Elsevier

Molecular Immunology

Volume 131, March 2021, Pages 191-200
Molecular Immunology

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

https://doi.org/10.1016/j.molimm.2020.12.038Get rights and content

Highlights

  • Elucidating the mechanism of NLRP3 activation in disease.

  • Five types of interactive functional crosstalk mechanism between lncRNAs and NLRP3 inflammasome are interpreted.

  • Functional crosstalk mechanism between LncRNAs and NLRP3 inflammasomes in different diseases were systematically summarized.

Abstract

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.

Introduction

Inflammation underlies numerous pathological processes involving the action of various inflammatory cells and inflammatory activating factors and serves as the body's initial adaptive response to external stimuli, stress, tumours, metabolic diseases, and allergies(Medzhitov, 2008, 2010). Long noncoding RNAs (lncRNAs) do not encode any proteins 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 specific 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 transcription factors for DNA regulation and epigenetic regulation of chromatin 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 inflammatory genes and the activation of signalling pathways found abnormal lncRNA expression (Liao et al., 2018). Pyridine-containing domain 3 (NLRP3), also called the inflammasome, is a multimolecular protein complex that performs a specific function for the cytoplasm in the host's innate immunity by secreting inflammatory factors and promoting maturation, such as for interleukin-1β (IL-1β) and interleukin-18 (IL-18)(He et al., 2016a; Zhou et al., 2011). Seasonable activation of the NLRP3 inflammasome has a positive effect on the body; excessive activation of the NLRP3 inflammasome causes programmed cell death termed ‘pyroptosis’ via the secretion of large amounts of proinflammatory cytokines and triggers a highly inflammatory form of caspase-1(Van Opdenbosch and Lamkanfi, 2019). There are usually two signal modes of NLRP3 inflammasome activation: the host's 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 inflammasome in the activation of inflammatory pathways has brought to the forefront new insights into the prophylaxis, diagnosis, and treatment of various diseases(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 inflammasome under different conditions.

Section snippets

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 modifications, and polyadenylation. RNA polymerase 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

The regulatory relationship between lncRNAs and NLRP3 inflammasomes

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 inflammation 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 and NLRP3 in inflammatory diseases

The biological and clinical roles of lncRNAs and the NLRP3 inflammasome have recently been studied extensively in the inflammatory response and inflammatory cascade reaction(Haque et al., 2020; He et al., 2019). Zhang et al. speculated that the lncRNA Neat1 regulated activation of inflammasomes 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

Conclusion

In summary, the interaction of the NLRP3 inflammasome and lncRNAs has been most intensively investigated in various diseases, including cancer, pyroptosis, Alzheimer's disease, neuroinflammation, cardiovascular diseases, I/R injury, and inflammatory diseases. There is increasing evidence that lncRNAs mainly regulate the NLRP3 inflammasome associated with pathophysiological processes pre- and posttranscription. We still know very little about their precise regulatory mechanisms during the

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.

Author:Wenqiang Tao Contribution:Review and revise the papers.

Author:Rui Xiao Contribution:Review and revise the papers Author:Jianguo Zhang Contribution:Review and edit the article. Author:Qiang Shao Contribution:Review and revise the papers.

Author:Wei Dai Contribution:Review and revise the papers.

Author: Keqian Qian

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 conflict of interest in this study. All authors declare that they have no conflict 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 Affiliated Hospital of Nanchang University.

References (121)

  • J.A. Heward et al.

    Long non-coding RNAs in the regulation of the immune response

    Trends Immunol.

    (2014)
  • S.-x. Hou et al.

    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.

    (2014)
  • J. Hu et al.

    LncRNA ANRIL promotes NLRP3 inflammasome activation in uric acid nephropathy through miR-122-5p/BRCC3 axis

    Biochimie

    (2019)
  • M. Lamkanfi et al.

    Nlrp3: an immune sensor of cellular stress and infection

    Int. J. Biochem. Cell Biol.

    (2010)
  • X. Li et al.

    Long noncoding RNA MALAT1 regulates renal tubular epithelial pyroptosis by modulated miR-23c targeting of ELAVL1 in diabetic nephropathy

    Exp. Cell Res.

    (2017)
  • K. Liao et al.

    The research progress of LncRNA involved in the regulation of inflammatory diseases

    Mol. Immunol.

    (2018)
  • X. Liu et al.

    Nuclear factor E2-Related Factor-2 negatively regulates NLRP3 inflammasome activity by inhibiting reactive oxygen species-induced NLRP3 priming

    Antioxid. Redox Signal.

    (2016)
  • A. Lu et al.

    Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes

    Cell

    (2014)
  • R. Medzhitov

    Inflammation 2010: new adventures of an old flame

    Cell

    (2010)
  • J.-W. Nam et al.

    Incredible RNA: dual functions of coding and noncoding

    Mol. Cells

    (2016)
  • J.-H. Park et al.

    Lipopolysaccharide directly stimulates Th17 differentiation in vitro modulating phosphorylation of RelB and NF-κB1

    Immunol. Lett.

    (2015)
  • L. Salmena et al.

    A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language?

    Cell

    (2011)
  • Y.A. Samra et al.

    Cepharanthine and Piperine ameliorate diabetic nephropathy in rats: role of NF-κB and NLRP3 inflammasome

    Life Sci.

    (2016)
  • K. Schroder et al.

    The inflammasomes

    Cell

    (2010)
  • Y. Song et al.

    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.

    (2019)
  • N. Van Opdenbosch et al.

    Caspases in cell death, inflammation, and disease

    Immunity

    (2019)
  • Y.H. Wang et al.

    Isosibiricin inhibits microglial activation by targeting the dopamine D1/D2 receptor-dependent NLRP3/caspase-1 inflammasome pathway

    Acta Pharmacol. Sin.

    (2019)
  • G.-C. Wu et al.

    Emerging role of long noncoding RNAs in autoimmune diseases

    Autoimmun. Rev.

    (2015)
  • T. Wu et al.

    The BET family member BRD4 interacts with OCT4 and regulates pluripotency gene expression

    Stem Cell Reports

    (2015)
  • L.-M. Wu et al.

    Atorvastatin inhibits pyroptosis through the lncRNA NEXN-AS1/NEXN pathway in human vascular endothelial cells

    Atherosclerosis

    (2020)
  • F.G. Bauernfeind et al.

    Cutting edge: NF-κB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression

    J. Immunol.

    (2009)
  • Y. Bordon

    Trans-Golgi network breaks away to activate NLRP3

    Nat. Rev. Immunol.

    (2019)
  • C.N. Brocker et al.

    Long non-coding RNA Gm15441 attenuates hepatic inflammasome activation in response to metabolic stress

    bioRxiv

    (2019)
  • W.K. Chen et al.

    lnc RNA s: novel players in intervertebral disc degeneration and osteoarthritis

    Cell Prolif.

    (2017)
  • J.C. de Oliveira et al.

    Long non‐coding RNAs in cancer: another layer of complexity

    J. Gene Med.

    (2019)
  • T. Derrien et al.

    The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression

    Genome Res.

    (2012)
  • J. Ding et al.

    Pore-forming activity and structural autoinhibition of the gasdermin family

    Nature

    (2016)
  • Y. Dombrowski et al.

    Cytosolic DNA triggers inflammasome activation in keratinocytes in psoriatic lesions

    Sci. Transl. Med.

    (2011)
  • J.A. Duncan et al.

    Cryopyrin/NALP3 binds ATP/dATP, is an ATPase, and requires ATP binding to mediate inflammatory signaling

    Proc. Natl. Acad. Sci.

    (2007)
  • J.M. Engreitz et al.

    Local regulation of gene expression by lncRNA promoters, transcription and splicing

    Nature

    (2016)
  • J.M. Engreitz et al.

    Long non-coding RNAs: spatial amplifiers that control nuclear structure and gene expression

    Nat. Rev. Mol. Cell Biol.

    (2016)
  • S. Ergun et al.

    Oncocers: ceRNA-mediated cross-talk by sponging miRNAs in oncogenic pathways

    Tumor Biol.

    (2015)
  • L. Franchi et al.

    Sensing and reacting to microbes through the inflammasomes

    Nat. Immunol.

    (2012)
  • L. Freeman et al.

    NLR members NLRC4 and NLRP3 mediate sterile inflammasome activation in microglia and astrocytes

    J. Exp. Med.

    (2017)
  • J. Guo et al.

    Long noncoding RNA: an emerging player in diabetes and diabetic kidney disease

    Clin. Sci.

    (2019)
  • T. Gutschner et al.

    The hallmarks of cancer: a long non-coding RNA point of view

    RNA Biol.

    (2012)
  • M. Guttman et al.

    Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals

    Nature

    (2009)
  • M.R. Hadjicharalambous et al.

    Long non-coding RNAs and the innate immune response

    Noncoding RNA

    (2019)
  • M.E. Haque et al.

    Targeting the microglial NLRP3 inflammasome and its role in Parkinson’s disease

    Mov. Disord.

    (2020)
  • Y. He et al.

    NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux

    Nature

    (2016)
  • Cited by (0)

    View full text