SlideShare a Scribd company logo
1 of 8
Download to read offline
International Immunopharmacology 96 (2021) 107608
Available online 12 April 2021
1567-5769/© 2021 Elsevier B.V. All rights reserved.
Review
Inflammatory mediators in various molecular pathways involved in the
development of pulmonary fibrosis
M. Fathimath Muneesa, Sadiya B. Shaikh, T.M. Jeena, Yashodhar P. Bhandary *
Yenepoya Research Centre, Yenepoya University, Deralakatte, Mangalore 575018, Karnataka, India
A R T I C L E I N F O
Keywords:
Inflammation
Idiopathic pulmonary fibrosis
Cytokines
Epithelial-mesenchymal transition
Fibrinolytic system
Apoptosis
A B S T R A C T
Idiopathic pulmonary fibrosis (IPF) is a type of interstitial lung disease (ILD) that is marked by scarring of lung
tissue, ultimately leading to respiratory failure. The survival rate of IPF is disappointing and to date demonstrates
a clinical quandary. The exact etiology of the disease remains under discussion. According to the recent hy­
pothesis, inflammatory mediators cause severe damage to the alveolar epithelium leading to the impairment of
the alveolar structure. The role of inflammation in the development of the IPF has been controversial for years.
There are two schools of thought regarding the role of inflammation. One group of researchers claims that cell
death and fibroblast dysfunction are the primary causes and inflammation is just a secondary cause of IPF. The
other group claims inflammation to be the primary cause. Studies using human subjects have also reported
inflammation as a critical element in IPF. Inflammatory cytokines serve a major role in commencing the in­
flammatory response in the lungs. Several cytokines are reported to be involved in different molecular mecha­
nisms underlying IPF, some of which also contribute additionally by acting as growth factors. The present review
addressed to explore the contribution of various inflammatory cytokines, growth factors, and various other in­
flammatory molecules activating the major molecular pathways involved during the development of IPF.
1. Introduction
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung
disease (ILD) that involves the irreversible scarring of the tissue because
of inflammation, the impaired wound healing process, and ultimately
fibrosis. Fibrosis is due to the excessive deposition of extracellular ma­
trix (ECM) proteins like fibronectin and collagens in the interstitial space
and alveolar septa [1]. This results in the alveolar structural damage
reducing the number of functional alveoli and thereby resulting in
impaired gas exchange. Thus the disease is associated with reduced lung
function and gradual respiratory failure.
The first step of diagnosis of IPF involves ruling out the known causes
of ILDs. The next step is to check for the usual interstitial pneumonia
(UIP) pattern on High-resolution computer tomography (HRCT) and
Abbreviations: AIM2, Absent In Melanoma; AKT, Also known as Protein Kinase B (PKB); ARDS, Acute Respiratory Distress Syndrome; ASC, Apoptosis-Associated
Speck-Like Protein Containing A Caspase-Recruitment Domain; ATII, Alveolar Type II; BAL, Bronchoalveolar Lavage Fluid; bFGF, Basic Fibroblast Growth Factor;
CCL, CC Chemokine Ligand; CCN3, Cellular Communication Network Factor 3; CCR, CC Chemokine Receptor; CD28, Cluster Of Differentiation 28; CXCL, Chemokine
(C-X-C Motif) Ligand; CXCR, Chemokine (C-X-C Motif) Receptor; dNLR, Derived Neutrophil To Lymphocyte Ratio; ECM, Extracellular Matrix; EMT, Epithelial-
Mesenchymal Transition; ERK, Extracellular Signal-Regulated Kinases; FVC, Forced Vital Capacity; GDF, Growth/Differentiation Factor; HRCT, High-Resolution
Computed Tomography; ICAM-1, Intercellular Adhesion Molecule 1; IFN-γ, Interferon Gamma; IGF, Insulin-Like Growth Factor; IKK, Inhibitor Of NF-Κb Kinase; IL,
Interleukin; ILD, Interstitial Lung Disease; IPF, Idiopathic Pulmonary Fibrosis; JAK, Janus Kinase; LPS, Lipopolysaccharide; MAPK, Mitogen-Activated Protein Kinase;
MCP, Monocyte Chemoattractant Protein; M-CSF, Macrophage Colony-Stimulating Factor; MMP, Matrix Metalloproteinases; NF-KB, Nuclear Factor Kappa B (NF-Κb);
NLRP3, NLR Family Pyrin Domain Containing 3; NOD, Nucleotide-Binding And Oligomerization Domain; NUR77, Also Known As Nerve Growth Factor IB (NGFIB);
PAI-1, Plasminogen Activator Inhibitor; PBMCs, Peripheral Blood Mononuclear Cells; PDGF, Platelet-Derived Growth Factor; PGE2, Prostaglandin E2; PI3K, Phos­
phoinositide 3-Kinase; PMN, Polymorphonuclear Leukocytes; SIRI, Systemic Inflammation Response Index; SMA, Smooth Muscle Actin; SMAD, Mothers Against
Decapentaplegic Homolog; SNAIL-1, Zinc finger protein SNAI1; STAT, Signal Transducer And Activator Of Transcription; TGF-β, Transforming Growth Factor-Beta;
TIMP, Tissue Inhibitors Of Matrix Metalloproteinases; TLR, Toll-Like Receptor; TNF-α, Tumor Necrosis Factor; TRAF, Tumor Necrosis Factor Receptor–Associated
Factor; UIP, Usual Interstitial Pneumonia; uPA, Urokinase Plasminogen Activator; uPAR, Urokinase Plasminogen Activator Receptor; Wnt, Wingless-Related Inte­
gration Site.
* Corresponding author.at: Yenepoya Research Centre, Yenepoya University, Mangalore 575 018, Karnataka, India.
E-mail address: yash28bhandary@gmail.com (Y.P. Bhandary).
Contents lists available at ScienceDirect
International Immunopharmacology
journal homepage: www.elsevier.com/locate/intimp
https://doi.org/10.1016/j.intimp.2021.107608
Received 10 December 2020; Received in revised form 23 February 2021; Accepted 21 March 2021
International Immunopharmacology 96 (2021) 107608
2
surgical lung biopsy. The characteristic histopathological feature in
surgical lung biopsies of IPF patients is the prominent fibrotic foci whose
appearance is due to the accumulated fibroblasts and myofibroblasts.
The median survival rate of an IPF patient after diagnosis is 3–5 years
[2]. Currently, there are no clinically effective treatment options against
IPF except lung transplantation [3]. The molecular mechanisms under­
lying the disease are also not clearly understood. But in the last few
years, researchers across the world have reached significant landmarks
associated with the disease. Initially, the inflammation process triggered
by an injury was considered to be the primary cause for the initiation
and progression of fibrosis. When there is an injury to the alveoli, the
resident macrophages release cytokines to attract fibroblasts, epithelial
and endothelial cells for promoting wound healing. But if there is
persistent injury, the neutrophils and the monocytes are recruited to the
sites. These inflammatory cells generate reactive oxygen species which
leads to an imbalance in oxidants and anti-oxidants. Eventually, the
inflammatory cells gathered secrete growth factors and other mediators
contributing to fibrosis [4,5]. But currently, the role of inflammation is a
controversial topic among researchers as many studies showed results
contradicting the inflammation theory. Various cellular processes play a
prime role in the pathogenesis of lung fibrosis (Fig. 1). The present re­
view discusses the role of inflammatory molecules in the core molecular
pathways involved during the development of lung fibrosis. Here, we
also highlight the controversial role of inflammation in IPF.
2. Inflammatory mediators in epithelial-mesenchymal
transition
The fibrotic foci in the parenchyma of a fibrotic lung are accumu­
lated with mesenchymal cells. These mesenchymal cells include fibro­
blasts and myofibroblasts, which are highly proliferative and produce an
excessive amount of ECM. The origin of the mesenchymal cells may be
the circulating fibrocytes or alveolar epithelial cells (AEC) that under­
went epithelial-mesenchymal transition (EMT) [6]. EMT is a process in
which the epithelial cells undergo morphological change and attain
motility due to the change in cytoskeletal arrangements and dissociation
of membrane-associated adherens junctions and desmosomes.
Myofibroblasts (or activated fibroblasts) and macrophages secrete
chemokines and growth factors that activate the ERK-MAPK (extrac­
ellularsignal-regulated kinase-mitogen activated protein kinase),
Smads, and the PI3K-AKT (Phosphatidylinositol-3-kinase-Protein kinase
B) signaling pathways to induce EMT [7]. Cytokines like transforming
growth factor-β (TGF-β), tumor necrosis factor (TNF-α), and interleukin-
1β (IL-1β) are reported to induce EMT in the mesenchymal (myofibro­
blast-like) phenotype in the fibrotic foci [8]. TGF-β is the most studied
and most crucial cytokine playing role in this process. High expression of
TGF-β in bronchoalveolar lavage (BAL) fluid and at the regions of
epithelial cells lining the fibrotic foci have been reported [9]. TGF-β
mediated EMT is through Mothers against decapentaplegic homolog 2
(SMAD-2) pathway but not MAPK pathways. TGF-β induces elevated
expression of matrix metalloproteinase 2 (MMP-2). This could disinte­
grate the basement membrane and in turn, induce motility of the cells
[10]. Shaikh et al. 2020., reported the potential contribution of TGF-β1
promoting p53-fibrinolytic system during alveolar EMT in A549 cells
[11].There are notable similarities between lung fibrosis and lung can­
cer concerning genetic and epigenetic alterations, EMT, fibroblasts in­
vasion, and some signal transduction pathways [12,13].
Elevated levels of IL-17A are reported in IPF and bleomycin-induced
pulmonary fibrosis (BLM-PF) models [14,15]. IL-17A mediates EMT
through TGF-β mediated ERK1/2 and Smad2/3 activation [16]. Studies
also reported the participation of IL-17A in activation of TGF-β via smad
dependent and independent signaling in alveolar epithelial cells during
ALI in vivo [17]. In a recent study, it was reported that IL-17A and
growth/differentiation factor 15 (GDF15) induced EMT of lung epithe­
lial cells in response to cigarette smoke [18]. IL-6 promotes growth and
EMT in CD133 + A549 cells by targeting Hhg (Hedgehog protein), Bcl-2
(B cell lymphoma 2), and Erk/MEK [19,20]. Zhang et al.,demonstrated
that inhibition of IL-18 in BLM-PF led to an increase in epithelial marker,
E-cadherin. The study also concluded that IL-18 regulated EMT by up-
regulating Snail-1 [21].
NOD-like receptor protein 3 (NLRP3) inflammasome is a multi­
protein complex that activates caspase-1 which in turn leads to the
secretion of pro-inflammatory cytokines, IL-1β and IL-18, and their
maturation. The components of this protein complex are a sensor
molecule, NLRP3, an adaptor molecule apoptosis-associated speck-like
protein containing a caspase recruitment domain (ASC), and pro-cas­
pase-1[22,23]. It is found to be involved in acute respiratory distress
syndrome (ARDS), chronic obstructive pulmonary disease (COPD), IPF,
asthma, and silicosis [24]. NLRP3 inflammasome mediates EMT through
TAK1-MAPK-Snail/NF-κB pathway [25]. IL-1β can activate EMT related
pathways like Wnt/GSK3β (Wingless-Related Integration Site/glycogen
synthase kinase 3β) and TGF-/Smad signaling pathways [26,27]. The
expression and activation of another inflammasome named absent in
melanoma 2 (AIM2) are enhanced in IPF peripheral blood mononuclear
cells (PBMCs) as well as in a lung fibrosis exacerbation model [28,29].
AIM2, upon activation in PBMCs, causes the release of pro-fibrotic cy­
tokines IL-1α, IL-18, and TGF-β in a TLR4-/caspase-1-/caspase-8-/cal­
pain-independent fashion and also releases caspase-4 [29]. Recent
studies reports that AIM2 inflammasome-dependent inflammation plays
apivotal role during progression of pulmonary fibrosis [30].
Fig 1. Cellular factors responsible for lung fibrosis: Various cellular processes like apoptosis of alveolar epithelial cells, EMT, inflammatory cells infiltration,
myofibroblast invasion or differentiation, and DNA damage play a major role in the pathogenesis of lung fibrosis.
M. Fathimath Muneesa et al.
International Immunopharmacology 96 (2021) 107608
3
CXCR3 is the receptor for chemokine ligand CXCL9. Beirne et al.,
demonstrated the elevated expression of CXCR3 in the proximity of cells
that have undergone EMT in IPF surgical lung biopsies. Their study
concluded the antifibrotic activity of CXCL9 via abrogation of the TGFβ
signaling pathway by the reduction of phosphorylation of Smad2 and
Smad3 in AECs [31]. The expression of IL-22, belonging to the IL-10
family, is shown to be reduced in BLM-PF mice models. IL-22 inhibits
EMT of AECs [32].
In a study conducted on HCl-induced epithelial remodeling, it was
found that monocytes interact with epithelial cells through ICAM-1
(Intracellular adhesion molecule-1) and enhance EMT and induce the
release of IL-8 and PDGF [33]. IL-8 interacts with CXCR1/CXCR2 to
activate the MAPK pathway and is shown to favor fibrosis [34,35].
IL-37 is an antifibrotic interleukin [36–38]. IL-37 and CCL22 co-
localizes and inhibits proliferation and EMT by abrogating IL-6/Signal
Transducer and Activator of Transcription-3 (STAT3) signaling
pathway in A549 cells [37,39]. IL-37 also binds to Smad3 and down­
regulates different signaling pathways, including focal adhesion kinase
(FAK), STAT, p53, MAP kinase p38α, and mTOR (mammalian target of
rapamycin)[40]. IL-27 is also a negative regulator of EMT in AEC
through the JAK/STAT and TGF-β/Smad signalling pathways [41].
3. Inflammatory mediators in apoptosis of epithelial cells
Apoptosis plays a significant role in the pathogenesis of PF. Epithelial
cells undergo apoptosis in patients with IPF and also in PF animal
models. Intratracheal administration of BLM-induced elevates expres­
sion of fas on alveolar epithelium [42]. The abnormal activation of AEC
is sufficient to initiate fibrotic activities [43]. Alveolar type II (AT-II)
epithelial cells are the ones that undergo apoptosis. AT-II cells have the
capability of being transformed into type 1 cells [44]. Consistent with
the theory, it was proved that collagen deposition could be reduced by
the installation of apoptotic inhibitors in animal models [45]. The
presence of epithelial cell apoptosis in normal lungs suggests that
apoptosis can probably be an initiating event in fibrogenesis [46].
Epithelial cells are important because they produce mediators that are
antifibrotic. For instance, prostaglandin E2 (PGE2) produced by
epithelial cells inhibit fibroblast proliferation. Plasminogen activators
and metalloproteinases expressed by them aid in fibrin and ECM
degradation [47]. Epithelial cells probably play as a barrier to cytokines
secreted by activated macrophages and thereby protecting the under­
lying tissue. Thus apoptosis of epithelial cells could play a crucial role in
the onset of fibrosis [48].
TNF-α induces AEC apoptosis dependent on angiotensin II in the
BLM-PF model [49,50]. It activates the IKK ( IκB kinase ) and NF-KB
(nuclear factor kappa light chain enhancer of activated B cells) com­
plex. CD28 (Cluster of differentiation-23) is a costimulatory protein for
the activation of T-cells expressed on antigen-presenting cells. CD28
deficiency abrogates blast exposure-induced lung tissue apoptosis [49].
In multiple myeloma, the CD28 pathway is associated with the phos­
phorylation of PI3K/Akt and inactivation of the FoxO1 [51]. CD28 in­
duces the expression of IL-17A and other cytokines, IL-6 and IL-8, in
multiple sclerosis T lymphocytes [52]. It activates RelA/NF-κB which in
turn induces IL-6 expression that leads to the activation and nuclear
translocation of tyrosine-phosphorylated STAT3 (pSTAT3). pSTAT3 as­
sociates with RelA/NF-κB by binding to the specific sequences in the
proximal promoter of the human IL-17A gene, thereby inducing its
expression. Class 1A PI3K regulates CD28-mediated RelA/NF-κB and
STAT3 recruitments and trans-activation of IL-17A promoter [53]. IL-
17A induces the expression of p53 augmenting apoptosis of AEC
[54,55].
Elevated levels of nephroblastoma overexpressed protein (NOV; also
known as CCN3), an inflammatory mediator, was observed in the
plasma of patients with severe pneumonia-induced ARDS when
compared to the healthy controls [56]. CCN3 brings about pro-apoptotic
effects on AECs by the activation of the TGF-β signaling pathway, NF-κB
signaling pathway, and Bcl-2/caspase-3 pathway [57]. CCN3 could play
a pro-fibrotic role by inducing apoptosis of AECs and the destruction of
epithelial integrity, ultimately leading to IPF. The inflammatory cyto­
kines such as IL-17A, IL-6, TNF-α, IL-33, and IL-37 act via various
signaling pathways through cell surface receptors playing a pivotal role
in regulating alveolar apoptosis and EMT causing alveolar damage and
ultimately leading to the development of lung fibrosis (Fig. 2).
4. Inflammatory mediators in the impaired fibrinolytic system
Extravascular accumulation of fibrin is implicated in the progression
of PF [58]. When an injury occurs in the lung tissue, the vascular
permeability increases; due to which the coagulation and fibrinolytic
factors leak out to the impaired tissue [59]. Thrombosis and fibrin
deposition in the alveolar space are common features in a fibrotic lung
[60].
The balance between fibrinolysis and coagulation plays a critical role
in tissue homeostasis. The main components of the lung fibrinolytic
system are urokinase plasminogen activators (uPA), urokinase plas­
minogen activator receptor (uPAR), and plasminogen activator inhibitor
Type-1 (PAI-1). uPA and uPAR promote fibrinolysis whereas PAI-1 in­
hibits fibrinolysis. Increased level of PAI-1 due to inflammatory pro­
cesses is observed in pleural effusion [61]. Park et al., observed
increased levels of PAI-1 in the plasma and the injured lungs of patients
with PF. The study also demonstrated PAI-1 as a potential “don’t eat me
signals” for viable and apoptotic neutrophils [62]. The persistence of
apoptotic neutrophils in the fibrotic lung may worsen the condition by
releasing intracellular toxic substances to the surroundings through
necrosis or autolysis of the apoptotic bodies.
Regulation of coagulation and fibrinolytic system by inflammatory
mediators is studied extensively in certain pathological conditions like
sepsis. Stimulation of human adipocytes with IL-β or TNF-α or TGF-β
elevates the expression of PAI-1 [63]. Chiu et al., suggested that the
increased expression of pro-inflammatory cytokines may lead to fibri­
nolytic imbalance and result in fibrin deposition in the pleura of the
pediatric para-pneumonic lung [64]. Akpan et al., noted that interferon-
γ (IFN-γ) and IL-10 increased fibrinolysis inactive tuberculosis which
involves chronic inflammation [65]. A recent study proved that IL-17A
enhanced the expression of PAI-1 and p53 [55] and downregulates the
expression of uPA and uPAR in acute lung injury (ALI) in vivo [54]. p53
is reported to destabilize the mRNAs of uPA and uPAR and stabilization
of mRNA of PAI-1[66,67].Tiwari et al., stated that PAI-1 attracts poly­
morphonuclear (PMN) cells and other inflammatory cells to alveolar
space through induction of expression of chemokines by AECs [68]. IL-
1β and IL-12 have also been suggested to play important roles in the
fibrinolytic system [69]. IGF-II (Insulin-like growth factor-II) impairs
MMP and tissue inhibitor of metalloproteinases (TIMP) balance [70]. IL-
5 stimulates eosinophils to adhere to fibrinogen and results in degran­
ulation which ultimately leads to the secretion of IL-13, IL1a, IL-4, TNF,
and CCL24 [71]. Soluble fibrinogen interacts with CD11b on neutrophils
and activates focal adhesion kinase and ERK1/2 to regulate degranula­
tion, phagocytosis, and apoptosis [72]. TGF-β is a strong inducer of PAI-
1. Downstream molecules of the TGF-β signaling pathway, SMAD 3 and
4 proteins, bind to consensus sites in the TGF beta-inducible elements in
the promoter of PAI-1 promoter [73]. Numerous inflammatory mole­
cules induce PAI-1 expression further giving rise to various inflamma­
tory responses such as fibrin deposition, activation of macrophages,
ECM deposition, increasing fibronectin and collagen levels during lung
fibrosis (Fig. 3).
Lipopolysaccharides (LPS) mediated expression of PAI-1 is
augmented by TNF-α.Nuclear receptor-77 (Nur77) is a transcriptional
factor generated in response to TNF-α. Nur77 induces transcription
of PAI-1 by binding to NGFI-B responsive element (NBRE) in the PAI-1
gene in a ligand-independent mechanism [73]. IL-33 participates in
the production of profibrotic cytokines and modulates the expression of
collagen IV, MMP-9, TIMP-1, tumor necrosis factor receptor (TNFR)-
M. Fathimath Muneesa et al.
International Immunopharmacology 96 (2021) 107608
4
associated factor 6 (TRAF-6), and NF-κB. It creates an imbalance be­
tween MMP-9 and TIMP-1 and thereby modulating the deposition of
MMP [74].
PAI-1 also influences inflammatory responses. It can increase the
infiltration of neutrophils, macrophages, and myofibroblasts; thereby
contributing to the progression of fibrosis (Fig. 4) [75,76]. In short,
fibrinolytic system is capable of regulating inflammatory responses and
vice-versa.
5. Inflammatory mediators in accumulation of myofibroblasts
Accumulation of fibroblasts is one of the significant observations in
PF. The fibroblasts in fibrotic lung express α-smooth muscle actin
(α-SMA) and ECM, which leads to damage to the alveolar architecture
[77]. They are key sources of collagen and profibrotic cytokines like
TGF-β. The role of inflammatory cytokines in the proliferation and
accumulation of fibroblasts has been implicated in several studies. Han
et al. noted that basic fibroblast growth factor (bFGF) induced activation
of ERK5 by enhancing its phosphorylation. ERK5 is an important protein
for cell proliferation, differentiation, and tissue repair. bFGF-induces
PAI-1 and cell proliferation in lung fibroblasts [78]. TGF-β is the most
reported growth factor cytokine which exhibits the property of induc­
tion of differentiation into myofibroblasts. TGF-β is elevated in human
subjects with IPF as well as in animal models of PF. It promotes the
activation and differentiation of normal lung fibroblasts to fibrotic lung
fibroblasts [79,80]. Fibrotic lung fibroblasts express α-SMA and ECM in
excess which in turn leads to changes in the alveolar architecture.
There is increased expression of CCR7 by fibroblasts, mononuclear,
and epithelial cells in IPF lung [81]. The ligand of CCR7, CCL21, en­
hances the proliferation and migration of fibroblasts. Immuno-
neutralization of either of the proteins inhibits migration of fibro­
blasts. This indicates that the migration of fibroblasts is dependent on
both, the ligand and the receptor [82]. CXCL16/CXCR6 accelerates
fibrosis by augmenting proliferation, migration, and collagen synthesis
of human pulmonary fibroblasts through the activation of the PI3K/
AKT/FOXO3a signaling pathway [83]. IL-11, a cytokine from the IL-6
Fig 2. Inflammatory cytokines mediated pathways activating alveolar apoptosis and EMT leading to the pathogenesis of pulmonary fibrosis: Pro-Inflammatory
cytokines are responsible for activation of alveolar inflammatory pathways. IL-17AR operates IL-17A activating the prime modulator of EMT TGF-β1 activating
smad2/3 complex leading to phosphorylation of smad 2/3 in the nucleus. IL-17A also induces EGFR stimulating MAPK/ERK 1/2 elevating the expression of p53 and
favors its translocation in the nucleus inducing phosphorylation of p53 causing alveolar injury. IL-6R facilitates the binding of P-JAK stimulating P-STAT1 and P-
STAT3 further translocating in the nucleus promoting JAK-STAT pathway activation. The receptor of TNF-α binds to TRAF2 elevating IKK expressions forming the
IKK-NFKB complex inducing phosphorylation of NFKB in the nucleus. TRAF6 binds to the receptor of IL-33 activating MAPK and NFKB further expressing NFKB
phosphorylation in the nucleus, however, this mechanism is found to be blocked by the anti-inflammatory cytokine IL-37. All the above cytokines are responsible to
induce alveolar epithelial injury and activate the biological process such as apoptosis and EMT leading to the pathogenesis of pulmonary fibrosis. On the other hand,
BLM injury stimulates NLRP3 inflammasome activating pro-caspase 3 and active caspase 3 promoting alveolar apoptosis causing lung epithelial injury ultimately
leading to pulmonary fibrosis.
M. Fathimath Muneesa et al.
International Immunopharmacology 96 (2021) 107608
5
family, has mitogenic property and inhibits fas-induced apoptosis of
lung fibroblasts [84,85]. Oncostatin M, another cytokine of the IL-6
family, also has mitogenic property and induces collagen expression in
lung fibroblasts. Oncostatin also inhibits the expression of α-SMA
induced by the TGFβ signaling pathway [86,87]. IGF-II induces trans­
differentiation of normal lung fibroblasts into myofibroblasts by acti­
vating the TGFβ receptor [70]. As reported by Tager et al., CXCL10
inhibits the accumulation of fibroblasts and thereby plays a protective
role in PF. It indicates that overexpression of CXCL10 could resolve PF
[88].
6. Inflammatory mediators in immune cell infiltration
Elevated levels of cytokines that play a role in chemotaxis and acti­
vation of neutrophils, lymphocytes, and monocytes are observed in tis­
sue and fluid from the lungs of IPF patients. IL-8, monocyte
chemoattractant protein (MCP-1/CCL2), and macrophage inflammatory
protein-1α (MIP-1α/CCL3) are examples of these cytokines. Baran et al.,
(2007), reported elevated levels of macrophage colony-stimulating
factor (M− CSF) in the fluid from the lungs of IPF patients. The study
also demonstrated M− CSF mediated recruitment of macrophages in the
BLM-PF model [89].Cytokine-like-factor-1(CLF-1) augments the infil­
tration of CD4 + T cells in BLM-induced fibrotic lung [90]. Recent study
demonstrated the potentiality of the pro-inflammatory cytokine IL-17A
in elevation of AMPKα/COX-2 expressions via enhancing inflammatory
molecules like NF-κB-p65, NF-κB-p105, CXCL1 and IL-1β etc [91]. IL-6,
an activator of JAK-STAT pathway, promotes migration and infiltration
by inducing the expression of MMP1 and MMP-9 through PGE-2 and
COX2 signaling [92]. IL-1β upregulates CXCL5 and causes infiltration of
neutrophils and macrophages. CXCL5 is, in turn, a substrate for MMPs 2
and 9 which activate CXCL5 by cleaving it [93]. Anti TNF-α adminis­
tration in nitrogen mustard induced pulmonary injury model, reduced
the numbers of M1 macrophages Ym1+
M2 macrophages in the lung
[94]. In conclusion, inflammatory mediators like CXCL5, IL-1β and TNF-
α play potential role in infiltration of immune cells which in turn leads to
enhanced inflammation and gradually contributes to fibrosis.
7. Therapeutics targeting inflammation
Inflammation can worsen the condition in PF and thereby leading to
early mortality. The therapy against ILD includes anti-inflammatory and
immunosuppressive agents, with anti-fibrotic agents introduced lately
for the treatment of IPF. Treatment with anti-inflammatory drugs even
before the process of irreversible parenchymal fibrosis could probably
ease IPF. Pirfenidone used widely against IPF is an immunosuppressant
with anti-inflammatory as well as anti-fibrotic features [95]. It can also
modulate cross-talk between DCs and T-cells to a reduce the production
of pro-inflammatory cytokines, decrease Th-cell proliferation and im­
pairs Th2-cell polarization [96–98]. Nintedanib, another widely used
drug against IPF, reduces BAL lymphocytes and neutrophils but not
macrophages. It also lowers the levels of interleukin-1b, keratinocyte
chemoattractant, TIMP-1, and lung collagen [99]. Thalidomide, pos­
sessing immunomodulatory potentials, could improve cough and res­
piratory quality in IPF patients [100]. Anti-inflammatory and anti-
fibrotic properties of dabigatranetexilate resolve pulmonary fibrosis in
experimental models. It reduces the infiltration of inflammatory cells
and pro-fibrotic protein expression [101]. Statin therapy, which is
mainly used to manage cardiovascular disorders, reduces inflammation
and release of pro-inflammatory cytokines in lungs. It is also potent in
inhibiting fibroblast activity and hinders the advancement of the disease
to fibrotic stage; thereby reducing mortality. There are also studies
showing adverse effect of statins against ILDs [102–106]. A high dose of
a non-steroidal anti-inflammatory drug, ibuprofen is reported to slow
down the progression of lung disease in cystic fibrosis [107]. Cortico­
steroids when used at the active stage of inflammation could bring about
a significant response in delaying the progress of the disease [108].
Fig 3. Inflammatory mediators in the fibrinolytic system leading to pulmonary
fibrosis: Inflammatory irritants induce reactive oxygen species (ROS) activating
the inflammatory cytokines IL-6, IL-17A, and TNF-α increasing the expression
levels of p53 further recruiting the fibrinolytic molecules. ie, decrease expres­
sions of uPA and uPAR and increase levels of PAI-1 promoting fibrin deposition
causing fibroblast accumulation eventually causing pulmonary fibrosis.
Fig 4. PAI-1 influences various inflammatory responses: Various inflammatory
cytokines like IL-6, IL-1β, TGF-β1, IL-17A, and TNF-α elevates expression levels
of PAI-1 stimulating several inflammatory responses like fibrin deposition, in­
duction of macrophages and PMN, activation of myofibroblast, ECM deposition,
and increase in fibronectin and collagen levels favors in the development of
pulmonary fibrosis.
M. Fathimath Muneesa et al.
International Immunopharmacology 96 (2021) 107608
6
These medications are efficient against inflammation, but have potential
side effects and some of their benefits are unproven. Generous resolution
of inflammation in lungs is extremely important, as compromised res­
olution could lead to chronic inflammatory stage and ultimately fibrosis.
8. The controversy of the role of inflammation in IPF
The role of inflammation in PF has been a topic of discussion for
years and is still under controversy. Since there is no effective response
to the immunosuppressive medications in PF patients, some experts
claim that inflammation doesn’t play a role in the pathogenesis of the
disease. But several studies involving human subjects as well as animal
models delineate the role of inflammation in the fibrotic process. Some
researchers state that inflammation is just a secondary cause for the
disease. Ga67
uptake is used as a measure of inflammation at a specific
site. Mura et al., observed that although there is a prominent decline in
lung function parameters during the progression of PF, there was no
significant correlation between Ga67
uptake and the disease progression.
They concluded that inflammation is prominent in advanced stages of
fibrosis but does not play a pivotal role in the progression of the disease
[109]. A similar study conducted by Grijm et al., also indicates that
effective reduction in inflammation (as assessed using Ga67
scintig­
raphy) did not improve the clinical outcome of the disease [110]. Pa­
tients suffering from IPF have periods of an acute decline in lung
function parameters known as acute exacerbations. Increased levels of
neutrophils, pro-inflammatory cytokines, and M2 cytokines (an indica­
tion of macrophage activation) were noted during acute exacerbations
in BALF of IPF patients. The study concluded that acute exacerbation is
not a subsidiary event but is mediated by M2 macrophage activation
[111]. In a very recent study, it was reported that blood inflammation
indexes like neutrophil to lymphocyte ratio (NLR), derived neutrophil to
lymphocyte ratio (dNLR), monocyte to lymphocyte ratio (MLR), sys­
temic inflammation response index (SIRI), and aggregate index of sys­
temic inflammation (AISI) independently associated with the existence
of IPF after adjusting for age, gender, body mass index and smoking
status [113].
In a study by Balestro et al., pulmonary fibrosis patients were clas­
sified into slow or rapid progressors of PF based on the FVC fall per year.
The morphometric analysis of lung explants of slow progressors with
acute exacerbation and rapid progressors with or without acute exac­
erbations in PF showed significant inflammatory cell infiltrates. They
also observed that the number of innate immune cells (neutrophils and
macrophages) and that of adaptive immune cells (CD4+, CD8+, and B
cells) were high in rapid progressors than in slow progressors, pointing
at the influencing role of inflammation in the progression of PF [112].
In this review, we advocate that inflammation has a decisive role in
IPF. Delineating the mechanisms of the disease by using high throughput
technologies, the understanding of the disease pathogenesis will be
concrete and wider and also open the way for novel therapeutics and
diagnostics.
9. Conclusion
Inflammation does play role in the development of the disease. This
is demonstrated in different animal models as well as in studies in
human IPF patients. As the models used and modes of induction of the
disease in these models differ from study to study, there is non-
uniformity in the investigation and results obtained. Numerous cyto­
kines are shown to be involved in the disease. But their mechanistic role
in the disease is not studied extensively. With the usage of sophisticated
technologies in biochemistry and molecular biology, we expect delin­
eation of the mechanisms underlying the disease and thereby, probably,
the discovery of novel diagnostic and therapeutic approaches for pa­
tients suffering from IPF.
Authors contributions
The manuscript has been prepared and reviewed by Ms. Fathimath
Muneesa M in correspondence with Dr. Yashodhar Prabhakar Bhandary.
Sadiya B Shaikh added some more points and prepared the figures.
Jeena TM added some information and reviewed the entire manuscript.
Acknowledgments
Authors would like to acknowledge Yenepoya Research Centre,
Yenepoya University, Department of Biotechnology Grant (BT/
PR25198/MED/30/1896/2017), and Indian Council of Medical
Research (No.59/12/2015/Online/BMS/TRM) for supporting our
research.
References
[1] J.K. Burgess, T. Mauad, G. Tjin, J.C. Karlsson, G. Westergren-Thorsson, The
extracellular matrix - the under-recognized element in lung disease? J. Pathol.
[Internet]. 240 (4) (2016) 397–409. https://www.ncbi.nlm.nih.gov/pubmed
/27623753.
[2] G. Raghu, H.R. Collard, J.J. Egan, F.J. Martinez, J. Behr, K.K. Brown, et al., An
official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-
based guidelines for diagnosis and management, Available from, Am. J. Respir.
Crit. Care Med. [Internet]. 183 (6) (2011 Mar 15) 788–824, https://www.ncbi.
nlm.nih.gov/pubmed/21471066.
[3] K.D. Kistler, L. Nalysnyk, P. Rotella, D. Esser, Lung transplantation in idiopathic
pulmonary fibrosis: a systematic review of the literature, BMC Pulm. Med.
[Internet]. 16 (14) (2014 Aug) 139. https://www.ncbi.nlm.nih.gov/pubmed
/25127540.
[4] W.R. Coward, G. Saini, G. Jenkins, The pathogenesis of idiopathic pulmonary
fibrosis, Ther. Adv. Respir. Dis. 4 (6) (2010 Dec) 367–388.
[5] J.S. Zolak, J.A. de Andrade, Idiopathic pulmonary fibrosis, Immunol. Allergy
Clin. North Am. 32 (4) (2012 Nov) 473–485.
[6] B.C. Willis, J.M. Liebler, K. Luby-Phelps, A.G. Nicholson, E.D. Crandall, R.M. du
Bois, et al., Induction of epithelial-mesenchymal transition in alveolar epithelial
cells by transforming growth factor-beta1: potential role in idiopathic pulmonary
fibrosis, Am. J. Pathol. [Internet]. 166 (5) (2005 May) 1321–1332. https://www.
ncbi.nlm.nih.gov/pubmed/15855634.
[7] M. Li, B. Liu, Epithelial to mesenchymal transition in the progression of
tubulointerstitial fibrosis, Chin Med. J. (Engl). 120 (21) (2007 Nov) 1925–1930.
[8] F. Rieder, S.P. Kessler, G.A. West, S. Bhilocha, C. de la Motte, T.M. Sadler, et al.,
Inflammation-induced endothelial-to-mesenchymal transition: a novel
mechanism of intestinal fibrosis, Am. J. Pathol. [Internet]. 179 (5) (2011)
2660–2673, 2011/09/21.
[9] Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, et al.
Alveolar epithelial cell mesenchymal transition develops <em>in vivo</
em> during pulmonary fibrosis and is regulated by the extracellular matrix.
Proc Natl Acad Sci [Internet]. 2006 Aug 29;103(35):13180 LP – 13185. Available
from: http://www.pnas.org/content/103/35/13180.abstract.
[10] H. Kasai, J.T. Allen, R.M. Mason, T. Kamimura, Z. Zhang, TGF-beta1 induces
human alveolar epithelial to mesenchymal cell transition (EMT), Respir. Res.
[Internet]. 6 (1) (2005) 56. https://www.ncbi.nlm.nih.gov/pubmed/15946381.
[11] Shaikh SB, Prabhu A, Bhandary YP. Curcumin targets p53-fibrinolytic system in
TGF-β1 mediated alveolar epithelial mesenchymal transition in alveolar epithelial
cells. Endocr Metab Immune Disord Drug Targets. 2020 Sep.
[12] C. Vancheri, Idiopathic pulmonary fibrosis: An altered fibroblast proliferation
linked to cancer biology, Proc. Am. Thorac. Soc. 9 (3) (2012) 153–157.
[13] T. Kinoshita, T. Goto, Molecular Mechanisms of Pulmonary Fibrogenesis and Its
Progression to Lung Cancer: A Review, Int. J. Mol. Sci. [Internet]. 20 (6) (2019)
1461. https://www.ncbi.nlm.nih.gov/pubmed/30909462.
[14] Wang A, Wang F, Yin Y, Zhang M, Chen P. Dexamethasone reduces serum level of
IL-17 in Bleomycin-A5-induced rats model of pulmonary fibrosis. Artif Cells,
Nanomedicine, Biotechnol [Internet]. 2018 May 19;46(4):783–7. Available from:
https://doi.org/10.1080/21691401.2017.1339051.
[15] C. Huang, Y. Li, X. Fan, Y. Ma, M. Zhang, W. Wang, IL-17A promotes pulmonary
inflammation in rats with pulmonary fibrosis induced by bleomycin, Xi Bao Yu
Fen Zi Mian Yi Xue Za Zhi [Internet]. 30 (4) (2014) 366–370. http://europepmc.
org/abstract/MED/24721403.
[16] Wang T, Liu Y, Zou J-F, Cheng Z-S. Interleukin-17 induces human alveolar
epithelial to mesenchymal cell transition via the TGF-β1 mediated Smad2/3 and
ERK1/2 activation. PLoS One [Internet]. 2017 Sep 5;12(9):e0183972–e0183972.
Available from: https://www.ncbi.nlm.nih.gov/pubmed/28873461.
[17] S.B. Shaikh, S.G. Bhat, Y.P. Bhandary, Curcumin attenuates IL-17A mediated
pulmonary SMAD dependent and non-dependent mechanism during acute lung
injury in vivo, Mol. Biol. Rep. 47 (7) (2020 Jul) 5643–5649.
[18] G. Jiang, C.-T. Liu, W.-D. Zhang, IL-17A and GDF15 are able to induce epithelial-
mesenchymal transition of lung epithelial cells in response to cigarette smoke,
Exp. Ther. Med. [Internet]. 16 (1) (2018) 12–20. https://www.ncbi.nlm.nih.
gov/pubmed/29977354.
M. Fathimath Muneesa et al.
International Immunopharmacology 96 (2021) 107608
7
[19] S.O. Lee, X. Yang, S. Duan, Y. Tsai, L.R. Strojny, P. Keng, et al., IL-6 promotes
growth and epithelial-mesenchymal transition of CD133+ cells of non-small cell
lung cancer, Oncotarget 7 (6) (2016 Feb) 6626–6638.
[20] Y. Shintani, A. Fujiwara, T. Kimura, T. Kawamura, S. Funaki, M. Minami, et al.,
IL-6 Secreted from Cancer-Associated Fibroblasts Mediates Chemoresistance in
NSCLC by Increasing Epithelial-Mesenchymal Transition Signaling, J. Thorac.
Oncol. Off. Publ. Int. Assoc. Study Lung Cancer. 11 (9) (2016 Sep) 1482–1492.
[21] L.-M. Zhang, Y. Zhang, C. Fei, J. Zhang, L. Wang, Z.-W. Yi, et al., Neutralization of
IL-18 by IL-18 binding protein ameliorates bleomycin-induced pulmonary fibrosis
via inhibition of epithelial-mesenchymal transition, Biochem. Biophys. Res.
Commun. [Internet]. 508 (2) (2019) 660–666. http://www.sciencedirect.com/sci
ence/article/pii/S0006291X18325610.
[22] V. Petrilli, C. Dostert, D.A. Muruve, J. Tschopp, The inflammasome: a danger
sensing complex triggering innate immunity, Curr. Opin. Immunol. 19 (6) (2007
Dec) 615–622.
[23] S. Kesavardhana, T.-D. Kanneganti, Mechanisms governing inflammasome
activation, assembly and pyroptosis induction, Int. Immunol. 29 (5) (2017 May)
201–210.
[24] J.W. Pinkerton, R.Y. Kim, A.A.B. Robertson, J.A. Hirota, L.G. Wood, D.A. Knight,
et al., Inflammasomes in the lung, Mol. Immunol. 86 (2017 Jun) 44–55.
[25] Li X, Yan X, Wang Y, Wang J, Zhou F, Wang H, et al. NLRP3 inflammasome
inhibition attenuates silica-induced epithelial to mesenchymal transition (EMT)
in human bronchial epithelial cells. Exp Cell Res [Internet]. 2018;362(2):489–97.
Available from: https://doi.org/10.1016/j.yexcr.2017.12.013.
[26] P. Kaler, L. Augenlicht, L. Klampfer, Macrophage-derived IL-1beta stimulates Wnt
signaling and growth of colon cancer cells: a crosstalk interrupted by vitamin D3,
Oncogene 28 (44) (2009 Nov) 3892–3902.
[27] J. Wang, L. Bao, B. Yu, Z. Liu, W. Han, C. Deng, et al., Interleukin-1β Promotes
Epithelial-Derived Alveolar Elastogenesis via αvβ6 Integrin-Dependent TGF-β
Activation, Cell Physiol. Biochem. [Internet] 36 (6) (2015) 2198–2216. https
://www.karger.com/DOI/10.1159/000430185.
[28] Cho SJ, Moon J-S, Nikahira K, Yun HS, Harris R, Hong KS, et al. GLUT1-
dependent glycolysis regulates exacerbation of fibrosis via AIM2 inflammasome
activation. Thorax [Internet]. 2020 Mar 1;75(3):227 LP – 236. Available from:
http://thorax.bmj.com/content/75/3/227.abstract.
[29] M. Terlizzi, A. Molino, C. Colarusso, C. Donovan, P. Imitazione, P. Somma, et al.,
Activation of the absent in melanoma 2 inflammasome in peripheral blood
mononuclear cells from Idiopathic pulmonary fibrosis patients leads to the release
of pro-fibrotic mediators, Front. Immunol. (2018) 1–12, 9(APR).
[30] S.J. Cho, M. Lee, H.W. Stout-Delgado, J.-S. Moon, DROSHA-Dependent miRNA
and AIM2 Inflammasome Activation in Idiopathic Pulmonary Fibrosis, Int. J. Mol.
Sci. 21 (5) (2020 Feb).
[31] O’Beirne SL, Walsh SM, Fabre A, Reviriego C, Worrell JC, Counihan IP, et al.
CXCL9 Regulates TGF-β1-Induced Epithelial to Mesenchymal Transition in
Human Alveolar Epithelial Cells. J Immunol [Internet]. 2015/08/12. 2015 Sep
15;195(6):2788–96. Available from: https://www.ncbi.nlm.nih.gov/pubmed/
26268659.
[32] M. Liang, J. Wang, H. Chu, X. Zhu, H. He, Q. Liu, et al., Interleukin-22 Inhibits
Bleomycin-Induced Pulmonary, Fibrosis (2013).
[33] Chen Q, Luo AA, Qiu H, Han B, Ko BH-K, Slutsky AS, et al. Monocyte interaction
accelerates HCl-induced lung epithelial remodeling. BMC Pulm Med [Internet].
2014;14(1):135. Available from: https://doi.org/10.1186/1471-2466-14-135.
[34] D.D. Browning, W.C. Diehl, M.H. Hsu, I.U. Schraufstatter, R.D. Ye, Autocrine
regulation of interleukin-8 production in human monocytes, Am. J. Physiol. Lung
Cell. Mol. Physiol. 279 (6) (2000 Dec) L1129–L1136.
[35] E.M. Bruscia, P.-X. Zhang, E. Ferreira, C. Caputo, J.W. Emerson, D. Tuck, et al.,
Macrophages directly contribute to the exaggerated inflammatory response in
cystic fibrosis transmembrane conductance regulator-/- mice, Am. J. Respir. Cell
Mol. Biol. 40 (3) (2009 Mar) 295–304.
[36] Park sung woo, Kim CR, Lee EH, Kim JH, Park JS, Lee JH, et al. Intranasal
administration of IL-37 attenuates bleomycin induced experimental lung injury/
fibrosis. FASEB J [Internet]. 2013 Apr 1;27(1_supplement):1166.12-1166.12.
Available from: https://www.fasebj.org/doi/abs/10.1096/fasebj.27.1_
supplement.1166.12.
[37] Y. Li, Q. Gao, K. Xu, X. Peng, X. Yuan, W. Jiang, et al., Interleukin-37 Attenuates
Bleomycin-Induced Pulmonary Inflammation and Fibrosis in Mice, Inflammation
41 (5) (2018 Oct) 1772–1779.
[38] M.S. Kim, A.R. Baek, J.H. Lee, A.S. Jang, D.J. Kim, S.S. Chin, et al., IL-37
Attenuates Lung Fibrosis by Inducing Autophagy and Regulating TGF-beta1
Production in Mice, J. Immunol. 203 (8) (2019 Oct) 2265–2275.
[39] Y.-H. Chen, B.-Y. Zhou, X.-J. Wu, J.-F. Xu, J.-A. Zhang, Y.-H. Chen, et al., CCL22
and IL-37 inhibit the proliferation and epithelial-mesenchymal transition process
of NSCLC A549 cells, Oncol. Rep. 36 (4) (2016 Oct) 2017–2024.
[40] M.F. Nold, C.A. Nold-Petry, J.A. Zepp, B.E. Palmer, P. Bufler, C.A. Dinarello, IL-37
is a fundamental inhibitor of innate immunity, Nat. Immunol. 11 (11) (2010 Nov)
1014–1022.
[41] Z. Dong, W. Tai, W. Lei, Y. Wang, Z. Li, T. Zhang, IL-27 inhibits the TGF-β1-
induced epithelial-mesenchymal transition in alveolar epithelial cells, BMC Cell
Biol. 17 (2016 Mar) 7.
[42] Drakopanagiotakis F, Xifteri A, Polychronopoulos V, Bouros D. Apoptosis in lung
injury and fibrosis. Eur Respir J [Internet]. 2008 Dec 1;32(6):1631 LP – 1638.
Available from: http://erj.ersjournals.com/content/32/6/1631.abstract.
[43] M. Kasper, K. Barth, Potential contribution of alveolar epithelial type I cells to
pulmonary fibrosis, Biosci. Rep. [Internet]. 37 (6) (2017). BSR20171301.
Available from.
[44] M. Plataki, A.V. Koutsopoulos, K. Darivianaki, G. Delides, N.M. Siafakas,
D. Bouros, Expression of Apoptotic and Antiapoptotic Markers in Epithelial Cells
in Idiopathic Pulmonary Fibrosis, Available from, Chest [Internet]. 127 (1)
(2005) 266–274, http://www.sciencedirect.com/science/article/pii/S0012369
21532403X.
[45] B.D. Uhal, R. Wang, J. Laukka, J. Zhuang, V. Soledad-Conrad, G. Filippatos,
Inhibition of amiodarone-induced lung fibrosis but not alveolitis by angiotensin
system antagonists, Pharmacol. Toxicol. 92 (2) (2003 Feb) 81–87.
[46] J.V. Barbas-Filho, M.A. Ferreira, A. Sesso, R.A. Kairalla, C.R. Carvalho, V.
L. Capelozzi, Evidence of type II pneumocyte apoptosis in the pathogenesis of
idiopathic pulmonary fibrosis (IFP)/usual interstitial pneumonia (UIP), J. Clin.
Pathol. 54 (2) (2001 Feb) 132–138.
[47] P.D. Bozyk, B.B. Moore, Prostaglandin E2 and the pathogenesis of pulmonary
fibrosis, Am. J. Respir. Cell Mol. Biol. 45 (3) (2011 Sep) 445–452.
[48] B.D. Uhal, The role of apoptosis in pulmonary fibrosis, Eur. Respir. Rev.
[Internet]. 17 (109) (2008), 138 LP – 144. Available from:.
[49] X. Li, H. Zhang, V. Soledad-Conrad, J. Zhuang, B.D. Uhal, Bleomycin-induced
apoptosis of alveolar epithelial cells requires angiotensin synthesis de novo, Am.
J. Physiol. Lung Cell. Mol. Physiol. 284 (3) (2003 Mar) L501–L507.
[50] R. Wang, G. Alam, A. Zagariya, C. Gidea, H. Pinillos, O. Lalude, et al., Apoptosis
of lung epithelial cells in response to TNF-alpha requires angiotensin II generation
de novo, J. Cell. Physiol. 185 (2) (2000 Nov) 253–259.
[51] Murray ME, Gavile CM, Nair JR, Koorella C, Carlson LM, Buac D, et al. CD28-
mediated pro-survival signaling induces chemotherapeutic resistance in multiple
myeloma. Blood [Internet]. 2014/04/29. 2014 Jun 12;123(24):3770–9. Available
from: https://www.ncbi.nlm.nih.gov/pubmed/24782505.
[52] C. Camperio, M. Muscolini, E. Volpe, D. Di Mitri, R. Mechelli, M.C. Buscarinu, et
al., CD28 ligation in the absence of TCR stimulation up-regulates IL-17A and pro-
inflammatory cytokines in relapsing-remitting multiple sclerosis T lymphocytes,
Immunol. Lett. 158 (1–2) (2014) 134–142.
[53] Kunkl M, Mastrogiovanni M, Porciello N, Caristi S, Monteleone E, Arcieri S, et al.
CD28 Individual Signaling Up-regulates Human IL-17A Expression by Promoting
the Recruitment of RelA/NF-κB and STAT3 Transcription Factors on the Proximal
Promoter. Front Immunol [Internet]. 2019 Apr 24;10:864. Available from:
https://www.ncbi.nlm.nih.gov/pubmed/31068940.
[54] M.M. Gouda, Y.P. Bhandary, Curcumin down-regulates IL-17A mediated p53-
fibrinolytic system in bleomycin induced acute lung injury in vivo, J. Cell.
Biochem. 119 (9) (2018 Sep) 7285–7299.
[55] M.M. Gouda, A. Prabhu, Y.P. Bhandary, Curcumin alleviates IL-17A-mediated
p53-PAI-1 expression in bleomycin-induced alveolar basal epithelial cells, J. Cell.
Biochem. 119 (2) (2018 Feb) 2222–2230.
[56] C. Chen, L. Shi, Y. Li, X. Wang, S. Yang, Disease-specific dynamic biomarkers
selected by integrating inflammatory mediators with clinical informatics in ARDS
patients with severe pneumonia, Cell Biol. Toxicol. 32 (3) (2016 Jun) 169–184.
[57] H. Zhu, H. Huang, D. Wu, N. Dong, L. Dong, C. Chen, et al., Regulatory
mechanism of NOV/CCN3 in the inflammation and apoptosis of lung epithelial
alveolar cells upon lipopolysaccharide stimulation, Mol. Med. Rep. (2019) 1–9.
[58] Schuliga M. The Inflammatory Actions of Coagulant and Fibrinolytic Proteases in
Disease. 2015;2015.
[59] McKeown S, Richter AG, O'Kane C, McAuley DF, Thickett DR. MMP
expression and abnormal lung permeability are important determinants of
outcome in IPF. Eur Respir J [Internet]. 2009 Jan 1;33(1):77 LP – 84. Available
from: http://erj.ersjournals.com/content/33/1/77.abstract.
[60] V. Navaratnam, A.W. Fogarty, T. McKeever, N. Thompson, G. Jenkins, S.
R. Johnson, et al., Presence of a prothrombotic state in people with idiopathic
pulmonary fibrosis: a population-based case-control study, Thorax 69 (3) (2014
Mar) 207–215.
[61] D. Zentina, I. Stukena, A. Krams, A. Lejnieks, PAI-1 Level Differences in Malignant
Plural Effusion, Parapneumonic Pleuritis, and Cardiac Hydrothorax, Medicina
(Kaunas) [Internet]. 55 (9) (2019) 567. Available from:.
[62] Y.-J. Park, G. Liu, E.F. Lorne, X. Zhao, J. Wang, Y. Tsuruta, et al., PAI-1 inhibits
neutrophil efferocytosis, Proc. Natl. Acad. Sci. U S A. 105 (33) (2008 Aug)
11784–11789.
[63] M. Birgel, H. Gottschling-Zeller, K. Röhrig, H. Hauner, Role of cytokines in the
regulation of plasminogen activator inhibitor-1 expression and secretion in newly
differentiated subcutaneous human adipocytes, Arterioscler. Thromb. Vasc. Biol.
20 (6) (2000 Jun) 1682–1687.
[64] C.-Y. Chiu, K.-S. Wong, J.-L. Huang, M.-H. Tasi, T.-Y. Lin, S.-Y. Hsieh,
Proinflammatory cytokines, fibrinolytic system enzymes, and biochemical indices
in children with infectious para-pneumonic effusions, Pediatr. Infect. Dis. J. 27
(8) (2008 Aug) 699–703.
[65] P.A. Akpan, J.O. Akpotuzor, E.E. Osim, The role of cytokines in fibrinolysis: A
case study of active tuberculosis, J. Infect. Dis. Med. Microbiol. 1 (1) (2017) 1–5.
[66] Shetty P, Velusamy T, Bhandary YP, Shetty RS, Liu M-C, Shetty S. Urokinase
expression by tumor suppressor protein p53: a novel role in mRNA turnover. Am
J Respir Cell Mol Biol [Internet]. 2008/04/03. 2008 Sep;39(3):364–72. Available
from: https://pubmed.ncbi.nlm.nih.gov/18390474.
[67] Shetty S, Velusamy T, Idell S, Shetty P, Mazar AP, Bhandary YP, et al. Regulation
of urokinase receptor expression by p53: novel role in stabilization of uPAR
mRNA. Mol Cell Biol [Internet]. 2007/06/04. 2007 Aug;27(16):5607–18.
Available from: https://pubmed.ncbi.nlm.nih.gov/17548471.
[68] N. Tiwari, A.S. Marudamuthu, Y. Tsukasaki, M. Ikebe, J. Fu, S. Shetty, p53- and
PAI-1-mediated induction of C-X-C chemokines and CXCR2: importance in
pulmonary inflammation due to cigarette smoke exposure, Am. J. Physiol. Lung
Cell. Mol. Physiol. 310 (6) (2016 Mar) L496–L506.
M. Fathimath Muneesa et al.
International Immunopharmacology 96 (2021) 107608
8
[69] M. Birgel, H. Gottschling-Zeller, K. Röhrig, H. Hauner, Role of cytokines in the
regulation of plasminogen activator inhibitor-1 expression and secretion in newly
differentiated subcutaneous human adipocytes, Arterioscler. Thromb. Vasc. Biol.
20 (6) (2000) 1682–1687.
[70] Garrett SM, Hsu E, Thomas JM, Pilewski JM, Feghali-Bostwick C. Insulin-like
growth factor (IGF)-II- mediated fibrosis in pathogenic lung conditions. PLoS One
[Internet]. 2019;14(11):1–21. Available from: https://doi.org/10.1371/journal.
pone.0225422.
[71] M.E. Coden, L.F. Loffredo, T. Matthew, B.M. Jeong, K. Nam, B.S. Bochner, et al.,
Fibrinogen Is a Specific Trigger for, Cytolytic. Eosinophil. Degranulat. (2019).
[72] Rubel C, Fernández GC, Rosa FA, Gómez S, Bompadre MB, Coso OA, et al. Soluble
Fibrinogen Modulates Neutrophil Functionality Through the Activation of an
Extracellular Signal-Regulated Kinase-Dependent Pathway. J Immunol [Internet].
2002 Apr 1;168(7):3527 LP – 3535. Available from: http://www.jimmunol.org/
content/168/7/3527.abstract.
[73] S. Dennler, S. Itoh, D. Vivien, P. ten Dijke, S. Huet, J.M. Gauthier, Direct binding
of Smad3 and Smad4 to critical TGF beta-inducible elements in the promoter of
human plasminogen activator inhibitor-type 1 gene, EMBO J. 17 (11) (1998 Jun)
3091–3100.
[74] L. Wu, Z. Luo, J. Zheng, P. Yao, Z. Yuan, X. Lv, et al., IL-33 Can Promote the
Process of Pulmonary Fibrosis by Inducing the Imbalance Between MMP-9 and
TIMP-1, Inflammation 41 (3) (2018 Jun) 878–885.
[75] B. Thapa, Y.H. Kim, H.-J. Kwon, D.-S. Kim, The LRP1-independent mechanism of
PAI-1-induced migration in CpG-ODN activated macrophages, Int. J. Biochem.
Cell Biol. 49 (2014 Apr) 17–25.
[76] Y. Tashiro, C. Nishida, K. Sato-Kusubata, M. Ohki-Koizumi, M. Ishihara, A. Sato,
et al., Inhibition of PAI-1 induces neutrophil-driven neoangiogenesis and
promotes tissue regeneration via production of angiocrine factors in mice, Blood
119 (26) (2012 Jun) 6382–6393.
[77] R.T. Kendall, C.A. Feghali-Bostwick, Fibroblasts in fibrosis: novel roles and
mediators, Front. Pharmacol. 5 (2014) 123.
[78] J.-H. Han, A.-R. Hwang, D.-H. Nam, S. Kim, H.C. Choi, J.H. Lim, et al., ERK5
regulates basic fibroblast growth factor-induced type 1 plasminogen activator
inhibitor expression and cell proliferation in lung fibroblasts, Available from, Life
Sci. [Internet] 135 (2015) 1–8, http://europepmc.org/abstract/MED/26032256.
[79] N.W. Todd, I.G. Luzina, S.P. Atamas, Molecular and cellular mechanisms of
pulmonary fibrosis, Fibrogenesis Tissue Repair. 5 (1) (2012 Jul) 11.
[80] I.E. Fernandez, O. Eickelberg, The impact of TGF-beta on lung fibrosis: from
targeting to biomarkers, Proc. Am. Thorac. Soc. 9 (3) (2012 Jul) 111–116.
[81] E.S. Choi, E.M. Pierce, C. Jakubzick, K.J. Carpenter, S.L. Kunkel, H. Evanoff, et al.,
Focal interstitial CC chemokine receptor 7 (CCR7) expression in idiopathic
interstitial pneumonia, Available from, J. Clin. Pathol. [Internet]. 59 (1) (2006
Jan) 28–39, https://www.ncbi.nlm.nih.gov/pubmed/16394278.
[82] Pierce EM, Carpenter K, Jakubzick C, Kunkel SL, Evanoff H, Flaherty KR, et al.
Idiopathic pulmonary fibrosis fibroblasts migrate and proliferate to CC
chemokine ligand 21. Eur Respir J [Internet]. 2007 Jun 1;29(6):1082 LP – 1093.
Available from: http://erj.ersjournals.com/content/29/6/1082.abstract.
[83] Ma Z, Yu R, Zhu Q, Sun L, Jian L, Wang X, et al. CXCL16/CXCR6 axis promotes
bleomycin-induced fibrotic process in MRC-5 cells via the PI3K/AKT/FOXO3a
pathway. Int Immunopharmacol [Internet]. 2020;81:106035. Available from:
http://europepmc.org/abstract/MED/31753588.
[84] Y.P. Moodley, N.L.A. Misso, A.K. Scaffidi, M. Fogel-Petrovic, R.J. McAnulty, G.
J. Laurent, et al., Inverse effects of interleukin-6 on apoptosis of fibroblasts from
pulmonary fibrosis and normal lungs, Am. J. Respir. Cell Mol. Biol. 29 (4) (2003
Oct) 490–498.
[85] Y.P. Moodley, A.K. Scaffidi, N.L. Misso, C. Keerthisingam, R.J. McAnulty, G.
J. Laurent, et al., Fibroblasts isolated from normal lungs and those with idiopathic
pulmonary fibrosis differ in interleukin-6/gp130-mediated cell signaling and
proliferation, Available from, Am. J. Pathol. [Internet] 163 (1) (2003 Jul)
345–354, https://www.ncbi.nlm.nih.gov/pubmed/12819039.
[86] A.K. Scaffidi, S.E. Mutsaers, Y.P. Moodley, R.J. McAnulty, G.J. Laurent, P.
J. Thompson, et al., Oncostatin M stimulates proliferation, induces collagen
production and inhibits apoptosis of human lung fibroblasts, Br. J. Pharmacol.
136 (5) (2002 Jul) 793–801.
[87] D.V. Pechkovsky, C.M. Prele, J. Wong, C.M. Hogaboam, R.J. McAnulty, G.
J. Laurent, et al., STAT3-mediated signaling dysregulates lung fibroblast-
myofibroblast activation and differentiation in UIP/IPF, Am. J. Pathol. 180 (4)
(2012 Apr) 1398–1412.
[88] A.M. Tager, R.L. Kradin, P. LaCamera, S.D. Bercury, G.S.V. Campanella, C.
P. Leary, et al., Inhibition of pulmonary fibrosis by the chemokine IP-10/CXCL10,
Am. J. Respir. Cell Mol. Biol. 31 (4) (2004 Oct) 395–404.
[89] C.P. Baran, J.M. Opalek, S. McMaken, C.A. Newland, J.M.J. O’Brien, M.
G. Hunter, et al., Important roles for macrophage colony-stimulating factor, CC
chemokine ligand 2, and mononuclear phagocytes in the pathogenesis of
pulmonary fibrosis, Am. J. Respir. Crit. Care Med. 176 (1) (2007 Jul) 78–89.
[90] Kass DJ, Yu G, Loh KS, Savir A, Borczuk A, Kahloon R, et al. Cytokine-like factor 1
gene expression is enriched in idiopathic pulmonary fibrosis and drives the
accumulation of CD4+ T cells in murine lungs: evidence for an antifibrotic role in
bleomycin injury. Am J Pathol [Internet]. 2012/03/16. 2012 May;180(5):
1963–78. Available from: https://www.ncbi.nlm.nih.gov/pubmed/22429962.
[91] S.B. Shaikh, B.Y. Prabhakar, Effect of curcumin on IL-17A mediated pulmonary
AMPK kinase/cyclooxygenase-2 expressions via activation of NFκB in bleomycin-
induced acute lung injury in vivo, Available from, Int. Immunopharmacol.
[Internet]. 85 (2020), 106676, http://europepmc.org/abstract/MED/32535538.
[92] Moradi S, Ziaei R, Foshati S, Mohammadi H, Nachvak SM, Rouhani MH. Jo ur na l
P re ro of. Complement Ther Med [Internet]. 2019;102211. Available from:
https://doi.org/10.1016/j.ctim.2019.102211.
[93] Song J, Wu C, Zhang X, Sorokin LM. In Vivo Processing of CXCL5 (LIX) by Matrix
Metalloproteinase (MMP)-2 and MMP-9 Promotes Early Neutrophil Recruitment
in IL-1β–Induced Peritonitis. J Immunol [Internet]. 2013 Jan 1;190(1):401 LP –
410. Available from: http://www.jimmunol.org/content/190/1/401.abstract.
[94] R. Malaviya, V.R. Sunil, A. Venosa, V.L. Verissimo, J.A. Cervelli, K.N. Vayas, et
al., Attenuation of Nitrogen Mustard-Induced Pulmonary Injury and Fibrosis by
Anti-Tumor Necrosis Factor-α Antibody, Toxicol. Sci. [Internet]. 148 (1) (2015)
71–88. Available from: https://pubmed.ncbi.nlm.nih.gov/26243812.
[95] H. Oku, T. Shimizu, T. Kawabata, M. Nagira, I. Hikita, A. Ueyama, et al.,
Antifibrotic action of pirfenidone and prednisolone: Different effects on
pulmonary cytokines and growth factors in bleomycin-induced murine
pulmonary fibrosis, Eur. J. Pharmacol. 590 (1–3) (2008) 400–408.
[96] J. Navarro-Partida, A.B. Martinez-Rizo, J. Gonzalez-Cuevas, G. Arrevillaga-Boni,
V. Ortiz-Navarrete, J. Armendariz-Borunda, Pirfenidone restricts Th2
differentiation in vitro and limits Th2 response in experimental liver fibrosis,
Available from, Eur. J. Pharmacol. [Internet]. 678 (1) (2012) 71–77, https://
www.sciencedirect.com/science/article/pii/S0014299911015597.
[97] P. Bizargity, K. Liu, L. Wang, W.W. Hancock, G.A. Visner, Inhibitory effects of
pirfenidone on dendritic cells and lung allograft rejection, Available from,
Transplantation [Internet] 94 (2) (2012 Jul 27) 114–122, https://pubmed.ncbi.
nlm.nih.gov/22743547.
[98] G.A. Visner, F. Liu, P. Bizargity, H. Liu, K. Liu, J. Yang, et al., Pirfenidone inhibits
T-cell activation, proliferation, cytokine and chemokine production, and host
alloresponses, Available from, Transplantation [Internet]. 88 (3) (2009 Aug 15)
330–338, https://pubmed.ncbi.nlm.nih.gov/19667934.
[99] Wollin L, Maillet I, Quesniaux V, Holweg A, Ryffel B. Antifibrotic and Anti-
inflammatory Activity of the Tyrosine Kinase Inhibitor Nintedanib in
Experimental Models of Lung Fibrosis. J Pharmacol Exp Ther [Internet]. 2014
May 1;349(2):209 LP – 220. Available from: http://jpet.aspetjournals.org/
content/349/2/209.abstract.
[100] M.R. Horton, V. Santopietro, L. Mathew, K.M. Horton, A.J. Polito, M.C. Liu, et al.,
Thalidomide for the treatment of cough in idiopathic pulmonary fibrosis: a
randomized trial, Ann. Intern. Med. 157 (6) (2012 Sep) 398–406.
[101] G.S. Bogatkevich, A. Ludwicka-Bradley, P.J. Nietert, T. Akter, J. van Ryn, R.
M. Silver, Antiinflammatory and antifibrotic effects of the oral direct thrombin
inhibitor dabigatran etexilate in a murine model of interstitial lung disease,
Arthritis Rheum. 63 (5) (2011 May) 1416–1425.
[102] A.B. Fernández, R.H. Karas, A.A. Alsheikh-Ali, P.D. Thompson, Statins and
interstitial lung disease: a systematic review of the literature and of food and drug
administration adverse event reports, Chest 134 (4) (2008 Oct) 824–830.
[103] J.-F. Xu, G.R. Washko, K. Nakahira, H. Hatabu, A.S. Patel, I.E. Fernandez, et al.,
Statins and pulmonary fibrosis: the potential role of NLRP3 inflammasome
activation, Am. J. Respir. Crit. Care Med. 185 (5) (2012 Mar) 547–556.
[104] N. Saad, P. Camus, S. Suissa, P. Ernst, Statins and the risk of interstitial lung
disease: a cohort study, Thorax 68 (4) (2013 Apr) 361–364.
[105] S. Vedel-Krogh, S.F. Nielsen, B.G. Nordestgaard, Statin Use Is Associated with
Reduced Mortality in Patients with Interstitial Lung Disease, PLoS ONE 10 (10)
(2015) e0140571.
[106] M. Kreuter, F. Bonella, T.M. Maher, U. Costabel, P. Spagnolo, D. Weycker, et al.,
Effect of statins on disease-related outcomes in patients with idiopathic
pulmonary fibrosis, Thorax 72 (2) (2017) 148–153.
[107] Lands LC, Stanojevic S. Oral non-steroidal anti-inflammatory drug therapy for
lung disease in cystic fibrosis. Cochrane database Syst Rev. 2016;4(4):CD001505.
[108] K.R. Flaherty, G.B. Toews, J.P. Lynch 3rd, E.A. Kazerooni, B.H. Gross, R.
L. Strawderman, et al., Steroids in idiopathic pulmonary fibrosis: a prospective
assessment of adverse reactions, response to therapy, and survival, Am. J. Med.
110 (4) (2001 Mar) 278–282.
[109] M. Mura, G. Belmonte, S. Fanti, P. Contini, A.M.G. Pacilli, L. Fasano, et al.,
Inflammatory activity is still present in the advanced stages of idiopathic
pulmonary fibrosis, Respirology 10 (5) (2005 Nov) 609–614.
[110] K. Grijm, H.J. Verberne, F.H. Krouwels, F.R. Weller, H.M. Jansen, P. Bresser,
Semiquantitative 67Ga scintigraphy as an indicator of response to and prognosis
after corticosteroid treatment in idiopathic interstitial pneumonia, J. Nucl. Med.
46 (9) (2005 Sep) 1421–1426.
[111] J.C. Schupp, H. Binder, B. Jager, G. Cillis, G. Zissel, J. Muller-Quernheim, et al.,
Macrophage activation in acute exacerbation of idiopathic pulmonary fibrosis,
PLoS ONE 10 (1) (2015) e0116775.
[112] Balestro E, Calabrese F, Turato G, Lunardi F, Bazzan E, Marulli G, et al. Immune
Inflammation and Disease Progression in Idiopathic Pulmonary Fibrosis. PLoS
One [Internet]. 2016 May 9;11(5):e0154516. Available from: https://doi.org/
10.1371/journal.pone.0154516.
[113] A. Zinellu, P. Paliogiannis, E. Sotgiu, S. Mellino, A.A. Mangoni, E. Zinellu, et al.,
Blood Cell Count Derived Inflammation Indexes in Patients with Idiopathic
Pulmonary Fibrosis, Lung 198 (5) (2020) 821–827, https://doi.org/10.1007/
s00408-020-00386-7.
M. Fathimath Muneesa et al.

More Related Content

Similar to Inflammatory mediators in various molecular pathways involved in the development of pulmonary fibrosis pdf.pdf

Thesis-Final Draft
Thesis-Final DraftThesis-Final Draft
Thesis-Final DraftEmer Shelly
 
Neutrophil extracellular trap
Neutrophil extracellular trapNeutrophil extracellular trap
Neutrophil extracellular trapNainshi Bhatt
 
Mast cells in health and disease
Mast cells in health and disease  Mast cells in health and disease
Mast cells in health and disease Pannaga Kumar
 
Communicable Disease_ Review Malaria paper
Communicable Disease_ Review Malaria paperCommunicable Disease_ Review Malaria paper
Communicable Disease_ Review Malaria paperThant Zin
 
Dr William Barnes - The I Factor - Inflammation, Immunity, Illness
Dr William Barnes - The I Factor - Inflammation, Immunity, IllnessDr William Barnes - The I Factor - Inflammation, Immunity, Illness
Dr William Barnes - The I Factor - Inflammation, Immunity, IllnessDr William Barnes
 
Immuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal diseaseImmuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal diseaseGanesh Nair
 
Innate immune response
Innate immune responseInnate immune response
Innate immune responseReyam Ahmed
 
Smoking related idiopathic interstitial pneumonia a review 2016
Smoking related idiopathic interstitial pneumonia a review 2016Smoking related idiopathic interstitial pneumonia a review 2016
Smoking related idiopathic interstitial pneumonia a review 2016Rosy Olmos Tufiño
 
Asthma advances in pathophysiology
Asthma advances in pathophysiologyAsthma advances in pathophysiology
Asthma advances in pathophysiologyYosunii
 

Similar to Inflammatory mediators in various molecular pathways involved in the development of pulmonary fibrosis pdf.pdf (20)

Defective phagocytosis in airways disease
Defective phagocytosis in airways diseaseDefective phagocytosis in airways disease
Defective phagocytosis in airways disease
 
Thesis-Final Draft
Thesis-Final DraftThesis-Final Draft
Thesis-Final Draft
 
Corticosteriods
CorticosteriodsCorticosteriods
Corticosteriods
 
Neutrophil extracellular trap
Neutrophil extracellular trapNeutrophil extracellular trap
Neutrophil extracellular trap
 
Immunity & Inflammation
Immunity & InflammationImmunity & Inflammation
Immunity & Inflammation
 
periodontitis-and-oral-cancer.pdf
periodontitis-and-oral-cancer.pdfperiodontitis-and-oral-cancer.pdf
periodontitis-and-oral-cancer.pdf
 
periodontitis and oral cancer.pptx
periodontitis and oral cancer.pptxperiodontitis and oral cancer.pptx
periodontitis and oral cancer.pptx
 
Mast cells in health and disease
Mast cells in health and disease  Mast cells in health and disease
Mast cells in health and disease
 
Sepsis
SepsisSepsis
Sepsis
 
plospathogen
plospathogenplospathogen
plospathogen
 
Communicable Disease_ Review Malaria paper
Communicable Disease_ Review Malaria paperCommunicable Disease_ Review Malaria paper
Communicable Disease_ Review Malaria paper
 
PM2012-808260
PM2012-808260PM2012-808260
PM2012-808260
 
Dr William Barnes - The I Factor - Inflammation, Immunity, Illness
Dr William Barnes - The I Factor - Inflammation, Immunity, IllnessDr William Barnes - The I Factor - Inflammation, Immunity, Illness
Dr William Barnes - The I Factor - Inflammation, Immunity, Illness
 
Immuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal diseaseImmuno microbial pathogenesis of periodontal disease
Immuno microbial pathogenesis of periodontal disease
 
rcmb%2E2015-0009oc
rcmb%2E2015-0009ocrcmb%2E2015-0009oc
rcmb%2E2015-0009oc
 
Innate immune response
Innate immune responseInnate immune response
Innate immune response
 
Immunity
Immunity Immunity
Immunity
 
chronic inflamation
chronic inflamationchronic inflamation
chronic inflamation
 
Smoking related idiopathic interstitial pneumonia a review 2016
Smoking related idiopathic interstitial pneumonia a review 2016Smoking related idiopathic interstitial pneumonia a review 2016
Smoking related idiopathic interstitial pneumonia a review 2016
 
Asthma advances in pathophysiology
Asthma advances in pathophysiologyAsthma advances in pathophysiology
Asthma advances in pathophysiology
 

Recently uploaded

Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...
Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...
Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...akbard9823
 
定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一
定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一
定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一Fs
 
Git and Github workshop GDSC MLRITM
Git and Github  workshop GDSC MLRITMGit and Github  workshop GDSC MLRITM
Git and Github workshop GDSC MLRITMgdsc13
 
How is AI changing journalism? (v. April 2024)
How is AI changing journalism? (v. April 2024)How is AI changing journalism? (v. April 2024)
How is AI changing journalism? (v. April 2024)Damian Radcliffe
 
VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...
VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...
VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...aditipandeya
 
Denver Web Design brochure for public viewing
Denver Web Design brochure for public viewingDenver Web Design brochure for public viewing
Denver Web Design brochure for public viewingbigorange77
 
VIP Call Girls Kolkata Ananya 🤌 8250192130 🚀 Vip Call Girls Kolkata
VIP Call Girls Kolkata Ananya 🤌  8250192130 🚀 Vip Call Girls KolkataVIP Call Girls Kolkata Ananya 🤌  8250192130 🚀 Vip Call Girls Kolkata
VIP Call Girls Kolkata Ananya 🤌 8250192130 🚀 Vip Call Girls Kolkataanamikaraghav4
 
Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝soniya singh
 
Russian Call girls in Dubai +971563133746 Dubai Call girls
Russian  Call girls in Dubai +971563133746 Dubai  Call girlsRussian  Call girls in Dubai +971563133746 Dubai  Call girls
Russian Call girls in Dubai +971563133746 Dubai Call girlsstephieert
 
AlbaniaDreamin24 - How to easily use an API with Flows
AlbaniaDreamin24 - How to easily use an API with FlowsAlbaniaDreamin24 - How to easily use an API with Flows
AlbaniaDreamin24 - How to easily use an API with FlowsThierry TROUIN ☁
 
Call Girls South Delhi Delhi reach out to us at ☎ 9711199012
Call Girls South Delhi Delhi reach out to us at ☎ 9711199012Call Girls South Delhi Delhi reach out to us at ☎ 9711199012
Call Girls South Delhi Delhi reach out to us at ☎ 9711199012rehmti665
 
VIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call Girl
VIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call GirlVIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call Girl
VIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call Girladitipandeya
 
Chennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts service
Chennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts serviceChennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts service
Chennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts servicevipmodelshub1
 
Call Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on Delivery
Call Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on DeliveryCall Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on Delivery
Call Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on Deliverybabeytanya
 
定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一
定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一
定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一Fs
 
VIP Kolkata Call Girl Kestopur 👉 8250192130 Available With Room
VIP Kolkata Call Girl Kestopur 👉 8250192130  Available With RoomVIP Kolkata Call Girl Kestopur 👉 8250192130  Available With Room
VIP Kolkata Call Girl Kestopur 👉 8250192130 Available With Roomdivyansh0kumar0
 

Recently uploaded (20)

Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...
Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...
Sushant Golf City / best call girls in Lucknow | Service-oriented sexy call g...
 
定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一
定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一
定制(AUT毕业证书)新西兰奥克兰理工大学毕业证成绩单原版一比一
 
Rohini Sector 26 Call Girls Delhi 9999965857 @Sabina Saikh No Advance
Rohini Sector 26 Call Girls Delhi 9999965857 @Sabina Saikh No AdvanceRohini Sector 26 Call Girls Delhi 9999965857 @Sabina Saikh No Advance
Rohini Sector 26 Call Girls Delhi 9999965857 @Sabina Saikh No Advance
 
Git and Github workshop GDSC MLRITM
Git and Github  workshop GDSC MLRITMGit and Github  workshop GDSC MLRITM
Git and Github workshop GDSC MLRITM
 
Call Girls In South Ex 📱 9999965857 🤩 Delhi 🫦 HOT AND SEXY VVIP 🍎 SERVICE
Call Girls In South Ex 📱  9999965857  🤩 Delhi 🫦 HOT AND SEXY VVIP 🍎 SERVICECall Girls In South Ex 📱  9999965857  🤩 Delhi 🫦 HOT AND SEXY VVIP 🍎 SERVICE
Call Girls In South Ex 📱 9999965857 🤩 Delhi 🫦 HOT AND SEXY VVIP 🍎 SERVICE
 
How is AI changing journalism? (v. April 2024)
How is AI changing journalism? (v. April 2024)How is AI changing journalism? (v. April 2024)
How is AI changing journalism? (v. April 2024)
 
sasti delhi Call Girls in munirka 🔝 9953056974 🔝 escort Service-
sasti delhi Call Girls in munirka 🔝 9953056974 🔝 escort Service-sasti delhi Call Girls in munirka 🔝 9953056974 🔝 escort Service-
sasti delhi Call Girls in munirka 🔝 9953056974 🔝 escort Service-
 
VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...
VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...
VIP 7001035870 Find & Meet Hyderabad Call Girls Dilsukhnagar high-profile Cal...
 
Denver Web Design brochure for public viewing
Denver Web Design brochure for public viewingDenver Web Design brochure for public viewing
Denver Web Design brochure for public viewing
 
VIP Call Girls Kolkata Ananya 🤌 8250192130 🚀 Vip Call Girls Kolkata
VIP Call Girls Kolkata Ananya 🤌  8250192130 🚀 Vip Call Girls KolkataVIP Call Girls Kolkata Ananya 🤌  8250192130 🚀 Vip Call Girls Kolkata
VIP Call Girls Kolkata Ananya 🤌 8250192130 🚀 Vip Call Girls Kolkata
 
Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Uttam Nagar Delhi 💯Call Us 🔝8264348440🔝
 
Russian Call girls in Dubai +971563133746 Dubai Call girls
Russian  Call girls in Dubai +971563133746 Dubai  Call girlsRussian  Call girls in Dubai +971563133746 Dubai  Call girls
Russian Call girls in Dubai +971563133746 Dubai Call girls
 
AlbaniaDreamin24 - How to easily use an API with Flows
AlbaniaDreamin24 - How to easily use an API with FlowsAlbaniaDreamin24 - How to easily use an API with Flows
AlbaniaDreamin24 - How to easily use an API with Flows
 
Call Girls South Delhi Delhi reach out to us at ☎ 9711199012
Call Girls South Delhi Delhi reach out to us at ☎ 9711199012Call Girls South Delhi Delhi reach out to us at ☎ 9711199012
Call Girls South Delhi Delhi reach out to us at ☎ 9711199012
 
VIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call Girl
VIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call GirlVIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call Girl
VIP 7001035870 Find & Meet Hyderabad Call Girls LB Nagar high-profile Call Girl
 
Chennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts service
Chennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts serviceChennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts service
Chennai Call Girls Alwarpet Phone 🍆 8250192130 👅 celebrity escorts service
 
young call girls in Uttam Nagar🔝 9953056974 🔝 Delhi escort Service
young call girls in Uttam Nagar🔝 9953056974 🔝 Delhi escort Serviceyoung call girls in Uttam Nagar🔝 9953056974 🔝 Delhi escort Service
young call girls in Uttam Nagar🔝 9953056974 🔝 Delhi escort Service
 
Call Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on Delivery
Call Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on DeliveryCall Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on Delivery
Call Girls In Mumbai Central Mumbai ❤️ 9920874524 👈 Cash on Delivery
 
定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一
定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一
定制(UAL学位证)英国伦敦艺术大学毕业证成绩单原版一比一
 
VIP Kolkata Call Girl Kestopur 👉 8250192130 Available With Room
VIP Kolkata Call Girl Kestopur 👉 8250192130  Available With RoomVIP Kolkata Call Girl Kestopur 👉 8250192130  Available With Room
VIP Kolkata Call Girl Kestopur 👉 8250192130 Available With Room
 

Inflammatory mediators in various molecular pathways involved in the development of pulmonary fibrosis pdf.pdf

  • 1. International Immunopharmacology 96 (2021) 107608 Available online 12 April 2021 1567-5769/© 2021 Elsevier B.V. All rights reserved. Review Inflammatory mediators in various molecular pathways involved in the development of pulmonary fibrosis M. Fathimath Muneesa, Sadiya B. Shaikh, T.M. Jeena, Yashodhar P. Bhandary * Yenepoya Research Centre, Yenepoya University, Deralakatte, Mangalore 575018, Karnataka, India A R T I C L E I N F O Keywords: Inflammation Idiopathic pulmonary fibrosis Cytokines Epithelial-mesenchymal transition Fibrinolytic system Apoptosis A B S T R A C T Idiopathic pulmonary fibrosis (IPF) is a type of interstitial lung disease (ILD) that is marked by scarring of lung tissue, ultimately leading to respiratory failure. The survival rate of IPF is disappointing and to date demonstrates a clinical quandary. The exact etiology of the disease remains under discussion. According to the recent hy­ pothesis, inflammatory mediators cause severe damage to the alveolar epithelium leading to the impairment of the alveolar structure. The role of inflammation in the development of the IPF has been controversial for years. There are two schools of thought regarding the role of inflammation. One group of researchers claims that cell death and fibroblast dysfunction are the primary causes and inflammation is just a secondary cause of IPF. The other group claims inflammation to be the primary cause. Studies using human subjects have also reported inflammation as a critical element in IPF. Inflammatory cytokines serve a major role in commencing the in­ flammatory response in the lungs. Several cytokines are reported to be involved in different molecular mecha­ nisms underlying IPF, some of which also contribute additionally by acting as growth factors. The present review addressed to explore the contribution of various inflammatory cytokines, growth factors, and various other in­ flammatory molecules activating the major molecular pathways involved during the development of IPF. 1. Introduction Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease (ILD) that involves the irreversible scarring of the tissue because of inflammation, the impaired wound healing process, and ultimately fibrosis. Fibrosis is due to the excessive deposition of extracellular ma­ trix (ECM) proteins like fibronectin and collagens in the interstitial space and alveolar septa [1]. This results in the alveolar structural damage reducing the number of functional alveoli and thereby resulting in impaired gas exchange. Thus the disease is associated with reduced lung function and gradual respiratory failure. The first step of diagnosis of IPF involves ruling out the known causes of ILDs. The next step is to check for the usual interstitial pneumonia (UIP) pattern on High-resolution computer tomography (HRCT) and Abbreviations: AIM2, Absent In Melanoma; AKT, Also known as Protein Kinase B (PKB); ARDS, Acute Respiratory Distress Syndrome; ASC, Apoptosis-Associated Speck-Like Protein Containing A Caspase-Recruitment Domain; ATII, Alveolar Type II; BAL, Bronchoalveolar Lavage Fluid; bFGF, Basic Fibroblast Growth Factor; CCL, CC Chemokine Ligand; CCN3, Cellular Communication Network Factor 3; CCR, CC Chemokine Receptor; CD28, Cluster Of Differentiation 28; CXCL, Chemokine (C-X-C Motif) Ligand; CXCR, Chemokine (C-X-C Motif) Receptor; dNLR, Derived Neutrophil To Lymphocyte Ratio; ECM, Extracellular Matrix; EMT, Epithelial- Mesenchymal Transition; ERK, Extracellular Signal-Regulated Kinases; FVC, Forced Vital Capacity; GDF, Growth/Differentiation Factor; HRCT, High-Resolution Computed Tomography; ICAM-1, Intercellular Adhesion Molecule 1; IFN-γ, Interferon Gamma; IGF, Insulin-Like Growth Factor; IKK, Inhibitor Of NF-Κb Kinase; IL, Interleukin; ILD, Interstitial Lung Disease; IPF, Idiopathic Pulmonary Fibrosis; JAK, Janus Kinase; LPS, Lipopolysaccharide; MAPK, Mitogen-Activated Protein Kinase; MCP, Monocyte Chemoattractant Protein; M-CSF, Macrophage Colony-Stimulating Factor; MMP, Matrix Metalloproteinases; NF-KB, Nuclear Factor Kappa B (NF-Κb); NLRP3, NLR Family Pyrin Domain Containing 3; NOD, Nucleotide-Binding And Oligomerization Domain; NUR77, Also Known As Nerve Growth Factor IB (NGFIB); PAI-1, Plasminogen Activator Inhibitor; PBMCs, Peripheral Blood Mononuclear Cells; PDGF, Platelet-Derived Growth Factor; PGE2, Prostaglandin E2; PI3K, Phos­ phoinositide 3-Kinase; PMN, Polymorphonuclear Leukocytes; SIRI, Systemic Inflammation Response Index; SMA, Smooth Muscle Actin; SMAD, Mothers Against Decapentaplegic Homolog; SNAIL-1, Zinc finger protein SNAI1; STAT, Signal Transducer And Activator Of Transcription; TGF-β, Transforming Growth Factor-Beta; TIMP, Tissue Inhibitors Of Matrix Metalloproteinases; TLR, Toll-Like Receptor; TNF-α, Tumor Necrosis Factor; TRAF, Tumor Necrosis Factor Receptor–Associated Factor; UIP, Usual Interstitial Pneumonia; uPA, Urokinase Plasminogen Activator; uPAR, Urokinase Plasminogen Activator Receptor; Wnt, Wingless-Related Inte­ gration Site. * Corresponding author.at: Yenepoya Research Centre, Yenepoya University, Mangalore 575 018, Karnataka, India. E-mail address: yash28bhandary@gmail.com (Y.P. Bhandary). Contents lists available at ScienceDirect International Immunopharmacology journal homepage: www.elsevier.com/locate/intimp https://doi.org/10.1016/j.intimp.2021.107608 Received 10 December 2020; Received in revised form 23 February 2021; Accepted 21 March 2021
  • 2. International Immunopharmacology 96 (2021) 107608 2 surgical lung biopsy. The characteristic histopathological feature in surgical lung biopsies of IPF patients is the prominent fibrotic foci whose appearance is due to the accumulated fibroblasts and myofibroblasts. The median survival rate of an IPF patient after diagnosis is 3–5 years [2]. Currently, there are no clinically effective treatment options against IPF except lung transplantation [3]. The molecular mechanisms under­ lying the disease are also not clearly understood. But in the last few years, researchers across the world have reached significant landmarks associated with the disease. Initially, the inflammation process triggered by an injury was considered to be the primary cause for the initiation and progression of fibrosis. When there is an injury to the alveoli, the resident macrophages release cytokines to attract fibroblasts, epithelial and endothelial cells for promoting wound healing. But if there is persistent injury, the neutrophils and the monocytes are recruited to the sites. These inflammatory cells generate reactive oxygen species which leads to an imbalance in oxidants and anti-oxidants. Eventually, the inflammatory cells gathered secrete growth factors and other mediators contributing to fibrosis [4,5]. But currently, the role of inflammation is a controversial topic among researchers as many studies showed results contradicting the inflammation theory. Various cellular processes play a prime role in the pathogenesis of lung fibrosis (Fig. 1). The present re­ view discusses the role of inflammatory molecules in the core molecular pathways involved during the development of lung fibrosis. Here, we also highlight the controversial role of inflammation in IPF. 2. Inflammatory mediators in epithelial-mesenchymal transition The fibrotic foci in the parenchyma of a fibrotic lung are accumu­ lated with mesenchymal cells. These mesenchymal cells include fibro­ blasts and myofibroblasts, which are highly proliferative and produce an excessive amount of ECM. The origin of the mesenchymal cells may be the circulating fibrocytes or alveolar epithelial cells (AEC) that under­ went epithelial-mesenchymal transition (EMT) [6]. EMT is a process in which the epithelial cells undergo morphological change and attain motility due to the change in cytoskeletal arrangements and dissociation of membrane-associated adherens junctions and desmosomes. Myofibroblasts (or activated fibroblasts) and macrophages secrete chemokines and growth factors that activate the ERK-MAPK (extrac­ ellularsignal-regulated kinase-mitogen activated protein kinase), Smads, and the PI3K-AKT (Phosphatidylinositol-3-kinase-Protein kinase B) signaling pathways to induce EMT [7]. Cytokines like transforming growth factor-β (TGF-β), tumor necrosis factor (TNF-α), and interleukin- 1β (IL-1β) are reported to induce EMT in the mesenchymal (myofibro­ blast-like) phenotype in the fibrotic foci [8]. TGF-β is the most studied and most crucial cytokine playing role in this process. High expression of TGF-β in bronchoalveolar lavage (BAL) fluid and at the regions of epithelial cells lining the fibrotic foci have been reported [9]. TGF-β mediated EMT is through Mothers against decapentaplegic homolog 2 (SMAD-2) pathway but not MAPK pathways. TGF-β induces elevated expression of matrix metalloproteinase 2 (MMP-2). This could disinte­ grate the basement membrane and in turn, induce motility of the cells [10]. Shaikh et al. 2020., reported the potential contribution of TGF-β1 promoting p53-fibrinolytic system during alveolar EMT in A549 cells [11].There are notable similarities between lung fibrosis and lung can­ cer concerning genetic and epigenetic alterations, EMT, fibroblasts in­ vasion, and some signal transduction pathways [12,13]. Elevated levels of IL-17A are reported in IPF and bleomycin-induced pulmonary fibrosis (BLM-PF) models [14,15]. IL-17A mediates EMT through TGF-β mediated ERK1/2 and Smad2/3 activation [16]. Studies also reported the participation of IL-17A in activation of TGF-β via smad dependent and independent signaling in alveolar epithelial cells during ALI in vivo [17]. In a recent study, it was reported that IL-17A and growth/differentiation factor 15 (GDF15) induced EMT of lung epithe­ lial cells in response to cigarette smoke [18]. IL-6 promotes growth and EMT in CD133 + A549 cells by targeting Hhg (Hedgehog protein), Bcl-2 (B cell lymphoma 2), and Erk/MEK [19,20]. Zhang et al.,demonstrated that inhibition of IL-18 in BLM-PF led to an increase in epithelial marker, E-cadherin. The study also concluded that IL-18 regulated EMT by up- regulating Snail-1 [21]. NOD-like receptor protein 3 (NLRP3) inflammasome is a multi­ protein complex that activates caspase-1 which in turn leads to the secretion of pro-inflammatory cytokines, IL-1β and IL-18, and their maturation. The components of this protein complex are a sensor molecule, NLRP3, an adaptor molecule apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and pro-cas­ pase-1[22,23]. It is found to be involved in acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), IPF, asthma, and silicosis [24]. NLRP3 inflammasome mediates EMT through TAK1-MAPK-Snail/NF-κB pathway [25]. IL-1β can activate EMT related pathways like Wnt/GSK3β (Wingless-Related Integration Site/glycogen synthase kinase 3β) and TGF-/Smad signaling pathways [26,27]. The expression and activation of another inflammasome named absent in melanoma 2 (AIM2) are enhanced in IPF peripheral blood mononuclear cells (PBMCs) as well as in a lung fibrosis exacerbation model [28,29]. AIM2, upon activation in PBMCs, causes the release of pro-fibrotic cy­ tokines IL-1α, IL-18, and TGF-β in a TLR4-/caspase-1-/caspase-8-/cal­ pain-independent fashion and also releases caspase-4 [29]. Recent studies reports that AIM2 inflammasome-dependent inflammation plays apivotal role during progression of pulmonary fibrosis [30]. Fig 1. Cellular factors responsible for lung fibrosis: Various cellular processes like apoptosis of alveolar epithelial cells, EMT, inflammatory cells infiltration, myofibroblast invasion or differentiation, and DNA damage play a major role in the pathogenesis of lung fibrosis. M. Fathimath Muneesa et al.
  • 3. International Immunopharmacology 96 (2021) 107608 3 CXCR3 is the receptor for chemokine ligand CXCL9. Beirne et al., demonstrated the elevated expression of CXCR3 in the proximity of cells that have undergone EMT in IPF surgical lung biopsies. Their study concluded the antifibrotic activity of CXCL9 via abrogation of the TGFβ signaling pathway by the reduction of phosphorylation of Smad2 and Smad3 in AECs [31]. The expression of IL-22, belonging to the IL-10 family, is shown to be reduced in BLM-PF mice models. IL-22 inhibits EMT of AECs [32]. In a study conducted on HCl-induced epithelial remodeling, it was found that monocytes interact with epithelial cells through ICAM-1 (Intracellular adhesion molecule-1) and enhance EMT and induce the release of IL-8 and PDGF [33]. IL-8 interacts with CXCR1/CXCR2 to activate the MAPK pathway and is shown to favor fibrosis [34,35]. IL-37 is an antifibrotic interleukin [36–38]. IL-37 and CCL22 co- localizes and inhibits proliferation and EMT by abrogating IL-6/Signal Transducer and Activator of Transcription-3 (STAT3) signaling pathway in A549 cells [37,39]. IL-37 also binds to Smad3 and down­ regulates different signaling pathways, including focal adhesion kinase (FAK), STAT, p53, MAP kinase p38α, and mTOR (mammalian target of rapamycin)[40]. IL-27 is also a negative regulator of EMT in AEC through the JAK/STAT and TGF-β/Smad signalling pathways [41]. 3. Inflammatory mediators in apoptosis of epithelial cells Apoptosis plays a significant role in the pathogenesis of PF. Epithelial cells undergo apoptosis in patients with IPF and also in PF animal models. Intratracheal administration of BLM-induced elevates expres­ sion of fas on alveolar epithelium [42]. The abnormal activation of AEC is sufficient to initiate fibrotic activities [43]. Alveolar type II (AT-II) epithelial cells are the ones that undergo apoptosis. AT-II cells have the capability of being transformed into type 1 cells [44]. Consistent with the theory, it was proved that collagen deposition could be reduced by the installation of apoptotic inhibitors in animal models [45]. The presence of epithelial cell apoptosis in normal lungs suggests that apoptosis can probably be an initiating event in fibrogenesis [46]. Epithelial cells are important because they produce mediators that are antifibrotic. For instance, prostaglandin E2 (PGE2) produced by epithelial cells inhibit fibroblast proliferation. Plasminogen activators and metalloproteinases expressed by them aid in fibrin and ECM degradation [47]. Epithelial cells probably play as a barrier to cytokines secreted by activated macrophages and thereby protecting the under­ lying tissue. Thus apoptosis of epithelial cells could play a crucial role in the onset of fibrosis [48]. TNF-α induces AEC apoptosis dependent on angiotensin II in the BLM-PF model [49,50]. It activates the IKK ( IκB kinase ) and NF-KB (nuclear factor kappa light chain enhancer of activated B cells) com­ plex. CD28 (Cluster of differentiation-23) is a costimulatory protein for the activation of T-cells expressed on antigen-presenting cells. CD28 deficiency abrogates blast exposure-induced lung tissue apoptosis [49]. In multiple myeloma, the CD28 pathway is associated with the phos­ phorylation of PI3K/Akt and inactivation of the FoxO1 [51]. CD28 in­ duces the expression of IL-17A and other cytokines, IL-6 and IL-8, in multiple sclerosis T lymphocytes [52]. It activates RelA/NF-κB which in turn induces IL-6 expression that leads to the activation and nuclear translocation of tyrosine-phosphorylated STAT3 (pSTAT3). pSTAT3 as­ sociates with RelA/NF-κB by binding to the specific sequences in the proximal promoter of the human IL-17A gene, thereby inducing its expression. Class 1A PI3K regulates CD28-mediated RelA/NF-κB and STAT3 recruitments and trans-activation of IL-17A promoter [53]. IL- 17A induces the expression of p53 augmenting apoptosis of AEC [54,55]. Elevated levels of nephroblastoma overexpressed protein (NOV; also known as CCN3), an inflammatory mediator, was observed in the plasma of patients with severe pneumonia-induced ARDS when compared to the healthy controls [56]. CCN3 brings about pro-apoptotic effects on AECs by the activation of the TGF-β signaling pathway, NF-κB signaling pathway, and Bcl-2/caspase-3 pathway [57]. CCN3 could play a pro-fibrotic role by inducing apoptosis of AECs and the destruction of epithelial integrity, ultimately leading to IPF. The inflammatory cyto­ kines such as IL-17A, IL-6, TNF-α, IL-33, and IL-37 act via various signaling pathways through cell surface receptors playing a pivotal role in regulating alveolar apoptosis and EMT causing alveolar damage and ultimately leading to the development of lung fibrosis (Fig. 2). 4. Inflammatory mediators in the impaired fibrinolytic system Extravascular accumulation of fibrin is implicated in the progression of PF [58]. When an injury occurs in the lung tissue, the vascular permeability increases; due to which the coagulation and fibrinolytic factors leak out to the impaired tissue [59]. Thrombosis and fibrin deposition in the alveolar space are common features in a fibrotic lung [60]. The balance between fibrinolysis and coagulation plays a critical role in tissue homeostasis. The main components of the lung fibrinolytic system are urokinase plasminogen activators (uPA), urokinase plas­ minogen activator receptor (uPAR), and plasminogen activator inhibitor Type-1 (PAI-1). uPA and uPAR promote fibrinolysis whereas PAI-1 in­ hibits fibrinolysis. Increased level of PAI-1 due to inflammatory pro­ cesses is observed in pleural effusion [61]. Park et al., observed increased levels of PAI-1 in the plasma and the injured lungs of patients with PF. The study also demonstrated PAI-1 as a potential “don’t eat me signals” for viable and apoptotic neutrophils [62]. The persistence of apoptotic neutrophils in the fibrotic lung may worsen the condition by releasing intracellular toxic substances to the surroundings through necrosis or autolysis of the apoptotic bodies. Regulation of coagulation and fibrinolytic system by inflammatory mediators is studied extensively in certain pathological conditions like sepsis. Stimulation of human adipocytes with IL-β or TNF-α or TGF-β elevates the expression of PAI-1 [63]. Chiu et al., suggested that the increased expression of pro-inflammatory cytokines may lead to fibri­ nolytic imbalance and result in fibrin deposition in the pleura of the pediatric para-pneumonic lung [64]. Akpan et al., noted that interferon- γ (IFN-γ) and IL-10 increased fibrinolysis inactive tuberculosis which involves chronic inflammation [65]. A recent study proved that IL-17A enhanced the expression of PAI-1 and p53 [55] and downregulates the expression of uPA and uPAR in acute lung injury (ALI) in vivo [54]. p53 is reported to destabilize the mRNAs of uPA and uPAR and stabilization of mRNA of PAI-1[66,67].Tiwari et al., stated that PAI-1 attracts poly­ morphonuclear (PMN) cells and other inflammatory cells to alveolar space through induction of expression of chemokines by AECs [68]. IL- 1β and IL-12 have also been suggested to play important roles in the fibrinolytic system [69]. IGF-II (Insulin-like growth factor-II) impairs MMP and tissue inhibitor of metalloproteinases (TIMP) balance [70]. IL- 5 stimulates eosinophils to adhere to fibrinogen and results in degran­ ulation which ultimately leads to the secretion of IL-13, IL1a, IL-4, TNF, and CCL24 [71]. Soluble fibrinogen interacts with CD11b on neutrophils and activates focal adhesion kinase and ERK1/2 to regulate degranula­ tion, phagocytosis, and apoptosis [72]. TGF-β is a strong inducer of PAI- 1. Downstream molecules of the TGF-β signaling pathway, SMAD 3 and 4 proteins, bind to consensus sites in the TGF beta-inducible elements in the promoter of PAI-1 promoter [73]. Numerous inflammatory mole­ cules induce PAI-1 expression further giving rise to various inflamma­ tory responses such as fibrin deposition, activation of macrophages, ECM deposition, increasing fibronectin and collagen levels during lung fibrosis (Fig. 3). Lipopolysaccharides (LPS) mediated expression of PAI-1 is augmented by TNF-α.Nuclear receptor-77 (Nur77) is a transcriptional factor generated in response to TNF-α. Nur77 induces transcription of PAI-1 by binding to NGFI-B responsive element (NBRE) in the PAI-1 gene in a ligand-independent mechanism [73]. IL-33 participates in the production of profibrotic cytokines and modulates the expression of collagen IV, MMP-9, TIMP-1, tumor necrosis factor receptor (TNFR)- M. Fathimath Muneesa et al.
  • 4. International Immunopharmacology 96 (2021) 107608 4 associated factor 6 (TRAF-6), and NF-κB. It creates an imbalance be­ tween MMP-9 and TIMP-1 and thereby modulating the deposition of MMP [74]. PAI-1 also influences inflammatory responses. It can increase the infiltration of neutrophils, macrophages, and myofibroblasts; thereby contributing to the progression of fibrosis (Fig. 4) [75,76]. In short, fibrinolytic system is capable of regulating inflammatory responses and vice-versa. 5. Inflammatory mediators in accumulation of myofibroblasts Accumulation of fibroblasts is one of the significant observations in PF. The fibroblasts in fibrotic lung express α-smooth muscle actin (α-SMA) and ECM, which leads to damage to the alveolar architecture [77]. They are key sources of collagen and profibrotic cytokines like TGF-β. The role of inflammatory cytokines in the proliferation and accumulation of fibroblasts has been implicated in several studies. Han et al. noted that basic fibroblast growth factor (bFGF) induced activation of ERK5 by enhancing its phosphorylation. ERK5 is an important protein for cell proliferation, differentiation, and tissue repair. bFGF-induces PAI-1 and cell proliferation in lung fibroblasts [78]. TGF-β is the most reported growth factor cytokine which exhibits the property of induc­ tion of differentiation into myofibroblasts. TGF-β is elevated in human subjects with IPF as well as in animal models of PF. It promotes the activation and differentiation of normal lung fibroblasts to fibrotic lung fibroblasts [79,80]. Fibrotic lung fibroblasts express α-SMA and ECM in excess which in turn leads to changes in the alveolar architecture. There is increased expression of CCR7 by fibroblasts, mononuclear, and epithelial cells in IPF lung [81]. The ligand of CCR7, CCL21, en­ hances the proliferation and migration of fibroblasts. Immuno- neutralization of either of the proteins inhibits migration of fibro­ blasts. This indicates that the migration of fibroblasts is dependent on both, the ligand and the receptor [82]. CXCL16/CXCR6 accelerates fibrosis by augmenting proliferation, migration, and collagen synthesis of human pulmonary fibroblasts through the activation of the PI3K/ AKT/FOXO3a signaling pathway [83]. IL-11, a cytokine from the IL-6 Fig 2. Inflammatory cytokines mediated pathways activating alveolar apoptosis and EMT leading to the pathogenesis of pulmonary fibrosis: Pro-Inflammatory cytokines are responsible for activation of alveolar inflammatory pathways. IL-17AR operates IL-17A activating the prime modulator of EMT TGF-β1 activating smad2/3 complex leading to phosphorylation of smad 2/3 in the nucleus. IL-17A also induces EGFR stimulating MAPK/ERK 1/2 elevating the expression of p53 and favors its translocation in the nucleus inducing phosphorylation of p53 causing alveolar injury. IL-6R facilitates the binding of P-JAK stimulating P-STAT1 and P- STAT3 further translocating in the nucleus promoting JAK-STAT pathway activation. The receptor of TNF-α binds to TRAF2 elevating IKK expressions forming the IKK-NFKB complex inducing phosphorylation of NFKB in the nucleus. TRAF6 binds to the receptor of IL-33 activating MAPK and NFKB further expressing NFKB phosphorylation in the nucleus, however, this mechanism is found to be blocked by the anti-inflammatory cytokine IL-37. All the above cytokines are responsible to induce alveolar epithelial injury and activate the biological process such as apoptosis and EMT leading to the pathogenesis of pulmonary fibrosis. On the other hand, BLM injury stimulates NLRP3 inflammasome activating pro-caspase 3 and active caspase 3 promoting alveolar apoptosis causing lung epithelial injury ultimately leading to pulmonary fibrosis. M. Fathimath Muneesa et al.
  • 5. International Immunopharmacology 96 (2021) 107608 5 family, has mitogenic property and inhibits fas-induced apoptosis of lung fibroblasts [84,85]. Oncostatin M, another cytokine of the IL-6 family, also has mitogenic property and induces collagen expression in lung fibroblasts. Oncostatin also inhibits the expression of α-SMA induced by the TGFβ signaling pathway [86,87]. IGF-II induces trans­ differentiation of normal lung fibroblasts into myofibroblasts by acti­ vating the TGFβ receptor [70]. As reported by Tager et al., CXCL10 inhibits the accumulation of fibroblasts and thereby plays a protective role in PF. It indicates that overexpression of CXCL10 could resolve PF [88]. 6. Inflammatory mediators in immune cell infiltration Elevated levels of cytokines that play a role in chemotaxis and acti­ vation of neutrophils, lymphocytes, and monocytes are observed in tis­ sue and fluid from the lungs of IPF patients. IL-8, monocyte chemoattractant protein (MCP-1/CCL2), and macrophage inflammatory protein-1α (MIP-1α/CCL3) are examples of these cytokines. Baran et al., (2007), reported elevated levels of macrophage colony-stimulating factor (M− CSF) in the fluid from the lungs of IPF patients. The study also demonstrated M− CSF mediated recruitment of macrophages in the BLM-PF model [89].Cytokine-like-factor-1(CLF-1) augments the infil­ tration of CD4 + T cells in BLM-induced fibrotic lung [90]. Recent study demonstrated the potentiality of the pro-inflammatory cytokine IL-17A in elevation of AMPKα/COX-2 expressions via enhancing inflammatory molecules like NF-κB-p65, NF-κB-p105, CXCL1 and IL-1β etc [91]. IL-6, an activator of JAK-STAT pathway, promotes migration and infiltration by inducing the expression of MMP1 and MMP-9 through PGE-2 and COX2 signaling [92]. IL-1β upregulates CXCL5 and causes infiltration of neutrophils and macrophages. CXCL5 is, in turn, a substrate for MMPs 2 and 9 which activate CXCL5 by cleaving it [93]. Anti TNF-α adminis­ tration in nitrogen mustard induced pulmonary injury model, reduced the numbers of M1 macrophages Ym1+ M2 macrophages in the lung [94]. In conclusion, inflammatory mediators like CXCL5, IL-1β and TNF- α play potential role in infiltration of immune cells which in turn leads to enhanced inflammation and gradually contributes to fibrosis. 7. Therapeutics targeting inflammation Inflammation can worsen the condition in PF and thereby leading to early mortality. The therapy against ILD includes anti-inflammatory and immunosuppressive agents, with anti-fibrotic agents introduced lately for the treatment of IPF. Treatment with anti-inflammatory drugs even before the process of irreversible parenchymal fibrosis could probably ease IPF. Pirfenidone used widely against IPF is an immunosuppressant with anti-inflammatory as well as anti-fibrotic features [95]. It can also modulate cross-talk between DCs and T-cells to a reduce the production of pro-inflammatory cytokines, decrease Th-cell proliferation and im­ pairs Th2-cell polarization [96–98]. Nintedanib, another widely used drug against IPF, reduces BAL lymphocytes and neutrophils but not macrophages. It also lowers the levels of interleukin-1b, keratinocyte chemoattractant, TIMP-1, and lung collagen [99]. Thalidomide, pos­ sessing immunomodulatory potentials, could improve cough and res­ piratory quality in IPF patients [100]. Anti-inflammatory and anti- fibrotic properties of dabigatranetexilate resolve pulmonary fibrosis in experimental models. It reduces the infiltration of inflammatory cells and pro-fibrotic protein expression [101]. Statin therapy, which is mainly used to manage cardiovascular disorders, reduces inflammation and release of pro-inflammatory cytokines in lungs. It is also potent in inhibiting fibroblast activity and hinders the advancement of the disease to fibrotic stage; thereby reducing mortality. There are also studies showing adverse effect of statins against ILDs [102–106]. A high dose of a non-steroidal anti-inflammatory drug, ibuprofen is reported to slow down the progression of lung disease in cystic fibrosis [107]. Cortico­ steroids when used at the active stage of inflammation could bring about a significant response in delaying the progress of the disease [108]. Fig 3. Inflammatory mediators in the fibrinolytic system leading to pulmonary fibrosis: Inflammatory irritants induce reactive oxygen species (ROS) activating the inflammatory cytokines IL-6, IL-17A, and TNF-α increasing the expression levels of p53 further recruiting the fibrinolytic molecules. ie, decrease expres­ sions of uPA and uPAR and increase levels of PAI-1 promoting fibrin deposition causing fibroblast accumulation eventually causing pulmonary fibrosis. Fig 4. PAI-1 influences various inflammatory responses: Various inflammatory cytokines like IL-6, IL-1β, TGF-β1, IL-17A, and TNF-α elevates expression levels of PAI-1 stimulating several inflammatory responses like fibrin deposition, in­ duction of macrophages and PMN, activation of myofibroblast, ECM deposition, and increase in fibronectin and collagen levels favors in the development of pulmonary fibrosis. M. Fathimath Muneesa et al.
  • 6. International Immunopharmacology 96 (2021) 107608 6 These medications are efficient against inflammation, but have potential side effects and some of their benefits are unproven. Generous resolution of inflammation in lungs is extremely important, as compromised res­ olution could lead to chronic inflammatory stage and ultimately fibrosis. 8. The controversy of the role of inflammation in IPF The role of inflammation in PF has been a topic of discussion for years and is still under controversy. Since there is no effective response to the immunosuppressive medications in PF patients, some experts claim that inflammation doesn’t play a role in the pathogenesis of the disease. But several studies involving human subjects as well as animal models delineate the role of inflammation in the fibrotic process. Some researchers state that inflammation is just a secondary cause for the disease. Ga67 uptake is used as a measure of inflammation at a specific site. Mura et al., observed that although there is a prominent decline in lung function parameters during the progression of PF, there was no significant correlation between Ga67 uptake and the disease progression. They concluded that inflammation is prominent in advanced stages of fibrosis but does not play a pivotal role in the progression of the disease [109]. A similar study conducted by Grijm et al., also indicates that effective reduction in inflammation (as assessed using Ga67 scintig­ raphy) did not improve the clinical outcome of the disease [110]. Pa­ tients suffering from IPF have periods of an acute decline in lung function parameters known as acute exacerbations. Increased levels of neutrophils, pro-inflammatory cytokines, and M2 cytokines (an indica­ tion of macrophage activation) were noted during acute exacerbations in BALF of IPF patients. The study concluded that acute exacerbation is not a subsidiary event but is mediated by M2 macrophage activation [111]. In a very recent study, it was reported that blood inflammation indexes like neutrophil to lymphocyte ratio (NLR), derived neutrophil to lymphocyte ratio (dNLR), monocyte to lymphocyte ratio (MLR), sys­ temic inflammation response index (SIRI), and aggregate index of sys­ temic inflammation (AISI) independently associated with the existence of IPF after adjusting for age, gender, body mass index and smoking status [113]. In a study by Balestro et al., pulmonary fibrosis patients were clas­ sified into slow or rapid progressors of PF based on the FVC fall per year. The morphometric analysis of lung explants of slow progressors with acute exacerbation and rapid progressors with or without acute exac­ erbations in PF showed significant inflammatory cell infiltrates. They also observed that the number of innate immune cells (neutrophils and macrophages) and that of adaptive immune cells (CD4+, CD8+, and B cells) were high in rapid progressors than in slow progressors, pointing at the influencing role of inflammation in the progression of PF [112]. In this review, we advocate that inflammation has a decisive role in IPF. Delineating the mechanisms of the disease by using high throughput technologies, the understanding of the disease pathogenesis will be concrete and wider and also open the way for novel therapeutics and diagnostics. 9. Conclusion Inflammation does play role in the development of the disease. This is demonstrated in different animal models as well as in studies in human IPF patients. As the models used and modes of induction of the disease in these models differ from study to study, there is non- uniformity in the investigation and results obtained. Numerous cyto­ kines are shown to be involved in the disease. But their mechanistic role in the disease is not studied extensively. With the usage of sophisticated technologies in biochemistry and molecular biology, we expect delin­ eation of the mechanisms underlying the disease and thereby, probably, the discovery of novel diagnostic and therapeutic approaches for pa­ tients suffering from IPF. Authors contributions The manuscript has been prepared and reviewed by Ms. Fathimath Muneesa M in correspondence with Dr. Yashodhar Prabhakar Bhandary. Sadiya B Shaikh added some more points and prepared the figures. Jeena TM added some information and reviewed the entire manuscript. Acknowledgments Authors would like to acknowledge Yenepoya Research Centre, Yenepoya University, Department of Biotechnology Grant (BT/ PR25198/MED/30/1896/2017), and Indian Council of Medical Research (No.59/12/2015/Online/BMS/TRM) for supporting our research. References [1] J.K. Burgess, T. Mauad, G. Tjin, J.C. Karlsson, G. Westergren-Thorsson, The extracellular matrix - the under-recognized element in lung disease? J. Pathol. [Internet]. 240 (4) (2016) 397–409. https://www.ncbi.nlm.nih.gov/pubmed /27623753. [2] G. Raghu, H.R. Collard, J.J. Egan, F.J. Martinez, J. Behr, K.K. Brown, et al., An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence- based guidelines for diagnosis and management, Available from, Am. J. Respir. Crit. Care Med. [Internet]. 183 (6) (2011 Mar 15) 788–824, https://www.ncbi. nlm.nih.gov/pubmed/21471066. [3] K.D. Kistler, L. Nalysnyk, P. Rotella, D. Esser, Lung transplantation in idiopathic pulmonary fibrosis: a systematic review of the literature, BMC Pulm. Med. [Internet]. 16 (14) (2014 Aug) 139. https://www.ncbi.nlm.nih.gov/pubmed /25127540. [4] W.R. Coward, G. Saini, G. Jenkins, The pathogenesis of idiopathic pulmonary fibrosis, Ther. Adv. Respir. Dis. 4 (6) (2010 Dec) 367–388. [5] J.S. Zolak, J.A. de Andrade, Idiopathic pulmonary fibrosis, Immunol. Allergy Clin. North Am. 32 (4) (2012 Nov) 473–485. [6] B.C. Willis, J.M. Liebler, K. Luby-Phelps, A.G. Nicholson, E.D. Crandall, R.M. du Bois, et al., Induction of epithelial-mesenchymal transition in alveolar epithelial cells by transforming growth factor-beta1: potential role in idiopathic pulmonary fibrosis, Am. J. Pathol. [Internet]. 166 (5) (2005 May) 1321–1332. https://www. ncbi.nlm.nih.gov/pubmed/15855634. [7] M. Li, B. Liu, Epithelial to mesenchymal transition in the progression of tubulointerstitial fibrosis, Chin Med. J. (Engl). 120 (21) (2007 Nov) 1925–1930. [8] F. Rieder, S.P. Kessler, G.A. West, S. Bhilocha, C. de la Motte, T.M. Sadler, et al., Inflammation-induced endothelial-to-mesenchymal transition: a novel mechanism of intestinal fibrosis, Am. J. Pathol. [Internet]. 179 (5) (2011) 2660–2673, 2011/09/21. [9] Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, et al. Alveolar epithelial cell mesenchymal transition develops <em>in vivo</ em> during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci [Internet]. 2006 Aug 29;103(35):13180 LP – 13185. Available from: http://www.pnas.org/content/103/35/13180.abstract. [10] H. Kasai, J.T. Allen, R.M. Mason, T. Kamimura, Z. Zhang, TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT), Respir. Res. [Internet]. 6 (1) (2005) 56. https://www.ncbi.nlm.nih.gov/pubmed/15946381. [11] Shaikh SB, Prabhu A, Bhandary YP. Curcumin targets p53-fibrinolytic system in TGF-β1 mediated alveolar epithelial mesenchymal transition in alveolar epithelial cells. Endocr Metab Immune Disord Drug Targets. 2020 Sep. [12] C. Vancheri, Idiopathic pulmonary fibrosis: An altered fibroblast proliferation linked to cancer biology, Proc. Am. Thorac. Soc. 9 (3) (2012) 153–157. [13] T. Kinoshita, T. Goto, Molecular Mechanisms of Pulmonary Fibrogenesis and Its Progression to Lung Cancer: A Review, Int. J. Mol. Sci. [Internet]. 20 (6) (2019) 1461. https://www.ncbi.nlm.nih.gov/pubmed/30909462. [14] Wang A, Wang F, Yin Y, Zhang M, Chen P. Dexamethasone reduces serum level of IL-17 in Bleomycin-A5-induced rats model of pulmonary fibrosis. Artif Cells, Nanomedicine, Biotechnol [Internet]. 2018 May 19;46(4):783–7. Available from: https://doi.org/10.1080/21691401.2017.1339051. [15] C. Huang, Y. Li, X. Fan, Y. Ma, M. Zhang, W. Wang, IL-17A promotes pulmonary inflammation in rats with pulmonary fibrosis induced by bleomycin, Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi [Internet]. 30 (4) (2014) 366–370. http://europepmc. org/abstract/MED/24721403. [16] Wang T, Liu Y, Zou J-F, Cheng Z-S. Interleukin-17 induces human alveolar epithelial to mesenchymal cell transition via the TGF-β1 mediated Smad2/3 and ERK1/2 activation. PLoS One [Internet]. 2017 Sep 5;12(9):e0183972–e0183972. Available from: https://www.ncbi.nlm.nih.gov/pubmed/28873461. [17] S.B. Shaikh, S.G. Bhat, Y.P. Bhandary, Curcumin attenuates IL-17A mediated pulmonary SMAD dependent and non-dependent mechanism during acute lung injury in vivo, Mol. Biol. Rep. 47 (7) (2020 Jul) 5643–5649. [18] G. Jiang, C.-T. Liu, W.-D. Zhang, IL-17A and GDF15 are able to induce epithelial- mesenchymal transition of lung epithelial cells in response to cigarette smoke, Exp. Ther. Med. [Internet]. 16 (1) (2018) 12–20. https://www.ncbi.nlm.nih. gov/pubmed/29977354. M. Fathimath Muneesa et al.
  • 7. International Immunopharmacology 96 (2021) 107608 7 [19] S.O. Lee, X. Yang, S. Duan, Y. Tsai, L.R. Strojny, P. Keng, et al., IL-6 promotes growth and epithelial-mesenchymal transition of CD133+ cells of non-small cell lung cancer, Oncotarget 7 (6) (2016 Feb) 6626–6638. [20] Y. Shintani, A. Fujiwara, T. Kimura, T. Kawamura, S. Funaki, M. Minami, et al., IL-6 Secreted from Cancer-Associated Fibroblasts Mediates Chemoresistance in NSCLC by Increasing Epithelial-Mesenchymal Transition Signaling, J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer. 11 (9) (2016 Sep) 1482–1492. [21] L.-M. Zhang, Y. Zhang, C. Fei, J. Zhang, L. Wang, Z.-W. Yi, et al., Neutralization of IL-18 by IL-18 binding protein ameliorates bleomycin-induced pulmonary fibrosis via inhibition of epithelial-mesenchymal transition, Biochem. Biophys. Res. Commun. [Internet]. 508 (2) (2019) 660–666. http://www.sciencedirect.com/sci ence/article/pii/S0006291X18325610. [22] V. Petrilli, C. Dostert, D.A. Muruve, J. Tschopp, The inflammasome: a danger sensing complex triggering innate immunity, Curr. Opin. Immunol. 19 (6) (2007 Dec) 615–622. [23] S. Kesavardhana, T.-D. Kanneganti, Mechanisms governing inflammasome activation, assembly and pyroptosis induction, Int. Immunol. 29 (5) (2017 May) 201–210. [24] J.W. Pinkerton, R.Y. Kim, A.A.B. Robertson, J.A. Hirota, L.G. Wood, D.A. Knight, et al., Inflammasomes in the lung, Mol. Immunol. 86 (2017 Jun) 44–55. [25] Li X, Yan X, Wang Y, Wang J, Zhou F, Wang H, et al. NLRP3 inflammasome inhibition attenuates silica-induced epithelial to mesenchymal transition (EMT) in human bronchial epithelial cells. Exp Cell Res [Internet]. 2018;362(2):489–97. Available from: https://doi.org/10.1016/j.yexcr.2017.12.013. [26] P. Kaler, L. Augenlicht, L. Klampfer, Macrophage-derived IL-1beta stimulates Wnt signaling and growth of colon cancer cells: a crosstalk interrupted by vitamin D3, Oncogene 28 (44) (2009 Nov) 3892–3902. [27] J. Wang, L. Bao, B. Yu, Z. Liu, W. Han, C. Deng, et al., Interleukin-1β Promotes Epithelial-Derived Alveolar Elastogenesis via αvβ6 Integrin-Dependent TGF-β Activation, Cell Physiol. Biochem. [Internet] 36 (6) (2015) 2198–2216. https ://www.karger.com/DOI/10.1159/000430185. [28] Cho SJ, Moon J-S, Nikahira K, Yun HS, Harris R, Hong KS, et al. GLUT1- dependent glycolysis regulates exacerbation of fibrosis via AIM2 inflammasome activation. Thorax [Internet]. 2020 Mar 1;75(3):227 LP – 236. Available from: http://thorax.bmj.com/content/75/3/227.abstract. [29] M. Terlizzi, A. Molino, C. Colarusso, C. Donovan, P. Imitazione, P. Somma, et al., Activation of the absent in melanoma 2 inflammasome in peripheral blood mononuclear cells from Idiopathic pulmonary fibrosis patients leads to the release of pro-fibrotic mediators, Front. Immunol. (2018) 1–12, 9(APR). [30] S.J. Cho, M. Lee, H.W. Stout-Delgado, J.-S. Moon, DROSHA-Dependent miRNA and AIM2 Inflammasome Activation in Idiopathic Pulmonary Fibrosis, Int. J. Mol. Sci. 21 (5) (2020 Feb). [31] O’Beirne SL, Walsh SM, Fabre A, Reviriego C, Worrell JC, Counihan IP, et al. CXCL9 Regulates TGF-β1-Induced Epithelial to Mesenchymal Transition in Human Alveolar Epithelial Cells. J Immunol [Internet]. 2015/08/12. 2015 Sep 15;195(6):2788–96. Available from: https://www.ncbi.nlm.nih.gov/pubmed/ 26268659. [32] M. Liang, J. Wang, H. Chu, X. Zhu, H. He, Q. Liu, et al., Interleukin-22 Inhibits Bleomycin-Induced Pulmonary, Fibrosis (2013). [33] Chen Q, Luo AA, Qiu H, Han B, Ko BH-K, Slutsky AS, et al. Monocyte interaction accelerates HCl-induced lung epithelial remodeling. BMC Pulm Med [Internet]. 2014;14(1):135. Available from: https://doi.org/10.1186/1471-2466-14-135. [34] D.D. Browning, W.C. Diehl, M.H. Hsu, I.U. Schraufstatter, R.D. Ye, Autocrine regulation of interleukin-8 production in human monocytes, Am. J. Physiol. Lung Cell. Mol. Physiol. 279 (6) (2000 Dec) L1129–L1136. [35] E.M. Bruscia, P.-X. Zhang, E. Ferreira, C. Caputo, J.W. Emerson, D. Tuck, et al., Macrophages directly contribute to the exaggerated inflammatory response in cystic fibrosis transmembrane conductance regulator-/- mice, Am. J. Respir. Cell Mol. Biol. 40 (3) (2009 Mar) 295–304. [36] Park sung woo, Kim CR, Lee EH, Kim JH, Park JS, Lee JH, et al. Intranasal administration of IL-37 attenuates bleomycin induced experimental lung injury/ fibrosis. FASEB J [Internet]. 2013 Apr 1;27(1_supplement):1166.12-1166.12. Available from: https://www.fasebj.org/doi/abs/10.1096/fasebj.27.1_ supplement.1166.12. [37] Y. Li, Q. Gao, K. Xu, X. Peng, X. Yuan, W. Jiang, et al., Interleukin-37 Attenuates Bleomycin-Induced Pulmonary Inflammation and Fibrosis in Mice, Inflammation 41 (5) (2018 Oct) 1772–1779. [38] M.S. Kim, A.R. Baek, J.H. Lee, A.S. Jang, D.J. Kim, S.S. Chin, et al., IL-37 Attenuates Lung Fibrosis by Inducing Autophagy and Regulating TGF-beta1 Production in Mice, J. Immunol. 203 (8) (2019 Oct) 2265–2275. [39] Y.-H. Chen, B.-Y. Zhou, X.-J. Wu, J.-F. Xu, J.-A. Zhang, Y.-H. Chen, et al., CCL22 and IL-37 inhibit the proliferation and epithelial-mesenchymal transition process of NSCLC A549 cells, Oncol. Rep. 36 (4) (2016 Oct) 2017–2024. [40] M.F. Nold, C.A. Nold-Petry, J.A. Zepp, B.E. Palmer, P. Bufler, C.A. Dinarello, IL-37 is a fundamental inhibitor of innate immunity, Nat. Immunol. 11 (11) (2010 Nov) 1014–1022. [41] Z. Dong, W. Tai, W. Lei, Y. Wang, Z. Li, T. Zhang, IL-27 inhibits the TGF-β1- induced epithelial-mesenchymal transition in alveolar epithelial cells, BMC Cell Biol. 17 (2016 Mar) 7. [42] Drakopanagiotakis F, Xifteri A, Polychronopoulos V, Bouros D. Apoptosis in lung injury and fibrosis. Eur Respir J [Internet]. 2008 Dec 1;32(6):1631 LP – 1638. Available from: http://erj.ersjournals.com/content/32/6/1631.abstract. [43] M. Kasper, K. Barth, Potential contribution of alveolar epithelial type I cells to pulmonary fibrosis, Biosci. Rep. [Internet]. 37 (6) (2017). BSR20171301. Available from. [44] M. Plataki, A.V. Koutsopoulos, K. Darivianaki, G. Delides, N.M. Siafakas, D. Bouros, Expression of Apoptotic and Antiapoptotic Markers in Epithelial Cells in Idiopathic Pulmonary Fibrosis, Available from, Chest [Internet]. 127 (1) (2005) 266–274, http://www.sciencedirect.com/science/article/pii/S0012369 21532403X. [45] B.D. Uhal, R. Wang, J. Laukka, J. Zhuang, V. Soledad-Conrad, G. Filippatos, Inhibition of amiodarone-induced lung fibrosis but not alveolitis by angiotensin system antagonists, Pharmacol. Toxicol. 92 (2) (2003 Feb) 81–87. [46] J.V. Barbas-Filho, M.A. Ferreira, A. Sesso, R.A. Kairalla, C.R. Carvalho, V. L. Capelozzi, Evidence of type II pneumocyte apoptosis in the pathogenesis of idiopathic pulmonary fibrosis (IFP)/usual interstitial pneumonia (UIP), J. Clin. Pathol. 54 (2) (2001 Feb) 132–138. [47] P.D. Bozyk, B.B. Moore, Prostaglandin E2 and the pathogenesis of pulmonary fibrosis, Am. J. Respir. Cell Mol. Biol. 45 (3) (2011 Sep) 445–452. [48] B.D. Uhal, The role of apoptosis in pulmonary fibrosis, Eur. Respir. Rev. [Internet]. 17 (109) (2008), 138 LP – 144. Available from:. [49] X. Li, H. Zhang, V. Soledad-Conrad, J. Zhuang, B.D. Uhal, Bleomycin-induced apoptosis of alveolar epithelial cells requires angiotensin synthesis de novo, Am. J. Physiol. Lung Cell. Mol. Physiol. 284 (3) (2003 Mar) L501–L507. [50] R. Wang, G. Alam, A. Zagariya, C. Gidea, H. Pinillos, O. Lalude, et al., Apoptosis of lung epithelial cells in response to TNF-alpha requires angiotensin II generation de novo, J. Cell. Physiol. 185 (2) (2000 Nov) 253–259. [51] Murray ME, Gavile CM, Nair JR, Koorella C, Carlson LM, Buac D, et al. CD28- mediated pro-survival signaling induces chemotherapeutic resistance in multiple myeloma. Blood [Internet]. 2014/04/29. 2014 Jun 12;123(24):3770–9. Available from: https://www.ncbi.nlm.nih.gov/pubmed/24782505. [52] C. Camperio, M. Muscolini, E. Volpe, D. Di Mitri, R. Mechelli, M.C. Buscarinu, et al., CD28 ligation in the absence of TCR stimulation up-regulates IL-17A and pro- inflammatory cytokines in relapsing-remitting multiple sclerosis T lymphocytes, Immunol. Lett. 158 (1–2) (2014) 134–142. [53] Kunkl M, Mastrogiovanni M, Porciello N, Caristi S, Monteleone E, Arcieri S, et al. CD28 Individual Signaling Up-regulates Human IL-17A Expression by Promoting the Recruitment of RelA/NF-κB and STAT3 Transcription Factors on the Proximal Promoter. Front Immunol [Internet]. 2019 Apr 24;10:864. Available from: https://www.ncbi.nlm.nih.gov/pubmed/31068940. [54] M.M. Gouda, Y.P. Bhandary, Curcumin down-regulates IL-17A mediated p53- fibrinolytic system in bleomycin induced acute lung injury in vivo, J. Cell. Biochem. 119 (9) (2018 Sep) 7285–7299. [55] M.M. Gouda, A. Prabhu, Y.P. Bhandary, Curcumin alleviates IL-17A-mediated p53-PAI-1 expression in bleomycin-induced alveolar basal epithelial cells, J. Cell. Biochem. 119 (2) (2018 Feb) 2222–2230. [56] C. Chen, L. Shi, Y. Li, X. Wang, S. Yang, Disease-specific dynamic biomarkers selected by integrating inflammatory mediators with clinical informatics in ARDS patients with severe pneumonia, Cell Biol. Toxicol. 32 (3) (2016 Jun) 169–184. [57] H. Zhu, H. Huang, D. Wu, N. Dong, L. Dong, C. Chen, et al., Regulatory mechanism of NOV/CCN3 in the inflammation and apoptosis of lung epithelial alveolar cells upon lipopolysaccharide stimulation, Mol. Med. Rep. (2019) 1–9. [58] Schuliga M. The Inflammatory Actions of Coagulant and Fibrinolytic Proteases in Disease. 2015;2015. [59] McKeown S, Richter AG, O'Kane C, McAuley DF, Thickett DR. MMP expression and abnormal lung permeability are important determinants of outcome in IPF. Eur Respir J [Internet]. 2009 Jan 1;33(1):77 LP – 84. Available from: http://erj.ersjournals.com/content/33/1/77.abstract. [60] V. Navaratnam, A.W. Fogarty, T. McKeever, N. Thompson, G. Jenkins, S. R. Johnson, et al., Presence of a prothrombotic state in people with idiopathic pulmonary fibrosis: a population-based case-control study, Thorax 69 (3) (2014 Mar) 207–215. [61] D. Zentina, I. Stukena, A. Krams, A. Lejnieks, PAI-1 Level Differences in Malignant Plural Effusion, Parapneumonic Pleuritis, and Cardiac Hydrothorax, Medicina (Kaunas) [Internet]. 55 (9) (2019) 567. Available from:. [62] Y.-J. Park, G. Liu, E.F. Lorne, X. Zhao, J. Wang, Y. Tsuruta, et al., PAI-1 inhibits neutrophil efferocytosis, Proc. Natl. Acad. Sci. U S A. 105 (33) (2008 Aug) 11784–11789. [63] M. Birgel, H. Gottschling-Zeller, K. Röhrig, H. Hauner, Role of cytokines in the regulation of plasminogen activator inhibitor-1 expression and secretion in newly differentiated subcutaneous human adipocytes, Arterioscler. Thromb. Vasc. Biol. 20 (6) (2000 Jun) 1682–1687. [64] C.-Y. Chiu, K.-S. Wong, J.-L. Huang, M.-H. Tasi, T.-Y. Lin, S.-Y. Hsieh, Proinflammatory cytokines, fibrinolytic system enzymes, and biochemical indices in children with infectious para-pneumonic effusions, Pediatr. Infect. Dis. J. 27 (8) (2008 Aug) 699–703. [65] P.A. Akpan, J.O. Akpotuzor, E.E. Osim, The role of cytokines in fibrinolysis: A case study of active tuberculosis, J. Infect. Dis. Med. Microbiol. 1 (1) (2017) 1–5. [66] Shetty P, Velusamy T, Bhandary YP, Shetty RS, Liu M-C, Shetty S. Urokinase expression by tumor suppressor protein p53: a novel role in mRNA turnover. Am J Respir Cell Mol Biol [Internet]. 2008/04/03. 2008 Sep;39(3):364–72. Available from: https://pubmed.ncbi.nlm.nih.gov/18390474. [67] Shetty S, Velusamy T, Idell S, Shetty P, Mazar AP, Bhandary YP, et al. Regulation of urokinase receptor expression by p53: novel role in stabilization of uPAR mRNA. Mol Cell Biol [Internet]. 2007/06/04. 2007 Aug;27(16):5607–18. Available from: https://pubmed.ncbi.nlm.nih.gov/17548471. [68] N. Tiwari, A.S. Marudamuthu, Y. Tsukasaki, M. Ikebe, J. Fu, S. Shetty, p53- and PAI-1-mediated induction of C-X-C chemokines and CXCR2: importance in pulmonary inflammation due to cigarette smoke exposure, Am. J. Physiol. Lung Cell. Mol. Physiol. 310 (6) (2016 Mar) L496–L506. M. Fathimath Muneesa et al.
  • 8. International Immunopharmacology 96 (2021) 107608 8 [69] M. Birgel, H. Gottschling-Zeller, K. Röhrig, H. Hauner, Role of cytokines in the regulation of plasminogen activator inhibitor-1 expression and secretion in newly differentiated subcutaneous human adipocytes, Arterioscler. Thromb. Vasc. Biol. 20 (6) (2000) 1682–1687. [70] Garrett SM, Hsu E, Thomas JM, Pilewski JM, Feghali-Bostwick C. Insulin-like growth factor (IGF)-II- mediated fibrosis in pathogenic lung conditions. PLoS One [Internet]. 2019;14(11):1–21. Available from: https://doi.org/10.1371/journal. pone.0225422. [71] M.E. Coden, L.F. Loffredo, T. Matthew, B.M. Jeong, K. Nam, B.S. Bochner, et al., Fibrinogen Is a Specific Trigger for, Cytolytic. Eosinophil. Degranulat. (2019). [72] Rubel C, Fernández GC, Rosa FA, Gómez S, Bompadre MB, Coso OA, et al. Soluble Fibrinogen Modulates Neutrophil Functionality Through the Activation of an Extracellular Signal-Regulated Kinase-Dependent Pathway. J Immunol [Internet]. 2002 Apr 1;168(7):3527 LP – 3535. Available from: http://www.jimmunol.org/ content/168/7/3527.abstract. [73] S. Dennler, S. Itoh, D. Vivien, P. ten Dijke, S. Huet, J.M. Gauthier, Direct binding of Smad3 and Smad4 to critical TGF beta-inducible elements in the promoter of human plasminogen activator inhibitor-type 1 gene, EMBO J. 17 (11) (1998 Jun) 3091–3100. [74] L. Wu, Z. Luo, J. Zheng, P. Yao, Z. Yuan, X. Lv, et al., IL-33 Can Promote the Process of Pulmonary Fibrosis by Inducing the Imbalance Between MMP-9 and TIMP-1, Inflammation 41 (3) (2018 Jun) 878–885. [75] B. Thapa, Y.H. Kim, H.-J. Kwon, D.-S. Kim, The LRP1-independent mechanism of PAI-1-induced migration in CpG-ODN activated macrophages, Int. J. Biochem. Cell Biol. 49 (2014 Apr) 17–25. [76] Y. Tashiro, C. Nishida, K. Sato-Kusubata, M. Ohki-Koizumi, M. Ishihara, A. Sato, et al., Inhibition of PAI-1 induces neutrophil-driven neoangiogenesis and promotes tissue regeneration via production of angiocrine factors in mice, Blood 119 (26) (2012 Jun) 6382–6393. [77] R.T. Kendall, C.A. Feghali-Bostwick, Fibroblasts in fibrosis: novel roles and mediators, Front. Pharmacol. 5 (2014) 123. [78] J.-H. Han, A.-R. Hwang, D.-H. Nam, S. Kim, H.C. Choi, J.H. Lim, et al., ERK5 regulates basic fibroblast growth factor-induced type 1 plasminogen activator inhibitor expression and cell proliferation in lung fibroblasts, Available from, Life Sci. [Internet] 135 (2015) 1–8, http://europepmc.org/abstract/MED/26032256. [79] N.W. Todd, I.G. Luzina, S.P. Atamas, Molecular and cellular mechanisms of pulmonary fibrosis, Fibrogenesis Tissue Repair. 5 (1) (2012 Jul) 11. [80] I.E. Fernandez, O. Eickelberg, The impact of TGF-beta on lung fibrosis: from targeting to biomarkers, Proc. Am. Thorac. Soc. 9 (3) (2012 Jul) 111–116. [81] E.S. Choi, E.M. Pierce, C. Jakubzick, K.J. Carpenter, S.L. Kunkel, H. Evanoff, et al., Focal interstitial CC chemokine receptor 7 (CCR7) expression in idiopathic interstitial pneumonia, Available from, J. Clin. Pathol. [Internet]. 59 (1) (2006 Jan) 28–39, https://www.ncbi.nlm.nih.gov/pubmed/16394278. [82] Pierce EM, Carpenter K, Jakubzick C, Kunkel SL, Evanoff H, Flaherty KR, et al. Idiopathic pulmonary fibrosis fibroblasts migrate and proliferate to CC chemokine ligand 21. Eur Respir J [Internet]. 2007 Jun 1;29(6):1082 LP – 1093. Available from: http://erj.ersjournals.com/content/29/6/1082.abstract. [83] Ma Z, Yu R, Zhu Q, Sun L, Jian L, Wang X, et al. CXCL16/CXCR6 axis promotes bleomycin-induced fibrotic process in MRC-5 cells via the PI3K/AKT/FOXO3a pathway. Int Immunopharmacol [Internet]. 2020;81:106035. Available from: http://europepmc.org/abstract/MED/31753588. [84] Y.P. Moodley, N.L.A. Misso, A.K. Scaffidi, M. Fogel-Petrovic, R.J. McAnulty, G. J. Laurent, et al., Inverse effects of interleukin-6 on apoptosis of fibroblasts from pulmonary fibrosis and normal lungs, Am. J. Respir. Cell Mol. Biol. 29 (4) (2003 Oct) 490–498. [85] Y.P. Moodley, A.K. Scaffidi, N.L. Misso, C. Keerthisingam, R.J. McAnulty, G. J. Laurent, et al., Fibroblasts isolated from normal lungs and those with idiopathic pulmonary fibrosis differ in interleukin-6/gp130-mediated cell signaling and proliferation, Available from, Am. J. Pathol. [Internet] 163 (1) (2003 Jul) 345–354, https://www.ncbi.nlm.nih.gov/pubmed/12819039. [86] A.K. Scaffidi, S.E. Mutsaers, Y.P. Moodley, R.J. McAnulty, G.J. Laurent, P. J. Thompson, et al., Oncostatin M stimulates proliferation, induces collagen production and inhibits apoptosis of human lung fibroblasts, Br. J. Pharmacol. 136 (5) (2002 Jul) 793–801. [87] D.V. Pechkovsky, C.M. Prele, J. Wong, C.M. Hogaboam, R.J. McAnulty, G. J. Laurent, et al., STAT3-mediated signaling dysregulates lung fibroblast- myofibroblast activation and differentiation in UIP/IPF, Am. J. Pathol. 180 (4) (2012 Apr) 1398–1412. [88] A.M. Tager, R.L. Kradin, P. LaCamera, S.D. Bercury, G.S.V. Campanella, C. P. Leary, et al., Inhibition of pulmonary fibrosis by the chemokine IP-10/CXCL10, Am. J. Respir. Cell Mol. Biol. 31 (4) (2004 Oct) 395–404. [89] C.P. Baran, J.M. Opalek, S. McMaken, C.A. Newland, J.M.J. O’Brien, M. G. Hunter, et al., Important roles for macrophage colony-stimulating factor, CC chemokine ligand 2, and mononuclear phagocytes in the pathogenesis of pulmonary fibrosis, Am. J. Respir. Crit. Care Med. 176 (1) (2007 Jul) 78–89. [90] Kass DJ, Yu G, Loh KS, Savir A, Borczuk A, Kahloon R, et al. Cytokine-like factor 1 gene expression is enriched in idiopathic pulmonary fibrosis and drives the accumulation of CD4+ T cells in murine lungs: evidence for an antifibrotic role in bleomycin injury. Am J Pathol [Internet]. 2012/03/16. 2012 May;180(5): 1963–78. Available from: https://www.ncbi.nlm.nih.gov/pubmed/22429962. [91] S.B. Shaikh, B.Y. Prabhakar, Effect of curcumin on IL-17A mediated pulmonary AMPK kinase/cyclooxygenase-2 expressions via activation of NFκB in bleomycin- induced acute lung injury in vivo, Available from, Int. Immunopharmacol. [Internet]. 85 (2020), 106676, http://europepmc.org/abstract/MED/32535538. [92] Moradi S, Ziaei R, Foshati S, Mohammadi H, Nachvak SM, Rouhani MH. Jo ur na l P re ro of. Complement Ther Med [Internet]. 2019;102211. Available from: https://doi.org/10.1016/j.ctim.2019.102211. [93] Song J, Wu C, Zhang X, Sorokin LM. In Vivo Processing of CXCL5 (LIX) by Matrix Metalloproteinase (MMP)-2 and MMP-9 Promotes Early Neutrophil Recruitment in IL-1β–Induced Peritonitis. J Immunol [Internet]. 2013 Jan 1;190(1):401 LP – 410. Available from: http://www.jimmunol.org/content/190/1/401.abstract. [94] R. Malaviya, V.R. Sunil, A. Venosa, V.L. Verissimo, J.A. Cervelli, K.N. Vayas, et al., Attenuation of Nitrogen Mustard-Induced Pulmonary Injury and Fibrosis by Anti-Tumor Necrosis Factor-α Antibody, Toxicol. Sci. [Internet]. 148 (1) (2015) 71–88. Available from: https://pubmed.ncbi.nlm.nih.gov/26243812. [95] H. Oku, T. Shimizu, T. Kawabata, M. Nagira, I. Hikita, A. Ueyama, et al., Antifibrotic action of pirfenidone and prednisolone: Different effects on pulmonary cytokines and growth factors in bleomycin-induced murine pulmonary fibrosis, Eur. J. Pharmacol. 590 (1–3) (2008) 400–408. [96] J. Navarro-Partida, A.B. Martinez-Rizo, J. Gonzalez-Cuevas, G. Arrevillaga-Boni, V. Ortiz-Navarrete, J. Armendariz-Borunda, Pirfenidone restricts Th2 differentiation in vitro and limits Th2 response in experimental liver fibrosis, Available from, Eur. J. Pharmacol. [Internet]. 678 (1) (2012) 71–77, https:// www.sciencedirect.com/science/article/pii/S0014299911015597. [97] P. Bizargity, K. Liu, L. Wang, W.W. Hancock, G.A. Visner, Inhibitory effects of pirfenidone on dendritic cells and lung allograft rejection, Available from, Transplantation [Internet] 94 (2) (2012 Jul 27) 114–122, https://pubmed.ncbi. nlm.nih.gov/22743547. [98] G.A. Visner, F. Liu, P. Bizargity, H. Liu, K. Liu, J. Yang, et al., Pirfenidone inhibits T-cell activation, proliferation, cytokine and chemokine production, and host alloresponses, Available from, Transplantation [Internet]. 88 (3) (2009 Aug 15) 330–338, https://pubmed.ncbi.nlm.nih.gov/19667934. [99] Wollin L, Maillet I, Quesniaux V, Holweg A, Ryffel B. Antifibrotic and Anti- inflammatory Activity of the Tyrosine Kinase Inhibitor Nintedanib in Experimental Models of Lung Fibrosis. J Pharmacol Exp Ther [Internet]. 2014 May 1;349(2):209 LP – 220. Available from: http://jpet.aspetjournals.org/ content/349/2/209.abstract. [100] M.R. Horton, V. Santopietro, L. Mathew, K.M. Horton, A.J. Polito, M.C. Liu, et al., Thalidomide for the treatment of cough in idiopathic pulmonary fibrosis: a randomized trial, Ann. Intern. Med. 157 (6) (2012 Sep) 398–406. [101] G.S. Bogatkevich, A. Ludwicka-Bradley, P.J. Nietert, T. Akter, J. van Ryn, R. M. Silver, Antiinflammatory and antifibrotic effects of the oral direct thrombin inhibitor dabigatran etexilate in a murine model of interstitial lung disease, Arthritis Rheum. 63 (5) (2011 May) 1416–1425. [102] A.B. Fernández, R.H. Karas, A.A. Alsheikh-Ali, P.D. Thompson, Statins and interstitial lung disease: a systematic review of the literature and of food and drug administration adverse event reports, Chest 134 (4) (2008 Oct) 824–830. [103] J.-F. Xu, G.R. Washko, K. Nakahira, H. Hatabu, A.S. Patel, I.E. Fernandez, et al., Statins and pulmonary fibrosis: the potential role of NLRP3 inflammasome activation, Am. J. Respir. Crit. Care Med. 185 (5) (2012 Mar) 547–556. [104] N. Saad, P. Camus, S. Suissa, P. Ernst, Statins and the risk of interstitial lung disease: a cohort study, Thorax 68 (4) (2013 Apr) 361–364. [105] S. Vedel-Krogh, S.F. Nielsen, B.G. Nordestgaard, Statin Use Is Associated with Reduced Mortality in Patients with Interstitial Lung Disease, PLoS ONE 10 (10) (2015) e0140571. [106] M. Kreuter, F. Bonella, T.M. Maher, U. Costabel, P. Spagnolo, D. Weycker, et al., Effect of statins on disease-related outcomes in patients with idiopathic pulmonary fibrosis, Thorax 72 (2) (2017) 148–153. [107] Lands LC, Stanojevic S. Oral non-steroidal anti-inflammatory drug therapy for lung disease in cystic fibrosis. Cochrane database Syst Rev. 2016;4(4):CD001505. [108] K.R. Flaherty, G.B. Toews, J.P. Lynch 3rd, E.A. Kazerooni, B.H. Gross, R. L. Strawderman, et al., Steroids in idiopathic pulmonary fibrosis: a prospective assessment of adverse reactions, response to therapy, and survival, Am. J. Med. 110 (4) (2001 Mar) 278–282. [109] M. Mura, G. Belmonte, S. Fanti, P. Contini, A.M.G. Pacilli, L. Fasano, et al., Inflammatory activity is still present in the advanced stages of idiopathic pulmonary fibrosis, Respirology 10 (5) (2005 Nov) 609–614. [110] K. Grijm, H.J. Verberne, F.H. Krouwels, F.R. Weller, H.M. Jansen, P. Bresser, Semiquantitative 67Ga scintigraphy as an indicator of response to and prognosis after corticosteroid treatment in idiopathic interstitial pneumonia, J. Nucl. Med. 46 (9) (2005 Sep) 1421–1426. [111] J.C. Schupp, H. Binder, B. Jager, G. Cillis, G. Zissel, J. Muller-Quernheim, et al., Macrophage activation in acute exacerbation of idiopathic pulmonary fibrosis, PLoS ONE 10 (1) (2015) e0116775. [112] Balestro E, Calabrese F, Turato G, Lunardi F, Bazzan E, Marulli G, et al. Immune Inflammation and Disease Progression in Idiopathic Pulmonary Fibrosis. PLoS One [Internet]. 2016 May 9;11(5):e0154516. Available from: https://doi.org/ 10.1371/journal.pone.0154516. [113] A. Zinellu, P. Paliogiannis, E. Sotgiu, S. Mellino, A.A. Mangoni, E. Zinellu, et al., Blood Cell Count Derived Inflammation Indexes in Patients with Idiopathic Pulmonary Fibrosis, Lung 198 (5) (2020) 821–827, https://doi.org/10.1007/ s00408-020-00386-7. M. Fathimath Muneesa et al.