ArticlePDF AvailableLiterature Review

Long non-coding RNA (lncRNA): A potential therapeutic target in acute lung injury

Authors:

Abstract

Acute lung injury (ALI) and its severe form acute respiratory distress syndrome (ARDS) are the major cause of ICU death worldwide. ALI/ARDS is characterized by severe hypoxemia and inflammation that leads to poor lung compliance. Despite many advances in understanding and management, ALI/ARDS is still causing significant morbidity and mortality. Long non-coding RNA (lncRNA) is a fast-growing topic in lung inflammation and injury. lncRNA is a class of non-coding RNA having a length of more than 200 nucleotides. It has been a center of research for understanding the pathophysiology of various diseases in the past few years. Multiple studies have shown that lncRNAs are abundant in acute lung injury/injuries in mouse models and cell lines. By targeting these long non-coding RNAs, many investigators have demonstrated the alleviation of ALI in various mouse models. Therefore, lncRNAs show great promise as a therapeutic target in ALI. This review provides the current state of knowledge about the relationship between lncRNAs in various biological processes in acute lung injury and its use as a potential therapeutic target.
REVIEW ARTICLE
Long non-coding RNA (lncRNA): A potential
therapeutic target in acute lung injury
Almaz Zaki
b,1
, M. Shadab Ali
a,1
, Vijay Hadda
a,
*,
Syed Mansoor Ali
c,
**, Anita Chopra
d
, Tasneem Fatma
b
a
Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences,
New Delhi, 110029, India
b
Department of Biosciences, Jamia Millia Islamia University, New Delhi, 110025, India
c
Department of Biotechnology, Jamia Millia Islamia University, New Delhi, 110025, India
d
Lab Oncology, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, 110029, India
Received 18 March 2021; received in revised form 15 July 2021; accepted 17 July 2021
KEYWORDS
Acute lung injury;
Apoptosis;
Inflammation;
Long non-coding RNA
(lncRNA);
Macrophage
polarization
Abstract Acute Lung Injury (ALI) and its severe form Acute Respiratory Distress Syndrome
(ARDS) are the major cause of ICU death worldwide. ALI/ARDS is characterized by severe hyp-
oxemia and inflammation that leads to poor lung compliance. Despite many advances in under-
standing and management, ALI/ARDS is still causing significant morbidity and mortality. Long
non-coding RNA (lncRNA) is a fast-growing topic in lung inflammation and injury. lncRNA is a
class of non-coding RNA having a length of more than 200 nucleotides. It has been a center
of research for understanding the pathophysiology of various diseases in the past few years.
Multiple studies have shown that lncRNAs are abundant in acute lung injury/injuries in mouse
models and cell lines. By targeting these long non-coding RNAs, many investigators have
demonstrated the alleviation of ALI in various mouse models. Therefore, lncRNAs show great
promise as a therapeutic target in ALI. This review provides the current state of knowledge
about the relationship between lncRNAs in various biological processes in acute lung injury
and its use as a potential therapeutic target.
Copyright ª2021, Chongqing Medical University. Production and hosting by Elsevier B.V. This is
an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/
by-nc-nd/4.0/).
* Corresponding author. Room no. 8, Porta Cabin, New Private Ward, Department of Pulmonary, Critical Care and Sleep Medicine, AIIMS,
Ansari Nagar, New Delhi, 110029, India.
** Corresponding author. Room no. 509, Department of Biotechnology, Ramanujan Block, Jamia Millia Islamia University, Jamia Nagar, New
Delhi, 110025, India.
E-mail addresses: almazzaki1122@gmail.com (A. Zaki), ms1993ali@gmail.com (M.S. Ali), vijayhadda@yahoo.com (V. Hadda), smansoor@
jmi.ac.in (S.M. Ali), chopraanita2005@gmail.com (A. Chopra), tfatima@jmi.ac.in (T. Fatma).
Peer review under responsibility of Chongqing Medical University.
1
AZ and MSA have contributed equally and are joint first authors.
+MODEL
Please cite this article as: A. Zaki, M.S. Ali, V. Hadda et al., Long non-coding RNA (lncRNA): A potential therapeutic target in acute lung
injury, Genes &Diseases, https://doi.org/10.1016/j.gendis.2021.07.004
https://doi.org/10.1016/j.gendis.2021.07.004
2352-3042/Copyright ª2021, Chongqing Medical University. Production and hosting by Elsevier B.V. This is an open access article under the
CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Available online at www.sciencedirect.com
ScienceDirect
journal homepage: http://ees.elsevier.com/gendis/default.asp
Genes &Diseases xxx (xxxx) xxx
Introduction
Acute lung injury (ALI) is a syndrome that comprises severe
hypoxemic respiratory failure along with bilateral lung in-
filtrates and is also linked with pulmonary as well as non-
pulmonary risk factors.
1
It is characterized by severe
inflammation that causes serious damage to the alveoli and
results in poor lung compliance.
2
ALI is associated with
significant morbidity and mortality, and its severe form,
i.e., acute respiratory distress syndrome (ARDS), has even
higher mortality rates. Globally, it affects around 3 million
people annually and is responsible for 10 percent of ICU
admissions.
3
Despite recent advancements in understand-
ing and management of the disease, patients suffering from
ALI/ARDS end up with a poor prognosis. Therefore, a new
approach for better understanding and effective treatment
strategies is urgently needed to mitigate the disease and
overcome the economic burden it causes.
Long non-coding RNA (lncRNA) is a fast-growing topic in
lung inflammation and injury.
4
In human, less than 3% of
genome codes for proteins, while the rest of the genomic
part is consists of either introns or intergenic DNA.
5
Among
all the transcribed parts of the genome, most are tran-
scribed into non-coding RNA (ncRNA)
6
and are likely to be
non-functional.
7
However, a certain part of it plays a vital
role in gene regulation. ncRNA have been further divided
into multiple categories such as miRNA, snoRNA, piRNA, and
lncRNA based on their size/nucleotide length.
8
Long non-
coding RNA or lncRNA is the “non-coding RNA that ex-
ceeds 200 nucleotides in length”
9
and exhibits diverse roles
and functions in many important biological processes.
Earlier, lncRNAs were considered as a by-product of the
transcription process. However, with more research/study
focusing on the lncRNA, it became evident that these RNA
molecules play a crucial role in regulating many
physiological processes, including immunity,
10
inflamma-
tion, proliferation, cell differentiation, and cell sur-
vival.
11,12
In recent years, advances in next-generation
sequencing (NGS) technologies have prompted an eruption
of newly discovered lncRNAs, particularly in humans.
13
Long non-coding RNA regulates gene expression at
transcriptional, post-transcriptional, epigenetic, and chro-
matin levels and activates or constrains the expression of
target genes by directly binding to them or by recruiting
transcription factors (Fig. 1 showing lncRNA activity in
cellular environment).
14
Multiple studies suggest that dys-
regulation of lncRNA is associated with many human dis-
eases such as focal ischemia, cancer, neurodegenerative
diseases, and respiratory diseases.
15e18
Among respiratory
diseases, lncRNAs have been implicated in usual interstitial
pneumonia, chronic obstructive pulmonary diseases, lung
cancer, pulmonary arterial hypertension, and ALI.
19
The
scope of the present review will be limited to acute lung
injury/acute respiratory distress syndrome.
lncRNA in acute lung injury
lncRNA has provided new insights on the pathogenesis of
ALI. It will aid the investigators in developing a novel
therapeutic target for the treatment of acute lung injury.
So far, many studies have discovered the role of various
lncRNAs as a potential therapeutic target in acute lung
injury, such as MALAT1, NEAT1, TUG1, THRILL, etc. (see
Table 1). Some of the important lncRNAs are discussed
below.
MALAT1
MALAT1 (Metastasis Associated Lung Adenocarcinoma
Transcript 1) is 6.5 kb, abundant, evolutionary conserved,
and stable lncRNA that was found overexpressed in solid
Figure 1 General description of lncRNA activity in the regulation of cellular activities. (A) Expression of lncRNA in response to an
external stimulus. (B) It regulates the promoter and enhancer region, (C) lncRNA providing a platform for transcriptional activity.
(D) It also controls the chromatin remodeling; (E) alternate splicing via spliceosome, (F) translation and protein modification, and
(G) microRNA regulation in many cellular activities by lncRNA.
A. Zaki, M.S. Ali, V. Hadda et al.
+MODEL
2
Table 1 lncRNAs involved in ALI/ARDS.
lncRNAs Model and/or cells Expression Function Molecular targets References
TUG1 LPS-induced ALI Murine,
PMVECs
Decreased Alleviates inflammation and
apoptosis
miR-34 b-5p/GAB1
41
MALAT1 LPS-induced ALI in
HPMEC, Rat ALI/ARDS
model, sepsis patients
Increased Decreased apoptosis and
inflammation
miR-181a-5p,
ICAM-1, miR-425
42e44
CASC2 A549 cell line Decreased Increased viability, decreased
apoptosis, and inflammation
miR-27b/TAB2 axis
38
NEAT1 LPS-induced ALI mice
models and A549 cell line
Increased Enhanced cell viability and
reduced lactate dehydrogenase
release, apoptosis, and
caspase-3/9 activity.
Decreased inflammation
HMGB1/RAGE
signaling, miR-
944/TRIM37
32,36
THRIL Septic-induced ALI in
C57BL/6 mice model and
MPVECs.
Increased Decreases the inflammation
and apoptosis
miR-424/ROCK2
25
SNHG5 LPS-induced mouse
model, A549 cell line
Decreased It inhibits inflammation and
promotes the cell viability
miR-205/COMMD1
45
PRNCR1 LPS-treated PVEC and
mouse model
Increased Attenuates expressions of
TLR4, NF-kB, and inflammatory
cytokines and increases cell
viability
miR-330-5p/TLR4
axis
46
Hsp4 LPS-induced MLE-12 cells Decreased Reduced apoptosis mTOR signaling,
miR-466 m-3p/
DNAjb6
47
lncRNA-5657 CLP-induced ALI mouse
model, patients with
sepsis-induced lung
injury, NR8383 cell line
Increased Alleviated inflammatory
response
spns2
48
CLMAT3 Monocytes isolated from
blood samples of healthy
controls and ALI
patients, U937
macrophage cell line,
C57BL/6 mice Model
Increased Alleviates pro-inflammatory
cytokines
CtBP2-p300-NF-kB
complex
49
MEG3 LPS-induced ALI mouse
alveolar and macrophage
NR8383 cells
Increased Improves inflammatory
response
microRNA-7b (miR-
7b)/NLR pyrin
domain containing
3 (NLRP3)
50
CASC9 LPS-induced HSAECs as
well sepsis-induced lung
injury mouse model
Decreased Increased the viability of
HSAECs
miR-195-5p/PDK4
axis
51
XIST Lung transplant patients,
PCI-induced rat model
Increased Alleviation of inflammation and
apoptosis, cessation of NET
formation
miR-21/IL-12 A
52
GAS5 LPS-induced murine
alveolar epithelial cell
line MLE-12
Decreased Decreased cell inflammatory
responses and apoptosis
miR-429/DUSP1
axis
53
SNGH14 LPS-induced ALI mouse
model
Increased Reduces the levels of pro-
inflammatory cytokines IL-18,
IL-1b, TNF-a, and IL-6 and
inhibits MH-S cell viability.
miR-34c-3p/WISP1
54
H19 LPS-induced ARDS in rats
and MH-S cells
Increased Pulmonary injury,
inflammation, and fibrosis
miR-423-5p/
FOXA1
55
Mirt2 LPS induced primary
cultured peritoneal
macrophages,
RAW264.7 cells
Increased Regulate macrophage
polarization and inflammation
TRAF6, NF-kB,
stat6 and MAPK
56
Genes &Diseases xxx (xxxx) xxx
+MODEL
3
tumors and associated with proliferation, progression,
metastasis, migration, invasion, survival, and acts as a
prognostic marker of cancers.
20
Investigation in LPS
treated macrophages showed upregulation of MALAT1,
while it was downregulated in IL4 treated cells and showed
contrasting expression in differentially treated macro-
phages. MALAT1 knockdown ameliorated LPS-induced M1
macrophage activation involved in lung injury and inflam-
mation, while IL-4 induced M2 macrophages activation was
enhanced in MALAT1 knockdown cells known for inflam-
mation resolution.
21,22
THRIL
TNF and HNRNPL related immunoregulatory long non-
coding RNA or THRIL have been previously reported as a
critical regulator in inflammatory diseases. The expression
level of THRIL increases in coronary heart disease patients
with systemic inflammation.
23
In a recent study, over-
expression of THRIL has been observed in sepsis patients.
More strikingly, it positively correlates with the increased
risk of ARDS, disease severity, inflammation, and high
mortality.
24
Chen et al reported overexpression of THRIL in
the tissues of sepsis-induced ALI mice, which was further
validated in the sepsis cell model. They also reported
attenuation in lung injury and increased survival when
THRIL was downregulated. It indicates that THRIL prompts
acute lung injury in sepsis.
25
THRIL binds to the promoter region of the TNF-aforming
RNA-protein complex with hnRPL, inducing the expression
of TNF-a, thus imposes an inflammatory response.
26
Chen
et al
25
reported a dramatic decrease in the protein level of
TNF-a, IL-1b, IL-6, and the number of macrophages and
neutrophils counts by silencing THRIL, indicating that inhi-
bition of THRIL relieved sepsis-induced inflammation and
ALI. Apoptosis is a crucial feature for inducing organ dam-
age.
27
Significant apoptosis occurs in sepsis-induced ALI,
and inhibition of THRIL can suppress the apoptosis and
further alleviate the ALI induced by sepsis.
25
Evidence
suggested that suppressed immune response may be linked
with sepsis-induced ALI. Downregulating the THRIL may
enhance immune response that can alleviate sepsis-induced
ALI as suggested by inhibition of alveolar macrophage
apoptosis.
28
Therefore, lncRNA THRIL can serve as a po-
tential therapeutic target for acute lung injury induced by
sepsis.
NEAT1
In recent years, the role of Nuclear Paraspeckle Assembly
Transcript 1 (NEAT1) has been vastly explored in multiple
diseases such as cancer and inflammatory diseases.
29
NEAT1
has also been implicated in inflammatory diseases. For
instance, it was found upregulated in rats having chronic
constriction injury, while knockdown of NEAT1 resulted in
suppression of neuroinflammation as well as neuropathic
pain behaviors.
30
In brain injury, NEAT1 has anti-
inflammatory and anti-apoptotic functions.
31
Zhou et al
32
observed a very high level of NEAT1
expression in LPS induced ALI in mouse model. Huang et al
33
also reported remarkably high levels of NEAT1 in sepsis
patients, which was associated with augmented disease
severity and poor prognosis. Alveolar epithelial cells-II
(AECs) are the main injury sites in ALI/ARDS. It maintains
the alveolar-capillary barrier integrity and produces sur-
factant substances that decrease the pulmonary surface
tension. In ALI, this mechanism is impaired and results in
lower lung compliance. Therefore, protecting AECs from
injury represents a vital therapeutic strategy against ALI
induced by sepsis.
34,35
Zhou et al investigated the role of NEAT1 on the LPS-
exposed adenocarcinoma cell line. They discovered that
cessation of NEAT1 reduces the adverse function of LPS on
the availability of AECs. Additionally, NEAT1 inhibition
restrained apoptosis, caspase-3/9 activity, and release of
LDH in LPS-induced ALI, suggesting its protective roles in
ameliorating LPS induced AECs damage and apoptosis.
32
Inflammation and cytokine storm is the leading cause of
mortality and morbidity in ALI/ARDS. Controlling the in-
flammatory response could provide a vital therapeutic
target in sepsis-induced ALI. Suppression of NEAT1 signifi-
cantly reduces the pro-inflammatory cytokines IL-6, IL-1b,
and TNF-a.
32
Zhou et al
32
also reported that restoration of the
HMGB1-RAGE pathway, which is excessively activated in
sepsis models and patients, reverses the anti-inflammatory
and anti-injury efficacy of NEAT1 knockdown in AECs
exposed LPS. Evidence also suggested that NEAT1, like
other lncRNA, regulates the activities of specific down-
stream proteins such as TRIM37.
36,37
CASC2
Cancer susceptibility candidate 2 (CASC2) is located on
chromosome 10 and has been implicated as a crucial gene
involved in the pathogenesis of many types of malignancies,
including glioma and endometrial cancers.
38
Liu et al
39
discovered that CASC2 was downregulated in A549 cell
lines treated with LPS, while accumulation of CASC2
attenuated LPS-induced ALI in in vitro and in vivo condi-
tions. Ji L et al
40
established bronchopulmonary dysplasia
(BPD) model by giving 14 days of hyperoxia treatment to
neonates mice. Furthermore, lncRNA CASC2 has been found
reduced in murine lung tissue while CAV1 protein was found
elevated.
The mechanistic function of lncRNA in acute
lung injury
lncRNA-mediated cell signaling regulation and
acute lung injury
Cell signaling is vital in various cellular and physiological
processes. Many cell signaling pathways have been discov-
ered that play a pivotal role in various biological processes
and gene expression. A single signaling pathway often
consists of multiple signaling molecules that ultimately
affect gene expression by regulating the activity of tran-
scription factors directly or indirectly. Shreds of evidence
from recent studies suggest that lncRNAs are involved in
various cell signaling pathways in many diseases.
12,57
However, few studies are available that focus on the role
of these lncRNAs in acute lung injury. Here we have dis-
cussed some pathways that have been studied so far.
Study in the past demonstrated that MALAT1 could
worsen sepsis-induced inflammation and cardiac
A. Zaki, M.S. Ali, V. Hadda et al.
+MODEL
4
insufficiency by regulating p38 MAPK/p65 NF-kB signaling
pathway.
58
Lin et al used three study groups in mice: the
control group, LPS group, and LPSþMALAT1 group, to
investigate the role of lncRNA MALAT1. They checked
various factors, including the expression level of p38MAPK/
P65 NF-kB signaling pathway-related protein in the lung
tissue and the BAL fluid of the mouse and reported that
knockdown of MALAT1 leads to reversal of the abnormal
increase in the phosphorylation of p38 MAPK and p65 NF-
kB
59
Likewise, Zhu et al
60
also demonstrated that MALAT1
via interaction with p65 blocks LPS-induced activation of
NF-kB and repressed LPS-induced inflammatory injury in
WI-38 cell line.
In a separate study on the mechanism of NEAT1 in ALI,
investigators corroborated the activation of HMGB1/RAGE
and NF-kB signaling in A549 cell lines treated with LPS.
Suppression of NEAT1 represses this pathway while reac-
tivates the HMGB1/RAGE signaling through HMGB1 over-
expression and dampened the anti-inflammatory/injury
effects of NEAT1 knockdown, implying it as a potential
therapeutic target for this condition.
32
lncRNA sponging miRNA in acute lung injury
miRNA is 18e22 nucleotides small non-coding RNA that
regulate gene expression post-transcriptionally, typically
binding at 30UTR of target messenger RNA (mRNA). On
account of acute lung injury, miRNA act as a potential
biomarker in the diagnosis and severity of the acute lung
injury disease.
61
In brief, biogenesis of miRNA begins in the
nucleus where RNA polymerase II transcribes pri-miRNA
(2000 bp long, double-stranded, capped, and poly-
adenylated) as a precursor of miRNA, which is further
cleaved and processed by RNase III-like endonuclease,
drosha, and transported as pre-miRNA to the cytoplasm
through exportin-5. In the cytoplasm, pre-miRNA is further
processed, producing 25 nucleotides miRNA duplex (miR-
NAemiRNA*) by the action of cytoplasmic RNase III Dicer.
One strand of miRNA duplex is degraded, and the remaining
one gets accommodated into an RNA-induced silencing
complex (RISC) incorporated with Argonaut protein. miRNA
loaded with RISC-Ago complex has seed region which
complementarily binds to 30or 50UTR regions of target
mRNA for its degradation resulting negative regulation.
62,63
It is estimated that approximately 60% of human genes are
targeted and regulated by miRNA, and they can serve as a
therapeutic target in various diseases.
64
The developing
horizon of research identified and reported that lncRNA is
an endogenous sponge to miRNAs that can control the
expression of genes by targeting miRNAs in acute lung
injury. In a study, downregulation of miR-34 b-5p by over-
expressed TUG1 was protective in sepsis-induced acute
lung injury in mice model.
41
Elevated MALAT1 was found to
be associated with acute lung injury that downregulated
miR-181a-5p expression.
42
Similarly, increased level of
THRIL downregulated miR-424 in sepsis-induced lung injury
where knockdown of THRIL potentially enhanced miR-424
and markedly increased the survival rate of septic mice.
25
LPS treated A549 cell lines showed a decreased amount of
lnc-CASC2 with a raised level of miR-27 b. Overexpression
of lnc-CASC2 further downregulated miR-27 b, preventing
LPS induced acute lung injury.
38
Jinyuan et al
54
performed
in vivo and in vitro studies and found aggravated expression
of lncRNA SNHG14 adversely influenced microRNA-34c-3p
expression in LPS induced acute lung injury models. It is
well established that lncRNA regulates miRNA by acting as
an agonist or antagomir. Furthermore, miRNA processing,
their transport to mRNA, and even outside the cell could be
controlled by lncRNA.
65e67
Role of lncRNA in different biological processes
in acute lung injury
Pyroptosis
Pyroptosis is a newly discovered cell death process stimu-
lated by both infectious and non-infectious sources,
including factors produced in the host during myocardial
infarction.
68
It is distinct from other forms of cell death
both morphologically and mechanistically. The defining
feature of pyroptosis is its dependency on Caspase 1, which
mediates this type of cell death. It is not involved in
apoptosis, and neither its degradation ceases apoptosis.
69
In recent decades lncRNAs were found to be associated
with pyroptosis. Pyroptosis plays a key role in the inflam-
matory resolution of acute lung inflammation. Guo
et al demonstrated that inflammasomes activation and
inflammasome-dependent pyroptosis is associated with
acute lung inflammation.
70
Several lncRNAs have been
recognized as a key regulator in acute lung inflammation,
either by modulating miRNAs/downstream molecules or by
directly regulating pyroptosis.
71,72
Luo et al investigated
the molecular mechanism and also looked for the candidate
lncRNA and mRNA involved in pyroptosis in LPS induced
acute lung injury. They found 1503 differentially expressed
lncRNA and observed that lncRNA4344 sponged miR-138-5p
and promoted pyroptosis in inflammatory responses by
targeting NLRP3 in LPS-induced ALI.
73
MALAT1 and NEAT1 have been the most widely reported
lncRNAs in acute lung injury, and it is also associated with
pyroptosis (See Fig. 2). Apart from these lncRNAs, many
other lncRNAs are associated with pyroptosis regulation,
and still, many more are being identified. Targeting these
lncRNAs associated with pyroptosis could provide a poten-
tial therapeutic target.
Apoptosis
Apoptosis is a highly conserved “programmed cell death”
found across all metazoans and is crucial for the develop-
ment, maintenance of tissue homeostasis, and prevention
of cancer.
74
Inadequate apoptosis may result in autoim-
munity or cancer. On the other hand, augmented or
heightened apoptosis may lead to degenerative diseases.
75
Blebbing, cell shrinking, nuclear fragmentation, and
apoptotic body formation are some of the characteristic
morphological changes during apoptosis.
76
Our lung is a
complex structure made up of various types of cells, such as
Genes &Diseases xxx (xxxx) xxx
+MODEL
5
endothelial cells, epithelial cells, fibroblasts, and different
leukocytes. Apoptosis of these cells can either exacerbate
or ameliorate lung injury subject to cell types involved.
77
In
the lungs of ALI patients, apoptosis of neutrophils is
delayed, and various soluble factors, including GM-CSF,
mediate this effect. In contrast, epithelial apoptosis is
likely to be mediated by sFasL.
77
The role of lncRNAs in acute lung injury and apoptosis
has been studied by many researchers in the last few years.
The results obtained by Liu et al
42
demonstrated that
downregulation of lncRNA MALAT1 significantly decreased
the Fas level and apoptosis in the HPMEC cell line and ALI
mouse model. Li et al
38
showed that accumulation of
lncRNA CASC2 attenuated the increased apoptosis in LPS
induced A549 cells. Ji et al
47
in a similar study found that
overexpression of Hsp4 lncRNA significantly reverted LPS
induced apoptosis in MLE12 cell lines. Together, these
outcomes clearly show that regulation of lncRNAs could
reduce ALI-induced apoptosis in cell lines and mouse
models.
Until now, studies have primarily focused on the role of
lncRNAs in apoptosis during ALI in general. There are two
different apoptosis pathways: intrinsic (mitochondrial-
dependent pathway) and extrinsic (death receptor-
dependent pathway).
78
The Bcl-2 family proteins regulate
the intrinsic pathway in addition to pro-apoptotic proteins
and anti-apoptotic proteins. This pathway is ignited by
the mitochondrial damage that induces the secretion of
cytochrome-c that activates the Caspase-3, which is the
ultimate factor responsible for apoptosis. On the other
hand, the extrinsic pathway is initiated by the interaction
of Fas ligand with Fas receptor and TNF-ligand with TNF-
receptor that activates the Caspase-8 culminating with
the activation of Caspase-3 and initiation of apoptosis.
79
Therefore, the apoptotic pathway associated with the ef-
fects of lncRNAs on ALI remains enigmatic, and further
studies are needed to find out more about its role.
Autophagy
In different forms of stress, including nutrient scarcity,
growth factor deprivation, infectious condition, and hyp-
oxia, cells degrade themselves to recycle nutrients essen-
tial for cellular functions by an intracellular catabolic
process known as autophagy. Autophagy initiates with the
formation of double-membrane vesicles, the autophago-
some, that encapsulates or engulfs damaged organelles,
distorted macromolecules, and different cytoplasmic
debris and ends up with the fusion with lysosomes, termed
as autolysosome, for degradation.
80,81
The autophagic flux
is the measure of cargo delivery by autophagosomes to
autolysosomes.
82
The main purpose of autophagy is to
provide cellular essentials for cell thriving. Poorly regulated
autophagy processes are linked with various diseases,
including acute lung injury. Although the role of autophagy
in the context of acute lung injury is still indefinite and
remains uncertain. However, increased pro-inflammatory
cytokines in LPS treated mice manifested acute lung
Figure 2 The roles of lncRNAs involved in the various biological process during acute lung injury. lncRNAs act via targeting their
downstream molecules, such as miRNA/proteins. TUG1 act via regulating their downstream target miR-34 b-5p/GAB1. MALAT1 has
more than one downstream target molecules including miR-181a-5p, ICAM-1, miR-425. CASC2 acts via regulating miR-27b/TAB2
axis. NEAT1 has HMGB1/RAGE signaling as its downstream target. THRIL regulates the activity of miR-424/ROCK2 for functional
effects. Mirt2 controls TRAF6, NF-kB, stat6 and MAPK signaling pathways. The target of PRNCR1 is miR-330-5p/TLR4 axis. In
contrast, XIST acts via regulating the activity of miR-21/IL-12 A.
A. Zaki, M.S. Ali, V. Hadda et al.
+MODEL
6
injury. Alveolar epithelial cells of LPS treated mice showed
a significant increase in the expression of autophagy marker
LC3B. In addition, administration of autophagy inhibitors 3-
methyladenine (3-MA) and chloroquine (CLQ) potentially
decreased total protein in BALF and lung injury score, thus
alleviating lung injury.
83
Stimulation of WI-38 human lung
fibroblasts with LPS triggers the expression of the lncRNA
HAGLROS and thereby causing cell injury.
Furthermore, downregulation of HAGLROS assuaged LPS
induced cellular injury by increasing cell viability and
curbing autophagy.
39
lncRNAs regulating autophagy in acute
lung injury are not fully characterized. Therefore, elabo-
rated research is needed to elucidate the role of lncRNA in
autophagy linked with ALI.
Inflammation
Inflammation is the sequential response by the body’s im-
mune system against harmful stimuli, including invading
infectious agents, toxic substances, damaged cells, or
irradiation, consequently eliminating noxious substances
and initiation of the healing process by the body. There-
fore, inflammation is categorized as a protective mecha-
nism essential for health.
84,85
Inflammation is known to play
an essential role in the pathophysiology of ALI. Various re-
ports have exhibited that lncRNA exerts an essential func-
tion in inflammatory processes. Excessive inflammation
could initiate a cascade to exacerbate further lung damage
leading to macrophage activation and polarization, neu-
trophils recruitment, and release of pro-inflammatory cy-
tokines such as TNF-a, IL-1b, IL-6, and ICAM1.
86
lncRNA-
5657 gets upregulated in LPS treated alveolar macrophage
cell lines and in lung tissue of CLP rats.
Furthermore, substantially overexpressed lncRNA-5657
results in the production of pro-inflammatory cytokines
including TNF-a, IL-1b, and IL-6, whereas the reverse was
noticed after silencing of lncRNA-5657.
48
In a study,
knockdown of lncRNA MALAT1 improved LPS induced acute
lung injury in mice by decreasing the level of inflammatory
cytokines. In addition, the number of neutrophils and
macrophages in BALF was found to be reduced.
59
Mice with
acute lung injury showed decreased expression of lncRNA
CASC2, AQP1, and increased miR-144-3p proportional to
inflammation in lung tissue. Alternatively, overexpression
of CASC2 by transfection in LPS treated A549 cell lines
restored AQP1 protein and ameliorated acute lung injury.
87
Zhiming et al reported upregulation of lncRNA SNHG16 and
pro-inflammatory cytokines such as (IL-6, IL-1b, and TNF-a)
in WI-38 cells after treatment with LPS. Downregulation of
SNHG16 significantly increased cell viability and propor-
tionally reduced the expression of IL-6, IL-1b, and TNF-a.
88
lncRNA effect on macrophage polarization in acute
lung injury
Macrophages are versatile and multifaceted innate immune
cells involved in the first line of defense. Their ability to
detect, engulf and eliminate intruding pathogens make
them prominent in an inflammatory response
89
However,
they can be subdivided into two functional categories (M1
and M2 macrophages) depending on specific external
stimuli and signals. M1 macrophages are the classically
activated subtype of macrophages that are involved in pro-
inflammatory response, on the other hand, alternatively
activated M2 macrophages are responsible for the anti-
inflammatory and angiogenic properties
90
. M1 macro-
phages are characterized by their ability to express pro-
inflammatory cytokines, including TNF-a and IL-1band
iNOS (inducible NO synthase).
91
Besides, M2 macrophages
release anti-inflammatory cytokines such as IL-10 and TGF-
b, including characteristic markers (Arg-1, Ym1, Fizz1)
expression.
92
Macrophage polarization plays a crucial role in the eti-
ology of many multifactorial diseases such as atheroscle-
rosis, sepsis, autoimmune diseases, cancer, tissue injury
and repair.
93
Differentially expressed lncRNA in polarized
macrophages (M1 and M2) provide a conspicuous result in
their involvement in inflammation and regulation.
94
In a
study, overexpression of MALAT1 in LPS treated mouse
BMDM was reported. Further studies also reported similar
observations in human monocytic THP-1 and human pe-
ripheral blood mononuclear cell-derived macrophages.
MALAT1 demonstrated its role in regulating clec16a (C-type
lectin domain family 16 member A) protein known for its
role in activating pro-inflammatory macrophages. MALAT1
knockdown cells were observed with substantially
decreased clec16a transcript. Extending work showed that
clec16a knockdown proportionally decreased in MALAT1
knockout mice that distinctly showed the role of MALAT1 in
macrophage activation and inflammation.
21
LPS stimulated
primary cultured peritoneal macrophages (PCPM) showed
Mirt2 lncRNA upregulation, whereas the opposite was
observed in IL-4 treated cells. Knockdown of inflammatory
pathways downstream signaling proteins such as MyD88 and
TRIF partially inhibited Mirt2 expression. Immunoprecipi-
tation assays revealed Mirt2 interaction with TRAF6 that
regulates inflammatory response. Mirt2 also regulates M2
polarization via PPARgand STAT-6 independent pathway
56
The horizon of lncRNAs can be exploited to evaluate their
intricate role in macrophage polarization and their thera-
peutic potential in acute lung injury.
Ferroptosis
Ferroptosis is a type of cell death induced by a small
molecule called erastin; it inhibits the import of cystine,
leading to depletion of glutathione and inactivation of
phospholipid peroxidase glutathione peroxidase 4 (GPX4).
95
Cell death induced by erastin in ferroptosis is distinct from
other forms of cell death such as apoptosis, necrosis, and
autophagy based on morphological, biochemical, and ge-
netic characteristics.
95
Many diseases have been reported
to be associated with ferroptosis. Recently, the role of
ferroptosis in lung diseases have also been reported.
96,97
Liu et al
98
demonstrated that Fer-1 (ferroptosis inhibitor)
alleviates the LPS-induced ALI as well as the inflammatory
responses in vivo efficiently by regulating ferroptosis.
lncRNA seems to play an essential role in the regulation
of ferroptosis. Interaction between many lncRNAs and
various cancer cells has been demonstrated, including lung
cancer cells.
99
However, the role of lncRNA in the regula-
tion of ferroptosis during acute lung injury has not been
Genes &Diseases xxx (xxxx) xxx
+MODEL
7
explored. Exploring the role of lncRNA in this area could
provide better therapeutic targets to manage ALI patients.
Efferocytosis
Efferocytosis is a process in which apoptotic cells are
cleared by the phagocytic cells. It is vital for normal
development, tissue repair and homeostatic cell turnover.
Apoptotic cells comprise many potential autoantigens and
alarmins like adenosine, HSP proteins, HMG box-1 proteins.
Secretion of these molecules during necrosis leads to mor-
tality in the appropriate sepsis model, and their long-term
persistence can also induce autoimmunity.
100
Dysfunctional
efferocytosis has been reported in many lung diseases such
as asthma, COPD, and cystic fibrosis; This led to the pro-
posal that promoting efferocytosis might arrest the pro-
gression of the disease.
101
Dysfunctional efferocytosis affects the prognosis in
ALI/ARDS murine model. During gut ischemia-reperfusion
in mice, the concentration of MFG-E8, an efferocytotic
opsonin, decreases significantly while intraperitoneal
administration of the same decrease inflammatory cyto-
kines in the lung and also alleviates lung injury leading to
increased survival.
102
In the LPS-induced ALI model, mice
lacking MFG-E8 showed enhanced neutrophil infiltration,
inflammatory cytokines, and reduced survival.
103
The role of lncRNAs in efferocytosis has not been well
studied. However, a recent study by Simion et al showed the
role of macrophage-specific lncRNA MAARS in atherosclerosis
and demonstrated that knockdown of MAARS is associated
with increased expression of MerTK, a crucial receptor for
efferocytosis, both in vivo and in bone marrow-derived
macrophages (BMDMs). Additionally, silencing of MAARS
increased the expression of mRNA SIRT1, an anti-apoptotic
HuR target gene known to increase the macrophage-
dependent efferocytosis.
104
Conclusion and future perspective
Acute lung injury is one of the major causes of ICU death
around the world. The current treatment strategy is not
effective enough to mitigate disease morbidity and mor-
tality. New therapeutic targets are needed urgently that
can effectively reduce the disease burden. In the last few
decades, lncRNAs provided a new dimension to researchers.
Studies have successfully demonstrated these molecules as
novel potential targets in acute lung injury/ARDS/sepsis.
These RNAs are involved in various pathophysiological pro-
cesses such as apoptosis, inflammation, cell signaling
pathways etc. However, the mechanisms underlying these
processes are still needed to be explored extensively.
Moreover, most of these studies are based on mouse models
and cell lines, and only a few are based on patient’s sam-
ples. Therefore, the role of these lncRNAs is needed to be
established in a larger sample size and to find its role as a
therapeutic target in human subjects.
Conflict of Interests
The authors declare that there is no conflict of interests.
Acknowledgements
We would like to thank All India Institute of Medical Sci-
ences and Jamia Millia Islamia, New Delhi for providing
infrastructure, journal access, and internet facilities. We
acknowledge the Indian Council of Medical Research (ICMR)
for providing fellowship to AZ and MSA.
References
1. Rubenfeld GD, Caldwell E, Peabody E, et al. Incidence and
outcomes of acute lung injury. N Engl J Med. 2005;353(16):
1685e1693.
2. Dushianthan A, Grocott MP, Postle AD, Cusack R. Acute res-
piratory distress syndrome and acute lung injury. Postgrad
Med. 2011;87(1031):612e622.
3. McNicholas BA, Rooney GM, Laffey JG. Lessons to learn from
epidemiologic studies in ARDS. Curr Opin Crit Care. 2018;
24(1):41e48.
4. Mathy NW, Chen XM. Long non-coding RNAs (lncRNAs) and
their transcriptional control of inflammatory responses. J Biol
Chem. 2017;292(30):12375e12382.
5. Elgar G, Vavouri T. Tuning in to the signals: noncoding
sequence conservation in vertebrate genomes. Trends Genet.
2008;24(7):344e352.
6. Uchida S, Dimmeler S. Long noncoding RNAs in cardiovascular
diseases. Circ Res. 2015;116(4):737e750.
7. Palazzo AF, Lee ES. Non-coding RNA: what is functional and
what is junk? Front Genet. 2015;6:e2.
8. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet.
2011;12(12):861e874.
9. Cao J. The functional role of long non-coding RNAs and epi-
genetics. Biol Proced Online. 2014;16:e11.
10. Hadjicharalambous MR, Lindsay MA. Long non-coding RNAs
and the innate immune response. Noncoding RNA. 2019;5(2):
e34.
11. Marques-Rocha JL, Samblas M, Milagro FI, Bressan J,
Martı
´
nez JA, Marti A. Noncoding RNAs, cytokines, and
inflammation-related diseases. Faseb J. 2015;29(9):
3595e3611.
12. Chen J, Ao L, Yang J. Long non-coding RNAs in diseases
related to inflammation and immunity. Ann Transl Med. 2019;
7(18):e494.
13. Ma L, Cao J, Liu L, et al. Lncbook: a curated knowledgebase of
human long non-coding RNAs. Nucleic Acids Res. 2019;47(D1):
D128eD134.
14. Li Z, Li X, Chen C, et al. Long non-coding RNAs in nucleus
pulposus cell function and intervertebral disc degeneration.
Cell Prolif. 2018;51(5), e12483.
15. Dharap A, Nakka VP, Vemuganti R. Effect of focal ischemia on
long noncoding RNAs. Stroke. 2012;43(10):2800e2802.
16. Wu P, Zuo X, Deng H, Liu X, Liu L, Ji A. Roles of long noncoding
RNAs in brain development, functional diversification and
neurodegenerative diseases. Brain Res Bull. 2013;97:69e80.
17. Schmitt AM, Chang HY. Long noncoding RNAs in cancer path-
ways. Canc Cell. 2016;29(4):452e463.
18. Booton R, Lindsay MA. Emerging role of microRNAs and long
noncoding RNAs in respiratory disease. Chest. 2014;146(1):
193e204.
19. Groot M, Zhang D, Jin Y. Long non-coding RNA review and
implications in lung diseases. JSM Bioinform Genom Proteom.
2018;3(2), e1033.
20. Zhao M, Wang S, Li Q, Ji Q, Guo P, Liu X. MALAT1: a long non-
coding RNA highly associated with human cancers (review).
Oncol Lett. 2018;16(1):19e26.
A. Zaki, M.S. Ali, V. Hadda et al.
+MODEL
8
21. Cui H, Banerjee S, Guo S, et al. Long noncoding RNA Malat1
regulates differential activation of macrophages and response
to lung injury. JCI Insight. 2019;4(4), e124522.
22. Saqib U, Sarkar S, Suk K, Mohammad O, Baig MS, Savai R.
Phytochemicals as modulators of M1-M2 macrophages in
inflammation. Oncotarget. 2018;9(25):17937e17950.
23. Qi H, Shen J, Zhou W. Up-regulation of long non-coding RNA
THRIL in coronary heart disease: prediction for disease risk,
correlation with inflammation, coronary artery stenosis, and
major adverse cardiovascular events. J Clin Lab Anal. 2020;
34(5), e23196.
24. Wang Y, Fu X, Yu B, Ai F. Long non-coding RNA THRIL predicts
increased acute respiratory distress syndrome risk and posi-
tively correlates with disease severity, inflammation, and
mortality in sepsis patients. J Clin Lab Anal. 2019;33(6),
e22882.
25. Chen H, Hu X, Li R, et al. LncRNA THRIL aggravates sepsis-
induced acute lung injury by regulating miR-424/ROCK2
axis. Mol Immunol. 2020;126:111e119.
26. Li Z, Chao TC, Chang KY, et al. The long noncoding RNA THRIL
regulates TNFaexpression through its interaction with
hnRNPL. Proc Natl Acad Sci U S A. 2014;111(3):1002e1007.
27. Wang Z, Wang SP, Shao Q, et al. Brain-derived neurotrophic
factor mimetic, 7,8-dihydroxyflavone, protects against
myocardial ischemia by rebalancing optic atrophy 1 process-
ing. Free Radic Biol Med. 2019;145:187e197.
28. Yang L, Zhang Z, Zhuo Y, et al. Resveratrol alleviates sepsis-
induced acute lung injury by suppressing inflammation and
apoptosis of alveolar macrophage cells. Am J Transl Res.
2018;10(7):1961e1975.
29. Yu X, Li Z, Zheng H, Chan MT, Wu WK. NEAT1: a novel cancer-
related long non-coding RNA. Cell Prolif. 2017;50(2):e12329.
30. Xia LX, Ke C, Lu JM. NEAT1 contributes to neuropathic pain
development through targeting miR-381/HMGB1 axis in CCI
rat models. J Cell Physiol. 2018;233(9):7103e7111.
31. Zhong J, Jiang L, Huang Z, et al. The long non-coding RNA
Neat1 is an important mediator of the therapeutic effect of
bexarotene on traumatic brain injury in mice. Brain Behav
Immun. 2017;65:183e194.
32. Zhou H, Wang X, Zhang B. Depression of lncRNA NEAT1 an-
tagonizes LPS-evoked acute injury and inflammatory response
in alveolar epithelial cells via HMGB1-RAGE signaling. Mediat
Inflamm. 2020;2020, e8019467.
33. Huang Q, Huang C, Luo Y, He F, Zhang R. Circulating lncRNA
NEAT1 correlates with increased risk, elevated severity and
unfavorable prognosis in sepsis patients. Am J Emerg Med.
2018;36(9):1659e1663.
34. Tojo K, Tamada N, Nagamine Y, Yazawa T, Ota S, Goto T.
Enhancement of glycolysis by inhibition of oxygen-sensing
prolyl hydroxylases protects alveolar epithelial cells from
acute lung injury. Faseb J. 2018;32(4):2258e2268.
35. Li C, Huang Y, Yao X, et al. Lugrandoside attenuates LPS-
induced acute respiratory distress syndrome by anti-
inflammation and anti-apoptosis in mice. Am J Transl Res.
2016;8(12):5557e5568.
36. Chen C, Zhang H, Ge M, Ye J, Li R, Wang D. LncRNA NEAT1 acts
as a key regulator of cell apoptosis and inflammatory response
by the miR-944/TRIM37 axis in acute lung injury. J Pharmacol
Sci. 2021;145(2):202e212.
37. Yang Y, Yang L, Liu Z, Wang Y, Yang J. Long noncoding RNA
NEAT 1 and its target microRNA-125a in sepsis: correlation
with acute respiratory distress syndrome risk, biochemical
indexes, disease severity, and 28-day mortality. J Clin Lab
Anal. 2020;34(12), e23509.
38. Li X, Mo J, Li J, Chen Y. lncRNA CASC2 inhibits
lipopolysaccharide-induced acute lung injury via miR-
27b/TAB2 axis. Mol Med Rep. 2020;22(6):5181e5190.
39. Liu M, Han T, Shi S, Chen E. Long noncoding RNA HAGLROS
regulates cell apoptosis and autophagy in
lipopolysaccharides-induced WI-38cells via modulating miR-
100/NF-kB axis. Biochem Biophys Res Commun. 2018;500(3):
589e596.
40. Ji L, Liu Z, Dong C, Wu D, Yang S, Wu L. LncRNA CASC2 targets
CAV1 by competitively binding with microRNA-194-5p to
inhibit neonatal lung injury. Exp Mol Pathol. 2021;118,
e104575.
41. Qiu N, Xu X, He Y. LncRNA TUG1 alleviates sepsis-induced
acute lung injury by targeting miR-34b-5p/GAB1. BMC Pulm
Med. 2020;20(1):e49.
42. Liu Y, Wang X, Li P, et al. Targeting MALAT1 and miRNA-181a-
5p for the intervention of acute lung injury/acute respiratory
distress syndrome. Respir Med. 2020;175, e106210.
43. Yao MY, Zhang WH, Ma WT, Liu QH, Xing LH, Zhao GF. Long
non-coding RNA MALAT1 exacerbates acute respiratory
distress syndrome by upregulating ICAM-1 expression via
microRNA-150-5p downregulation. Aging (Albany NY). 2020;
12(8):6570e6585.
44. Wang L, Liu J, Xie W, et al. Overexpression of MALAT1 relates
to lung injury through sponging miR-425 and promoting cell
apoptosis during ARDS. Canc Res J. 2019;2019, e1871394.
45. Wang J, Zhang Y, Zhang L. Long non-coding RNA SNHG5 sup-
presses the development of acute respiratory distress syn-
drome by targeting miR-205/COMMD1 axis. Mol Cell Biochem.
2021;476(2):1063e1074.
46. Yu Y, Sun H, Zhu L, Ji L, Liu H. Downregulating lncRNA PRNCR1
ameliorates LPS-induced pulmonary vascular endothelial cell
injury by modulating miR-330-5p/TLR4 axis. J Biochem Mol
Toxicol. 2021;35(2), e22644.
47. Ji Q, Pan C, Wang J, et al. Long non-coding RNA Hsp4 allevi-
ates lipopolysaccharide-induced apoptosis of lung epithelial
cells via miRNA-466m-3p/DNAjb6 axis. Exp Mol Pathol. 2020;
117, e104547.
48. Liu F, Hu S, Zhao N, et al. LncRNA-5657 silencing alleviates
sepsis-induced lung injury by suppressing the expression of
spinster homology protein 2. Int Immunopharm. 2020;88,
e106875.
49. Chen Z, Dong WH, Qiu ZM, Li QG. The monocyte-derived
exosomal CLMAT3 activates the CtBP2-p300-NF-kB transcrip-
tional complex to induce proinflammatory cytokines in ALI.
Mol Ther Nucleic Acids. 2020;21:1100e1110.
50. Liao H, Zhang S, Qiao J. Silencing of long non-coding RNA
MEG3 alleviates lipopolysaccharide-induced acute lung injury
by acting as a molecular sponge of microRNA-7b to modulate
NLRP3. Aging (Albany NY). 2020;12(20):20198e20211.
51. Wang HR, Guo XY, Liu XY, Song X. Down-regulation of lncRNA
CASC9 aggravates sepsis-induced acute lung injury by regu-
lating miR-195-5p/PDK4 axis. Inflamm Res. 2020;69(6):
559e568.
52. Li J, Wei L, Han Z, Chen Z, Zhang Q. Long non-coding RNA X-
inactive specific transcript silencing ameliorates primary graft
dysfunction following lung transplantation through microRNA-
21-dependent mechanism. EBioMedicine. 2020;52, e102600.
53. Li J, Liu S. LncRNA GAS5 suppresses inflammatory responses
and apoptosis of alveolar epithelial cells by targeting miR-
429/DUSP1. Exp Mol Pathol. 2020;113, e104357.
54. Zhu J, Bai J, Wang S, Dong H. Down-regulation of long non-
coding RNA SNHG14 protects against acute lung injury
induced by lipopolysaccharide through microRNA-34c-3p-
dependent inhibition of WISP1. Respir Res. 2019;20(1):e233.
55. Mu X, Wang H, Li H. Silencing of long noncoding RNA H19 al-
leviates pulmonary injury, inflammation, and fibrosis of acute
respiratory distress syndrome through regulating the
microRNA-423-5p/FOXA1 axis. Exp Lung Res. 2021;47(4):
183e197.
Genes &Diseases xxx (xxxx) xxx
+MODEL
9
56. Du M, Yuan L, Tan X, et al. The LPS-inducible lncRNA Mirt2 is a
negative regulator of inflammation. Nat Commun. 2017;8(1),
e2049.
57. Peng WX, Koirala P, Mo YY. LncRNA-mediated regulation of
cell signaling in cancer. Oncogene. 2017;36(41):5661e5667.
58. Wei S, Liu Q. Long noncoding RNA MALAT1 modulates sepsis-
induced cardiac inflammation through the miR-150-5p/NF-kB
axis. Int J Clin Exp Pathol. 2019;12(9):3311e3319.
59. Lin LP, Niu GH, Zhang XQ. Influence of lncRNA MALAT1 on
septic lung injury in mice through p38 MAPK/p65 NF-kB
pathway. Eur Rev Med Pharmacol Sci. 2019;23(3):1296e1304.
60. Zhu W, Men X. Negative feedback of NF-kB signaling by long
noncoding RNA MALAT1 controls lipopolysaccharide-induced
inflammation injury in human lung fibroblasts WI-38. J Cell
Biochem. 2020;121(2):1945e1952.
61. Jiang ZF, Zhang L, Shen J. MicroRNA: potential biomarker and
target of therapy in acute lung injury. Hum Exp Toxicol. 2020;
39(11):1429e1442.
62. Du T, Zamore PD. Beginning to understand microRNA function.
Cell Res. 2007;17(8):661e663.
63. O’Connell RM, Rao DS, Baltimore D. MicroRNA regulation of
inflammatory responses. Annu Rev Immunol. 2012;30:
295e312.
64. Strub GM, Perkins JA. MicroRNAs for the pediatric otolaryn-
gologist. Int J Pediatr Otorhinolaryngol. 2018;112:195e207.
65. Paneru B, Ali A, Al-Tobasei R, Kenney B, Salem M. Crosstalk
among lncRNAs, microRNAs and mRNAs in the muscle
“degradome” of rainbow trout. Sci Rep. 2018;8(1), e8416.
66. Li DS, Ainiwaer JL, Sheyhiding I, Zhang Z, Zhang LW. Identi-
fication of key long non-coding RNAs as competing endoge-
nous RNAs for miRNA-mRNA in lung adenocarcinoma. Eur Rev
Med Pharmacol Sci. 2016;20(11):2285e2295.
67. Paraskevopoulou MD, Hatzigeorgiou AG. Analyzing
MiRNAeLncRNA interactions. Methods Mol Biol. 2016;1402:
271e286.
68. Frantz S, Ducharme A, Sawyer D, et al. Targeted deletion of
caspase-1 reduces early mortality and left ventricular dila-
tation following myocardial infarction. J Mol Cell Cardiol.
2003;35(6):685e694.
69. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death
and inflammation. Nat Rev Microbiol. 2009;7(2):99e109.
70. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of
action, role in disease, and therapeutics. Nat Med. 2015;
21(7):677e687.
71. Li J, Tian H, Yang J, Gong Z. Long noncoding RNAs regulate
cell growth, proliferation, and apoptosis. DNA Cell Biol. 2016;
35(9):459e470.
72. Ba
¨r C, Chatterjee S, Thum T. Long noncoding RNAs in car-
diovascular pathology, diagnosis, and therapy. Circulation.
2016;134(19):1484e1499.
73. Luo D, Liu F, Zhang J, et al. Comprehensive analysis of
LncRNA-mRNA expression profiles and the ceRNA network
associated with pyroptosis in LPS-induced acute lung injury. J
Inflamm Res. 2021;14:413e428.
74. Singh R, Letai A, Sarosiek K. Regulation of apoptosis in health
and disease: the balancing act of BCL-2 family proteins. Nat
Rev Mol Cell Biol. 2019;20(3):175e193.
75. Hassan M, Watari H, Abualmaaty A, Ohba Y, Sakuragi N.
Apoptosis and molecular targeting therapy in cancer. BioMed
Res Int. 2014;2014, e150845.
76. Grosse J, Warnke E, Wehland M, et al. Mechanisms of
apoptosis in irradiated and sunitinib-treated follicular thyroid
cancer cells. Apoptosis. 2014;19(3):480e490.
77. Lu Q, Harrington EO, Rounds S. Apoptosis and lung injury. Keio
J Med. 2005;54(4):184e189.
78. Zhang L, Wang H. Long non-coding RNA in CNS injuries: a new
target for therapeutic intervention. Mol Ther Nucleic Acids.
2019;17:754e766.
79. Czabotar PE, Lessene G, Strasser A, Adams JM. Control of
apoptosis by the BCL-2 protein family: implications for
physiology and therapy. NatRevMolCellBiol. 2014;15(1):
49e63.
80. Dikic I, Elazar Z. Mechanism and medical implications of
mammalian autophagy. Nat Rev Mol Cell Biol. 2018;19(6):
349e364.
81. Mohsin M, Tabassum G, Ahmad S, Ali S, Ali Syed M. The role of
mitophagy in pulmonary sepsis. Mitochondrion. 2021;59:
63e75.
82. Ogata M, Hino S, Saito A, et al. Autophagy is activated for cell
survival after endoplasmic reticulum stress. Mol Cell Biol.
2006;26(24):9220e9231.
83. Guo L, Wu X, Zhao S, Zhang X, Qian G, Li S. Autophagy inhi-
bition protects from alveolar barrier dysfunction in LPS-
induced ALI mice by targeting alveolar epithelial cells.
Respir Physiol Neurobiol. 2021;283, e103532.
84. Medzhitov R. Inflammation 2010: new adventures of an old
flame. Cell. 2010;140(6):771e776.
85. Ferrero-Miliani L, Nielsen OH, Andersen PS, Girardin SE.
Chronic inflammation: importance of NOD2 and NALP3 in
interleukin-1beta generation. Clin Exp Immunol. 2007;147(2):
227e235.
86. Goodman RB, Pugin J, Lee JS, Matthay MA. Cytokine-medi-
ated inflammation in acute lung injury. Cytokine Growth
Factor Rev. 2003;14(6):523e535.
87. Li H, Shi H, Gao M, Ma N, Sun R. Long non-coding RNA CASC2
improved acute lung injury by regulating miR-144-3p/AQP1
axis to reduce lung epithelial cell apoptosis. Cell Biosci.
2018;8:e15.
88. Zhou Z, Zhu Y, Gao G, Zhang Y. Long noncoding RNA SNHG16
targets miR-146a-5p/CCL5 to regulate LPS-induced WI-38 cell
apoptosis and inflammation in acute pneumonia. Life Sci.
2019;228:189e197.
89. Khan S, Masood M, Gaur H, Ahmad S, Syed MA. Long non-
coding RNA: an immune cells perspective. Life Sci. 2021;
271, e119152.
90. Li C, Xu MM, Wang K, Adler AJ, Vella AT, Zhou B. Macrophage
polarization and meta-inflammation. Transl Res. 2018;191:
29e44.
91. Obaid M, Udden SMN, Deb P, Shihabeddin N, Zaki MH,
Mandal SS. LncRNA HOTAIR regulates lipopolysaccharide-
induced cytokine expression and inflammatory response in
macrophages. Sci Rep. 2018;8(1), e15670.
92. Khan MJ, Singh P, Dohare R, et al. Inhibition of miRNA-34a
promotes M2 macrophage polarization and improves LPS-
induced lung injury by targeting Klf4. Genes. 2020;11(9):
e966.
93. Murray PJ. Macrophage polarization. Annu Rev Physiol. 2017;
79:541e566.
94. Huang Z, Luo Q, Yao F, et al. Identification of differentially
expressed long non-coding RNAs in polarized macrophages.
Sci Rep. 2016;6, e19705.
95. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of
ferroptotic cancer cell death by GPX4. Cell. 2014;156(1e2):
317e331.
96. Li X, Zhuang X, Qiao T. Role of ferroptosis in the process of
acute radiation-induced lung injury in mice. Biochem Biophys
Res Commun. 2019;519(2):240e245.
97. Li X, Duan L, Yuan S, Zhuang X, Qiao T, He J. Ferroptosis in-
hibitor alleviates Radiation-induced lung fibrosis (RILF) via
down-regulation of TGF-b1. J Inflamm. 2019;16:e11.
A. Zaki, M.S. Ali, V. Hadda et al.
+MODEL
10
98. Liu P, Feng Y, Li H, et al. Ferrostatin-1 alleviates
lipopolysaccharide-induced acute lung injury via inhibiting
ferroptosis. Cell Mol Biol Lett. 2020;25:e10.
99. Zhang X, Wang L, Li H, Zhang L, Zheng X, Cheng W. Crosstalk
between noncoding RNAs and ferroptosis: new dawn for over-
coming cancer progression. Cell Death Dis. 2020;11(7), e580.
100. McCubbrey AL, Curtis JL. Efferocytosis and lung disease.
Chest. 2013;143(6):1750e1757.
101. Henson PM, Cosgrove GP, Vandivier RW. Apoptosis and cell
homeostasis in chronic obstructive pulmonary disease. Proc
Am Thorac Soc. 2006;3(6):512e516.
102. Cui T, Miksa M, Wu R, et al. Milk fat globule epidermal growth
factor 8 attenuates acute lung injury in mice after intestinal
ischemia and reperfusion. Am J Respir Crit Care Med. 2010;
181(3):238e246.
103. Aziz M, Matsuda A, Yang WL, Jacob A, Wang P. Milk fat
globule-epidermal growth factor-factor 8 attenuates neutro-
phil infiltration in acute lung injury via modulation of CXCR2.
J Immunol. 2012;189(1):393e402.
104. Simion V, Zhou H, Haemmig S, et al. A macrophage-specific
lncRNA regulates apoptosis and atherosclerosis by tethering
HuR in the nucleus. Nat Commun. 2020;11(1):e6135.
Genes &Diseases xxx (xxxx) xxx
+MODEL
11
... Another major class of non-coding RNA is lncRNA which are more than 200 nucleotides in length and is the second most widely studied ncRNA in human diseases after miRNA. Multiple studies have shown the dysregulation of lncRNA in brotic diseases, including pulmonary brosis as well as in acute lung injury [81], [82]. ...
... MALAT1 is a prominent lncRNA involved in many diseases including acute lung injury [82]. MALAT1 has been reported in macrophage activation and associated with pulmonary brosis. ...
Preprint
Full-text available
Pulmonary fibrosis is the major manifestation of idiopathic interstitial pneumonia as well as post-COVID-19 complications. The pathogenesis of PF is a complex molecular process that involves many types of cells, proteins, genes, and regulatory elements. The non-coding RNA is the main regulatory element in this process which mainly includes; miRNA, circRNA, and lncRNA. These regulatory elements control the expression of many important genes and various pathways that are involved in the pathogenesis of pulmonary fibrosis. Identification and molecular mechanisms by which these non-coding RNA molecules works are very important because they do not only help to understand the molecular basis of the disease but could also serve as a potential diagnostic/prognostic marker as well as therapeutic targets. In this review, we have provided the latest findings and discussed the role of these regulatory elements in various biological processes and pathways involved in the pathogenesis of pulmonary fibrosis associated with IIPs and Covid-19.
... Earlier studies have proved the possible role of lncRNAs in different respiratory diseases such as ALI [10], eosinophilic asthma [11], pneumonia [12], cigarette smoke-associated airway inflammatory disorder [13], etc. Crucial roles of lncRNAs have been identified in the pathogenesis of ALI [14,15]. LncRNAs not only possess a key role in the pathogenesis of ALI but also retain great potentials for the diagnosis [16] and treatment of ALI [17]. ...
Article
Full-text available
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), represent an acute stage of lung inflammation where the alveolar epithelium loses its functionality. ALI has a devastating impact on the population as it not only has a high rate of incidence, but also has high rates of morbidity and mortality. Due to the involvement of multiple factors, the pathogenesis of ALI is complex and is not fully understood yet. Long noncoding RNAs (lncRNAs) are a group of non-protein-coding transcripts longer than 200 nucleotides. Growing evidence has shown that lncRNAs have a decisive role in the pathogenesis of ALI. LncRNAs can either promote or hinder the development of ALI in various cell types in the lungs. Mechanistically, current studies have found that lncRNAs play crucial roles in the pathogenesis of ALI via the regulation of small RNAs (e.g., microRNAs) or downstream proteins. Undoubtedly, lncRNAs not only have the potential to reveal the underlying mechanisms of ALI pathogenesis but also serve as diagnostic and therapeutic targets for the therapy of ALI.
Article
Objective Endothelial glycocalyx (EG) maintains vascular homeostasis and is destroyed after one‐lung ventilation (OLV)‐induced lung injury. Long noncoding RNAs (lncRNAs) are critically involved in various lung injuries. This study aimed to investigate the role and regulatory mechanism of KCNQ1 overlapping transcript 1 (KCNQ1OT1) in OLV‐induced lung injury and LPS‐induced type II alveolar epithelial cell (AECII) apoptosis. Methods The rat OLV model was established, and the effects of KCNQ1OT1 on OLV‐induced ALI in vivo were explored. Bax and Caspase‐3 expression in rat lung tissues was measured by immunochemistry (IHC). AECIIs were isolated from rat lungs and treated with LPS or normal saline (control) for in vitro analysis. The expression of KCNQ1OT1, miR‐129‐5p, and HMGB1 was measured by quantitative real‐time PCR (qRT‐PCR) or Western blot (WB). Cell proliferation and apoptosis were examined by 3‐(4,5)‐dimethylthiahiazo (‐z‐y1)‐3,5‐di‐ phenytetrazoliumromide (MTT) and flow cytometry. The downstream targets of KCNQ1OT1 were predicted by bioinformatics, and the binding relationship between KCNQ1OT1 and miR‐129‐3p was verified by dual‐luciferase reporter assays. The potential target of miR‐129‐5p was further explored on the Targetscan website and revealed to target HMGB1. Enzyme‐linked immunosorbent assay (ELISA) or WB was adopted to determine the levels of IL‐1β, TNF‐α, MDA, SOD, heparanase (HPA), matrix metalloproteinase 9 (MMP9), heparan sulfate (HS) and syndecan‐1 (SDC‐1). Results KCNQ1OT1 and HMGB1 were up‐regulated during OLV‐induced lung injury, and their expression was positively correlated. KCNQ1OT1 knockdown reduced OLV‐induced pulmonary edema and lung epithelial cell apoptosis, increased vascular permeability, reduced IL‐1β, TNF‐α, MDA, and SOD levels and glycocalyx markers by targeting miR‐129‐5p or upregulating HMGB1. Overexpressing KCNQ1OT1 promoted cell apoptosis, reduced cell proliferation, aggravated inflammation and oxidative stress, and up‐regulated HMGB1, HPA and MMP9 in LPS‐treated AECIIs, while the HMGB1 silencing showed the opposite effects. MiR‐129‐5p mimics partially eliminated the KCNQ1OT1‐induced effects, while recombinant HMGB1 restored the effects of miR‐129‐5p overexpression on AECIIs. Additionally, KCNQ1OT1 was demonstrated to promote the activation of the p38 MAPK/Akt/ERK signaling pathways in AECIIs via HMGB1. Conclusion KCNQ1OT1 knockdown alleviated AECII apoptosis and EG damage during OLV by targeting miR‐129‐5p/HMGB1 to inactivate the p38 MAPK/Akt/ERK signaling. The findings of our study might deepen our understanding of the molecular basis in OLV‐induced lung injury and provide clues for the targeted disease management.
Article
Full-text available
Background: Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a common respiratory disease characterized by persistent hypoxemia and an uncontrolled inflammatory response. Valsartan, an angiotensin II type 1 receptor antagonist, is clinically used to treat hypertension and has anti-inflammatory and antioxidant effects on gefitinib-induced pneumonia in rats. However, the potential therapeutic effects of valsartan on lipopolysaccharide (LPS)-induced ALI remain unclear. This study investigated the protective role of valsartan in LPS-induced ALI and its underlying mechanisms. Methods: LPS-treated BEAS-2B cells and ALI mouse model were established. BEAS-2B cells were treated with LPS (10 μg/mL) for 24h, with or without valsartan (20, 40, and 80 µM). For ALI mouse models, LPS (5 mg/kg) was administered through intratracheal injection to treat the mice for 24h, and valsartan (10 or 30 mg/kg) was injected intraperitoneally twice 2 h before and 12 h after the LPS injection. Pulmonary functional parameters were examined by an EMKA pulmonary system. Hematoxylin and eosin staining, flow cytometry, CCK-8 assay, qRT-PCR, ELISA, immunofluorescence, Western blotting, and related commercial kits were used to assess the pathological damage to the lungs, neutrophil recruitment in the lung tissue and bronchoalveolar lavage fluid (BALF), cell viability, inflammation, oxidative activity, and mucus production, respectively. Potential mechanisms were further explored using network pharmacology and Western blotting. Results: Valsartan rescued LPS-reduced cell viability of BEAS-2B cells, improved the pulmonary function, ameliorated pathological lung injury in mice with ALI, ameliorated LPS-induced neutrophil recruitment in BALF and lung tissue of mice, attenuated oxidative stress by increasing the level of SOD and decreasing that of MDA and GSSG, inhibited LPS-induced MUC5AC overproduction, decreased the LPS-induced increase in expression of pro-inflammatory cytokines/chemokines including TNF-α, IL-6, IL-1β, CXCL-1 and CXCL-2, and restored the expression of anti-inflammatory IL-10. Mechanistic studies showed that valsartan inhibits LPS-induced phosphorylation of nuclear factor-kappa B (NF-κΒ) and mitogen-activated protein kinases (MAPKs) including P38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK) in both LPS-treated cells and the mouse model of ALI. Conclusion: Valsartan protects against LPS-induced ALI by attenuating oxidative stress, reducing MUC5AC production, and attenuating the inflammatory response that may involve MAPK and NF-κΒ pathways.
Article
Full-text available
Sepsis is a systemic inflammatory disease caused by severe infections that involves multiple systemic organs, among which the lung is the most susceptible, leaving patients highly vulnerable to acute lung injury (ALI). Refractory hypoxemia and respiratory distress are classic clinical symptoms of ALI caused by sepsis, which has a mortality rate of 40%. Despite the extensive research on the mechanisms of ALI caused by sepsis, the exact pathological process is not fully understood. This article reviews the research advances in the pathogenesis of ALI caused by sepsis by focusing on the treatment regimens adopted in clinical practice for the corresponding molecular mechanisms. This review can not only contribute to theories on the pathogenesis of ALI caused by sepsis, but also recommend new treatment strategies for related injuries.
Article
Full-text available
Ferroptosis, a programmed cell death discovered in recent years, is an iron-dependent lipid peroxidation accumulation. Unlike other modes of cell death (autophagy, necroptosis, pyroptosis, cuproptosis, etc.), ferroptosis has unique morphological characteristics and plays an important role in a variety of diseases. In recent years, there has been great progress in the study of ferroptosis. Studies have found that ferroptosis is associated with acute lung injury (ALI), a condition with a high mortality rate and limited treatment options. This paper summarizes the mechanism of ferroptosis from the perspectives of iron metabolism, lipid metabolism, amino acid metabolism, and glutathione metabolism. It also discusses the research progress of ferroptosis in ALI in order to find new directions for the prevention and treatment of this condition.
Article
Full-text available
Exposure to toxicants/stressors has been linked to the development of many human diseases. They could affect various cellular components, such as DNA, proteins, lipids, and non-coding RNAs (ncRNA), thereby triggering various cellular pathways, particularly oxidative stress, inflammatory responses, and apoptosis, which can contribute to pathophysiological states. Accordingly, modulation of these pathways has been the focus of numerous investigations for managing related diseases. The involvement of various ncRNAs, such as small interfering RNA (siRNA), microRNAs (miRNA), and long non-coding RNAs (lncRNA), as well as various proteins and peptides in mediating these pathways, provides many target sites for pharmaceutical intervention. In this regard, various oligonucleotide- and protein/peptide-based therapies have been developed to treat toxicity-induced diseases, which have shown promising results in vitro and in vivo. This comprehensive review provides information about various aspects of toxicity-related diseases including their causing factors, main underlying mechanisms and intermediates, and their roles in pathophysiological states. Particularly, it highlights the principles and mechanisms of oligonucleotide- and protein/peptide-based therapies in the treatment of toxicity-related diseases. Furthermore, various issues of oligonucleotides and proteins/peptides for clinical usage and potential solutions are discussed.
Article
Acute lung injury (ALI) and its extreme manifestation, acute respiratory distress syndrome (ARDS), are life-threatening diseases in intensive care units. LncRNA THRIL plays a crucial role in regulating the inflammatory response; however, the potential function of THRIL in ALI/ARDS and the associated mechanism remain unclear. In our study, we found that THRIL was upregulated in the serum of ALI/ARDS patients, and its increased expression was positively correlated with the inflammatory cytokines IL-17. In LPS-induced A549 cells, knockdown of THRIL inhibited the release of the proinflammatory cytokines TNF-α, IL-1β, IL-17, and IL-6, decreased the number of monodansylcadaverine-positive cells and LC3-II with immunofluorescence staining, decreased the expression of autophagy marker ATG7 and Beclin1, and increased expression of p62. Mechanistically, the transcription factor AP-1 bound directly to the THRIL promoter region and activated its transcription by c-Jun upon LPS exposure. Moreover, m6A modification of THRIL was increased in LPS-treated A549 cells, and METTL14 knockdown significantly abolished m6A modification and reduced stabilization of THRIL mRNA. In conclusion, our findings reveal that THRIL, transcriptionally activated by AP-1 and modified by METTL14-mediated m6A modification, induces autophagy in LPS-treated A549 cells, suggesting the potential application of THRIL for ALI/ARDS therapy.
Article
Full-text available
Long non-coding RNAs (lncRNAs) participate in acute lung injury (ALI). However, their latent biological function and molecular mechanism have not been fully understood. In the present study, the global expression profiles of lncRNAs and mRNAs between the control and lipopolysaccharide (LPS)-stimulated groups of human normal lung epithelial cells (BEAS-2B) were determined using high-throughput sequencing. Overall, a total of 433 lncRNAs and 183 mRNAs were differentially expressed. A lncRNA–mRNA co-expression network was established, and then the top 10 lncRNAs were screened using topological methods. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis results showed that the key lncRNAs targeting mRNAs were mostly enriched in the inflammatory-related biological processes. Gene set variation analysis and Pearson’s correlation coefficients confirmed the close correlation for the top 10 lncRNAs with inflammatory responses. A protein–protein interaction network analysis was conducted based on the key lncRNAs targeting mRNAs, where IL-1β, IL-6, and CXCL8 were regarded as the hub genes. A competing endogenous RNA (ceRNA) modulatory network was created with five lncRNAs, thirteen microRNAs, and twelve mRNAs. Finally, real-time quantitative reverse transcription-polymerase chain reaction was employed to verify the expression levels of several key lncRNAs in BEAS-2B cells and human serum samples.
Article
Pulmonary disease such as chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis and pulmonary hypertension are the leading cause of deaths. More importantly, lung diseases are on the rise and environmental factors induced epigenetic modifications are major players on this increased prevalence. It has been reported that dysregulation of genes involved in epigenetic regulation such as the histone deacetylase (HDACs) and histone acetyltransferase (HATs) play important role in lung health and pulmonary disease pathogenesis. Inflammation is an essential component of respiratory diseases. Injury and inflammation trigger release of extracellular vesicles that can act as epigenetic modifiers through transfer of epigenetic regulators such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), proteins and lipids, from one cell to another. The immune dysregulations caused by the cargo contents are important contributors of respiratory disease pathogenesis. N6 methylation of RNA is also emerging to be a critical mechanism of epigenetic alteration and upregulation of immune responses to environmental stressors. Epigenetic changes such as DNA methylation are stable and often long term and cause onset of chronic lung conditions. These epigenetic pathways are also being utilized for therapeutic intervention in several lung conditions.
Article
Full-text available
Purpose To explore the molecular mechanism and search for candidate lncRNA and mRNA associated with pyroptosis in the gene expression profile of LPS-induced acute lung injury (ALI). Methods We investigated lncRNA and mRNA expression in lipopolysaccharide (LPS)-induced ALI at an early stage. RNA sequencing (RNA-Seq) was carried out to analyze lncRNA and mRNA expression profiles between the LPS-induced and control groups. We used bioinformatics analysis to predict target genes of early differential lncRNAs among obtained the differential mRNAs. Results A total of 78 lncRNAs and 248 mRNAs were upregulated at 2 hours and downregulated at 9 hours, and 21 lncRNAs and 107 mRNAs were downregulated at 2 and upregulated at 9 hours in early ALI models. We predicted 7 cis-and trans-regulated target genes of the top 20 lncRNAs. Gene Ontology (GO) analysis indicated that the target genes for the screened lncRNAs were most enriched in three-terms: regulation of protein serine/threonine kinase activity, pertussis, and cellular response to LPS. Additionally, target genes of lncRNAs were the top three enriched in pertussis, osteoclast differentiation, and cAMP signaling pathways with Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. We also identified vital mRNAs and lncRNAs. Protein-protein interaction (PPI) network analysis suggested that Tnf, Jun, and Atf3 were the top three key genes. Hub lncRNA4344 (NONRATT004344.2) and cis-regulated target mRNA (NLRP3) were validated in vitro. Finally, luciferase assay results confirmed that lncRNA4344 sponged miR‐138-5p to promote pyroptosis in inflammatory responses to LPS‐induced acute lung injury by targeting NLRP3. Conclusion Based on analysis of lncRNA and mRNA expression profiles by RNA-Seq and experimental verification, this study is the first to reveal that lncRNA4344 sponged miR‐138-5p to promote pyroptosis in inflammatory responses of LPS‐induced acute lung injury by targeting NLRP3. These newly identified lncRNA, miRNA, and mRNA might be novel potential targets for early treatment and prevention in early ALI.
Article
Full-text available
Background ALI/ARDS is a severe lung injury leading to refractory respiratory failure, accounting for high morbidity and mortality. However, therapeutic approaches are rather limited. Targeting long non-coding RNA MALAT1 and microRNA miR-181a-5p might be potential option for ALI/ARDS intervention. Objective We aimed to investigate the role of MALAT and miR-181a-5p in the pathogenesis of ALI/ARDS, and test the therapeutic effects of targeting MALAT and miR-181a-5p for ALI/ARDS intervention in vitro. Methods MALAT1 and miR-181a-5p levels were measured in plasma from ALI/ARDS patients. In vitro human pulmonary microvascular endothelial cell (HPMEC) injury was induced by LPS treatment, and molecular targets of MALAT1 and miR-181a-5p were explored by molecular biology approaches, mainly focusing on cell apoptosis and vascular inflammation. Interaction between MALAT1 and miR-181a-5p was also detected. Finally, the effects of targeting MALAT1 and miR-181a-5p for ALI/ARDS intervention were validated in a rat ALI/ARDS model. Results MALAT1 upregulation and miR-181a-5p downregulation were observed in ALI/ARDS patients. Transfection of mimic miR-181a-5p into HPMECs revealed decreased Fas and apoptosis, along with reduced inflammatory factors. Fas was proved to be a direct target of miR-181a-5p. Similar effects were also present upon MALAT1 knockdown. As for the interaction between MALAT1 and miR-181a-5p, MALAT1 knockdown increased miR-181a-5p expression. Knocking down of MALAT1 and miR-181a-5p could both improve the outcome in ALI/ARDS rats. Conclusion MALAT1 antagonism or miR-181a-5p could both be potential therapeutic strategies for ALI/ARDS. Mechanistically, miR-181a-5p directly inhibits Fas and apoptosis, along with reduced inflammation. MALAT1 negatively regulates miR-181a-5p.
Article
Full-text available
Acute lung injury (ALI), a common complication of sepsis, is characterized by the impairment and injury of pulmonary function. The nuclear factor kappa-B (NF-κB) pathway is activated in ALI. Tripartite motif-containing 37 (TRIM37) can activate the NF-κB pathway and is closely associated with inflammation. The purpose of our study is to reveal the role of TRIM37 in ALI. The present study revealed that TRIM37 presented high levels in lung tissues of ALI mice, and knockdown of TRIM37 alleviated lipopolysaccharide (LPS)-induced lung injury, inflammatory response, and cell apoptosis in vivo. In addition, knockdown of TRIM37 inhibited the inflammatory response, and cell apoptosis of LPS-treated WI-38 cells. Mechanistically, miR-944 was identified to bind with and negatively regulate TRIM37. Furthermore, NEAT1 was indicated to act as a competitive endogenous RNA to promote TRIM37 expression by sequestering miR-944. Detailly, NEAT1 bound with miR-944, negatively modulated miR-944 expression, and positively modulated TRIM37 expression. The rescue assays suggested that overexpression of TRIM37 rescued the influence of NEAT1 knockdown on cell apoptosis and inflammatory response. Overall, NEAT1 facilitated cell apoptosis and inflammatory response of WI-38 cells by the miR-944/TRIM37 axis in sepsis-induced ALI, implying that NEAT1 may provide a novel insight for the treatment of sepsis-induced ALI.
Article
Full-text available
Long non-coding RNAs (lncRNAs) are emerging regulators of pathophysiological processes including atherosclerosis. Using RNA-seq profiling of the intima of lesions, here we identify a macrophage-specific lncRNA MAARS (Macrophage-Associated Atherosclerosis lncRNA Sequence). Aortic intima expression of MAARS increases by 270-fold with atherosclerotic progression and decreases with regression by 60%. MAARS knockdown reduces atherosclerotic lesion formation by 52% in LDLR−/− mice, largely independent of effects on lipid profile and inflammation, but rather by decreasing macrophage apoptosis and increasing efferocytosis in the vessel wall. MAARS interacts with HuR/ELAVL1, an RNA-binding protein and important regulator of apoptosis. Overexpression and knockdown studies verified MAARS as a critical regulator of macrophage apoptosis and efferocytosis in vitro, in an HuR-dependent manner. Mechanistically, MAARS knockdown alters HuR cytosolic shuttling, regulating HuR targets such as p53, p27, Caspase-9, and BCL2. These findings establish a mechanism by which a macrophage-specific lncRNA interacting with HuR regulates apoptosis, with implications for a broad range of vascular disease states.
Article
Full-text available
Previous studies have reported the important roles of long non-coding RNAs (lncRNAs) in acute respiratory distress syndrome (ARDS). Here, we focus on the role and regulatory mechanism of lncRNA SNHG5 in ARDS. LPS was used to induce mice to establish ARDS model in vivo and to induce A549 cells to establish ARDS model in vitro. qRT-PCR was performed to determine the expressions of SNHG5, miR-205, and inflammatory cytokines. MTT assay was applied to detect cell viability. Dual-luciferase reporter (DLR) assay was performed to test the interactions among SNHG5, miR-205 and COMMD1. Western blot was used to detect the protein expression of COMMD1. Lung injury was evaluated by evaluating the score of lung injury, lung wet/dry weight ratio, and myeloperoxidase (MPO) activity. SNHG5 was downregulated, while miR-205 was upregulated in the serum of ARDS patients and lung tissues of LPS-induced mice. Upregulation of SNHG5 or down-regulation of miR-205 inhibited inflammation and promoted the viability of LPS-induced A549 cells. SNHG5 alleviated the lung injury of ARDS mice. MiR-205 was a target of SNHG5 and inversely correlated with SNHG5. COMMD1 was targeted by miR-205, and was positively regulated by SNHG5. MiR-205 mimics or sh-COMMD1 reversed the promoting effect of SNHG5 on cell viability and the suppressing effect of SNHG5 on inflammation in cellular model of ARDS. Meantime, miR-205 mimics reversed the relieving effect of SNHG5 on lung injury in mouse model of ARDS. SNHG5 acted as a sponge for miR-205 to ameliorate LPS-induced ARDS by regulating COMMD1.
Article
Full-text available
Long non‑coding RNA (lncRNA) cancer susceptibility candidate 2 (CASC2) has been reported to exert an important role in acute lung injury (ALI). The present study aimed to investigate the potential underlying mechanism of CASC2 in ALI progression. Reverse transcription‑quantitative PCR was conducted to examine the expression of CASC2, microRNA (miR/miRNA)‑27b and TGF‑β activated kinase 1 and MAP3K7‑binding protein 2 (TAB2) in A549 cells. Cell viability and apoptosis were analyzed using 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide assay and flow cytometry. Enzyme‑linked immunosorbent assay was used to measure the levels of inflammatory‑related cytokines to assess the inflammatory response, including interleukin‑1β (IL‑1β), IL‑6 and tumor necrosis factor α (TNF‑α). The binding sites of miR‑27b in CASC2 or TAB2 were predicted using LncBase or microT‑CDS software, following which dual‑luciferase reporter and RNA binding protein immunoprecipitation assays were performed to confirm the target relationship between miR‑27b and CASC2 or TAB2. The protein expression of TAB2 was detected by western blotting. The decreased viability, and increased apoptosis and inflammatory responses were attenuated by the accumulation of CASC2 in lipopolysaccharide (LPS)‑stimulated A549 cells. CASC2 could directly bind to miR‑27b in A549 cells. CASC2 protected A549 cells from LPS‑triggered injury by downregulating miR‑27b. TAB2 was a target of miR‑27b in A549 cells. The influence of miR‑27b depletion was reversed by the silencing of TAB2 in an ALI cell model. CASC2 could increase the expression of TAB2 by serving as a competing endogenous RNA of miR‑27b in A549 cells. Collectively, the results suggested that CASC2 attenuated LPS‑induced injury in the ALI cell model by modulating the miR‑27b/TAB2 axis.
Article
Sepsis is a systemic inflammatory disease with an unacceptably high mortality rate caused by an infection or trauma that involves both innate and adaptive immune systems. Inflammatory events activate different downstream pathways leading to tissue damage and ultimately multi-organ failure. Mitochondria are responsible for cellular energy, thermoregulation, metabolite biosynthesis, intracellular calcium regulation, and cell death. Damaged mitochondria induce the high Ca2+ influx through mitochondrial calcium uniporter (MCU). It also generates excessive Reactive oxygen species (ROS) and releases mtDNA into the cytoplasm, which causes induction of NLRP3 inflammasome and apoptosis. Mitophagy (Autophagy of damaged mitochondria) controls mitochondrial dynamics and function. It also maintains cellular homeostasis. This review is about how pulmonary sepsis affects the body. What is the aftermath of sepsis, and how mitophagy affects Acute Lung Injury and macrophage polarisation to overcome the damages.
Article
Purpose: This study aimed to explore the regulatory effects and mechanisms of long noncoding RNA H19 (H19) on pulmonary injury, inflammation, and fibrosis of acute respiratory distress syndrome (ARDS). Materials and methods: A rat model of ARDS was established by intratracheal instillation of 2 mg/kg lipopolysaccharide (LPS). qRT-PCR was performed to detect the expression of H19, miR-423-5p, tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, monocyte chemoattractant protein (MCP)-1, and vascular endothelial growth factor (VEGF). Histology score was assessed by hematoxylin-eosin (HE) staining. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of proinflammatory cytokines and the content of VEGF in bronchoalveolar lavage fluid (BALF). The lung fibrosis was evaluated using western blot and Masson's trichrome staining. Dual-luciferase reporter gene assay was used for confirming the relationship between miR-423-5p and H19/FOXA1 in alveolar macrophage cells (MH-S) and alveolar epithelial cells (MLE-12). The regulatory effects of H19/miR-423-5p/FOXA1 axis on the inflammation and fibrosis were further analyzed in LPS-induced MH-S cells. Results: The expression of H19 and FOXA1 was significantly up-regulated, while the expression of miR-423-5p was down-regulated in LPS-induced ARDS rats. Silencing of H19 decreased the mRNA expression of TNF-α, IL-1β, IL-6, MCP-1, and VEGF, the contents of TNF-α, IL-1β, IL-6, and VEGF in BALF, and histology score in LPS-induced ARDS rats. H19 knockdown also reduced the fibrosis scores and the protein expression of vimentin and α-SMA, and elevated the protein expression of E-cadherin in LPS-induced ARDS rats. Furthermore, silencing of miR-423-5p and overexpression of FOXA1 reversed the inhibitory effects of si-H19 on the inflammation and fibrosis of LPS-induced MH-S cells. Conclusions: Silencing of H19 relieved the pulmonary injury, inflammation and fibrosis of LPS-induced ARDS in rats. Silencing of H19 also alleviated the inflammation and fibrosis of LPS-induced MH-S cells through regulating the miR-423-5p/FOXA1 axis.
Article
Long non-coding RNAs (lncRNAs) were considered as accumulated genetic waste until they were found to be gene expression regulators by highly sensitive modern genomics platforms. It is a huge class of non-coding transcripts with an arbitrary length of >200 nucleotides, which has gained much attention in the past few years. Increasing evidence from several experimental studies unraveled the expression of lncRNA linked to immune response and disease progression. However, only a small number of lncRNAs have robust evidence of their function. Differential expression of lncRNAs in different immune cells is also evident. In this review, we focused on how lncRNAs expression assist in shaping immune cells (Macrophages, Dendritic cells, NK cells, T cells, B cells, eosinophils, neutrophils, and microglial cells) function and their response to the diseased conditions. Emerging evidence revealed lncRNAs may serve as key regulators in the innate and adaptive immune response system. So, the molecular mechanism insight into the function of lncRNAs in immune response may contribute to the development of potential therapeutic targets for various disease treatments. Therefore, it is imperative to explore the expression of lncRNAs and understand its relevance associated with the immune system.
Article
Long non-coding RNAs (lncRNAs) are vital regulators of different biological processes during bronchopulmonary dysplasia (BPD). This study was conducted to probe the biological roles of lncRNA CASC2 in the pathogenesis of BPD and neonatal lung injury. Firstly, a hyperoxia-induced mouse model with BPD was established. LncRNAs with differential expression in lung tissues of normal and BPD mice were analyzed by microarray. An adenovirus vector overexpressing CASC2 was constructed and its functions on BPD symptoms in model mice were analyzed. Gain- and loss-of function studies of CASC2 were performed in a bronchial epithelial cell line BEAS-2B to determine its role in cell apoptosis and proliferation under normoxic and hyperoxic conditions. The downstream mechanical molecules of lncRNA CASC2 were predicted on bioinformatics systems and confirmed by luciferase assays. The functional interactions among lncRNA CASC2, miR-194-5p, and CAV1 in BPD were determined by rescue experiments. Consequently, lncRNA CASC2 was found to be poorly expressed in BPD mice. Besides, overexpressed CASC2 was found to relieve the symptoms of BPD in neonatal mice and suppress apoptosis as well as promote proliferation in hyperoxia-induced BEAS-2B cells. Importantly, CASC2 was found to regulate CAV1 expression by competitively binding to miR-194-5p and downregulate the activity of the TGF-β1 signaling pathway, thereby suppressing lung injury. Either miR-194-5p upregulation or CAV1 downregulation blocked the roles of CASC2. To sum up, this study evidenced that CASC2 alleviates hyperoxia-induced lung injury in mouse and cell models with the involvement of a miR-194-5p-CAV1 crosstalk and the TGF-β1 inactivation.