ArticlePDF AvailableLiterature Review

Functional crosstalk between Long non-coding RNAs and the NLRP3 inflammasome in the regulation of diseases

Authors:
  • The Second Affiliated Hospital ofNanchang University

Abstract and Figures

Emerging evidence has indicated that long noncoding RNAs (lncRNAs) are involved in various pathophysiological processes of disease, such as cancer occurrence, viral invasion, and inflammatory damage. The main inflammatory body component, nod-like receptor protein 3 (NLRP3), is the trigger point of inflammatory reactions and inflammation-related diseases and coordinates the body's response to inflammation. At present, increasing evidence shows that the interaction of lncRNAs and the NLRP3 inflammasome plays an important role in the inflammatory response and different diseases. This may be involved in the development and progression of various diseases by activating signalling pathways and a variety of molecular regulatory mechanisms—this article reviews progress in research on the relationship between lncRNAs and the NLRP3 inflammasome under different conditions.
Content may be subject to copyright.
Molecular Immunology 131 (2021) 191–200
Available online 11 January 2021
0161-5890/© 2021 Elsevier Ltd. All rights reserved.
Review
Functional crosstalk between Long non-coding RNAs and the NLRP3
inammasome in the regulation of diseases
Deqiang Luo
a
,
b
, Fen Liu
a
, Jianguo Zhang
a
, Qiang Shao
a
, Wenqiang Tao
a
, Rui Xiao
a
, Wei Dai
b
,
Kejian Qian
a
,
*
a
Department of Intensive Care Unit, The First Afliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Dong Lake District, Nanchang, Jiangxi Province,
330000, China
b
Department of Intensive Care Unit, the Fifth Peoples Hospital of Shangrao City, No. 1 Jiannan Road, Xin Zhou District, Shangrao 334000, China
ARTICLE INFO
Keywords:
Inammatory inammasome
Inammatory diseases
NLRP3
Long-chain non-coding RNA
ABSTRACT
Emerging evidence has indicated that long noncoding RNAs (lncRNAs) are involved in various pathophysio-
logical processes of disease, such as cancer occurrence, viral invasion, and inammatory damage. The main
inammatory body component, nod-like receptor protein 3 (NLRP3), is the trigger point of inammatory re-
actions and inammation-related diseases and coordinates the bodys response to inammation. At present,
increasing evidence shows that the interaction of lncRNAs and the NLRP3 inammasome plays an important role
in the inammatory response and different diseases. This may be involved in the development and progression of
various diseases by activating signalling pathways and a variety of molecular regulatory mechanismsthis
article reviews progress in research on the relationship between lncRNAs and the NLRP3 inammasome under
different conditions.
1. Introduction
Inammation underlies numerous pathological processes involving
the action of various inammatory cells and inammatory activating
factors and serves as the bodys initial adaptive response to external
stimuli, stress, tumours, metabolic diseases, and allergies(Medzhitov,
2008, 2010). Long noncoding RNAs (lncRNAs) do not encode any pro-
teins longer than 200 nucleotides and were previously regarded as
non-functional molecular noise RNA(Gutschner and Diederichs, 2012;
Mendell et al., 2004). However, certain annotated lncRNAs that bind to
ribosomes and contain a translation region that can translate peptides
have gradually been discovered(Nam et al., 2016). LncRNAs regulate
related genes by directly or indirectly binding to specic DNA/RNA or
protein sites in the cytoplasm or nucleus(Moura et al., 2014). In contrast
to miRNAs and circRNAs, lncRNAs have well-conserved sequences and
structures and cannot participate in various regulatory mechanisms
(Kornfeld and Brüning, 2014). In different biological environments, they
perform particular functions and regulate complex natural mechanism;
e.g., acting (i) as scaffolds to polymerize protein complexs, (ii) as
molecular sponges for ceRNA protagonists, (iii) as host genes of various
RNAs (iv) to guide the mRNA degradation process, and isolate of tran-
scription factors for DNA regulation and epigenetic regulation of chro-
matin conformation, and (v) as protectors of mRNA through the
prevention of degradation by binding miRNA(Hadjicharalambous and
Lindsay, 2019; Kornfeld and Brüning, 2014; Morlando and Fatica,
2018). Larg high-quality sequencing platform data sets have indicated
that the emergence of many inammatory genes and the activation of
signalling pathways found abnormal lncRNA expression (Liao et al.,
2018). Pyridine-containing domain 3 (NLRP3), also called the inam-
masome, is a multimolecular protein complex that performs a specic
function for the cytoplasm in the hosts innate immunity by secreting
inammatory factors and promoting maturation, such as for inter-
leukin-1β (IL-1β) and interleukin-18 (IL-18)(He et al., 2016a; Zhou et al.,
2011). Seasonable activation of the NLRP3 inammasome has a positive
effect on the body; excessive activation of the NLRP3 inammasome
causes programmed cell death termed ‘pyroptosisvia the secretion of
large amounts of proinammatory cytokines and triggers a highly in-
ammatory form of caspase-1(Van Opdenbosch and Lamkan, 2019).
* Corresponding author at: Department of Intensive Care Unit, The First Afliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Dong Lake
District, Nanchang City, Jiangxi Province, 330000, China.
E-mail addresses: yydlt@163.com (D. Luo), 807792302@qq.com (F. Liu), drzhangjianguo@163.com (J. Zhang), 372661438@qq.com (Q. Shao), twqnc@139.com
(W. Tao), 421583336@qq.com (R. Xiao), 415556881@qq.com (W. Dai), ndyfyicu@email.ncu.edu.cn (K. Qian).
Contents lists available at ScienceDirect
Molecular Immunology
journal homepage: www.elsevier.com/locate/molimm
https://doi.org/10.1016/j.molimm.2020.12.038
Received 2 May 2020; Received in revised form 22 November 2020; Accepted 30 December 2020
Molecular Immunology 131 (2021) 191–200
192
There are usually two signal modes of NLRP3 inammasome activation:
the hosts crucial immune response to external stimuli regulates the
main checkpoints of the two signalling pathways(He et al., 2016a;
Swanson et al., 2019). A recent explosion in studies on the regulation of
the interaction between lncRNAs and the NLRP3 inammasome in the
activation of inammatory pathways has brought to the forefront new
insights into the prophylaxis, diagnosis, and treatment of various dis-
eases(Bordon, 2019; Haque et al., 2020; Ma et al., 2019; Nie et al., 2019;
Song et al., 2019a; Zhang et al., 2020a). Our article reviews recent
high-throughput studies of the relationship between lncRNAs and
NLRP3 inammasome under different conditions.
2. The molecular structure and biological function of lncRNAs
and the NLRP3 inammasome
2.1. The molecular structure and the role of lncRNAs
LncRNAs are poorly conserved and lack complete ORFs, longer than
200 nt(Quinn and Chang, 2016). As with messenger RNA (mRNA),
transcription for most lncRNAs is mediated by RNA polymerase II; this
transcription mechanism is completed by 5-terminal capping, Pol II
occupancy, histone modications, and polyadenylation. RNA polymer-
ase III can mediate part of lncRNA transcription(Pagano et al., 2007).
The traditional understanding regards lncRNAs as having no coding
function and being more poorly conserved than mRNAs, but they have
been shown to exert a productive regulatory function(Pagano et al.,
2007). Many different levels of lncRNA structure play specic roles in
every level of gene expression in particular environments. The
well-known star molecule nuclear lncRNA MALAT1 has a standard
genomic structure; RNA polymerase II regulates MALAT1 to form a
transcript-like 3end of a tRNA-like structure. Although no functional
units can be found in the primary structure of lncRNAs, there are many
hairpin structures in their sublevel construction. For example, lncRNA
SPRY4-IT1 originates from an intron of the SPRY4 gene, whose sec-
ondary structure is highly conserved in melanocytoma(Khaitan et al.,
2011). Therefore, we should gradually shift the dogma concept of mo-
lecular biology to lncRNAs, which only encode proteins. LncRNAs
participate in the regulation of protein-coding and epigenetic genes by
controlling processes such as RNA epigenetic modications, transcrip-
tional gene silencing, and posttranscriptional gene regulation(Martens
et al., 2004)(Fig. 1)(Salviano-Silva et al., 2018). LncRNAs suppress gene
transcription and expression by acting on encoding genes upstream or
downstream of promoter regions by various mechanisms, such as
interfering with RNA binding sites or as transcription factors that
interact with sequence-matching promoters of mRNA (Engreitz et al.,
2016a). LncRNAs also alter DNA structure and its transcription mech-
anisms through DNA methylation, the formation of protein trimers, and
heterochromatin formation, leading to suppressor disease gene silencing
(Wu et al., 2015b; Zhou et al., 2015). On the other hand, lncRNAs inhibit
the expression of disease-associated genes by targeting mRNA
Fig. 1. Long noncoding RNAs (lncRNAs) genomic diversity location relative to protein-coding genes and regulatory mechanisms of lncRNAs in the various cellular
substructures. (A) Nomenclature of different lncRNA genes (purple ellipses) according to their genomic locations relative to nearby coding genes (blue curves) or
exons of coding genes (blue ovals). (B) The molecular mechanisms of lncRNAs: (B1) as a part of the Barr body of somatic cells, such as lncRNA Xist; (B2) as enhancer
RNAs, playing an RNA-mediated role in cis or in trans; (B3) as decoys to structural proteins, affecting their binding to DNA sites, such as remodelling transcription
factors inuencing chromatin structure; (B4) as molecular guided signals, regulating gene expression through signalling pathways; (B5) inducing chromatin
remodelling at specic genomic loci; (B6) as molecular scaffolds, assembling different proteins and forming new protein complexes, which regulate gene tran-
scription; (B7) regulating catalytic activity of enzymes (such as kinases) and modulating their signal pathways; (B8) forming new transcripts by splicing initial
transcripts; (B9) as molecular sponges competing for binding microRNAs (miRNAs), inhibiting their function; (B10) targeting proteins and forming new molecular
complexes that can change their subcellular location; (B11) binding messenger RNAs (mRNAs) and promoting their degradation to improve their function. In
addition, lncRNAs can (B12) produce results by transferring to other cells via extracellular vesicles (EVs).
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
193
transcription factors or activating extra proteins to promote the
expression of target genes(Engreitz et al., 2016b). In addition, lncRNAs
also interact with mRNAs in the cytoplasm or nucleus by various bio-
logical regulatory mechanisms, such as by modulating their activity to
bind to small RNAs, affecting the chromatin structure as with chromo-
some circulation, and regulating subcellular localization (B¨
ohmdorfer
and Wierzbicki, 2015; Willingham et al., 2005).
2.2. The structure and function of the NLRP3 inammasome
2.2.1. The molecular structure of the inammasome
The NLRP3 inammasome is a protein whose gene contains nine
coding exons and located on chromosome 1q44. NLRP3 is found in the
cytoplasm, and is an essential component of the inammatory signalling
complex responding to microbes or external stimuli(Freeman et al.,
2017; Lamkan and Kanneganti, 2010). The well-known NLRP3
inammasome complex mainly includes three components: the ASC
adaptor, pro-caspase-1 effector, and NLRP3 receptor (Swanson et al.,
2019) (Fig. 2). As a tripartite protein, its core functional structure in-
cludes an amino-terminal pyrin domain (PYD), a central NACHT domain
(domain present in NAIP, CIITA, HET-E, and TP1), and a
carboxy-terminal leucine-rich repeat domain (LRR domain). NLRP3
self-association and function are closely related to the ATPase activity of
the NACHT domain(Duncan et al., 2007), which is thought to induce
autoinhibition. ASC contains two protein interaction domains: PYD and
the carboxy-terminal caspase recruitment domain (CARD). Caspase-1
has a sizeable central catalytic domain (p20), an amino-terminal
CARD, and a carboxy-terminal small catalytic subunit domain (p10).
Given stimulation, homotypic interactions between the NACHT domains
of NLRP3 are oligomerized (Fig. 1B). Next, the oligomeric NLRP3
interacting through PYD-PYD binding ASCs and nucleates helical ASC
lament formation. The familiar ASC speck is the coalescence of mul-
tiple ASC laments into a single macromolecular focal point(Lu et al.,
2014; Schmidt et al., 2016). Assembled ASCs enable proximity-induced
caspase-1 self-cleavage and activation after recruiting caspase-1 through
CARDCARD interactions. Caspase-1 can increase proteolytic activity
after binding on the p33 - p10 complex generated from ASC self--
cleavage(Wang et al., 2019b). Through interaction between CARD and
p20, ASC releases the p20p10 heterotetramer that terminates its
protease activity because it is unstable in cells (Wang et al., 2019b).
Recently, NIMA-related kinase 7 (NEK7), a known serine-threonine ki-
nase associated with mitosis, was found to play an essential role in
NLRP3 inammasome activation (He et al., 2016b). Only the inam-
masome sensor NLRC4 (including NOD, LRR, and CARD 4) and
interferon-inducing proteins AIM2, and NEK7 interact specically with
NLRP3. Under inammasome activation, the increased NEK7NLRP3
interaction leads to the assembly of NEK7 and NLRP3 into a complex
that is essential for caspase-1 activation and ASC speck formation(Shi
et al., 2016). Therefore, NEK7 seems to be a unique core component of
the NLRP3 inammasome.
2.2.2. The signalling pathway of inammasome activation
The activity of NLRP3 can be regulated by carboxy-terminal LRR self-
inhibitory properties. NLRP3 can sense microbial ligands and endoge-
nous alarm proteins. (e.g., via the amino-terminal PYD domain of
NLRP3) and can allow complementary pairing with the PYD domain of
ASC, the core NACHT domain responsible for self-oligomerization
(Lamkan and Dixit, 2009). To date, we know that the NLRP3 inam-
masome performs molecular functions in the cytoplasm of multiple
mammalian cells, such as inammation and immune-related cells T
cells and B cells, dendritic cells, monocytes, macrophages, granulocytes,
etc.(Ahmad et al., 2013; Dombrowski et al., 2011; Sandanger et al.,
2013). The assembly of the NLRP3 inammasome is triggered by a series
of internal and external activators, including PAMPs, such as dsRNA,
viral RNA, LPS, MDP, decomposition products of the cell wall compo-
nent peptidoglycan, and bacteria(Kanneganti et al., 2006; Xu et al.,
2013). In addition to the most studied PAMPs, the NLRP3 inammasome
is activated by diverse compounds, toxins, microorganisms, the
little-known maltoxin, and Nigerian toxic aerosols containing Bacillus
brevis(McCoy et al., 2010). There are two critical distinct activation steps
for the NLRP3 inammasome(Ozaki et al., 2015; Sutterwala et al., 2014;
Zhong et al., 2013) (Fig. 2). The rst step is NLRP3 inammasome
priming: nuclear factor kappa B (NF-κB) signalling is initially activated
when TLRs recognize many PAMPs or DAMPs, and then the priming
process can regulate the transcription of inactive NLRP3, pro-IL-1β, and
pro-IL-18(Bauernfeind et al., 2009; Franchi et al., 2012). This priming of
the NLRP3 inammasome is often induced by lipopolysaccharides in
Vetro(Park et al., 2015). The second step is the activation of the NLRP3
Fig. 2. A Two-Signal Activated Model for the
NLRP3 inammasome. Multiple microbial mol-
ecules or inammatory cytokines stimulate the
priming signal model (Model 1, left). The acti-
vation of NF-κB and subsequent increases in the
activity of NLRP3, caspase-1, pro-interleukin-1β
(pro-IL-1β), fas-mediated death domain protein
(FADD), etc., participate in the priming step by
regulating the NF-κB/NLRP3/IL-1β axis. Acti-
vation of the NLRP3 inammasome results in
multiple posttranslational modications and
interacting receptor ligands. The signal activa-
tion model (Model 2, right) is motivated by
various endogenous and exogenous stimuli,
including small RNA viruses, ATP, pore-forming
toxins, and particulate matter. The upregulation
of NLRP3 induces the activation of diverse
molecular and cellular signalling pathways,
including mitochondrial dysfunction, ionic ux,
reactive oxygen species (ROS) generation, and
lysosomal damage, which are involved in the
activation of the NLRP3 inammasome.
BRCA1/BRCA2, BRCC3-containing complex
subunit 3; JUN N-terminal kinase 1; IL-1R, IL-1β
receptor; JNK1, JUN N-terminal kinase 1; PKD,
protein kinase D; TLR, toll-like receptor; TNFR,
tumour necrosis factor receptor.
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
194
inammasome via ASC assembly, oligomerization, and binding with
pro-caspase-1 to form a complex by a different stimulus. Then, the
activated membrane sensor of the NLRP3 inammasome induces the
structural conversion of pro-caspase-1 to functional caspase-1, which
results in the secretion of a large number of inammatory end products
IL-18 and IL-1β(Kim et al., 2015; Ozaki et al., 2015). In recent years, it
has been recognized that the NLRP3 inammasome is regulated by
noncoding RNAs (miRNAs, lncRNAs, circRNAs, etc.) in the post-
transcriptional regulation of gene expression.
2.3. Pyroptosis and NLRP3
The inammasome secretes and induces the maturation of IL-1β/18,
and its activation triggers a form of inammatory cell death called
pyroptosis(Fig. 2). Cell rupture caused by pyroptosis also promotes
immature inammatory cytokines such as IL-1β(Heid et al., 2013).
IL-1β/18 and pyrokinesis initiate inammatory or anti-inammatory
effects in the body. More importantly, the release of IL-1β is indepen-
dent of the anti-inammatory effects of pyroptosis(Liu et al., 2016).
Pyroptosis and the secretion of cytokines (IL-1β and IL-18) should be
considered together as a potential inammatory mechanism in the
analysis of inammatory diseases. In the last decade, essential ndings
have revealed that gasdermin D (GSDMD) plays a mediating role in
pyroptosis. GSDMD features an amino-terminal cell death domain
(GSDMD
Nterm
), a central short linker region, and a carboxy-terminal
autoinhibition domain. GSDMD is released from caspase-1 cleavage to
escape intramolecular inhibition by removal of its carboxyl terminus(Shi
et al., 2015). GSDMD
Nterm
can oligomerize phosphatidylserine in the
inner leaet of the cell membrane and insert into the plasma membrane,
forming a 1014 nm pore containing 16 symmetrical protomers, thus
killing cells from within(Ding et al., 2016). Additionally, GSDMD
Nterm
exerts bactericidal activity when bound to cardiolipin in outer and inner
bacterial membranes. The exact mechanism of this direct bactericidal
activity in the infection process has not yet been investigated. Activated
NLRP3 triggers cardiolipin phenomena in the inner and outer mito-
chondrial membranes. However, it is unclear whether GSDMD
Nterm
can
migrate into mitochondria by binding to mitochondrial cardiolipin.
Another pathway of GSDMD-mediated pyroptosis is through uncon-
ventional secretion of IL-1β and IL-18 release(Lieberman et al., 2019).
Pyroptosis plays a vital role in the secretion of IL-1
α
in the
calmodulin-processed form, although the process does not determine its
biological activity. The ndings described above reveal that GSDMD is
induced biochemically to undergo pyroptosis downstream of inam-
masome activation. Additionally, due to the critical role of caspase -1 in
IL-1β processing and pyroptosis, caspase -8 functions in the induction of
IL-1β and IL-18 maturation and cell death in unregulated pathways
(Antonopoulos et al., 2015). However, some studies have conrmed
caspase-8-mediated maturation of pro-IL-1β and pro-IL-18, which can
occur through NLRP3-independent and NLRP3-dependent pathways.
Together, these studies suggest that pyroptosis exerts a unique biological
effect that promotes inammation and inammasomes in the body.
2.4. LncRNAs and NLRP3 mRNA share similar biogenesis pathways
Similar to histone markers of NLRP3 mRNA, the lncRNA transcrip-
tion is performed by NAPII. Most of its structural changes include
capping at the 3 ends and 5 ends with 7-methylguanosine, poly-
adenylation, and splicing(Derrien et al., 2012; Hon et al., 2017). Studies
have shown that Nonpolyadenylated lncRNAs, enhancer transcription,
and enhancer RNA may play regulatory roles during transcription, and
are not simply transcriptional noise(Natoli and Andrau, 2012). These
mechanisms are achieved by cleavage of ribonuclease P to generate
mature 3ends, forming snoRNAprotein complexes, or capping circular
structures at their ends(Memczak et al., 2013; Yang Zhang et al., 2014).
The important difference between lncRNAs and mRNAs is that lncRNAs
are poorly conserved due to their lack of long ORFs and low
protein-coding potential (Derrien et al., 2012). However, interestingly,
Ruiz-Orera et al. found that lncRNAs show very similar characteristics
and sequence constraints to functional coding genes and may play an
active role in de novo protein evolution(Ruiz-Orera et al., 2014).
3. The regulatory relationship between lncRNAs and NLRP3
inammasomes
The central rule of molecular biology is the transcription of genetic
information from DNA to RNA and its translation into proteins. LncRNAs
have been found to function in inammation and immune-related cells,
such as T lymphocytes, B lymphocytes, and dendritic cells (DCS)(Geng
and Tan, 2016). LncRNAs participate in all cell function stages,
including cell differentiation, development, metabolism, and immunity
(Wu et al., 2015a). Numerous high-quality research data sets have
indicated that lncRNAs are engaged in various pathophysiological pro-
cesses, such as pain, tumours, ageing, neurodegenerative diseases, and
cardiovascular diseases via NLRP3 inammasomes(Guo et al., 2019; Xu
et al., 2018; Zhang et al., 2019c). Proteins related to the pyroptosis
signalling pathway are activated via a direct or indirect mechanism,
interact with pathogens, and regulate body immune responses(Bordon,
2019; Haque et al., 2020).
3.1. lncRNAs regulate the expression of the NLRP3 inammasome
through the ceRNA mechanism
Research has increasingly focused on the complex interaction be-
tween NLRP3 and various RNA species, including diverse noncoding
RNA (miRNAs, circular RNAs, and lncRNAs) and protein-coding mRNAs
(Hadjicharalambous and Lindsay, 2019). LncRNAs can compete with
NLRP3 to bind miRNA sponges to regulate the expression of target
genes. The NLRP3 inammasome is a miRNA target gene in various
diseases(Rituparno et al., 2014; Tay et al., 2014)(Fig. 3A). The ceRNA
mechanism allows lncRNA transcripts and NLRP3 to regulate and
communicate to affect mRNA expression by competing for natural
microRNA sponges, which have no protein-coding ability but regulates
NLRP3 mRNA expression through posttranscriptional regulation
degradation(Jiang et al., 2017; Wang et al., 2019a). The discovery of
this novel RNA crosstalk interaction mechanism will help gain signi-
cant insight into the lncRNA regulatory mechanism and impact human
diseases occurrence and development. It has been reported that
miR-135a reversed lncRNA DANCR regulation in the entire process of
promoting pancreatic cancer cells, which was attained by degrading the
downstream protein of NLRP3 in the cytoplasm(Y. Tang et al., 2019). In
ischaemia-reperfusion-injured hearts, lncRNA MALAT1 sponges
miR-133 to increase transcriptional expression of NLRP3 inammasome
(Yu et al., 2018). Furthermore, MALAT1 also competitively binds
miR-22 to affect NLRP3 expression in high glucose-induced human
endothelial cell pyroptosis(Song et al., 2019a). In those patients with
laryngeal squamous cell carcinoma (LSCC), lncRNA RGMB-AS1 could
make it competitive for molecular sponge miR-22 to attenuate NLRP3
transcription expression, and its reduction can suppress LSCC progres-
sion. Therefore, lncRNA RGMB-AS1 may become a new focus in the
diagnosis, treatment, and prognosis of LSCC because of its progress
through the ceRNA mechanism(Xu and Xi, 2019). LncRNA 00339 can
regulate the miR-22-3p/NLRP3 axis to promote renal tubular epithelial
cell pyrocytosis. Mechanistically, LINC00339 upregulates NLRP3
expression by sponging miR-22-3p, which acts as a competitive endog-
enous RNA(Song et al., 2019b). Zhang et al. have conrmed that lncRNA
NEAT1 was able to compete for binding to miR-3076-3p as a molecular
decoy of the NLRP3 inammasome by ceRNA. At the same time, Neat1
was found not only to bind to pro-caspase-1 but also facilitate the as-
sembly of NLRP3, NLRC4, and AIM2 inammasomes in mouse macro-
phages; Neat1 also was able to induce the maturation of IL-1β and cell
pyroptosis by activating caspase-1; thus a series of effects promoted the
expression of tolerogenic phenotype in DCs(Song et al., 2019b). In
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
195
Parkinsons disease, Long noncoding RNA SNHG1 contributes to neu-
roinammation by modulating the lncRNA SNHG1/miR-7/NLRP3
pathway(Cao et al., 2018). Certain drugs, like melatonin, can affect
the expression of lncRNA MEG3, leading to inhibition of endothelial cell
pyroptosis via regulation of the miR-223/NLRP3 signalling axis(Zhang
et al., 2018). Experimental evidence has conrmed that the lncRNAs
described above play an integral role in the regulation of gene expres-
sion of both competitions for miRNAs, small non-coding regulators, and
mRNAs(Salmena et al., 2011). Two new noncoding RNA participants,
lncRNAs, and miRNAs further increase RNA interactions through the
ceRNA mechanism(Ergun and Oztuzcu, 2015). Related literature has
analysed and veried the mechanism of ceRNA interaction through
bioinformatics and experimental methods, and this indicates that the
role of ceRNA is one of the transcriptional regulatory functions of
lncRNAs, which is even a different or complementary mechanism from
traditional protein-coding function(Tay et al., 2014; Yang et al., 2016).
3.2. LncRNAs regulate the expression of NLRP3 via the NF-κB/NLRP3
inammasome pathway
There are two studies about lncRNAs that can regulate the activation
of the NLRP3 inammasome by interacting with NF-κB, which is a
crucial participant in all of the inammatory phases in diabetic ne-
phropathy (DN) and inammatory diseases(Samra et al., 2016; Yang
et al., 2014). These results suggest that the lncRNA/NF-κB/NLRP3
inammasome signalling pathway may be a novel functional mecha-
nism in many diseases (Fig. 3B). LncRNA Gm4419 upregulates proin-
ammatory cytokines via the lncRNA/NF-KB/NLRP3 central
inammatory axis in mesangial cells (MCs) of DN(Yi et al., 2017). In the
present study about the bovine mammary epithelial cells and the in-
ammatory response, the NF-κB pathway was activated to mediate
inammation by enhancing the expression of XIST, which was part of a
negative feedback loop in the regulation of the NF-κB/NLRP3 pathway
(Ma et al., 2019). In addition, XIST inhibits NF-κB phosphorylation and
NLRP3 inammatory production through a pathway by inhibiting E. coli
or S. aureus-induced activity(Ma et al., 2019). In the latest article, Zhang
et al. found that lncRNA-far1 was involved in NLRP3
inammasome-mediated pyroptosis and BDL- and CCl4-induced proin-
ammatory of M1 macrophages(Zhang et al., 2020b). Furthermore, the
results of animal experiments have revealed that silencing lnc-far1 also
signicantly inhibited NLRP3 inammasome-mediated pyroptosis acti-
vation of macrophages(Zhang et al., 2020b).
3.3. LncRNAs regulate the expression of NLRP3 by direct interaction of
lncRNAs and proteins
LncRNAs regulate the expression of NLRP3 by inhibiting the encoded
TXNIP, constituting a newly discovered regulatory mechanism. TXNIP is
a crucial intermediary mediator linking the regulation of lncRNA and
NLRP3 inammasome activation in oxidative- and endoplasmic reticu-
lum (ER) stress-related disease(Anthony and Wek, 2012)(Fig. 3C).
LncRNA Gm15441 expression can suppress its antisense transcript to
modulate the expression of TXNIP. TXNIP can be encoded by upregu-
lating lncRNA Gm15441, which results in a downregulated expression of
TXNIP activating the NLRP3 inammasome followed by cleavage of
caspase-1, in turn, promoting pro-IL-1β to induce the maturation of IL-1β
(Brocker et al., 2019). The study shows that the upregulated lncRNA
gene Gm15441 can directly bind the promoter of nuclear
receptor-peroxisome proliferator-activated receptor alpha (PPARA) to
activate its ligand. Thus, we understand this regulatory mechanism by
which liver PPARA directly up-regulates lncRNA Gm15441 expression
by inhibiting Txnip encoding, in turn, repressing the activation of the
NLRP3 inammasome during metabolic stress(Brocker et al., 2019). A
recent study result shows that lncRNA Gm14205 regulate the expression
of NLRP3 by inhibiting the encoded OXTR by direct interaction of
lncRNA and protein(Zhu and Tang, 2020). The results of these studies
demonstrate an innovative indirect, interactive mechanism between
lncRNAs and NLRP3, which supports the benecial effects of intestinal
fasting during inammation to reduce food irritation.
3.4. LncRNAs promote the assembly of the NLRP3 inammasome
LncRNAs function as regulators in promoting various inammatory
factors in the cytoplasm under certain stimulatory conditions(Fig. 3D).
NEAT1 also plays an essential role in regulating the expression of che-
mokines and inammatory factors (including IL-6 and CXCL10) by
indirectly regulating NLRP3 activation (Zhang et al., 2016). We note
that many stimuli affect the expression of the lncRNA Neat1 and activate
Fig. 3. Regulatory mechanisms between long
non-coding RNAs (lncRNAs) and NLRP3 in the
cytoplasm and nucleus: (A) as a molecular
sponge, inhibiting microRNA (miRNA) function
by competing for binding with them; (B) tar-
geting proteins to exert positive or negative
activity by forming new molecular complexes or
altering their subcellular location; (C) as a
decoy to structural proteins affecting their
binding DNA sites, such as with remodelling
transcription factors inuencing chromatin
structure; (D) as molecular scaffolds, assem-
bling different proteins and forming new pro-
tein complexes, which regulate gene
transcription; (E) binding messenger RNAs
(mRNAs) and promoting their degradation to
alter their effects.
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
196
inammasomes. NLRP3 inammasome assembly and activation can be
induced by many stimuli, including various viruses, extracellular ATP,
by-products of oxygen metabolism (e.g., ROS), potassium efux, frus-
trated phagocytosis, and phagolysosome disruption. (Martinon, 2010).
One current study demonstrated that Neat1 promoted the assembly of
NLRC4, AIM2, and especially NLRP3 inammasomes, thereby activating
inammatory factor expression(Zhang et al., 2019b). Neat1 also en-
hances mature caspase-1 tetramer structural stability and increases its
protease activity by binding target sites. Neat1 likely performs a func-
tion to facilitate recruitment, assembly, stabilization, caspase-1 matu-
ration, and secretion by assembling a largely proteinaceous signalling
platform in activated macrophages(Zhang et al., 2019b), most likely
previously unanticipated. Furthermore, a joint priming event leads to
inammasome assembly, such as by TLRs, which promote the activation
and expression of NLRP3 and pro-IL-1β(Schroder and Tschopp, 2010;
Tang et al., 2012). The interaction between lncRNA Neat1 and NLRP3
inammasomes establishes an indirect regulatory mechanism of size-
able multimeric protein complexes in the cytoplasm.
3.5. LncRNAs decrease NLRP3 expression by inhibiting its promoter
activity
The NLRP3 inammasome is a complex protein that includes a
leucine-rich repeat-containing protein family of intracellular sensors
and a nucleotide-binding domain that can activate the promoter region
via main domain changes(Cassel et al., 2009). Recent research data have
afrmed that the NLRP3 inammasome plays a critical role in the
metastasis and invasion of multiple cancers, such as breast cancer,
melanoma, hepatocellular carcinoma, etc., (Ahmad et al., 2013; Karki
et al., 2017). The long noncoding RNA XLOC_000647 can inhibit NLRP3
inammasome activation by downregulating NLRP3 expression in
pancreatic cancer, which can block pancreatic cancer cell proliferation,
metastasis, epithelial-mesenchymal transition-induced cell invasion,
and EMT(Hu et al., 2018). The up-regulation of lncRNA XLOC_000647
expression can reduce the luciferase activity of the NLRP3 promoter
region to achieve function in vitro. Therefore, the cited study found that
lncRNA XLOC_000647 may inhibit the entire progression of pancreatic
cancer (PC) by negatively regulating the activation of NLRP3. These
results expand our understanding of the mechanisms underlying the
roles of lncRNAs and the NLRP3 inammasome and provide insights
into the clinical diagnosis and treatment of various tumours. This study
provides a new indirect molecular regulatory mechanism in the inter-
action between lncRNAs and the NLRP3 inammasome (Fig. 3E).
4. LncRNAs and NLRP3 inammasomes in various diseases
4.1. LncRNAs and NLRP3 in inammatory diseases
The biological and clinical roles of lncRNAs and the NLRP3 inam-
masome have recently been studied extensively in the inammatory
response and inammatory cascade reaction(Haque et al., 2020; He
et al., 2019). Zhang et al. speculated that the lncRNA Neat1 regulated
activation of inammasomes by assembling NLRP3, NLRC4, and AIM2
components and promoted IL-1β induced pyroptosis by stabilizing the
mature caspase-1 in mouse macrophages(Zhang et al., 2019b). In the
common pathways of various inammatory activation signals, Neat1
usually participates in NLRP3 inammatory activation by changing its
subcellular position to function after being released from the speck and
transferred to the cytoplasm. These results indicate that Neat1 acts as a
common mediator of inammasome stimuli via the direct regulatory
mechanism between Neat1 and the NLRP3 inammasome. In another
study, results showed that NEAT1 knockdown generates a tolerogenic
phenotype in dendritic cells by inhibiting NLRP3 activity(Zhang et al.,
2019a). Mechanistically, RNA sequence analysis showed that silenced
NEAT1 mainly targets miR-3076-3p and affects its expression in DCs.
NEAT1 knockdown can also induce DCs to develop immune tolerance in
organ transplantation models and autoimmune disorder myocarditis.
Silencing of lncRNA XIST can upregulate proinammatory cytokines by
inducing the activation of E. coli or S. aureus-induced In bovine mam-
mary epithelial cell inammatory responses(Ma et al., 2019).
Additionally, XIST knockdown inhibited E. coli or S.aureus-induced
NF-κB phosphorylation and cell proliferation, suppressed cell viability,
and eventually `guided cell apoptosis upon activation of the NLRP3
inammasome. We know that the long non-coding RNA MALAT1 is
closely related to apoptosis. In the pyroptosis of high glucose-induced
endothelial cells, pyroptosis, lncRNA MALAT1 plays a role in the pro-
cess by partially promoting the expression and activation of NLRP3 by
competitively binding miR-22(Song et al., 2019a). These results reveal
that the interaction between lncRNAs and NLRP3 may be widely
involved in the progression of these inammatory diseases.
4.2. lncRNAs and NLRP3 in gastroesophageal cancer
A group of lncRNAs has been found to be associated with cancer by
various means(Arun et al., 2018; Choudhari et al., 2020; de Oliveira
et al., 2019; Spizzo et al., 2012). LncRNAs and NLRP3 are increasingly
involved in tumour formation, metabolism, treatment, targets, and
related models. Hu et al. found that overexpression of XLOC_000647
inuences clinical tumour stage, size, lymph node metastasis, and
overall survival(H. Hu et al., 2018). They also observed that the
expression level of XLOC_000647 was negatively associated with NLRP3
activity in vivo and in vitro. The results show that XLOC_000647
dramatically reduces NLRP3 promoter movement, decreasing cancer
proliferation, invasion, and EMT in vitro. Tang et al. also found that the
lncRNA DANCR could promote cancer cell proliferation, aggression, and
EMT by regulating the miR-135a/NLRP3 axis in pancreatic cancer cells
in animal experiments(Tang et al., 2019). LncRNAs may be a new target
for diagnosis and treatment. In gastric cancer (GC), some investigators
have revealed that lncRNA ADAMTS9-AS2 mediates GC cell pyroptosis
via regulation of the miR-223-3p/NLRP3 axis by acting as a tumour
suppressor and enhancing cisplatin sensitivity. The interaction between
lncRNA RGMB-AS1 and NLRP3 plays specic functional roles in LSCC.
In addition, the high expression of RGMB-AS1 is closely related to
advanced clinicopathological characteristics and poor prognostic out-
comes(Xu and Xi, 2019). The silencing of lncRNA RGMB-AS1 can sponge
miR-22 and upregulate NLRP3 inammasome expression levels, which
inhibits the proliferation and invasion of LSCC cells in vitro and sup-
presses tumour growth in vivo(Xu and Xi, 2019). Taken together,
revealing the mechanism of interaction of lncRNA and NLRP3 in
digestive cancer may bring new prospects for treating these diseases.
4.3. LncRNAs and NLRP3 in nephropathy
In recent years, the roles of lncRNAs have received increasing
attention. The typical clinical manifestations of uric acid nephropathy
(UAN) are hyperuric acid, oedema, oliguria, proteinuria, and hyper-
tension. UAN is a type of urate crystal deposition caused by purine
metabolism disorders and eventually leads to inammation activation
and induce immune-changing diseases(Hou et al., 2014). The long-chain
noncoding RNA ANRIL is involved in the NF-κB-mediated inammatory
response and its high expression has been shown in the serum of UAN
patients and kidney tissues of an adenine-induced rat model(Hu et al.,
2019). Hu et al. reported that ANRIL can act as a competitive sponge for
particular miRNA to reduce NLRP3 degradation and activate it to exert a
pathogenic effect via the miR-122-5p/BRCC3 axis in UAN patients(Hu
et al., 2019). In diabetic nephropathy, Li et al. revealed that MALAT1
promotes renal tubular epithelial cell apoptosis through the same ceRNA
mechanism, combined with the targeting of ELAVL1 by miR-23c to
regulate expression of its downstream protein NLRP3(Li et al., 2017). Yi
et al. also found that silencing lincRNA-Gm4419 could signicantly
downregulate the expression of NLRP3 inammasome-mediated
inammation via the NF-κB/NLRP3 axis in diabetic nephropathy(Yi
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
197
et al., 2017). The same regulatory mechanism of interaction between
lncRNA and NLRP3 inammation was found in calcium oxalateinduced
kidney stones by Song et al. Their research conclusion revealed that
LINC00339 could cause renal tubular epithelial pyroptosis via the
miR-22-3p/NLRP3 ceRNA (Song et al., 2019b). Therefore we speculate
that inammation resulting from the interaction of lncRNAs and NLRP3
plays various important roles in diabetic nephropathy. A better under-
standing of this mechanism of regulating diabetic nephropathy patho-
genesis by long noncoding RNAs may facilitate the development of a
potential therapeutic strategy and criteria for diagnosis.
4.4. LncRNAs and NLRP3 in neuroinammation
Neuroinammation may be closely related to inammatory re-
actions, neuro infections, neurodegenerative changes, and other related
diseases. The specic regulatory mechanism of noncoding RNAs is not
yet evident in the whole process. In neuroimmunology, recent research
evidence has found that lncRNAs could be involved in neuroimmune
functions, such as regulation of the mass production of neuro-
inammatory mediators and DNA-protein and RNA-RNA interactions to
control cell proliferation, invasion, differentiation, migration, and sur-
vival(Heward and Lindsay, 2014). Recent studies have conrmed that
changes in lncRNA expression act as an essential regulatory participant
not only in the adaptive process but also in the adaptive immune
response; lincRNA-Cox2, which is also known as Ptgs2, is located at the
51 kb locus near the functional gene Cox2, and its expression is upre-
gulated through Tlr4 stimulation-induced lincRNA-Cox2 in bone
marrow-derived dendritic cells(Guttman et al., 2009). Xue et al. found
that lincRNA-Cox2 was able to target NF-κB p65 to modulate NLRP3 and
ASC activity by enhancing its nuclear translocation and transcription
(Xue et al., 2019). The silencing of lincRNA-Cox2 can downregulate
NLRP3 inammasome expression by disrupting NLRP3-mediated ASC
complex formation under LPS-induced stimuli. Inhibition of NLRP3
inammasome activity leads to caspase-1 inactivation, reducing the
release of TIR domain-containing adapter-inducing interferon-β (TRIF);
therefore, the nal product derepresses TRIF-mediated ATG5-dependent
autophagy. In the progression of Parkinsons disease (PD), the lncRNA
Snhg1 can sponge endogenous RNA miR-7 to regulate activation of the
NLRP3 inammasome(Cao et al., 2018). These research results provide
a better understanding of the interactive regulatory mechanism between
lncRNAs and the NLRP3 inammasome, which may be a potential op-
portunity for prophylaxis, diagnosis, and therapeutic intervention in
neuroinammation-related diseases.
4.5. LncRNAs and NLRP3 in liver diseases
In a recent study, lncRNAs and NLRP3 were involved in the devel-
opment and evolution of hepatic brosis, closely associated with
pyroptosis, the nal programmed inammatory cell death process.
Zhang et al. found that M1 macrophage activation and pyroptosis could
be regulated by silencing lnc-Lfar1 through excessive pyroptosis of CCl4-
and BDL- induced proinammatory factors(Zhang et al., 2020b).
Mechanistically, lnc-far1 regulated the activation of LPS- and
IFN-γ-induced proinammatory factors in macrophages via the
NF-ĸB/NLRP3 axis(Zhang et al., 2020b). The lncRNA-Gm15441 can
prompt elevated mature caspase-1 and IL-1β cleavage due to its sus-
ceptibility to NLRP3 inammasome activation under metabolic and
external inammatory stimuli(Brocker et al., 2019). These experimental
ndings provide new evidence for interpreting this novel mechanism by
which how they attenuate hepatic inammasome activation in the
progression of hepatic brosis and hepatic inammasome diseases.
4.6. LncRNAs and NLRP3 in cardiovascular diseases
In cardiovascular diseases of the circulatory system, high-throughput
sequencing has detected and characterized abnormal lncRNA expression
under pathophysiological conditions(Zhang et al., 2019c,d). The inter-
action between the NLRP3 inammasome and lncRNAs is involved in
structural heart disease and coronary heart disease(Uchida and Dimm-
eler, 2015). Atorvastatin can inhibit pyroptosis, which is one kind of
programmed cell death by changing biomarker (such as NLRP3,
caspase-1, ASC, IL-1 β, IL-18, etc.) expression levels of the NLRP3
inammasome pathway in atherosclerosis (AS) (Wu et al., 2020). The
precise mechanism is that the upregulation of the lncRNA NEXN-AS1
expression levels under pathological conditions directly regulates its
target gene NEXN in HVECs(Wu et al., 2020). In another experiment on
aortic endothelium of melatonin-treated animals with AS, the expression
of pyroptosis-related genes was signicantly reduced after melatonin
treatment; these inammatory factors included NF-κB/GSDMD, NLRP3,
ASC, Caspase-1, IL-1β, and IL-18 in the whole pathway(Zhang et al.,
2018). Human aortic endothelial cell (HAECs) pyroptosis can be
enhanced by activation of the NLRP3 inammasome through
lncRNA-MEG3/miR-223/NLRP3 ceRNA network(Yong Zhang et al.,
2018). In this process, lncRNA-MEG3 functions as an endogenous
sponge sequence complementing miR-223 and inhibits its expression
(Zhang et al., 2018). In the process of cardiac broblast apoptosis and
cardiac brosis disease, the main regulatory factor, lncRNA-GAS5, was
found to play an important role. Further research results revealed that
DNMT1 of lncRNA GAS5 activated the NLRP3 inammasome in the
progression of cardiac broblast pyroptosis(She et al., 2020). These are
the regulatory mechanisms of cardiovascular tissue apoptosis under LPS
stimulation, and provide a new therapeutic target for cardiovascular
diseases.
4.7. LncRNAs and NLRP3 in retinal ischaemia/reperfusion
Wang et al. reported that lncRNAs could be involved in ischaemic/
reperfusion by their transcriptional control of inammatory responses
(Wan et al., 2019). There are core causes of visual impairment or
blindness by I/R injury based on diverse retinal diseases, such as
infection, glaucoma, central retinal artery stenosis or obstruction, and
diabetic retinopathy(Paul Kamdem et al., 2016). Abnormal expression
of various immune cells and inammatory cytokines is closely related to
all I/R injuries(Yang et al., 2015). Some investigators have found that
the lncRNA H19/miR-21/PDCD4 ceRNA net forms a ceRNA mechanism
to change PDCD4 expression to regulate cytokine overproduction,
neuronal lesions, and I/R-induced sterile inammation(Wan et al.,
2019). Additionally, the lncRNA H19 is also involved in microglial
pyroptosis, neuronal death, and mitochondrial dysfunction through this
ceRNET. This intermolecular regulation mode enriched the regulatory
mechanism between lncRNAs and NLRP3 and facilitated prophylaxis
and I/R injury treatment.
4.8. LncRNAs and NLRP3 in intervertebral disk degeneration
In intervertebral disc degeneration, some studies have veried that
the change in lncRNA expression level signicantly increased in the
course of disease(Chen et al., 2017; Li et al., 2018; Mi et al., 2018). In a
current study, overexpressed LINC00969 massively bound to
miR-335-3p, which led to aberrant downregulation of the expression of
miR-335-3p in the nucleus pulposus (NP) tissues and cells of interver-
tebral disk degeneration patients(Yu et al., 2019). LINC00969 also en-
hances NP cell apoptosis in tissues by acting as a molecular bridge,
competing for miR-335-3p to modulate TXNIP expression and NLRP3
inammasome activation in vitro(Yu et al., 2019). These results provide
a reference mechanism on the mutual interaction between LINC00969
and NLRP3 to regulate the expression of the NLRP3 inammasome and
participate in the treatment and diagnosis of intervertebral disk
degeneration.
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
198
5. Conclusion
In summary, the interaction of the NLRP3 inammasome and
lncRNAs has been most intensively investigated in various diseases,
including cancer, pyroptosis, Alzheimers disease, neuroinammation,
cardiovascular diseases, I/R injury, and inammatory diseases. There is
increasing evidence that lncRNAs mainly regulate the NLRP3 inam-
masome associated with pathophysiological processes pre- and post-
transcription. We still know very little about their precise regulatory
mechanisms during the complex pathophysiological evolution of dis-
eases, such as phagocytosis, adhesion, chemotaxis, exocytosis, and an-
tigen presentation. Furthermore, we also rarely know-how domains
accurately activate the NLRP3 inammasome: by protein interactions or
RNA interactions. These ndings will help us identify the functional
interaction between lncRNAs and NLRP3 according to sequence and
structure. Finally, with increasing discoveries related to lncRNAs and
disease development, and the mechanism of action being investigated,
lncRNA and NLRP3 interactions have new drug targets for allergies,
autoimmune diseases, cancer, pain, chronic inammation, and in-
fections. These research data provide insights into the mutual regulation
of lncRNAs and NLRP3 inammatory bodies and open up potential new
avenues to treat many diseases.
Author contribution statement
Author: Deqiang Luo Contribution: 1. Design research direction 2.
Write papers. Author: Fen Liu Contribution: 1. Collect references and
analysis 2. Review and revise the papers.
AuthorWenqiang Tao ContributionReview and revise the
papers.
AuthorRui Xiao ContributionReview and revise the papers
AuthorJianguo Zhang ContributionReview and edit the article.
AuthorQiang Shao ContributionReview and revise the papers.
AuthorWei Dai ContributionReview and revise the papers.
Author: Keqian Qian Contribution: 1. Review and revise the papers,
2.guidance article writing.
Funding
The study accepts the National Natural Science Foundation of China
(81560306 and 81460292) and Graduate Student Innovation Special
Fund Project of Jiangxi Province (YC2020-B033).
Ethical approval and consent to participate
This article does not involve any studies with patients or animals
performed by any of the authors.
Declaration of Competing Interest
There is no conict of interest in this study. All authors declare that
they have no conict of interest.
Acknowledgments
We are grateful to the personnel of all the hospitals who participated
in this article for their support during design and revision. This work was
funded by a grant from The First Afliated Hospital of Nanchang
University.
References
Ahmad, I., Muneer, K.M., Tamimi, I.A., Chang, M.E., Ata, M.O., Yusuf, N., 2013.
Thymoquinone suppresses metastasis of melanoma cells by inhibition of NLRP3
inammasome. Toxicol. Appl. Pharmacol. 270, 7076.
Anthony, T.G., Wek, R.C., 2012. TXNIP switches tracks toward a terminal UPR. Cell
Metab. 16, 135137.
Antonopoulos, C., Russo, H.M., El Sanadi, C., Martin, B.N., Li, X., Kaiser, W.J.,
Mocarski, E.S., Dubyak, G.R., 2015. Caspase-8 as an effector and regulator of NLRP3
inammasome signaling. J. Biol. Chem. 290, 2016720184.
Arun, G., Diermeier, S.D., Spector, D.L., 2018. Therapeutic targeting of long non-coding
RNAs in cancer. Trends Mol. Med. 24, 257277.
Bauernfeind, F.G., Horvath, G., Stutz, A., Alnemri, E.S., MacDonald, K., Speert, D.,
Fernandes-Alnemri, T., Wu, J., Monks, B.G., Fitzgerald, K.A., 2009. Cutting edge: NF-
κB activating pattern recognition and cytokine receptors license NLRP3
inammasome activation by regulating NLRP3 expression. J. Immunol. 183,
787791.
B¨
ohmdorfer, G., Wierzbicki, A.T., 2015. Control of chromatin structure by long
noncoding RNA. Trends Cell Biol. 25, 623632.
Bordon, Y., 2019. Trans-Golgi network breaks away to activate NLRP3. Nat. Rev.
Immunol. 19, 6869.
Brocker, C.N., Kim, D., Melia, T., Karri, K., Velenosi, T.J., Takahashi, S., Bonzo, J.A.,
Waxman, D.J., Gonzalez, F.J., 2019. Long non-coding RNA Gm15441 attenuates
hepatic inammasome activation in response to metabolic stress. bioRxiv, 675785.
Cao, B., Wang, T., Qu, Q., Kang, T., Yang, Q., 2018. Long noncoding RNA SNHG1
promotes neuroinammation in Parkinsons disease via regulating miR-7/NLRP3
pathway. Neuroscience 388, 118127.
Cassel, S.L., Joly, S., Sutterwala, F.S., 2009. The NLRP3 inammasome: a sensor of
immune danger signals. Semin. Immunol. 194198. Elsevier.
Chen, W.K., Yu, X.H., Yang, W., Wang, C., He, W.S., Yan, Y.G., Zhang, J., Wang, W.J.,
2017. lnc RNA s: novel players in intervertebral disc degeneration and osteoarthritis.
Cell Prolif. 50, e12313.
Choudhari, R., Sedano, M.J., Harrison, A.L., Subramani, R., Lin, K.Y., Ramos, E.I.,
Lakshmanaswamy, R., Gadad, S.S., 2020. Long noncoding RNAs in cancer: from
discovery to therapeutic targets. Adv. Clin. Chem. 105147. Academic Press Inc.
de Oliveira, J.C., Oliveira, L.C., Mathias, C., Pedroso, G.A., Lemos, D.S., Salviano-
Silva, A., Jucoski, T.S., Lobo-Alves, S.C., Zambalde, E.P., Cipolla, G.A., 2019. Long
non-coding RNAs in cancer: another layer of complexity. J. Gene Med. 21, e3065.
Derrien, T., Johnson, R., Bussotti, G., Tanzer, A., Djebali, S., Tilgner, H., Guernec, G.,
Martin, D., Merkel, A., Knowles, D.G., 2012. The GENCODE v7 catalog of human
long noncoding RNAs: analysis of their gene structure, evolution, and expression.
Genome Res. 22, 17751789.
Ding, J., Wang, K., Liu, W., She, Y., Sun, Q., Shi, J., Sun, H., Wang, D.-C., Shao, F., 2016.
Pore-forming activity and structural autoinhibition of the gasdermin family. Nature
535, 111116.
Dombrowski, Y., Peric, M., Koglin, S., Kammerbauer, C., G¨
oß, C., Anz, D., Simanski, M.,
Gl¨
aser, R., Harder, J., Hornung, V., 2011. Cytosolic DNA triggers inammasome
activation in keratinocytes in psoriatic lesions. Sci. Transl. Med. 3, 82ra38-82ra38.
Duncan, J.A., Bergstralh, D.T., Wang, Y., Willingham, S.B., Ye, Z., Zimmermann, A.G.,
Ting, J.P.-Y., 2007. Cryopyrin/NALP3 binds ATP/dATP, is an ATPase, and requires
ATP binding to mediate inammatory signaling. Proc. Natl. Acad. Sci. 104,
80418046.
Engreitz, J.M., Haines, J.E., Perez, E.M., Munson, G., Chen, J., Kane, M., McDonel, P.E.,
Guttman, M., Lander, E.S., 2016a. Local regulation of gene expression by lncRNA
promoters, transcription and splicing. Nature 539, 452455.
Engreitz, J.M., Ollikainen, N., Guttman, M., 2016b. Long non-coding RNAs: spatial
ampliers that control nuclear structure and gene expression. Nat. Rev. Mol. Cell
Biol. 17, 756.
Ergun, S., Oztuzcu, S., 2015. Oncocers: ceRNA-mediated cross-talk by sponging miRNAs
in oncogenic pathways. Tumor Biol. 36, 31293136.
Franchi, L., Mu˜
noz-Planillo, R., Nú˜
nez, G., 2012. Sensing and reacting to microbes
through the inammasomes. Nat. Immunol. 13, 325332.
Freeman, L., Guo, H., David, C.N., Brickey, W.J., Jha, S., Ting, J.P.-Y., 2017. NLR
members NLRC4 and NLRP3 mediate sterile inammasome activation in microglia
and astrocytes. J. Exp. Med. 214, 13511370.
Geng, H., Tan, X.-D., 2016. Functional diversity of long non-coding RNAs in immune
regulation. Genes Dis. 3, 7281.
Guo, J., Liu, Z., Gong, R., 2019. Long noncoding RNA: an emerging player in diabetes
and diabetic kidney disease. Clin. Sci. 133, 13211339.
Gutschner, T., Diederichs, S., 2012. The hallmarks of cancer: a long non-coding RNA
point of view. RNA Biol. 9, 703719.
Guttman, M., Amit, I., Garber, M., French, C., Lin, M.F., Feldser, D., Huarte, M., Zuk, O.,
Carey, B.W., Cassady, J.P., 2009. Chromatin signature reveals over a thousand
highly conserved large non-coding RNAs in mammals. Nature 458, 223227.
Hadjicharalambous, M.R., Lindsay, M.A., 2019. Long non-coding RNAs and the innate
immune response. Noncoding RNA 5, 34.
Haque, M.E., Akther, M., Jakaria, M., Kim, I.S., Azam, S., Choi, D.K., 2020. Targeting the
microglial NLRP3 inammasome and its role in Parkinsons disease. Mov. Disord.
35, 2033.
He, Y., Hara, H., Nú˜
nez, G., 2016a. Mechanism and regulation of NLRP3 inammasome
activation. Trends Biochem. Sci. 41, 10121021.
He, Y., Zeng, M.Y., Yang, D., Motro, B., Nú˜
nez, G., 2016b. NEK7 is an essential mediator
of NLRP3 activation downstream of potassium efux. Nature 530, 354357.
He, D., Zhen, J., Hu, J., Chen, J., Wei, X., 2019. Long non-coding RNAs and pyroptosis.
Clin. Chim. Acta 201208.
Heid, M.E., Keyel, P.A., Kamga, C., Shiva, S., Watkins, S.C., Salter, R.D., 2013.
Mitochondrial reactive oxygen species induces NLRP3-dependent lysosomal damage
and inammasome activation. J. Immunol. 191, 52305238.
Heward, J.A., Lindsay, M.A., 2014. Long non-coding RNAs in the regulation of the
immune response. Trends Immunol. 35, 408419.
Hon, C.-C., Ramilowski, J.A., Harshbarger, J., Bertin, N., Rackham, O.J., Gough, J.,
Denisenko, E., Schmeier, S., Poulsen, T.M., Severin, J., 2017. An atlas of human long
non-coding RNAs with accurate 5ends. Nature 543, 199204.
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
199
Hou, S.-x., Zhu, W.-j., Pang, M.-q., Jeffry, J., Zhou, L.-l., 2014. Protective effect of iridoid
glycosides from Paederia scandens (LOUR.) MERRILL (Rubiaceae) on uric acid
nephropathy rats induced by yeast and potassium oxonate. Food Chem. Toxicol. 64,
5764.
Hu, H., Wang, Y., Ding, X., He, Y., Lu, Z., Wu, P., Tian, L., Yuan, H., Liu, D., Shi, G., 2018.
Long non-coding RNA XLOC_000647 suppresses progression of pancreatic cancer
and decreases epithelial-mesenchymal transition-induced cell invasion by down-
regulating NLRP3. Mol. Cancer 17, 18.
Hu, J., Wu, H., Wang, D., Yang, Z., Dong, J., 2019. LncRNA ANRIL promotes NLRP3
inammasome activation in uric acid nephropathy through miR-122-5p/BRCC3 axis.
Biochimie 157, 102110.
Jiang, H., Ma, R., Zou, S., Wang, Y., Li, Z., Li, W., 2017. Reconstruction and analysis of
the lncRNAmiRNAmRNA network based on competitive endogenous RNA reveal
functional lncRNAs in rheumatoid arthritis. Mol. Biosyst. 13, 11821192.
Kanneganti, T.-D., ¨
Oz¨
oren, N., Body-Malapel, M., Amer, A., Park, J.-H., Franchi, L.,
Whiteld, J., Barchet, W., Colonna, M., Vandenabeele, P., 2006. Bacterial RNA and
small antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature 440,
233236.
Karki, R., Man, S.M., Kanneganti, T.-D., 2017. Inammasomes and cancer. Cancer
Immunol. Res. 5, 9499.
Khaitan, D., Dinger, M.E., Mazar, J., Crawford, J., Smith, M.A., Mattick, J.S., Perera, R.J.,
2011. The melanoma-upregulated long noncoding RNA SPRY4-IT1 modulates
apoptosis and invasion. Cancer Res. 71, 38523862.
Kim, E.H., Park, M.-J., Park, S., Lee, E.-S., 2015. Increased expression of the NLRP3
inammasome components in patients with Behçets disease. J. Inamm. 12, 41.
Kornfeld, J.-W., Brüning, J.C., 2014. Regulation of metabolism by long, non-coding
RNAs. Front. Genet. 5, 57.
Lamkan, M., Dixit, V.M., 2009. Inammasomes: guardians of cytosolic sanctity.
Immunol. Rev. 227, 95105.
Lamkan, M., Kanneganti, T.-D., 2010. Nlrp3: an immune sensor of cellular stress and
infection. Int. J. Biochem. Cell Biol. 42, 792795.
Li, X., Zeng, L., Cao, C., Lu, C., Lian, W., Han, J., Zhang, X., Zhang, J., Tang, T., Li, M.,
2017. Long noncoding RNA MALAT1 regulates renal tubular epithelial pyroptosis by
modulated miR-23c targeting of ELAVL1 in diabetic nephropathy. Exp. Cell Res. 350,
327335.
Li, Z., Li, X., Chen, C., Li, S., Shen, J., Tse, G., Chan, M.T., Wu, W.K., 2018. Long non-
coding RNA s in nucleus pulposus cell function and intervertebral disc degeneration.
Cell Prolif. 51, e12483.
Liao, K., Xu, J., Yang, W., You, X., Zhong, Q., Wang, X., 2018. The research progress of
LncRNA involved in the regulation of inammatory diseases. Mol. Immunol. 101,
182188.
Lieberman, J., Wu, H., Kagan, J.C., 2019. Gasdermin D activity in inammation and host
defense. Sci. Immunol. 4.
Liu, X., Zhang, X., Ding, Y., Zhou, W., Tao, L., Lu, P., Wang, Y., Hu, R., 2016. Nuclear
factor E2-Related Factor-2 negatively regulates NLRP3 inammasome activity by
inhibiting reactive oxygen species-induced NLRP3 priming. Antioxid. Redox Signal.
26, 28.
Lu, A., Magupalli, V.G., Ruan, J., Yin, Q., Atianand, M.K., Vos, M.R., Schr¨
oder, G.F.,
Fitzgerald, K.A., Wu, H., Egelman, E.H., 2014. Unied polymerization mechanism
for the assembly of ASC-dependent inammasomes. Cell 156, 11931206.
Ma, M., Pei, Y., Wang, X., Feng, J., Zhang, Y., Gao, M.Q., 2019. LncRNA XIST mediates
bovine mammary epithelial cell inammatory response via NF-κB/NLRP3
inammasome pathway. Cell Prolif. 52, e12525.
Martens, J.A., Laprade, L., Winston, F., 2004. Intergenic transcription is required to
repress the Saccharomyces cerevisiae SER3 gene. Nature 429, 571574.
Martinon, F., 2010. Signaling by ROS drives inammasome activation. Eur. J. Immunol.
40, 616619.
McCoy, A.J., Koizumi, Y., Higa, N., Suzuki, T., 2010. Differential regulation of caspase-1
activation via NLRP3/NLRC4 inammasomes mediated by aerolysin and type III
secretion system during Aeromonas veronii infection. J. Immunol. 185, 70777084.
Medzhitov, R., 2008. Origin and physiological roles of inammation. Nature 454,
428435.
Medzhitov, R., 2010. Inammation 2010: new adventures of an old ame. Cell 140,
771776.
Memczak, S., Jens, M., Elefsinioti, A., Torti, F., Krueger, J., Rybak, A., Maier, L.,
Mackowiak, S.D., Gregersen, L.H., Munschauer, M., 2013. Circular RNAs are a large
class of animal RNAs with regulatory potency. Nature 495, 333338.
Mendell, J.T., Shari, N.A., Meyers, J.L., Martinez-Murillo, F., Dietz, H.C., 2004.
Nonsense surveillance regulates expression of diverse classes of mammalian
transcripts and mutes genomic noise. Nat. Genet. 36, 10731078.
Mi, D., Cai, C., Zhou, B., Liu, X., Ma, P., Shen, S., Lu, W., Huang, W., 2018. Long
noncoding RNA FAF1 promotes intervertebral disc degeneration by targeting the
Erk signaling pathway. Mol. Med. Report. 17, 31583163.
Morlando, M., Fatica, A., 2018. Alteration of epigenetic regulation by long noncoding
RNAs in cancer. Int. J. Mol. Sci. 19, 570.
Moura, J., Børsheim, E., Carvalho, E., 2014. The role of micrornas in diabetic
complicationsspecial emphasis on wound healing. Genes 5, 926956.
Nam, J.-W., Choi, S.-W., You, B.-H., 2016. Incredible RNA: dual functions of coding and
noncoding. Mol. Cells 39, 367374.
Natoli, G., Andrau, J.-C., 2012. Noncoding transcription at enhancers: general principles
and functional models. Annu. Rev. Genet. 46, 119.
Nie, L., Zhang, P., Wang, Q., Zhou, X., Wang, Q., 2019. lncRNA-Triggered Macrophage
Inammaging Deteriorates Age-Related Diseases. Mediators Inamm. 2019,
4260369.
Ozaki, E., Campbell, M., Doyle, S.L., 2015. Targeting the NLRP3 inammasome in
chronic inammatory diseases: current perspectives. J. Inamm. Res. 8, 1527.
Pagano, A., Castelnuovo, M., Tortelli, F., Ferrari, R., Dieci, G., Cancedda, R., 2007. New
small nuclear RNA gene-like transcriptional units as sources of regulatory
transcripts. PLoS Genet. 3, e1.
Park, J.-H., Jeong, S.-Y., Choi, A.-J., Kim, S.-J., 2015. Lipopolysaccharide directly
stimulates Th17 differentiation in vitro modulating phosphorylation of RelB and NF-
κB1. Immunol. Lett. 165, 1019.
Paul Kamdem, J., Olalekan Abolaji, A., Olusola Elekofehinti, O., Olaposi Omotuyi, I.,
Ibrahim, M., Hassan, W., Vargas Barbosa, N., Onofre Souza, D., Batista Teixeira da
Rocha, J., 2016. Therapeutic potential of plant extracts and phytochemicals against
brain ischemia-reperfusion injury: a review. Nat. Prod. J. 6, 250284.
Quinn, J.J., Chang, H.Y., 2016. Unique features of long non-coding RNA biogenesis and
function. Nat. Rev. Genet. 17, 4762.
Rituparno, S., Suman, G., Shaoli, D., Subrata, B., Jayprokas, C., 2014. Competing
endogenous RNA: the key to posttranscriptional regulation. Sci. World J. 2014, 16.
Ruiz-Orera, J., Messeguer, X., Subirana, J.A., Alba, M.M., 2014. Long non-coding RNAs
as a source of new peptides. elife 3, e03523.
Salmena, L., Poliseno, L., Tay, Y., Kats, L., Pandol, P.P., 2011. A ceRNA hypothesis: the
Rosetta Stone of a hidden RNA language? Cell 146, 353358.
Salviano-Silva, A., Lobo-Alves, S.C., Almeida, R.C., Malheiros, D., Petzl-Erler, M.L., 2018.
Besides pathology: long non-coding RNA in cell and tissue homeostasis. Noncoding
RNA 4, 3.
Samra, Y.A., Said, H.S., Elsherbiny, N.M., Liou, G.I., El-Shishtawy, M.M., Eissa, L.A.,
2016. Cepharanthine and Piperine ameliorate diabetic nephropathy in rats: role of
NF-κB and NLRP3 inammasome. Life Sci. 157, 187199.
Sandanger, Ø., Ranheim, T., Vinge, L.E., Bliksøen, M., Alfsnes, K., Finsen, A.V., Dahl, C.
P., Askevold, E.T., Florholmen, G., Christensen, G., 2013. The NLRP3 inammasome
is up-regulated in cardiac broblasts and mediates myocardial
ischaemiareperfusion injury. Cardiovasc. Res. 99, 164174.
Schmidt, F.I., Lu, A., Chen, J.W., Ruan, J., Tang, C., Wu, H., Ploegh, H.L., 2016. A single
domain antibody fragment that recognizes the adaptor ASC denes the role of ASC
domains in inammasome assembly. J. Exp. Med. 213, 771790.
Schroder, K., Tschopp, J., 2010. The inammasomes. Cell 140, 821832.
She, Q., Shi, P., Xu, S.-S., Xuan, H.-Y., Tao, H., Shi, K.-H., Yang, Y., 2020. DNMT1
methylation of LncRNA GAS5 leads to cardiac broblast pyroptosis via affecting
NLRP3 Axis. Inammation 112.
Shi, J., Zhao, Y., Wang, K., Shi, X., Wang, Y., Huang, H., Zhuang, Y., Cai, T., Wang, F.,
Shao, F., 2015. Cleavage of GSDMD by inammatory caspases determines pyroptotic
cell death. Nature 526, 660665.
Shi, H., Wang, Y., Li, X., Zhan, X., Tang, M., Fina, M., Su, L., Pratt, D., Bu, C.H.,
Hildebrand, S., 2016. NLRP3 activation and mitosis are mutually exclusive events
coordinated by NEK7, a new inammasome component. Nat. Immunol. 17,
250258.
Song, Y., Yang, L., Guo, R., Lu, N., Shi, Y., Wang, X., 2019a. Long noncoding RNA
MALAT1 promotes high glucose-induced human endothelial cells pyroptosis by
affecting NLRP3 expression through competitively binding miR-22. Biochem.
Biophys. Res. Commun. 509, 359366.
Song, Z., Zhang, Y., Gong, B., Xu, H., Hao, Z., Liang, C., 2019b. Long noncoding RNA
LINC00339 promotes renal tubular epithelial pyroptosis by regulating the miR-22-
3p/NLRP3 axis in calcium oxalateinduced kidney stone. J. Cell. Biochem. 120,
1045210462.
Spizzo, R., Almeida, M.Ie., Colombatti, A., Calin, G.A., 2012. Long non-coding RNAs and
cancer: a new frontier of translational research? Oncogene 31, 45774587.
Sutterwala, F.S., Haasken, S., Cassel, S.L., 2014. Mechanism of NLRP3 inammasome
activation. Ann. N. Y. Acad. Sci. 1319, 8295.
Swanson, K.V., Deng, M., Ting, J.P.-Y., 2019. The NLRP3 inammasome: molecular
activation and regulation to therapeutics. Nat. Rev. Immunol. 19, 477489.
Tang, D., Kang, R., Coyne, C.B., Zeh, H.J., Lotze, M.T., 2012. PAMP s and DAMP s: signal
0s that spur autophagy and immunity. Immunol. Rev. 249, 158175.
Tang, Y., Cao, G., Zhao, G., Wang, C., Qin, Q., 2019. LncRNA differentiation antagonizing
non-protein coding RNA promotes proliferation and invasion through regulating
miR-135a/NLRP37 axis in pancreatic cancer. Invest. New Drugs 18.
Tay, Y., Rinn, J., Pandol, P.P., 2014. The multilayered complexity of ceRNA crosstalk
and competition. Nature 505, 344352.
Uchida, S., Dimmeler, S., 2015. Long noncoding RNAs in cardiovascular diseases. Circ.
Res. 116, 737750.
Van Opdenbosch, N., Lamkan, M., 2019. Caspases in cell death, inammation, and
disease. Immunity 50, 13521364.
Wan, P., Su, W., Zhang, Y., Li, Z., Deng, C., Li, J., Jiang, N., Huang, S., Long, E., Zhuo, Y.,
2019. LncRNA H19 initiates microglial pyroptosis and neuronal death in retinal
ischemia/reperfusion injury. Cell Death Differ. 116.
Wang, L., Zheng, Z., Feng, X., Zang, X., Ding, W., Wu, F., Zhao, Q., 2019a. circRNA/
lncRNA-miRNA-mRNA Network in Oxidized, Low-Density, Lipoprotein-Induced
Foam Cells. DNA Cell Biol. 38, 14991511.
Wang, Y.H., Lv, H.N., Cui, Q.H., Tu, P.F., Zeng, K.W., 2019b. Isosibiricin inhibits
microglial activation by targeting the dopamine D1/D2 receptor-dependent NLRP3/
caspase-1 inammasome pathway. Acta Pharmacol. Sin. 41, 18.
Willingham, A., Orth, A., Batalov, S., Peters, E., Wen, B., Aza-Blanc, P., Hogenesch, J.,
Schultz, P., 2005. A strategy for probing the function of noncoding RNAs nds a
repressor of NFAT. Science 309, 15701573.
Wu, G.-C., Pan, H.-F., Leng, R.-X., Wang, D.-G., Li, X.-P., Li, X.-M., Ye, D.-Q., 2015a.
Emerging role of long noncoding RNAs in autoimmune diseases. Autoimmun. Rev.
14, 798805.
Wu, T., Pinto, H.B., Kamikawa, Y.F., Donohoe, M.E., 2015b. The BET family member
BRD4 interacts with OCT4 and regulates pluripotency gene expression. Stem Cell
Reports 4, 390403.
D. Luo et al.
Molecular Immunology 131 (2021) 191–200
200
Wu, L.-M., Wu, S.-G., Chen, F., Wu, Q., Wu, C.-M., Kang, C.-M., He, X., Zhang, R.-Y.,
Lu, Z.-F., Li, X.-H., Xu, Y.-J., Li, L.-M., Ding, L., Bai, H.-L., Liu, X.-H., Hu, Y.-W.,
Zheng, L., 2020. Atorvastatin inhibits pyroptosis through the lncRNA NEXN-AS1/
NEXN pathway in human vascular endothelial cells. Atherosclerosis 293, 2634.
Xu, Z., Xi, K., 2019. LncRNA RGMB-AS1 promotes laryngeal squamous cell carcinoma
cells progression via sponging miR-22/NLRP3 axis. Biomed. Pharmacother. 118,
109222.
Xu, P., Wen, Z., Shi, X., Li, Y., Fan, L., Xiang, M., Li, A., Scott, M.J., Xiao, G., Li, S., 2013.
Hemorrhagic shock augments Nlrp3 inammasome activation in the lung through
impaired pyrin induction. J. Immunol. 190, 52475255.
Xu, Y.-J., Zheng, L., Hu, Y.-W., Wang, Q., 2018. Pyroptosis and its relationship to
atherosclerosis. Clin. Chim. Acta 476, 2837.
Xue, Z., Zhang, Z., Liu, H., Li, W., Guo, X., Zhang, Z., Liu, Y., Jia, L., Li, Y., Ren, Y., 2019.
lincRNA-Cox2 regulates NLRP3 inammasome and autophagy mediated
neuroinammation. Cell Death Differ. 26, 130145.
Yang, S.-M., Ka, S.-M., Wu, H.-L., Yeh, Y.-C., Kuo, C.-H., Hua, K.-F., Shi, G.-Y., Hung, Y.-
J., Hsiao, F.-C., Yang, S.-S., 2014. Thrombomodulin domain 1 ameliorates diabetic
nephropathy in mice via anti-NF-κB/NLRP3 inammasome-mediated inammation,
enhancement of NRF2 antioxidant activity and inhibition of apoptosis. Diabetologia
57, 424434.
Yang, Y., Salayandia, V.M., Thompson, J.F., Yang, L.Y., Estrada, E.Y., Yang, Y., 2015.
Attenuation of acute stroke injury in rat brain by minocycline promotes bloodbrain
barrier remodeling and alternative microglia/macrophage activation during
recovery. J. Neuroinammation 12, 26.
Yang, C., Di Wu, L.G., Liu, X., Jin, Y., Wang, D., Wang, T., Li, X., 2016. Competing
endogenous RNA networks in human cancer: hypothesis, validation, and
perspectives. Oncotarget 7, 1347913490.
Yi, H., Peng, R., Zhang, L.-y., Sun, Y., Peng, H.-m., Liu, H.-d., Yu, L.-j., Li, A.-l., Zhang, Y.-
j., Jiang, W.-h., 2017. LincRNA-Gm4419 knockdown ameliorates NF-κ B/NLRP3
inammasome-mediated inammation in diabetic nephropathy. Cell Death Dis. 8
e2583-e2583.
Yu, S.-y., Dong, B., Tang, L., Zhou, S.-h., 2018. LncRNA MALAT1 sponges miR-133 to
promote NLRP3 inammasome expression in ischemia-reperfusion injured heart. Int.
J. Cardiol. 254, 50.
Yu, L., Hao, Y., Xu, C., Zhu, G., Cai, Y., 2019. LINC00969 promotes the degeneration of
intervertebral disk by sponging miR-335-3p and regulating NLRP3 inammasome
activation. IUBMB Life 71, 611618.
Zhang, Y., Yang, L., Chen, L.-L., 2014. Life without A tail: new formats of long noncoding
RNAs. Int. J. Biochem. Cell Biol. 54, 338349.
Zhang, F., Wu, L., Qian, J., Qu, B., Xia, S., La, T., Wu, Y., Ma, J., Zeng, J., Guo, Q., 2016.
Identication of the long noncoding RNA NEAT1 as a novel inammatory regulator
acting through MAPK pathway in human lupus. J. Autoimmun. 75, 96104.
Zhang, Y., Liu, X., Bai, X., Lin, Y., Li, Z., Fu, J., Li, M., Zhao, T., Yang, H., Xu, R., 2018.
Melatonin prevents endothelial cell pyroptosis via regulation of long noncoding RNA
MEG3/miR-223/NLRP3 axis. J. Pineal Res. 64, e12449.
Zhang, M., Zheng, Y., Sun, Y., Li, S., Chen, L., Jin, X., Hou, X., Liu, X., Chen, Q., Li, J.,
2019a. Knockdown of NEAT1 induces tolerogenic phenotype in dendritic cells by
inhibiting activation of NLRP3 inammasome. Theranostics 9, 34253442.
Zhang, P., Cao, L., Zhou, R., Yang, X., Wu, M., 2019b. The lncRNA Neat1 promotes
activation of inammasomes in macrophages. Nat. Commun. 10, 117.
Zhang, X., Li, D.Y., Reilly, M.P., 2019c. Long intergenic noncoding RNAs in
cardiovascular diseases: challenges and strategies for physiological studies and
translation. Atherosclerosis 281, 180188.
Zhang, X., Wu, N., Wang, J., Li, Z., 2019d. LncRNA MEG3 inhibits cell proliferation and
induces apoptosis in laryngeal cancer via miR-23a/APAF-1 axis. J. Cell. Mol. Med.
23, 67086719.
Zhang, J., Liu, X., Wan, C., Liu, Y., Wang, Y., Meng, C., Zhang, Y., Jiang, C., 2020a.
NLRP3 inammasome mediates M1 macrophage polarization and IL-1β production
in inammatory root resorption. J. Clin. Periodontol. 47, 451460.
Zhang, K., Shi, Z., Zhang, M., Dong, X., Zheng, L., Li, G., Han, X., Yao, Z., Han, T.,
Hong, W., 2020b. Silencing lncRNA Lfar1 alleviates the classical activation and
pyoptosis of macrophage in hepatic brosis. Cell Death Dis. 11, 113.
Zhong, Z., Zhai, Y., Liang, S., Mori, Y., Han, R., Sutterwala, F.S., Qiao, L., 2013. TRPM2
links oxidative stress to NLRP3 inammasome activation. Nat. Commun. 4, 111.
Zhou, R., Yazdi, A.S., Menu, P., Tschopp, J., 2011. A role for mitochondria in NLRP3
inammasome activation. Nature 469, 221225.
Zhou, J., Yang, L., Zhong, T., Mueller, M., Men, Y., Zhang, N., Xie, J., Giang, K.,
Chung, H., Sun, X., 2015. H19 lncRNA alters DNA methylation genome wide by
regulating S-adenosylhomocysteine hydrolase. Nat. Commun. 6, 113.
Zhu, J., Tang, J., 2020. LncRNA Gm14205 induces astrocytic NLRP3 inammasome
activation via inhibiting oxytocin receptor in postpartum depression. Biosci. Rep. 40,
BSR20200672.
D. Luo et al.
... Non-coding RNAs (long non-coding RNA (lncRNA), microRNA (miRNA), and circular RNA (circRNA), etc.) are a group of endogenous cellular RNAs without protein-coding capacity; in recent years, these molecules have been shown to play essential roles in the pathophysiological processes of multiple diseases [13]. Numerous studies have found that lncRNAs regulate various physiological and pathological processes, such as inflammatory diseases, cardiovascular diseases, inflammatory responses, neuroinflammation, and Alzheimer's disease, by targeting the NLRP3 inflammasome [14][15][16][17][18][19][20][21][22][23][24]. miR-138-5p has been found to be a tumour suppressor in bladder [25], non-small cell lung [26], colorectal [27], malignant glioblastoma [28], and pancreatic cancer [29]. ...
... Cumulative evidence has shown that lncRNAs are implicated in various pathophysiological processes, such as ageing, tumours, cardiovascular disease, and neurodegenerative disease [14, 17, 19-23, 45, 46, 48-53]. To date, there are six types of regulatory mechanisms between lncRNA and NLRP3 [24]. DNA methyltransferases of lncRNAs affect NLRP3 chromatin reconstruction and modifications [16]. ...
Article
Full-text available
Acute lung injury (ALI) is a common lung pathology that is accompanied by alveolar macrophage (AM) activation and inflammatory response. This study investigated the role of the long non-coding RNA NONRATT004344 (hereafter named lncRNA NLRP3) in regulating the Nod-like receptor protein 3 (NLRP3)-triggered inflammatory response in early ALI and the underlying mechanism as well. We established LPS-induced ALI models to explore their interactive mechanisms in vitro and in vivo. Luciferase reporter assays were performed to determine that miR-138-5p could bind to lncRNA NLRP3 and NLRP3. We observed increased lncRNA NLRP3 expression, decreased miR-138-5p expression, NLRP3 inflammasome activation, and upregulated caspase-1, IL-1β, and IL-18 expression in the LPS-induced ALI model. Furthermore, lncRNA NLRP3 overexpression activated the NLRP3 inflammasome and promoted IL-1β and IL-18 secretion; the miR-138-5p mimic abolished these effects in vivo and in vitro. Consistently, miR-138-5p inhibition reversed the effects of lncRNA NLRP3 silencing on the expression of NLRP3-related molecules and inhibition of the NLRP3/caspase-1/IL-1β signalling pathway. Mechanistically, lncRNA NLRP3 sponging miR-138-5p facilitated NLRP3 activation through a competitive endogenous RNA (ceRNA) mechanism. In summary, our results suggested that lncRNA NLRP3 binding miR-138-5p promotes NLRP3-triggered inflammatory response via lncRNA NLRP3 / miR-138-5p/NLRP3 ceRNA network (ceRNET) and provides insights into the treatment of early ALI.
... Published studies have shown that the NLRP3 inflammasome triggers the activation of Caspase-1, which cleaves the inactivated cytokine precursors (IL-1β and IL-18) into proinflammatory cytokines (IL-1β and IL-18) [54]. The inflammasome plays a vital role in the activation of CASP1 and the maturation of IL-1β. ...
Article
Full-text available
Neuroinflammation is an important reason for the occurrence and development of cognitive impairment. The Lentiviral vector Hsp22 was constructed for intracerebroventricular injection pretreatment, LPS was used to induce the cognitive impairment model in mice, and the Morris water maze was used to examine the changes in cognitive behavior in mice. LPS was used to induce BV-2 microglial cells, and plasmid pretreatment was used to overexpress Hsp22. HE staining, Nissl staining, immunohistochemistry, immunofluorescence, ELISA and protein blotting were used to examine microglial activation, changes in inflammatory factors, changes in pathway proteins and apoptosis. The results showed that LPS induced microglial expression of NLRP3/Caspase-1/IL-1β signaling pathway protein Iba1, and the inflammatory protein and inflammatory factors IL-1β, IL-6 and TNF-α, the expression of Bax increased significantly, Bcl2 expression decreased, and the learning and memory abilities of mice decreased significantly. Preconditioning with the Hsp22-overexpressing lentivirus attenuated LPS-induced activation of hippocampal microglia, the expression of inflammatory factors and pathway proteins, and apoptosis, and improved cognitive impairment in mice. In addition, plasmid-mediated Hsp22 overexpression reversed LPS-induced inflammation. These findings suggest that Hsp22 overexpression is a promising method for the treatment of cognitive impairment.
... Meanwhile, the cytosolic mtDNA released from impaired mitochondria acts as "signal 2" to activate the NOD-like receptor protein 3 (NLRP3) inflammasome in macrophages [9,10]. The NLRP3 inflammasome has been reported to play an essential role in pyroptosis, a form of gasdermin D (GSDMD)-dependent programmed cell death mediated by inflammatory cysteinyl aspartate-specific proteases (caspases), which may contribute to ALI progression [9,[11][12][13]. However, the underlying mechanisms by which mtDNA activates the NLRP3 inflammasome in ALI have not been completely elucidated. ...
Article
Full-text available
Background The present study was designed to explore the potential regulatory mechanism between mitophagy and pyroptosis during sepsis-associated acute lung injury (ALI).Methods In vitro or in vivo models of sepsis-associated ALI were established by administering lipopolysaccharide (LPS) or performing caecal ligation and puncture (CLP) surgery. Pyroptosis levels were detected by electron microscopy, immunofluorescence, flow cytometry, western blotting and immunohistochemistry. Dual-luciferase reporter gene assay was applied to verify the targeting relationship between miR-138-5p and NLRP3. Methylation-specific PCR and chromatin immunoprecipitation assays were used to determine methylation of the miR-138-5p promoter. Mitophagy levels were examined by transmission electron microscopy and western blotting.ResultsNLRP3 inflammasome silencing alleviated alveolar macrophage (AM) pyroptosis and septic lung injury. In addition, we confirmed the direct targeting relationship between miR-138-5p and NLRP3. Overexpressed miR-138-5p alleviated AM pyroptosis and the pulmonary inflammatory response. Moreover, the decreased expression of miR-138-5p was confirmed to depend on promoter methylation, while inhibition of miR-138-5p promoter methylation attenuated AM pyroptosis and pulmonary inflammation. Here, we discovered that an increased cytoplasmic mtDNA content in sepsis-induced ALI models induced the methylation of the miR-138-5p promoter, thereby decreasing miR-138-5p expression, which may activate the NLRP3 inflammasome and trigger AM pyroptosis. Mitophagy, a form of selective autophagy that clears damaged mitochondria, reduced cytoplasmic mtDNA levels. Furthermore, enhanced mitophagy might suppress miR-138-5p promoter methylation and relieve the pulmonary inflammatory response, changes that were reversed by treatment with isolated mtDNA.Conclusions In summary, our study indicated that mitophagy induced the demethylation of the miR-138-5p promoter, which may subsequently inhibit NLRP3 inflammasome, AM pyroptosis and inflammation in sepsis-induced lung injury. These findings may provide a promising therapeutic target for sepsis-associated ALI.
... 16,17 It is used as an inflammasome marker and contributes to several diseases associated with the inflammatory response. 18,19 In TAO, T and B lymphocytes are thought to direct an inflammatory response that stimulates OF activation and leads to hyaluronan accumulation and the physiological characteristics of the disease. 20 Several studies have implicated the inflammatory process in the manifestation of TAO. ...
Article
Full-text available
Objective Thyroid-associated ophthalmopathy (TAO) is an autoimmune disease that involves the remodeling of orbit and periorbital tissues. Thyroid-stimulating hormone receptor (TSHR) and insulin-like growth factor 1 receptor (IGF-1R) may stimulate the activation of autoimmunity in TAO, but the exact mechanism is unclear. We investigated whether IGF-1R/TSHR modulation in TAO may involve microRNA regulation. Methods We conducted microarray analysis using RNA from the orbital connective tissue samples of 3 healthy and 3 patients with TAO. The involvement of differentially regulated microRNA in IGF-1R/TSHR modulation in TAO was evaluated in orbital fibroblasts (OFs) and female BALB/c mice. Results Using hierarchical cluster analysis, we identified that miR-143 was downregulated in TAO. The expression levels of miR-143 in OFs were significantly reduced under IL-1B stimulation. However, OF proliferation and inflammatory responses decreased when miR-143 is overexpressed. In contrast, the suppression of miR-143 increased levels of inflammatory markers (IL-6, IL-8, MCP1) and hyaluronan accumulation. Moreover, overexpression of miR-143 significantly lowers levels of IGF-1R and TSHR. A luciferase assay indicated that miR-143 targets the 3′-UTR of IGF-1R. Increases in the expression of IGF-1R increased the expression of the inflammasome marker NLRP3 and apoptotic marker cleaved caspase-1; however, miR-143 overexpression decreased levels of IGF-1R, TSHR, NLRP3, cleaved caspase 1, IL-1B, and IL-18. In a mouse model of TAO, overexpression of miR-143 significantly reduced levels of IGF-1R and attenuated the adipogenesis associated with TAO. Conclusion We found that miR-143 directly targets IGF-1R to alleviate the inflammatory response in TAO by indirectly decreasing levels of TSHR and inactivating NLRP3.
... Accordingly, its accurate regulation is of paramount importance. Along with various factors, the role that different non-coding RNAs, such as LncRNAs and Micro RNA, play in their regulation is indicated (Luo et al. 2021;Tezcan et al. 2019). In this article, the regulatory interaction of miR-223 with NLRP3 inflammasome is investigated and explained. ...
Article
Full-text available
Millions of people around the world are involved with COVID-19 due to infection with SARS-CoV-2. Virological features of SARS-CoV-2, including its genomic sequence, have been identified but the mechanisms governing COVID-19 immunopathogenesis have remained uncertain. miR-223 is a hematopoietic cell-derived miRNA that is implicated in regulating monocyte-macrophage differentiation, neutrophil recruitment, and pro-inflammatory responses. The miR-223 controls inflammation by targeting a variety of factors, including TRAF6, IKKα, HSP-70, FOXO1, TLR4, PI3K/AKT, PARP-1, HDAC2, ITGB3, CXCL2, CCL3, IL-6, IFN-I, STMN1, IL-1β, IL-18, Caspase-1, NF-κB, and NLRP3. The key role of miR-223 in regulating the inflammatory process and its antioxidant and antiviral role can suggest this miRNA as a potential regulatory factor in the process of COVID-19 immunopathogenesis.
Article
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in the innate immune system and is a three-part macromolecular complex comprising the NLRP3 protein, apoptosis-associated speck-like protein containing a CARD (ASC) and the cysteine protease pro-caspase-1. When the NLRP3 inflammasome is activated, it can produce interleukin (IL)- 1β and IL-18 and eventually lead to inflammatory cell pyroptosis. Related studies have demonstrated that the NLRP3 inflammasome can induce an immune response and is related to the occurrence and development of gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer. NLRP3 inflammasome inhibitors are beneficial for maintaining cellular homeostasis and tissue health and have been found effective in targeting some gynecological diseases. However, excessive inhibitor concentrations have been found to cause adverse effects. Therefore, proper control of NLRP3 inflammasome activity is critical. This paper summarizes the structure and function of the NLRP3 inflammasome and highlights the therapeutic potential of targeting it in gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer The application of NLRP3 inflammasome inhibitors is also discussed.
Article
Background and aim: The hyperactive nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome in the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a key factor for cytokine storm, chronic inflammation, and mortality in infected patients. On the subject of the regulation of the NLRP3-inflammasome activation, micro-ribonucleic acid (RNA)-223 (miR-223), among the major RNA molecules, has been thus far investigated in some inflammatory diseases along with interleukin-1 beta (IL-1β) and NLRP3. Against this background, the present study aimed to compare healthy individuals and patients with severe COVID-19 with reference to the alterations in the expression of the miR-223, NLRP3, and IL-1β axis and the serum IL-1β levels. Methods: In total, 40 patients with severe COVID-19, admitted to the Infectious Ward of Razi Hospital, Ahvaz, Iran, who were homogenous in terms of age (40 years old) and gender, were selected based on the inclusion and exclusion criteria. The real-time polymerase chain reaction (RT-PCR) technique was then applied to assess the expression of the miR-223, NLRP3, and IL-1β genes, and enzyme-linked immunosorbent assay (ELISA) was then utilized to evaluate the serum IL-1β levels, using patients' blood samples. Moreover, inflammatory biochemical markers of the participants were collected and recorded RESULTS: According to the study results, the IL-1β expression was 3.9 times higher in the patients with COVID-19, compared with the control group (p = 0.0005). The NLRP3 expression was also 6.04 times greater in the infected patients, compared with the healthy individuals (p < 0.0001). On the other hand, the miR-223 expression was 5.37 times lower in the case group, compared with the controls (p = 0.04). Conclusion: The study findings indicated the potential role of miR-223 and the dysregulation of NLRP3 inflammasome followed by IL-1β, as a regulatory factor in the pathogenesis of COVID-19, like that in other inflammatory diseases.
Article
Full-text available
Mortality and morbidity associated with COVID-19 continue to be significantly high worldwide, owing to the absence of effective treatment strategies. The emergence of different variants of SARS-CoV-2 is also a considerable source of concern and has led to challenges in the development of better prevention and treatment strategies, including vaccines. Immune dysregulation due to pro-inflammatory mediators has worsened the situation in COVID-19 patients. Inflammasomes play a critical role in modulating pro-inflammatory cytokines in the pathogenesis of COVID-19 and their activation is associated with poor clinical outcomes. Numerous preclinical and clinical trials for COVID-19 treatment using different approaches are currently underway. Targeting different inflammasomes to reduce the cytokine storm, and its associated complications, in COVID-19 patients is a new area of research. Non-coding RNAs, targeting inflammasome activation, may serve as an effective treatment strategy. However, the efficacy of these therapeutic agents is highly dependent on the delivery system. MicroRNAs and long non-coding RNAs, in conjunction with an efficient delivery vehicle, present a potential strategy for regulating NLRP3 activity through various RNA interference (RNAi) mechanisms. In this regard, the use of nanomaterials and other vehicle types for the delivery of RNAi-based therapeutic molecules for COVID-19 may serve as a novel approach for enhancing drug efficacy. The present review briefly summarizes immune dysregulation and its consequences, the roles of different non-coding RNAs in regulating the NLRP3 inflammasome, distinct types of vectors for their delivery, and potential therapeutic targets of microRNA for treatment of COVID-19.
Article
Inflammasomes are multiprotein complexes responding to various microbes and endogenous danger signals, contributing to initiating the innate protective response of inflammatory diseases. NLRP3 inflammasome is a crucial regulator of pro-inflammatory cytokines (IL-1β and IL-18) production through activating caspase-1. Non-coding RNAs (ncRNAs) are a class of RNA transcripts lacking the ability to encode peptides or proteins. Its dysregulation leads to the development and progression of inflammation in diseases. Recently, accumulating evidence has indicated that NLRP3 inflammasome activation could be modulated by ncRNAs (lncRNAs, miRNAs, and circRNAs) in a variety of inflammatory diseases. This review focuses on the substantial role and function of ncRNAs in the NLRP3 inflammasome activation, providing novel insight for the future therapeutic approach of inflammatory diseases.
Article
Full-text available
Exploring the molecular mechanisms that prevent inflammation during caloric restriction may yield promising therapeutic targets. During fasting, activation of the nuclear receptor per-oxisome proliferator-activated receptor α (PPARα) promotes the utilization of lipids as an energy source. Herein, we show that ligand activation of PPARα directly upregulates the long non-coding RNA gene Gm15441 through PPARα binding sites within its promoter. Gm15441 expression suppresses its antisense transcript, encoding thioredoxin interacting protein (TXNIP). This, in turn, decreases TXNIP-stimulated NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, caspase-1 (CASP1) cleavage, and proinflammatory interleukin 1β (IL1B) maturation. Gm15441-null mice were developed and shown to be more susceptible to NLRP3 inflammasome activation and to exhibit elevated CASP1 and IL1B cleavage in response to PPARα agonism and fasting. These findings provide evidence for a mechanism by which PPARα attenuates hepatic inflammasome activation in response to metabolic stress through induction of lncRNA Gm15441.
Article
Full-text available
Postpartum depression (PPD) is a kind of mental disorder characterized by persistent low emotions in puerperium. The most significant physiological change in postpartum is lactation which is regulated by oxytocin receptor (OXTR). However, whether OXTR is related to pathological process of PPD and the potential mechanism still remain unclear. In the present study, we prepared hormone-simulated pregnancy (HSP)-induced PPD mouse model and found that the protein level of OXTR in hippocampus of PPD model mice was downregulated and NLRP3 inflammasome was activated. We identified five lncRNAs related to PPD by transcriptome sequencing, including 3 up-regulated ones and 2 down-regulated ones. The five lncRNAs are associated with the signaling pathway of OXTR according to the bioinformatics analysis. Furthermore, we focused on one of the five lncRNAs, Gm14205, and found that it targeted OXTR which inhibited astrocytic NLRP3 inflammasome activation in hippocampal primary astrocytes. These findings illustrate that OXTR has protective effects in PPD by inhibiting NLRP3 inflammasome activation and provides a new strategy for targeting lncRNA Gm14205 in the pathogenesis of PPD.
Article
Full-text available
Hepatic fibrosis is a common pathological consequence of a sustained wound healing response to continuous liver injury, characterized by increased production and accumulation of extracellular matrix. If unresolved, the fibrotic process results in organ failure, and eventually death after the development of cirrhosis. It has been suggested that macrophages play central role in the progression of hepatic fibrosis, which is related to inflammation and pyroptosis, a novel programmed and proinflammatory cell death. However, it remains far less clear if, or how, lncRNAs regulates the activation and pyroptosis of macrophage in hepatic fibrosis. In the present study, we demonstrated that the liver-enriched lncRNA Lfar1, which has been reported to promote hepatic fibrosis through inducing hepatic stellate cells activation and hepatocytes apoptosis, was dysregulated during proinflammatory M1 activation and pyroptosis of macrophage. Our study revealed that silencing lnc-Lfar1 by a lentivirus-shRNA alleviated CCl4- and BDL-induced proinflammatory M1 macrophage activation and NLRP3 inflammasome-mediated pyroptosis. Furthermore, the in vitro experiments demonstrated that lnc-Lfar1 knockdown significantly suppressed LPS- and IFN-γ-induced proinflammatory activation of macrophages, and inhibited LPS/ATP- and LPS/Nigericin-induced NLRP3 inflammasome-mediated pyroptosis. Mechanistically, lnc-Lfar1 regulated LPS- and IFN-γ-induced proinflammatory activation of macrophages through the NF-ĸB pathway. All these data supported our conclusion that lnc-Lfar1 plays a vital role in controlling the activation and pyroptosis of macrophage, thus providing a possible therapeutic target against inflammation-related disorders including hepatic fibrosis.
Article
Full-text available
Cell death and inflammation play critical roles in cardiac fibrosis. During the fibrosis process, inflammation and tissue injury were triggered; however, the mechanisms initiating cardiac fibrosis and driving fibroblast pyroptosis remained largely unknown. In this study, we identified long non-coding RNA (LncRNA)-GAS5 as the key onset of cardiac fibroblast pyroptosis and cardiac fibrosis. Here, we detected ISO-induced cardiac fibrosis models and cardiac fibroblast pyroptosis model by stimulating with LPS. We found that the expression of pyroptosis-related proteins such as caspase 1, NLRP3, and DNMT1 was increased in cardiac fibrosis tissue, while the expression of GAS5 was decreased. The overexpressing of LncRNA GAS5 was shown to increase and inhibit cardiac fibroblast pyroptosis, as well as attenuate caspase 1 and NLRP3 expression in cardiac fibroblast. However, the silencing of GAS5 was also observed; it shows the opposite situation. Furthermore, further studies revealed that treatment of DNMT inhibitor, 5-aza-2-deoxycytidine, or downregulation of DNMT1 led to increased GAS5 expression by reversion of promoter hypermethylation in cardiac fibroblast. Importantly, we have demonstrated that DNMT1 methylation of LncRNA GAS5 leads to cardiac fibroblast pyroptosis via affecting NLRP3 axis. Our findings indicate a new regulatory mechanism for cardiac fibroblast pyroptosis under LPS stress, providing a novel therapeutic target for cardiac fibrosis. Graphical Abstract
Article
Full-text available
Aging and age-related diseases (ARDs) share basic mechanisms largely involving inflammation. A chronic, low-grade, subclinical inflammation called inflammaging occurs during aging. Autophagy defects, oxidative stresses, senescence-associated secretory phenotypes (SASPs), and DNA damage generally contribute to inflammaging and are largely regulated by numerous lncRNA through two-level vicious cycles disrupting cellular homeostasis: (1) inflammaging and the cellular senescence cascade and (2) autophagy defects, oxidative stress, and the SASP cascade. SASPs and inflammasomes simultaneously cause inflammaging. This review discusses the involvement of macrophage inflammaging in various ARDs and its regulation via lncRNA. Among macrophages, this phenomenon potentially impairs its immunosurveillance and phagocytosis mechanisms, leading to decreased recognition and clearance of malignant and senescent cells. Moreover, SASPs extracellularly manifest to induce paracrine senescence. Macrophage senescence escalates to organ level malfunction, and the organism is more prone to ARDs. By targeting genes and proteins or functioning as competing endogenous RNA (ceRNA), lncRNA regulates different phenomena including inflammaging and ARDs. The detailed mechanism warrants further elucidation to obtain pathological evidence of ARDs and potential treatment approaches.
Article
Aims: To explore the involvement of NOD-like receptor protein 3 (NLRP3) inflammasome and M1 macrophage in root resorption (RR). Methods: A rat RR model was established by excessive orthodontic force. After different force-loading time, the expression levels of NLRP3, caspase-1, and interleukin-1β (IL-1β) and distribution of M1 macrophages were analyzed by immunohistochemistry and immunofluorescence staining in vivo. Then, the mechanism of NLRP3 activation was further verified by macrophage and human periodontal ligament cell (hPDLC) co-culture system in vitro. The production levels of NLRP3, caspase-1, pro-caspase-1, and IL-1β in M1 macrophages in the co-culture system were detected by western blot, and the polarization of CD68+IL-1β+ M1 macrophages was detected by immunofluorescence staining. Results: In the rat RR model, NLRP3, caspase-1, IL-1β, and M1 macrophages were expressed in periodontal ligament, mainly concentrated around RR areas. Force-pretreated hPDLCs promoted M1 macrophage polarization and the production of NLRP3, caspase-1 and IL-1β in M1 macrophages in co-culture system. When MCC950, an inhibitor of NLRP3 inflammasome, was added, NLRP3 activation and M1 macrophage polarization were inhibited. Conclusions: In periodontal tissues, hPDLCs stimulated by force promoted M1 macrophage polarization and increased IL-1β production by activating NLRP3 inflammasome in M1 macrophages, thus initiating the occurrence of RR.
Article
Although long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) have been suggested to play important roles in the pathogenesis of diseases, atherosclerosis-related lncRNAs and circRNAs remain rarely reported. This study aimed to explore the underlying molecular mechanisms of atherosclerosis based on the competing endogenous RNA (ceRNA) regulatory hypothesis of lncRNAs and circRNAs. The expression profiles of circRNAs, lncRNAs, and mRNAs in human THP-1 macrophages treated with oxidized low-density lipoprotein (an in vitro atherosclerosis model), or not, were obtained from the Gene Expression Omnibus database under accession numbers GSE107522, GSE54666, and GSE54039, respectively. The present study identified 29 differentially expressed circRNAs in GSE107522, 544 differentially expressed genes (DEGs) in GSE54666, and 502 DEGs and 231 differentially expressed lncRNAs in GSE54039 datasets by using the Linear Models for Microarray Data method. Eight DEGs were found to be shared and expressed with the consistent trend in GSE54666 and GSE54039 datasets. Two of them (ASPH, aspartate beta-hydroxylase; and PDE3B, phosphodiesterase 3B) were suggested to be crucial based on functional enrichment, protein-protein interaction, and ceRNA network analyses. ASPH, through interaction with CACNA2D4 (calcium voltage-gated channel auxiliary subunit alpha2delta 4), may be associated with atherosclerosis by regulating the cellular response to calcium ion; and PDE3B may exert roles in negative regulation of angiogenesis through cross talk with ELMO1 (engulfment and cell motility 1). Furthermore, the expression of ASPH and PDE3B may be regulated by hsa_circ_0028198/hsa_circ_0092317/XIST-miR-543; PDE3B expression may be also modulated by hsa_circ_0092317/hsa_circ_0003546/H19/XIST-miR-326. In conclusion, our identified ceRNA interaction axes may possibly be important targets for treatment of atherosclerosis.
Article
Background and aims: Many clinical trials have demonstrated that statins convey protective effects against atherosclerosis independent of cholesterol-lowering capacities. Other evidence indicates that pyroptosis, a type of programmed cell death, is likely involved in atherosclerosis, but the effects and mechanisms of statins on pyroptosis must be further revealed. Methods: Here, we explored the effects and mechanisms of atorvastatin on pyroptosis in human vascular endothelial cells by quantitative real-time polymerase chain reaction and Western blot analyses. Results: Atorvastatin upregulated long non-coding RNA (lncRNA) NEXN-AS1 and the expression of NEXN at both the mRNA and protein levels in a concentration- and time-dependent manner. Atorvastatin inhibited pyroptosis by decreasing the expression levels of the canonical inflammasome pathway biomarkers NLRP3, caspase-1, GSDMD, IL-1β, and IL-18 at both the mRNA and protein levels. The promotion effects of atorvastatin on NEXN-AS1 and NEXN expression could be significantly abolished by knockdown of lncRNA NEXN-AS1 or NEXN, and its inhibitory effects on pyroptosis were also markedly offset by knock-down of lncRNA NEXN-AS1 or interference of NEXN. Conclusions: These results demonstrated that atorvastatin regulated pyroptosis via the lncRNA NEXN-AS1-NEXN pathway, which provides a new insight into the mechanism of how atorvastatin promotes non-lipid-lower effects against the development of atherosclerosis and gives new directions on how to reverse atherosclerosis.
Article
Long noncoding RNAs (lncRNAs) are defined as transcripts longer than 200 nucleotides that have no or only a low coding potential. They are involved in the progression of multiple diseases by the regulation of mechanisms related to epigenetic modifications and transcriptional and posttranscriptional processing. Recent studies have revealed an important function of lncRNAs in the regulation of pyroptosis, a type of programmed cell death associated with inflammatory responses that plays a critical role in many diseases. Through direct or indirect action on proteins related to the pyroptosis signaling pathway, lncRNAs are involved in the pathological processes related to cardiovascular diseases, kidney diseases, immune diseases and other diseases. Based on the expression characteristics of lncRNAs, this paper reviews the role of lncRNAs in regulating pyroptosis, aiming to provide new ideas for the research of lncRNAs regulating pyroptosis and treating pyroptosis-related diseases.