosis (1 × 107 reverse: 5′ -CTG TTA GGC TGG GAA AAG TTA GG-3′ ), hk2 (forward,
bacilli/well) for 48 h at the same conditions as infected MLE-15 cells. 5′ -CGG AAT GGG GAG CCT TTG G-3′ ; reverse: 5′ -GCC TTC CTT ATC
After 48 h, the media was collected and filtered with 0.22 𝜇M filter CGT TTC AAT GG-3′ ). Glut1 (forward, 5′ - ATG GAT CCC AGC AGC
(Corning Incorporated, New York, USA). AAG-3′ ; reverse: 5′ - CCA GTG TTA TAG CCG AAC TGC-3′ )

4.6.2 Coculture of MLE-15 cells and BMDCs 4.7.3 Western blotting


1× 106 MLE-15 cells were incubated for 4 h with H37Rv (MOI 1:10). HIF-1𝛼 expression in BMDC was determined by Western blotting
The wells were then washed twice with 1× PBS, and 200 𝜇g/ml gen- according to manufacturer’s instructions. Briefly, after transference,
tamicin was added to ensure the absence of extracellular bacilli. Then, the nitrocellulose membrane was incubated with monoclonal anti-HIF-
0.5 × 106 BMDC, predominantly immature CD11c+ CD11b+ , were 1𝛼 (Sigma–Aldrich, St Louis, MO, USA) at 4◦ C overnight, followed by
cocultured for 48 h with infected or uninfected MLE-15 cells. BMDCs incubation with anti-rabbit horseradish peroxidase-linked secondary
were analyzed for expression of surface activation markers (MHC-II, antibody (Sigma–Aldrich, St Louis, MO, USA) and developed using
CD80, CD86) and cytokines (IL-1𝛽, IL-6, and IL-10). enhanced chemiluminescence technique (ChemiDoc XRS System; Bio-
Rad Laboratories, Hercules, CA, USA). The membrane was stripped and
4.6.3 Coculture of BMDC with naive T cells reprobed with Gapdh as a loading control.
BMDCs (0.5 × 105 per well) were stimulated with NIC or IC for 24 h,
and cocultured for 96 h with CD4+ CD62L+ cells (1 × 105 per well) 4.7.4 Flow cytometry
stimulated with mAb anti-CD3 (1 𝜇g/ml; BD Biosciences, Franklin
Cell suspensions (1 × 106 cells) obtained from lungs or in vitro cul-
Lakes, NJ, USA) in U-bottom 96-well plates (BD Biosciences, Franklin
tures were incubated with supernatant of 2.4G2 cell lineage (con-
Lakes, NJ, USA). Naive CD4+ T cells were purified from spleen and
taining antibodies anti-Fc𝛾 RII/III) for 20 min at 4◦ C. Then, the cells
lymph nodes of naive C57BL/6 mice following the instructions of the
were incubated for 30 min at 4◦ C with monoclonal antibodies: F4/80
CD4+ CD62L+ T Cell isolation kit (Miltenyi Biotec, San Diego, CA, USA).
PercP Cy5.5 (Clone BM8- Cat No.:123127; Biolegend, San Diego,
Proliferation rate was evaluated in CD4+ Ki67+ cells.
CA, USA), CD11c-PECy7 (Clone N418 - Cat No.: 25-0114-82; eBio-
science, San Diego, CA, USA), CD11b BV711 (Clone M1/70 – Cat no.
4.7 Methods details 563168; BD Biosciences, Franklin Lakes, NJ, USA), CD11b APCcy7
(Clone M1/70 - Cat No.:557657; BD Biosciences, Franklin Lakes, NJ,
4.7.1 Cytokines and Griess assay
USA), MHC-II Alexa 488 (Clone M5/114.15.2 - Cat No.:562352; BD
The levels of cytokines in the supernatants of cell cultures were mea-
Biosciences, Franklin Lakes, NJ, USA), CD80 PE (Clone L307.4; Cat
sured using ELISA kits obtained from (BD Biosciences, Franklin Lakes,
No.:557227; BD Biosciences, Franklin Lakes, NJ, USA), CD86 PE (Clone
NJ, USA), BD OptEIATM Set (IL-10) and R&D Systems, Minneapolis, MN,
GL-1; Cat No.:553692; BD Biosciences, Franklin Lakes, NJ, USA), CCR7
USA (IL-6, IL-1𝛽), according to the manufacturer’s instructions. Griess
PE (Clone 4B12; Cat No.:560682; BD Biosciences, Franklin Lakes, NJ,
assay was performed to analyze nitrite production in the supernatants
USA), CD4 PEcy7 (Clone RM4-5; Cat No.:561099; BD Biosciences,
as previously described.17
Franklin Lakes, NJ, USA), Nos2 APC (Clone CXNFT; Cat No.:17-5920-
82; eBioscience, San Diego, CA, USA), Ki67 FITC (Clone SolA15; Cat
4.7.2 RNA extraction, cDNA, and q-PCR No.:11-5698-82; eBioscience, San Diego, CA, USA). For intracellular
RNA was extracted from 1 × 106 cells using illustra RNAspin Mini RNA staining, cells were previously permeabilized with 2% saponin. Stained
isolation kit (GE healthcare, Chicago IL, USA), and c-DNA was made cells were washed with PBS with 2% FBS (Thermo Fisher Scientific,
using High Capacity cDNA Reverse Transcription kit (Thermo Fisher Waltham, MA, USA) and fixed with PBS containing 1% formaldehyde
Scientific Baltics, UAB, Vilnius, Lithuania), following the manufacture’s (J.T. Baker Phillipsburg, NJ, USA). The samples (100,000 events) were
instructions. PCR was performed using Maxima SYBR Green/ROX assessed using a FACSCantoTM II cytometer (BD Biosciences, Franklin
qPCR Master Mix (2×) (Thermo Fisher Scientific, Waltham, MA, USA) Lakes, NJ, USA) and CellQuest software (BD Biosciences, Franklin
in the StepOnePlusTM Real-Time PCR System (Applied Biosystems, Lakes, NJ, USA). FlowJo software (v 7.6.5; Tree Star, Ashland, OR,
Foster City, CA, USA). The following conditions were used for amplifi- USA) was used for cell analysis. First, the cells were gated on FSC-A
cation: initial denaturation at 95◦ C for 10 min, proceeded by 40 cycles and FSC-H for exclusion of doublets, and then gated on FSC-A (size)
12 RODRIGUES ET AL .

and SSC-A (granularity) to select DC, which were characterized as ORCID


F4/80− CD11c+ CD11b+ /MHC-II+ .
Vânia Luiza Deperon Bonato

https://orcid.org/0000-0003-4189-2685
4.7.5 Mitochondrial respiration and extracellular
acidification assays
Mitochondrial respiration and ECARs were assessed using Seahorse
REFERENCES
Extracellular Flux XFe96 Analyzer (Seahorse Bioscience, Agilent
Technologies, Santa Clara, CA, USA). Before plating, the 96-well 1. World Health Organization. Global Tuberculosis Report. 2019. Available
at: http://apps.who.int/bookorders. Accessed: January 17, 2020.
Seahorse plate was coated for 2 h with poly-D-lysine solution. Initially,
2. Scriba TJ, Coussens AK, Fletcher HA. Human immunology of
100,000 DC were plated in each well with media. Twenty-four hours tuberculosis. Microbiol Spectr. 2017;5(1). https://doi.org/10.1128/
later, media was replaced by different conditioned or control media microbiolspec.TBTB2-0016-2016.
and maintained for 2 or 24 h, when the assay was performed. On the 3. Ward HE, Nicholas TE. Alveolar type I and type II cells. Aust N Z J Med.
day of the assay, media was changed to the XF Seahorse assay media 1984;14(5 Suppl 3):731-734.
4. Féréol S, Gabriel P, Fodil R, Bruno L. Cell mechanics of alveolar
and cells were maintained at 37◦ C on an incubator without CO2 for
epithelial cells (AECs) and macrophages (AMs). Respir Physiol Neurobiol.
1 h before being placed in the equipment. Four cycles were used to 2008;163(1–3):3-16.
measure basal OCR, followed by automatic drug injections delivered 5. Jain R, Barkauskas CE, Takeda N, et al. Plasticity of Hopx+ type I
in each well. First, oligomycin (1 𝜇g/ml; Sigma–Aldrich, St Louis, MO, alveolar cells to regenerate type II cells in the lung. Nat Commun.
2015;6:6727.
USA) was employed to obtain respiration-driven ATP synthesis and
6. Castranova V, Tucker JH, Miles PR. The alveolar type II epithelial
proton leak-driven respiration. After 3 cycles, Carbonyl cyanide cell: a multifunctional pneumocyte the alveolar type II epithelial cell:
chlorophenyl hydrazine (CCCP, 5 𝜇M; Sigma–Aldrich, St Louis, MO, a multifunctional pneumocyte. Toxicol Appl Pharmacol. 1988;93(3):
USA) was delivered to analyze maximal respiratory capacity. At final, 472-483.
7. Speth JM, Bourdonnay E, Penke LRK, et al. Alveolar epithelial cell–
rotenone (1 𝜇M; Sigma–Aldrich, St Louis, MO, USA) and antimycin
derived prostaglandin E2 serves as a request signal for macrophage
(1 𝜇M; Sigma–Aldrich, St Louis, MO, USA) were added to inhibit secretion of suppressor of cytokine signaling 3 during innate inflam-
complexes I and III and thus ablate mitochondrial oxygen consumption mation. J Immunol. 2016;196(12):5112-5120.
in order to determine nonmitochondrial respiration. The levels of 8. Zimmermann N, Saiga H, Houthuys E, et al. Syndecans promote
mycobacterial internalization by lung epithelial cells. Cell Microbiol.
extracellular acidification were measured simultaneously in the same
2016;18(12):1846-1856.
wells to account for the acidification produced by lactate production 9. Bermudez LE, Goodman J. Mycobacterium tuberculosis invades and
and secretion as a readout of glycolysis. replicates within type II alveolar cells. Infect Immun. 1996;64(4):1400-
1406.
10. Rivas-santiago B, Sada E, Tsutsumi V, et al. b -Defensin gene expres-
4.7.6 Statistical analysis sion during the course of experimental tuberculosis infection. Infect
Dis. 2006;194(5):697-701.
Data were presented as the mean ± SEM and analyzed by Prism 6.0
11. Rivas-Santiago B, Hernandez-Pando E, Carranza C, et al. Expression
(GraphPad Software, San Diego, CA). For the majority of in vitro assays, of cathelicidin LL-37 during Mycobacterium tuberculosis infection in
we used n = 4/group, and for in vivo assays we used n = 6/group. For human alveolar macrophages, monocytes, neutrophils, and epithelial
comparisons between experimental groups, 1-way ANOVA was per- cells. Infect Immun. 2008;76(3):935-941.
12. Sharma M, Roy S, Sharma S, Verma S. Pulmonary epithelial cells are
formed followed by Newman–Keuls multiple comparisons post-tests.
a source of interferon-𝛾 in response to Mycobacterium tuberculosis
Individual groups were compared using the Mann–Whitney for non- infection. Immunol Cell Biol. 2007;85(3):229-237.
parametric data or 2-tailed Student’s t-test for parametric data. Statis- 13. Lin Y, Zhang M, Barnes PF. Chemokine production by a human alve-
tically significant differences were indicated by P values ≤ 0.05. For in olar epithelial cell line in response to Mycobacterium tuberculosis.
Infect Immun. 1998;66(3):1121-1126. http://www.ncbi.nlm.nih.gov/
vivo studies, the was no exclusion from analysis.
pubmed/9488404%5Cnhttp://www.pubmedcentral.nih.gov/article
render.fcgi?artid=PMC108024. Available at:.
14. Wickremasinghe MI, Thomas LH, Friedland JS. Pulmonary epithe-
ACKNOWLEDGMENTS lial cells are a source of IL-8 in the response to Mycobacterium
tuberculosis: essential role of IL-1 from infected monocytes in
We thank Denise Ferraz, Izaira Brandão, and Wendy Martin Rios a NF-kappa B-dependent network. J Immunol. 1999;163(7):3936-
for technical support. We are also grateful to professor Niels Olsen 3947. http://www.ncbi.nlm.nih.gov/pubmed/10490995. Available at:.
Saraiva Câmara (ICB- USP) for allowing us to use the Seahorse equip- (Accessed: June 10, 2018).
15. Boggaram V, Gottipati KR, Wang X, Samten B. Early secreted anti-
ment (grant 2018/08479-7). This work was supported by grants from
genic target of 6 kDa (ESAT-6) protein of Mycobacterium tubercu-
FAPESP (grants 2017/16538-0, and 2017/21629-5). losis induces interleukin-8 (IL-8) expression in lung epithelial cells
via protein kinase signaling and reactive oxygen species. J Biol Chem.
2013;288(35):25500-25511.
DISCLOSURES 16. Nouailles G, Dorhoi A, Koch M, et al. CXCL5-secreting pulmonary
epithelial cells drive destructive neutrophilic inflammation in tubercu-
The authors declare no competing interests. losis. J Clin Invest. 2014;124(3):1268-1282.
RODRIGUES ET AL . 13

17. Roy S, Sharma S, Sharma M, Aggarwal R, Bose M. Induction of nitric activated to promote T cell responses. J Immunol. 2008;181(9):6473-
oxide release from the human alveolar epithelial cell line A549: an in 6480.
vitro correlate of innate immune response to Mycobacterium tubercu- 37. Smith VL, Cheng Y, Bryant BR, Schorey JS. Exosomes function in anti-
losis. Immunology. 2004;112(3):471-480. gen presentation during an in vivo Mycobacterium tuberculosis infec-
18. Roy S, Sharma S, Sharma M, Bose M. Differential signaling of tion. Sci Rep. 2017;7:43578.
inducible nitric oxide synthase induction in Mycobacterium tubercu- 38. Alvarez-Jiménez VD, Leyva-Paredes K, García-Martínez M, et al.
losis infected alveolar epithelial cell line A549 in response to cytokines Extracellular vesicles released from Mycobacterium tuberculosis-
IFN-𝛾, TNF-𝛼 and IL-1𝛽. Int J Mycobacteriol. 2014;3(1):17-24. Infected neutrophils promote macrophage autophagy and
19. Elkington PTG, Emerson JE, Lopez-Pascua LDC, et al. Mycobacterium decrease intracellular mycobacterial survival. Front Immunol. 2018;
tuberculosis up-regulates matrix metalloproteinase-1 secretion from 9:272.
human airway epithelial cells via a p38 MAPK switch. J Immunol. 39. Demangel C, Bean AG, Martin R, Feng CG, Kamath AT, Britton
2005;175:5333-5340. WJ. Protection against aerosol Mycobacterium tuberculosis infection
20. Lutay N, Håkansson G, Alaridah N, et al. Mycobacteria bypass mucosal usingMycobacterium bovis Bacillus Calmette Guérin-infected den-
NF-kB signalling to induce an epithelial anti-inflammatory IL-22 and dritic cells. Eur J Immunol. 1999;29(6):1972-1979.
IL-10 response. PLoS One. 2014;9(1):e86466. 40. Tascon RE, Soares CS, Ragno S, Stavropoulos E, Hirst EMA, Colston MJ.
21. Pasula R, Azad AK, Gardner JC, Schlesinger LS, McCormack FX. Mycobacterium tuberculosis-activated dendritic cells induce protec-
Keratinocyte growth factor administration attenuates murine pul- tive immunity in mice. Immunology. 2000;99(3):473-480.
monary mycobacterium tuberculosis infection through granulocyte- 41. Gonzalez-juarrero M, Orme IANM. Characterization of murine lung
macrophage colony-stimulating factor (GM-CSF)-dependent dendritic cells infected with Mycobacterium tuberculosis. Society.
macrophage activation and phagolysosome fusion. J Biol Chem. 2001;69(2):1127-1133.
2015;290(11):7151-7159. 42. Belton M, Brilha S, Manavaki R, et al. Hypoxia and tissue destruction in
22. Reuschl AK, Edwards MR, Parker R, et al. Innate activation of human pulmonary TB. Thorax. 2016;71(12):1145-1153.
primary epithelial cells broadens the host response to Mycobacterium 43. Baay-Guzman GJ, Duran-Padilla MA, Rangel-Santiago J, et al. Dual
tuberculosis in the airways. PLoS Pathog. 2017a;13(9):e1006577. role of hypoxia-inducible factor 1 𝛼 in experimental pulmonary tuber-
23. Reuschl AK, Edwards MR, Parker R, et al. Innate activation of human culosis: its implication as a new therapeutic target. Fut Microbiol.
primary epithelial cells broadens the host response to Mycobacterium 2018;13:785-798.
tuberculosis in the airways. PLoS Pathog. 2017b;13(9):e1006577. 44. Robbins RA, Barnes PJ, Springall DR, et al. Expression of inducible
24. Scordo JM, Olmo-Fontánez AM, Kelley HV, et al. The human lung nitric oxide in human lung epithelial cells. Biochem Biophys Res Commun.
mucosa drives differential Mycobacterium tuberculosis infection out- 1994;203(1):209-218.
come in the alveolar epithelium. Mucosal Immunol. 2019;12(3):795- 45. Kwon OJ, Kim JH, Kim HC, et al. Nitric oxide expression in airway
804. epithelial cells in response to tubercle bacilli stimulation. Respirology.
25. Henderson RA, Watkins SC, Flynn JL. Activation of human dendritic 1998;3(2):119-124.
cells following infection with Mycobacterium tuberculosis. J Immunol. 46. Mvubu NE, Pillay B, McKinnon LR, Pillay M. Mycobacterium tubercu-
1997;159(2):635-643. losis strains induce strain-specific cytokine and chemokine response in
26. Mihret A. The role of dendritic cells in Mycobacterium tuberculosis pulmonary epithelial cells. Cytokine. 2018;14:53-64.
infection. Virulence. 2012;3(7):654-659. 47. Balboa L, Romero MM, Yokobori N, et al. Mycobacterium tuberculo-
27. Garcia-Romo GS, Pedroza-González A, Aguilar-León D, et al. Airways sis impairs dendritic cell response by altering CD1b, DC-SIGN and MR
infection with virulent Mycobacterium tuberculosis delays the influx profile. Immunol Cell Biol. 2010;88(7):716-726.
of dendritic cells and the expression of costimulatory molecules in 48. Pearce EL, Pearce EJ. Metabolic pathways in immune cell activation
mediastinal lymph nodes. Immunology. 2004;112(4):661-668. and quiescence. Immunity. 2013;38(4):633-643.
28. Wolf AJ, Linas B, Trevejo-Nuñez GJ, et al. Mycobacterium tuberculosis 49. Krawczyk CM, Holowka T, Sun J, et al. Toll-like receptor-induced
infects dendritic cells with high frequency and impairs their function in changes in glycolytic metabolism regulate dendritic cell activation.
vivo. J Immunol. 2007;179(4):2509-2519. Blood. 2010;115(23):4742-4749.
29. Pearce EJ, Everts B. Dendritic cell metabolism. Nat Rev Immunol. 50. Courtnay R, Ngo DC, Malik N, et al. Cancer metabolism and the War-
2015;15(1):18-29. burg effect: the role of HIF-1 and PI3K. Mol Biol Rep. 2015;42(4):841-
30. Lawless SJ, Kedia-Mehta N, Walls JF, et al. Glucose represses dendritic 851.
cell-induced T cell responses. Nat Commun. 2017;8:15620. 51. Lu H, Forbes RA, Verma A. Hypoxia-inducible Factor 1 activation by
31. O’Neill LAJ, Pearce EJ. Immunometabolism governs dendritic cell and aerobic glycolysis implicates the warburg effect in carcinogenesis. J
macrophage function. J Exp Med. 2016;213(1):15-23. Biol Chem. 2002;277(26):23111-23115.
32. Kim J, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated 52. Shi L, Salamon H, Eugenin EA, et al. Infection with Mycobacterium
expression of pyruvate dehydrogenase kinase: a metabolic switch tuberculosis induces the Warburg effect in mouse lungs. Sci Rep.
required for cellular adaptation to hypoxia. Cell Metab. 2006;3(3): 2015;5(1):18176.
177-185. 53. Braverman J, Sogi KM, Benjamin D, Nomura DK, Stanley SA. HIF-1𝛼
33. Yeung SJ, Pan J, Lee MH. Roles of p53, Myc and HIF-1 in regu- is an essential mediator of IFN-𝛾-dependent immunity to Mycobac-
lating glycolysis — the seventh hallmark of cancer. Cell Mol Life Sci. terium tuberculosis. J Immunol. 2016;197(4):1287-1297.
2008;65(24):3981-3999. 54. Osada-Oka M, Goda N, Saiga H, et al. Metabolic adaptation to glycol-
34. Corcoran SE, O’Neill LAJ. HIF1𝛼 and metabolic reprogramming in ysis is a basic defense mechanism of macrophages for Mycobacterium
inflammation. J Clin Invest. 2016;126(10):3699-3707. tuberculosis infection. Int Immunol. 2019;31(12):781-793.
35. Li C, Wang Y, Li Y, et al. HIF1 𝛼-dependent glycolysis promotes 55. Elks PM, Brizee S, van der Vaart M, et al. Hypoxia inducible factor sig-
macrophage functional activities in protecting against bacterial and naling modulates susceptibility to mycobacterial infection via a nitric
fungal infection. Sci Rep. 2018;8:3603. oxide dependent mechanism. PLoS Pathog. 2013;9(12):e1003789.
36. Carvalho LP, Pearce EJ, Scott P. Functional dichotomy of dendritic cells 56. Guak H, Al Habyan S, Ma EH, et al. Glycolytic metabolism is essential
following interaction with Leishmania braziliensis: infected cells pro- for CCR7 oligomerization and dendritic cell migration. Nat Commun.
duce high levels of TNF-alpha, whereas bystander dendritic cells are 2018;9(1):2463.
14 RODRIGUES ET AL .

57. Chan J, Tanaka K, Carroll D, Flynn J, Bloom BR. Effects of nitric oxide 62. Rivas-Santiago B, Contreras JCL, Sada E, Hernández-Pando R. The
synthase inhibitors on murine infection with Mycobacterium tubercu- potential role of lung epithelial cells and beta-defensins in experimen-
losis. Infect Immun. 1995;63(2):736-740. tal latent tuberculosis. Scand J Immunol. 2008;67(5):448-452.
58. Macmicking JD, North RJ, LaCourse R, Mudgett JS, Shah SK, Nathan
CF. Identification of nitric oxide synthase as a protective locus
against tuberculosis (Mycobacterium tuberculosisinfectious disease). SUPPORTING INFORMATION
Proc Natl Acad Sci USA. 1997;94:5243–5248.
59. Bonato VLD, Gonçalves EDC, Soares EG, et al. Immune regulatory Additional information may be found online in the Supporting Informa-
effect of pHSP65 DNA therapy in pulmonary tuberculosis: activa-
tion section at the end of the article.
tion of CD8+ cells, interferon-gamma recovery and reduction of lung
injury. Immunology. 2004;113(1):130-138.
60. Bonato VLD, Goncalves EDC, Santos RR, Silva CL. Genetic aspects and
How to cite this article: Rodrigues TS, Alvarez ARP,
microenvironment affect expression of CD18 and VLA-4 in experimen-
tal tuberculosis. Scand J Immunol. 2002;56(2):185-194. Gembre AF, et al. Mycobacterium tuberculosis-infected
61. Sérgio CA, Bertolini TB, Gembre AF, Prado RQ, Bonato VLD. alveolar epithelial cells modulate dendritic cell function
CD11c(+) CD103(+) cells of Mycobacterium tuberculosis-infected through the HIF-1𝛼-NOS2 axis. J Leukoc Biol. 2020;1–14.
C57BL/6 but not of BALB/c mice induce a high frequency of
https://doi.org/10.1002/JLB.3MA0520-113R
interferon-𝛾- or interleukin-17-producing CD4(+) cells. Immunology.
2015;144(4):574-586.

You might also like