You are on page 1of 22

Relative contribution of IL-1α, IL-1β and TNF to the

host
response to Mycobacterium tuberculosis and
attenuated M. bovis BCG

Andi Nurul Ilmi


INTRODUCTION
• 1/3 of the global population considered to be infected

Tuberculosis
• Only 5–10% develop an active disease host immune system efficiently dealing with
the mycobacteria
• Immunodepression of the host  reactivation of latent tuberculosis infection

(TB)
• Neutralisation of TNF for the treatment of severe inflammatory diseases  reactivation
of latent tuberculosis and increased susceptibility to primary tuberculosis infection
• Coordinated innate and adaptive immune responses are required for efficient control of
M. tuberculosis infection

• Early studies  Protective role of the IL-1α/β pathway in M. tuberculosis infection

Studies
• Study by Mayer-Barber  myeloid cell populations co-expressing both IL-1α and IL-1β
that are regulated by Type I and Type 2 interferons
• IL-1β and downstream TNF production  caspase dependent restriction of intracellular
M. tuberculosis growt
INTRODUCTION
Relative contribution of IL-1α vs. IL-1β in the impairment of host response to acute M.
tuberculosis infection  in the absence of IL-1R1, and compare it to the well established
susceptibility of TNF deficient mice

We confirm that both TNF and IL-1 pathways are required to control M. tuberculosis

Presence of either IL-1α or IL-1β allows some control of acute M. tuberculosis infection

TNF is essential for the early control of infection by virulent or attenuated mycobacteria

IL-1 pathway is dispensable for controlling less virulent infection by M. bovis BCG.
RESULT: Cross-regulation of IL-1α and IL-1β release by
macrophages in response to mycobacteria

Mutual induction of IL-1α and IL-1β  after stimulation with LPS or turpentine

Bone marrow derived macrophages from mice deficient IL-1α and/IL-1 β, or IL-1R1  stimulated with M.
tuberculosis H37Rv / M. bovis BCG in vitro
• No IL-1α was detected in the IL-1a deficient mice (Fig. 1A) and no IL-1 β was detected in the IL-1β deficient mice (Fig. 1B).
• IL-1α  reduced in IL-1β deficient macrophages stimulated with TLR4 agonist LPS, M. tuberculosis H37Rv or M. bovis BCG
• IL-1β  reduced in the IL-1α deficient macrophages stimulated with LPS, M. tuberculosis H37Rv or M. bovis BCG.
• Absence of functional IL-1R1  little effect on the release of IL-1α or IL-1β (Fig. 1A and B)
• TNF release induced by mycobacteria was independent of the IL-1α/ β /IL-1R1 axis in vitro (Fig. 1C)
RESULT: Lethal M. tuberculosis infection in the absence of both IL-1α
and IL-1β, or TNF pathways
IL-1R1 deficient mice  extremely sensitive to virulent M. tuberculosis H37Rv infection

Mayer-Barber et al a rapid mortality of mice deficient for IL-1α and/or IL-1β, within 30–40 days post M. tuberculosis infection

Deficient for IL-1α or IL-1β  survived the acute phase of infection without bodyweight loss

Deficient for IL-1α and IL-1β  rapidly lost weight and succumbed within 5 weeks

Deficient for IL-1α or IL-1β  survived for the duration of the acute M. tuberculosis infection study (3 months) without clinical symptoms of infection.

Presence of either IL-1α or IL-1β allows some control of acute M. tuberculosis infection.
RESULT: Exacerbated pulmonary inflammation after M. tuberculosis
infection in the absence of IL-1α, IL-1β or IL-1R1, alike in the absence of
TNF
• IL-1α and IL-1β deficient mice  strongly increased lung weights, 5 weeks post-infection
• IL-1R1 deficient mice and TNF KO mice  increased lung weights at 4 weeks
• Deficient IL- 1α or IL-1β  less pulmonary inflammation on day 35 post-infection
• IL-1α or IL-1β deficient mice  survived to chronic infection and lung inflammation at2 or 3 months post-
Lung infection (Fig. 2F and G).
inflammation • Expression of IL-1α or IL-1β  helped controlling acute lung inflammation induced by M. tuberculosis infection.

• Mice deficient for IL-1α + IL-1β could not control the infection, similar to IL-1R1 deficient mice on day 35 post-
infection
• Deficiency in IL-1α or IL-1β  bacterial growth restriction on day 35 post-infection and a limited increase in
bacterial load at 2–3 months post-infection
Bacterial • Deficient in IL-1α+IL-1β  a rapid, exacerbated lung inflammation uncontrolled bacterial infection, as seen in
IL-1R1 or TNF deficient mice
loads
RESULT: Exacerbated
pulmonary inflammation after M.
tuberculosis infection in the
absence of IL-1α, IL-1β or IL-
1R1, alike in the absence of TNF
RESULT: Presence of either IL-1β or IL-1α prevents the acute necrotic
pneumonia seen after M. tuberculosis infection in IL-1 pathway defective mice,
but do not compensate for lack of TNF
Granuloma

Deficient for IL- Deficient IL-1α


1α and IL-1β or IL-1β

2–3 months
free alveolar post-
Macroscopic Microscopic limited oedema no necrosis
space infectionlung
pathology

extensive
reduction of increased
pleural large confluent severe massive MN neutrophil confluent abundant reduced free acid-fast bacilli
ventilated inflammatory oedema
adhesions nodules inflammation cell infiltrations necrosis and mycobacteria alveolar space abundance
alveolar spaces cell infiltration
oedema

• Absence of both IL-1α and IL-1β were similar to those seen in


The lung lesions mice deficient for IL-1R1 or TNF

• Recapitulates the acute necrotic pneumonia from defective IL-


Absence of IL-1α and IL-1β 1R1 or TNF pathways

• Limits infiltration of inflammatory cells, oedema and necrosis


Presence of IL-1α or IL-1β after acute M. tuberculosis infection.
Presence of IL-1α or IL-1b prevents acute necrotic Progressive pneumonia after 2–3 months M. tuberculosis infection
pneumonia in response to M. tuberculosis infection. in the absence of IL-1α or IL-1b
RESULT: Regulation of pulmonary cytokine expression by IL-1α/IL-1β
pathway after M. tuberculosis infection

Uncontrolled bacilli growth and pulmonary inflammation  deficient IL-1α and IL-1β

Complete absence of IL-1α + IL-1β  increased IFNg pulmonary levels, but decreased IL-12/23 p40 and IL-23 p19 expression

Absence of IL-1 pathway  on partial reduction of TNF pulmonary levels after in vivo M. tuberculosis infection

No reduction of IL-1α or IL-1β lung content in the absence of TNF.

The levels of IL- IL-1α and IL-1β TNF pulmonary


Lung IFNg levels IL-12/23 p40 levels IL-23 p19 levels
17A levels levels
• Highly elevated in • Reduced in • reduced in TNF- • reduced in • elevated in TNF- • reduced in the
deficient IL-1α and deficient IL-1α and deficient at 4 deficient IL-1α deficient at 4 absence of IL-1α
IL-1β or IL-1R1 at IL-1β weeks of infection and/or IL-1β or IL- weeks of infection and/or IL-1β, or
5 weeks after M. • Lower than wild- 1R1 at 5 weeks • elevated in IL-1R1 IL-1R1
tuberculosis type controls in post-infection deficient
infection deficient IL-1α and • in IL-1α or IL-1β • IL-1α levels were
• TNF deficient at 4 IL-1β deficient at 8 slightly increased
weeks • decreased in IL- weeks post in IL-1β deficient
• Highly elevated in 1R1 deficient or infection mice
IL-1β deficient IL-1α or IL-1β, 5 • IL-1β levels were
IL-1β sufficed to weeks after slightly increased
control IFNg over- infection. in IL-1α deficient
expression.
RESULT: Regulation of pulmonary cytokine expression by IL-1α/IL-1β
pathway after M. tuberculosis infection

Cytokine pulmonary levels in M. tuberculosis infected TNF or


IL-1 deficient mice.
RESULT: IL-1 pathway is dispensable for controlling attenuated M.
bovis BCG infection, while TNF is essential

Mice deficient for MyD88  highly susceptible to virulent M. tuberculosis infection while they controlled chronic M.
bovis BCG infection and less susceptible to M. tuberculosis

IL-1R1 pathway  controlling acute infection


•Previous reports deficiency IL-1α or IL-1β  early death after infection or survival up to 3–6 months at lower dose infection
•TNF deficient mice  rapidly lost weight and succumbed by 6 weeks of infection
•IL-1R1 and IL-1α+IL-1β deficient  survived systemic M. bovis BCG infection

Histologically
• Liver and lung inflammation were exacerbated in TNF deficient mice
• The pathology was much less severe in IL-1R1 or IL-1α plus IL-1β deficient mice

IL-1 pathway  essential for controlling virulent M. tuberculosis infection is dispensable for the early control
of attenuated M. bovis BCG infection
TNF is crucial, but IL-1α/IL-1β pathway is
dispensable for controlling M. bovis BCG
infection.
DISCUSSION
• Neutralization of TNF in the treatment of severe inflammatory reactivation of latent tuberculosis and increased
susceptibility to primary tuberculosis infection
• IL-1 has been neutralized using the IL-1R antagonist Anakinra with good safety record  short half-life administered
daily
Neutralization • Efficient and persistent IL-1 neutralization with long-lasting anti-IL-1 antibodies  reduced host resistance and flare-up
of M. tuberculosis infection.

• Both IL-1α and IL-1β are critically required for host resistance
• Absence of both IL-1α+IL-1β recapitulated the overwhelming infection and acute necrotic pneumonia seen in IL-1R1
deficient mice
Mayer-Barber • Presence of IL-1α or IL-1β  allowed to control acute M. tuberculosis infection and pulmonary inflammation,
et al

The presence of IL-1α or IL-1β is sufficient to trigger IL-1R pathway essential


component in the development of innate response to acute M. tuberculosis infection
DISCUSSION

Inter-dependence of IL-1α and IL-1β expression and/or release


• IL-1β acting as a shuttle for secretion of mature IL-1α

Bone marrow-derived macrophages


• Macrophage deficient for IL-1α  defective in IL-1β production in vitro
• Macrophages deficient IL-1α  reduction IL-1β release in response to mycobacterial antigen stimulation
Indirect IL-1 amplification loop unlikely
• Absence of functional IL-1R1 did not hamper the release of IL-1α or IL-1β.

Coproduction of IL-1α and IL-1β at the single cell level


• In distinct myeloid cell populations in the lung of M. tuberculosis infected mice
DISCUSSION
• Slightly increased pulmonary IL-1α levels in IL-1β deficient mice during acute infection
• Increased concentrations of IL-1α reported in BAL and lungs of IL-1β deficient mice
• Absence of IL-1α and IL-1β  increased pulmonary levels of IFNg for IL-1R1-deficient mice
• Absence of IL-1α and IL-1β  Increased IFNg levels reported in the BAL of these mice 4 weeks post-infection
Our report • Increased IFNg levels single absence of IL-1α or IL-1β, or of TNF consequence of the failure of the innate immune response

• BAL IL-1α levels unaffected in IL-1β deficient mice, and conversely, 4 weeks after M. tuberculosis infection,
• Ly6GnegCD11bpos myeloid cells producing IL-1α and/or IL-1β were halved in these mice
Recent
study

• Th1 promoting cytokines were upregulated in the absence of IL-1 or TNF pathways
• There was little effect on IL-12/ 23 p40 and rather a decrease in IL-23 p19 expression.
• Increased IL-1β levels in single IL-1α deficient mice  associated with a partial correction of IFNg and IL-23 p19 levels
Our model • IL-17 was strongly reduced in IL-1 deficient mice  contribute to the enhanced susceptibility of these mice
DISCUSSION
High bacterial burden associated with acute M. tuberculosis infection in mice lacking TNF or functional IL-1
pathway abundant PAMPs to trigger cytokine release directly.

vitaPAMPs Acute, exacerbated M. tuberculosis infection.

High levels of IL-1β and TNF  in the BAL and lung of TRIF/MyD88 double deficient mice classical
TLR agonists, nor TRIF-dependent vitaPAMPs are essential for the increase in cytokine release

DAMPs Released by dying or necrotic cells to entertain the inflammatory context in highly infected
lungs

DAMPsDo not seem essential for in vivo


DISCUSSION
High levels of IL-1β,
TNF, IFNg and TLR, MyD88-
Mice deficient MyD88 chemokines MCP1 dependent pathways
and MIP1a 5 weeks were not essential
after infection

Mice deficient for NLRP3, ASC or caspase-1,


control acute M.
or for NLRP2, AIM2 or purinergic receptor tuberculosis infection
P2X7

NLRP3 thiol
Immunopathology nitrosylation and
The control by NO of M. tuberculosis and IL-1β production inhibition of NLRP3
inflammasome

Reduced expression of iNOS in IL-1 defective susceptibility to M.


tuberculosis
mice 5 weeks post-infection infection.

Molecular •IL-1β directly activates host resistance to M. tuberculosis


•Similar to Tim3, the ligand of Galectin-9 expressed on M. tuberculosis infected macrophages
•IL-1β inhibited the replication of M. tuberculosis in infected macrophages, Increased TNF release and

mechanisms TNFR1 cell surface expression caspase-3 activation, apoptosis, and restriction of M. tuberculosis
growth
DISCUSSION
Hypothesised  requirement for IL-1 responses depends on the virulence of the mycobacteria.

• MyD88 deficient mice highly susceptible to virulent M. tuberculosis infection control chronic M. bovis BCG infection and less susceptible to M.
tuberculosis
•TNF deficient mice highly susceptible to virulent M. tuberculosis and attenuated M. bovis BCG

Different regulation of caspase-1 dependent IL-1β release by type I IFNs

•Absence of IL-1R1, or IL-1α plus IL-1β did not compromise the control of a M. bovis BCG infection

The IL-1 pathway is dispensable for the immediate, innate response to control M. bovis BCG infection

•IL-1β requires TNF pathway to inhibit M. tuberculosis replication


•T-cell derived TNF is important at later stages of the infection

TNF is essential for the early control of mycobacterial infection

• TNF antimicrobial effects are independent of IL-1β


•TNF of macrophage/neutrophil origin being crucial for the immediate response

TNF essential for the IL-1 pathway  not Long-lasting


Presence of IL-1a or IL-
early control of infection central for controlling neutralizing monoclonal
1b  allows some
by either virulent or less virulent antibodies preferable
control of acute M.
attenuated mycobacteria such as to target specifically IL-
tuberculosis infection
mycobacteria M. bovis BCG. 1b, or IL-1a
MATERIALS AND METHODS
Bacteria and infection Pulmonary infection with
clumping was disrupted M. tuberculosis H37Rv
• M.
M. tuberculosis
tuberculosis H37Rv
H37Rv diluted in sterile saline
(Pasteur) aliquots kept by 30 repeated
containing 0.05% Tween • Performed
Performed by
by delivering
delivering
Mice frozen at 80 C were thawed
20
aspirations through a 26
gauge needle
1600±300 CFU/lung

• Mice deficient for IL-1α or


IL-1β , or both IL-1α and
IL-1β , IL1-R1, or TNF Bacterial load in tissues
•• Evaluated at different time
Alternatively, M. bovis
inoculum size was
injected intravenously at BCG or fluorescent
• Backcrossed at least 7– points after infection with M.
tuberculosis
tuberculosis H37Rv
H37Rv 1066 CFU/mouse. GFP-expressing M.
bovis BCG
verified 24 h after
infection

10 times on C57BL/6
genetic background.
• For experiments, adult
(8–12 weeks old) animals Organs were weighed
and defined aliquots
Tenfold serial dilutions
Plated in duplicates onto
Middlebrook 7H11
Colonies were
enumerated at 3 weeks
were kept in isolators were homogenised in
PBS in a Dispomix
of organ homogenates
in 0.05% Tween 20
(Difco) agar plates
containing 10% OADC
and results are
expressed as log10
containing 0.9% NaCl
• The infected mice  homogeniser and incubated at 37C. CFU per organ

monitored regularly for


clinical status and
weighed twice weekly. Determination of IL-1a, The supernatants
Pulmonary cytokine
determination
Immediately frozen on
IL-1β , IL-12/IL-23p40, sterilised by
dry ice and stored at -80 •• Lung
Lung homogenates
homogenates were
were
IL-23p19, TNF and IFNg centrifugation through centrifuged
C centrifuged
levels by ELISA 0.22 mm filter
MATERIALS AND METHODS

Histopathological analysis Liver and lung from M. bovis


• Left lobe of lungs from M. Free alveolar space, lung
Two to 3-mm sections were BCG infected mice  H&E
tuberculosis infected mice Fixed in 4% phosphate cellular infiltration, oedema
stained with Hematoxylin staining and the slides
buffered formalin and and necrosis quantified
and Eosin and a modified scored for inflammatory cell
paraffin-embedded. using a semi-quantitative
Ziehl-Neelsen method. infiltration and granuloma
score
lesions.

Primary macrophage
cultures Isolated from femurs and Three days after washing
Macrophages were plated in Stimulated with LPS, heat
• Murine bone marrow cells differentiated into and re-culturing in fresh
96 well microculture plates killed M. tuberculosis
macrophages after culturing medium  contained a
(at 105 cells/well in H37Rv, live or heat-killed M.
at 1066 cells/ml for 7 days in homogenous population of
supplemented DMEM) bovis BCG (HKBCG).
DMEM macrophages.

Statistical analysis
Cell supernatants  • Determined with Graph Pad Differences between
harvested after 24 h of Prism multiple in vivo groups  Non-parametric Mann–
Values of P 0.05 
stimulation for IL-1a, IL-1b analysed by means of one- Whitney t-test analyzing in
significant Two-tailed
and TNF quantification by way non-parametric ANOVA vitro results
ELISA test
THANKYOU

You might also like