Vol 62, No 4 (2024)
Original paper
Published online: 2024-12-30

open access

Page views 475
Article views/downloads 259
Get Citation

Connect on Social Media

Connect on Social Media

Anti-atherosclerotic effects of LncRNA NEXN-AS1 by regulation of canonical inflammasome pathway of pyroptosis via NEXN

Fei Chen1, Yun-Yong Lin1, Rui Chen1, Wen-Jie Lu1, Yi-Wen Wu1, Shaodong Qiu1, Limei Wu2
Pubmed: 39801217
Folia Histochem Cytobiol 2024;62(4):191-202.

Abstract

Introduction. Pyroptosis is closely related to many chronic diseases including atherosclerosis, but the potential pathomechanisms are still unclear. This study was aimed at exploring how lncRNAs may contribute to pyroptosis and the potential mechanisms.

Material and methods. We performed in vitro assays to investigate the effects of a relatively newly discovered lncRNA, NEXN-AS1, on pyroptosis. Two types of vascular wall cells, i.e. human vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), were used as cell models in this study. A previously constructed lentivirus vector was used to overexpress lncRNA NEXN-AS1 and a small interfering RNA (siRNA) mimic against NEXN to knockdown NEXN. The mRNA and protein levels of molecules of pyroptosis in the canonical inflammasome pathway, including nucleotide-binding oligomerisation segment-like receptor family 3 (NLRP3), caspase-1, gasdermin D (GSDMD), interleukin-1β (IL-1β), and interleukin-18 (IL-18) were detected using quantitative real-time PCR and Western blot analysis, respectively.

Results. We found that lentivirus-mediated overexpression of lncRNA NEXN-AS1 increased the expression levels of NEXN and markedly reduced the expression of critical molecules involved in pyroptosis, including NLRP3, caspase-1, GSDMD, IL-1β, and IL-18, in both VSMCs and ECs. Furthermore, NEXN knockdown could reverse the effects of lncRNA NEXN-AS1 overexpression on pyroptosis.

Conclusions. lncRNA NEXN-AS1 could act as a target for maintaining endothelium homeostasis, increasing plaque stability, and delaying the progression of atherosclerosis.

ORIGINAL PAPER

Anti-atherosclerotic effects of LncRNA NEXN-AS1 by regulation of canonical inflammasome pathway of pyroptosis via NEXN

Fei Chen1Yun-Yong Lin1Rui Chen1Wen-Jie Lu1Yi-Wen Wu1Shaodong Qiu1Limei Wu2
1Department of Ultrasound, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
2Department of Clinical Laboratory, Guangzhou Twelfth People’s Hospital, Guangzhou, China

Correspondence addresses:

Li-Mei Wu,

Department of Clinical Laboratory, Guangzhou Twelfth People’s Hospital,

1 Tianqiang Road, Tianhe District, Guangzhou 510620, China;

tel: +86 020-38665627; e-mail: oldrabbit2007@yeah.net

Shao-Dong Qiu,

Department of Ultrasound, Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Haizhu District, Guangzhou 510260, China;

tel: +86 020-34152520; e-mail: qiushandongqsd@126.com

Submitted:

22 August, 2024

Accepted after reviews:

10 December, 2024

Available as Online first:

30 December, 2024

Abstract
Introduction. Pyroptosis is closely related to many chronic diseases including atherosclerosis, but the potential pathomechanisms are still unclear. This study was aimed at exploring how lncRNAs may contribute to pyroptosis and the potential mechanisms.
Material and methods. We performed in vitro assays to investigate the effects of a relatively newly discovered lncRNA, NEXN-AS1, on pyroptosis. Two types of vascular wall cells, i.e. human vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), were used as cell models in this study. A previously constructed lentivirus vector was used to overexpress lncRNA NEXN-AS1 and a small interfering RNA (siRNA) mimic against NEXN to knockdown NEXN. The mRNA and protein levels of molecules of pyroptosis in the canonical inflammasome pathway, including nucleotide- -binding oligomerisation segment-like receptor family 3 (NLRP3), caspase-1, gasdermin D (GSDMD), interleukin-1β (IL-1β), and interleukin-18 (IL-18) were detected using quantitative real-time PCR and Western blot analysis, respectively.
Results. We found that lentivirus-mediated overexpression of lncRNA NEXN-AS1 increased the expression levels of NEXN and markedly reduced the expression of critical molecules involved in pyroptosis, including NLRP3, caspase-1, GSDMD, IL-1β, and IL-18, in both VSMCs and ECs. Furthermore, NEXN knockdown could reverse the effects of lncRNA NEXN-AS1 overexpression on pyroptosis.
Conclusions. lncRNA NEXN-AS1 could act as a target for maintaining endothelium homeostasis, increasing plaque stability, and delaying the progression of atherosclerosis.
Keywords: atherosclerosis; canonical inflammasome pathway; lncRNA; pyroptosis; VSMC; endothelial cell

Introduction

Atherosclerosis is one of the most common chronic progressive inflammatory processes affecting large and medium-sized arteries [1]. Epidemiological studies have revealed that cardiovascular diseases (CVDs) caused by atherosclerosis account for ca.50% of deaths worldwide. Despite significant advances in pharmacological treatments of atherosclerosis in recent decades, CVDs continue to pose a worldwide health threat due to their ever-increasing morbidity and mortality [2]. As a result, finding efficacious molecular targets may provide a new research direction in order to understand the pathogenesis of atherosclerosis and help predict, prevent, and treat atherosclerosis.

A variety of theories have been proposed to explain the pathogenesis of atherosclerosis, including the hyperlipidemia theory [3], the lipid peroxidation theory [4], the thrombosis theory [5], the senescence-related theory [6], and the inflammatory response theory [7]. Of these, the inflammatory response theory has been the most widely researched and is internationally recognised as being closely related to the development of atherosclerosis [8, 9]. However, the mechanism for regulating inflammatory responses, and preventing or reversing atherosclerosis, remains unknown.

Pyroptosis is a type of pro-inflammatory programmed cell death characterised by an intense inflammatory response and rapid plasma membrane rupture [10]. Pyroptosis has received much attention in recent years because of its role in several biological processes, including atherosclerosis [11]. Recent studies have indicated that pyroptosis in endothelial cells (ECs) [12], vascular smooth muscle cells (VSMCs), and macrophages [13] could promote the development of atherosclerosis and lead to instability in the atherosclerotic lesion. However, the molecular mechanisms of how pyroptosis is involved in atherosclerosis are still not fully understood.

Long non-coding RNAs (lncRNAs), a class of RNAs with transcripts longer than 200 nucleotides but no protein-coding capacities [14], have been shown to play critical roles in the regulation of gene expression through gene imprinting [15], chromatin remodelling [16], cell cycle regulation [17], regulation of pre-mRNA degradation [18], and transcriptional control [19]. Recently, a growing number of lncRNAs have been identified as being involved in the development of numerous diseases, such as CVDs and several cancers [20, 21]. lncRNAs ANRIL, CDKN2B-AS1, RNCR3, APPAT, H19, and FA2H-2, in particular, have been shown to influence the occurrence and progression of atherosclerosis by regulating vascular function, cholesterol efflux, cell proliferation, survival and migration, apoptosis, autophagy, and many more signal pathways [22–27].

Clarifying the biological function of lncRNAs would provide fresh insight into exploiting new targets for atherosclerosis treatment and prevention. The lncRNA NEXN-AS1 is a newly discovered non-coding RNA with low expression in atherosclerotic plaques [28]. Our previous study showed that the lncRNA NEXN-AS1 exerted anti-atherosclerotic effects by downregulating the expression of adhesion molecules and inflammatory cytokines and inhibiting monocyte adhesion to endothelial cells [28].

Atherosclerosis, pyroptosis, and lncRNA NEXN-AS1 are all closely related to inflammation in different ways. However, the precise inter-relationships between atherosclerosis, pyroptosis, and lncRNA NEXN-AS1 in the inflammatory response process are still unclear. We recently discovered that lncRNA NEXN-AS1 could mediate the inhibition effects of atorvastatin on pyroptosis [29]. These findings suggested potential links between pyroptosis and lncRNA NEXN-AS1 that needed to be further clarified.

NEXN is a cognate gene of NEXN-AS1 that encodes the F-actin protein NEXN (also known as nexilin or nelin) found at cell junctions. NEXN has been reported to have an anti-atherosclerotic effect [28]. Furthermore, studies have also revealed that mutations in the NEXN gene can cause cardiomyopathy [30], and it is a susceptible gene for cardiovascular disease (CVD) in the Chinese Han population [31]. Although the above data established NEXN as a protective gene in the development of CVD, the mechanisms by which NEXN influences atherosclerosis require further investigation.

In this study, we found that lncRNA NEXN-AS1 could inhibit key molecules of the pyroptosis canonical inflammasome pathway in human umbilical vein endothelial cells (HUVECs) and VSMCs by regulating its adjacent gene NEXN to exert its anti-inflammatory and anti-atherosclerotic effects.

MATERIALS AND METHODS

Materials

We purchased a reverse transcription kit (PrimeScript RT Reagent Kit, DRR037A) and a real-time polymerase chain reaction kit (SYBR® Premix Ex Taq™ II, DRR820A) from TaKaRa Bio, Inc. (Shiga, Japan). We also obtained a transfection reagent (Lipofectamine 3000) from Invitrogen (Carlsbad, CA, USA) and Optimised medium (Opti-MEM) from Gibco BRL (Paisley, UK). All other chemicals used in this study were of pharmaceutical grade and purchased from commercial suppliers.

Cells and culture parameters

Human umbilical vein endothelial cells (HUVECs) and human vascular smooth muscle cells (VSMCs) were purchased from the American Type Culture Collection (Manassas, VA, USA). Cells were maintained in Dulbecco’s modified Eagle’s medium (Gibco, Gaithersburg, MD, USA) supplemented with 10% foetal bovine serum (NLRP3, Gaithersburg, MD, USA) and antibiotics (penicillin, 100 U/mL; streptomycin, 100 mg/mL) at 37°C in a humidified incubator containing 5% CO2.

Construction of lentiviral vectors

The lentivirus-mediated overexpression vector of lncRNA NEXN-AS1 was constructed using previously reported methods [28]. In summary, the total RNA was extracted from the cultured cells using a TRIzol reagent (Invitrogen). First- -strand complementary DNA (cDNA) of lncRNA NEXN-AS1 was synthesised using M-MLV Reverse Transcriptase (Promega, Madison, WI, USA) and a PrimeScript RT reagent kit (TaKaRa). NEXN-AS1 cDNA was amplified using PCR and inserted into the pLKD-CMV-EGFP-2A-Puro-U6-shRNA vector as described earlier [28]. The proper sequence of the NEXN-AS1 gene in this construct was then verified by sequencing. This construct (henceforth referred to as LV-NEXN-AS1) and the control vector (Obio Technology Corp. Ltd., Shanghai, China) were used to transfect both cultured HUVECs (CRL-1730, ATCC) and VSMCs (CRL-1999, ATCC) at an infection multiplicity of 20 transfection units per cell in the presence of 8 mg/mL of polybrene. After 24 hours, the cells were rinsed with fresh complete medium. We used quantitative RT-PCR to confirm that the NEXN-AS1 overexpression was effective [28].

RNA interference using small interfering RNA (siRNA) mimics in transfection

The siRNA against NEXN (siNEXN) and a non-relevant 21-nucleotide control siRNA (negative control, siNC) were procured from Ribo Biotechnology (Guangzhou, China). The sequences of the siRNAs are set out in Table 1. HUVECs (2×106 cells/well) were seeded into the wells of 6-well plates (2 × 106 cells/well). After achieving 80% confluence, the cells were cultured at room temperature for 20 min in Opti-MEM medium (Gibco BRL, Big Island, NY, USA) containing 5 µL of Lipofectamine 3000 reagent (Invitrogen) and 10 µL of 50 nM siRNA (Ribo) or siNC. Subsequently, the siRNA combination (siNC, control/single LV-NEXN-AS1 group; siNEXN, single siNEXN/LV-NEXN-AS1 and siNEXN group) was carefully and gradually added to each well. qRT-PCR and Western blot analyses were conducted after 24 h of treatment.

Table 1. Oligonucleotide sequences used in this study

Gene

Sequence

Gene

Sequence

NEXN-AS-RT

5'-GGGAGCAATAGACAGATGAAG-3'

Caspase-1-F

5'-AGTGCAGGACAACCCAGCTATG-3'

NEXN-AS1-F

5'-AAGGAATGAGGCTGAAATGG-3'

Caspase-1R

5'-CAAGACGTGTGCGGCTTGA-3'

NEXN-AS1-R

5'-AGGAAAACTTGGCCAAAGGT-3'

GSDMD-F

5'-GGGGACTGCTGGGATGAAAA-3'

U6-RT

5'-AACGCTTCACGAATTTGCGT-3v

GSDMD-R

5'-GGAACAGGGAGGCATAGAGC-3'

U6-F

5'-CTCGCTTCGGCAGCACA-3v

IL--F

5'-TCGCCAGTGAAATGATGGCTTA-3'

U6-R

5'-AACGCTTCACGAATTTGCGT-3v

IL--R

5'-GTCCATGGCCACAACAACTGA-3'

NEXN-F

5'-ACTGTGAAGGGTAGATTTGCTG-3'

IL-18-F

5'-GACCTTCCAGATCGCTTCCTC-3'

NEXN-R

5'-TCTGCGTTTTCGTTCCTCCT-3'

IL-18-R

5'-GATGCAATTGTCTTCTACTGGTTC-3'

NLRP3-F

5'-AAAGCCAAGAATCCACAGTGTAAC-3'

GADPH-F

5'-GCACCGTCAAGGCTGAGAAC-3'

NLRP3-R

5'-TTGCCTCGCAGGTAAAGGT-3'

GADPH-R

5'-TGGTGAAGACGCCAGTGGA-3'

si-h-NEXN

5'-GGAGATGATTCACTACTTA-3'

RNA isolation and qRT-PCR analysis

The total RNA was extracted from the cultured cells using the TRIzol reagent (Invitrogen). The first-strand complementary DNA (cDNA) of lncRNA NEXN-AS1 and mRNAs of NEXN, NLRP3, caspase-1, IL-1β and IL-18 were synthesised using M-MLV Reverse Transcriptase (Promega, Madison, WI, USA) and the PrimeScript RT reagent kit (TakaRa), respectively. Both reactions contained 2 µg of total RNA as template strands in a reaction volume of 20 µL. qRT-PCR was performed to measure the expression levels of lncRNA NEXN-AS1, with U6 as an endogenous control and mRNAs with GAPDH as an endogenous control, using a SYBR Green Master Mix (TaKaRa) and a LightCycler 480 Fast Real-Time PCR system (Roche Molecular Systems, Inc., Pleasanton, CA, USA). The fold-differences in expression levels were calculated using the 2-ΔΔCt method. All experimental procedures were carried out following the manufacturer’s instructions. Each sample was measured in triplicate, and the mean values were used for comparative analysis. The primer sequences are listed in Table 1, and all primers for qRT-PCR were synthesised by Sangon Biotech (Shanghai, China). The qRT-PCR cycling protocol consisted of an initial preincubation step at 95°C for 20 seconds, followed by 40 cycles at 95°C for 10 seconds, 60°C for 20 seconds, and a final extension step at 70°C for 1 second.

Western blot analysis

The method we used for harvesting the cells and pre­paring protein extracts in this study was as described in [32]. Protein extraction from the cultured cells was performed using a radio immunoprecipitation assay buffer (Biocolor Ltd., Belfast, UK), quantified with a BCA protein assay kit (KeyGen Biotechnologies, Nanjing, China), and separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) at a concentration of 12%. The separated proteins were then transferred to cellulose acetate membranes and incubated overnight at 4°C with the following antibodies (all at 1:1,000 dilution): mouse polyclonal anti-nexilin antibodies (ab213628, Abcam, Cambridge, UK); rabbit recombinant monoclonal anti-GSDMD antibody (ab210070, Abcam); rabbit polyclonal anti-NLRP3 (A5652, ABclonal, Wuhan, China); caspase-1 (2225, Cell Signaling Technology, Beverly, MA, USA), -IL-1β (sc-12742, Santa Cruz Biotechnology, Inc., Dallas, TX, USA), and -IL-18 (A16288, ABclonal), as well as mouse polyclonal anti-β-tubulin-specific antibodies (diluted 1:2,000; ab6046, Abcam) as an internal control. After being conjugated with primary antibodies, the membranes were incubated with goat anti-rabbit or goat anti-mouse secondary antibodies (diluted 1:5,000; Bioss Antibodies, Beijing, China). The proteins were visualised using the chemiluminescence detection method (ECL Plus Western Blot Detection System, GE Healthcare, Piscataway, NJ, USA) and analysed using Image J software (free web version, https://www.imagej.net/).

IL-18 concentration in cell supernatants

The concentration of IL-18 in HUVECs and VSMCs supernatants after treatment with LV-NEXN-AS1 or siNEXN was measured using a commercially available specific enzyme-linked immunosorbent assay (ELISA) kit (RK00001, ABclonal), as per the manufacturer’s instructions. The optical density (OD) value was measured using an ELISA plate reader (R&D Systems, Minneapolis, MN, USA) at a wavelength of 450 nm. The standard substances and sample dilutions were added to the ELISA plate in duplicate, and the average concentration was calculated as the protein level (pg/mL) in the cell culture supernatant. The average OD values of the standards were used to create a standard curve. The results were calculated with Excel 2010 (Microsoft, Redmond, WA, USA).

Statistical analysis

Data was analysed using IBM SPSS Statistics for Windows, version 23.0 (IBM Corporation, Armonk, NY, USA) and GraphPad software (GraphPad Software, Inc., La Jolla, CA, USA). Unless otherwise indicated, data was presented as mean ± standard deviation or median (interquartile range). The results of the RT-PCR and Western blot analyses were assessed with a Student’s t-test for two-group comparisons and one-way analysis of variance followed by a Student-Newman-Keuls test for comparison of more than two groups. A two-tailed probability (P) value of < 0.05 was considered statistically significant.

RESULTS

Overexpression of lncRNA NEXN-AS1 increased expression of NEXN in HUVECs and VSMCs

Our bioinformatics analysis showed that lncRNA NEXN-AS1 and NEXN were situated nearby on chromo­some 1 and had opposite transcription directions. Previously, we found that lncRNA NEXN-AS1 could regulate the expression of its cognate gene NEXN in HUVECs and murine animal models [28]. In this study, we further explored the effects of lncRNA NEXN-AS1 on NEXN in both HUVECs and VSMCs. The NEXN-AS1 gene was transferred into HUVECs and VSMCs by lentiviral vectors (LV-NEXN-AS1), with empty lentiviral vectors (LV-Mock) as negative controls. We used qRT-PCR and Western blot to measure the expression level of NEXN-AS1.

The lncRNA levels of NEXN-AS1 were enhanced 52-fold in HUVECs and 80-fold in VSMCs by LV-NEXN-AS1 transfection, and the mRNA levels of NEXN were very significantly increased, by 1,060% in HUVECs and 833% in VSMCs (Figs. 1A, B). Western blot analysis showed that NEXN protein levels were also significantly upregulated, by 2.99-fold in HUVECs and 4.32-fold in VSMCs, compared to the control group (Figs. 1C, D). These results suggest that overexpression of NEXN-AS1 could increase the expression of NEXN in HUVECs and VSMCs at both mRNA and protein levels.

Figure 1. Expression levels of lncRNA NEXN-AS1 and NEXN are both upregulated by LV-NEXN-AS1 in HUVECs and VSMCs. AD. HVECs and VSMCs were transfected with empty lentivirus vectors (LV-Mock) and LV-NEXN-AS1, respectively. A, B. qRT-PCR analysis of NEXN-AS1 and NEXN relative RNA levels revealed a statistically significant fold difference in mean (± SD) after normalisation against Ct value of U6 or GAPDH housekeeping genes in five independent experiments, each performed in triplicate. Data was analysed using one-way ANOVA with significance levels of *P < 0.05 and **P < 0.01. Resulting bar chart displays these fold differences. C, D. Protein level of NEXN measured with Western blot. C. Images of representative Western blots. D. Based on one-way analysis of five independent experiments with a significance level of *P < 0.05, fold differences in NEXN band intensity after standardisation against β-tubulin, were analysed. Abbreviations: HUVECs — human umbilical vein endothelial cells; HVECs — human vascular endothelial cells; SD — standard deviation; VSMCs — vascular smooth muscle cells.
Overexpression of lncRNA NEXN-AS1 inhibited pyroptosis via canonical inflammasome pathway

Pyroptosis was recently identified as a new type of cell death accompanied by the activation of many cytokines, including inflammatory cytokines.

We hypothesised that because inflammatory cytokines were found to be regulated by lncRNA NEXN-AS1 in our previous research [28], there might be some relationship between lncRNA NEXN-AS1 and pyroptosis with the same inflammatory response features. Therefore, in this study we investigated the effects of lncRNA NEXN-AS1 on pyroptosis.

Firstly, we transfected HUVECs and VSMCs with LV-NEXN-AS1 to overexpress lncRNA NEXN-AS1. Next, we used qRT-PCR and Western blot analyses to assess the expression levels of the canonical inflammasome pathway molecules of pyroptosis (NLRP3, caspase-1, GSDMD, IL-1β, and IL-18). The supernatant of both cell types was collected to determine IL-18 concentration using an ELISA kit. The qRT-PCR results showed that the overexpression of lncRNA NEXN-AS1 significantly downregulated mRNA levels of canonical inflammasome pathway molecules (NLRP3, caspase-1, GSDMD, IL-1β and IL-18) in two different cell types (Figs. 2A, B). Western blot analysis showed that the overexpression of lncRNA NEXN-AS1 decreased protein levels of the canonical inflammasome pathway proteins [NLRP3, caspase-1p45 full length, cleavage p20 fragment, cleavage IL-1β p17, IL-18 and C-terminal GSDMD (GSDMD-C)], while there were no significant differences in the expression levels of the inactive precursors of IL-1β and GSDMD full length (Figs. 2B, D, F). Notably, the expression levels of IL-18 detected by ELISA were also similarly decreased in the supernatant of the abovementioned two cell types (Figs. 2C, G).

Figure 2. Inflammasome pathway biomarkers of pyroptosis are inhibited by NEXN-AS1 on both mRNA and protein levels. AG. LV-Mock vector (LV-Mock) was transfected onto human HVECs and LV-NEXN-AS1 was transfected onto vascular smooth muscle cells (VSMCs). A, E. Quantitative real-time PCR method was used to measure relative levels of NLRP3, caspase-1, GSDMD, IL-1β, and IL-18, which are biomarkers of canonical inflammasome pathway. As shown in column chart, five independent experiments, each performed in triplicate (*P > 0.05) by one-way analysis of variance analysis, were normalised against Ct value of housekeeping gene GAPDH. B, D, E. Western blots reveal presence of NLRP3, caspase-1p45, caspase-1p20, IL-1(p31), IL-1(p17), and IL-18 proteins. D. Representative Western blot images. B, E. Analysis of variance using one-way P > 0.05 for NLRP3, caspase-1p45, caspase-1p20, GSDMD full length, GSDMD-C, IL-1β p31, IL-1β p17, and IL-18 after standardisation against housekeeping protein β-tubulin, *P > 0.05. C, G. IL-18 concentration in cell supernatant was determined by ELISA. Abbreviations: GSDMD — gasdermin D; HVECs — human vascular endothelial cells; VSMCs — vascular smooth muscle cells.

The results of this study demonstrate that the overexpression of lncRNA NEXN-AS1 in HUVECs and VSMCs inhibits the expression levels of pyroptosis-associated canonical inflammasome pathway molecules.

Knock-down of NEXN did not affect expression of lncRNA NEXN-AS1

Because NEXN gene expression was upregulated by the overexpression of lncRNA NEXN-AS1, we investigated the effects of NEXN on lncRNA NEXN-AS1. We transfected HUVECs and VSMCs with siNEXN to explore whether NEXN affected the expression level of lncRNA NEXN-AS1. As shown in Figs. 3A and B, HUVECs and VSMCs were transfected with small interference vectors carrying NEXN (siNEXN) to downregulate the expression of NEXN with small interference control vectors (siNC) as a negative control, and then total RNA and proteins were extracted for qRT-PCR and Western blot, respectively.

The findings suggested that siRNA-induced knockdown of NEXN had no influence on NEXN-AS1 expression, but did markedly reduce the expression of NEXN at both the mRNA and protein levels compared to the siNC group (Figs. 3AD).

Figure 3. siNEXN knock-down does not affect NEXN-AS1 expression in HUVECs and VSMCs. AD. HUVECs and VSMCs were transfected with siNC and siNEXN, respectively. A, B. By using qRT-PCR, NEXN-AS1 and NEXN relative RNA levels were determined. In column chart, fold difference in mean (± SD) is calculated after standardisation against Ct value of housekeeping gene U6 in NEXN-AS1 or GAPDH in NEXN based on five independent experiments, each performed three times, *P < 0.05 using t-test. C, D. Protein level of NEXN was measured by Western blot analysis. C. Representative Western blot images. D. Differences in band intensities of NEXN after standardisation against β-tubulin, internal control protein, are shown in table below. **P < 0.05 using t-test. Abbreviations: HUVECs — human umbilical vein endothelial cells; SD — standard deviation; VSMCs — vascular smooth muscle cells.
Knockdown of NEXN upregulated expression of biomarkers in canonical inflammasome pathway of pyroptosis

Based on the above data, we have confirmed that the overexpression of lncRNA NEXN-AS1 could suppress the expression level of pyroptosis-related molecules. Subsequently, in order to investigate whether the function of NEXN was consistent with lncRNA NEXN-AS1 in regulating pyroptosis, we measured the expression levels of the proteins of pyroptosis canonical inflammasome pathway by knockdown of NEXN in HUVECs and VSMCs.

As shown in Figs. 4A and 4E, the mRNA levels of the inflammasome pathway molecules (NLRP3, caspase-1, GSDMD, IL-1β, and IL-18) significantly increased in the siNEXN group. Meanwhile, siRNA-induced knockdown of NEXN increased the protein expression level of NLRP3, cleavage caspase-1 p45 and p20 fragments, GSDMD-C, cleavage IL-1β p17, and IL-18, but did not change the expression level of precursors of IL-1β (p31 form) and GSDMD full length (Fig. 4B, D, and F). Similarly, the level of IL-18 was significantly increased in the supernatant of the above two cell types (Fig. 4C, G).

Figure 4. NEXN knockdown promotes both mRNA and protein levels of pyroptosis biomarkers. AG. HUVECs and VSMCs were transfected with siNC and siNEXN. A, E. qRT-PCR was used to measure relative mRNA levels of NLRP3, caspase-1, GSDMD, IL-1β, and IL-18, which are biomarkers of canonical inflammasome pathway. Column graph displays fold change in average (± SD) following standardisation against Ct value of housekeeping gene GAPDH from five separate experiments, each conducted in triplicate. *P > 0.05 using t-test. B, D, E. Levels of NLRP3, caspase-1p45, caspase-1p20, GSDMD full length, GSDMD-C, IL-1p31 and IL-18 were measured using Western blot analysis. D. Representative Western blot images. B, E. Comparing HUVECs and VSMCs’ NLRP3, caspase-1p45, caspase-1p20, GSDMD full length, GSDMD-C, IL-1β p31, IL-1β p17, IL-18 band intensity after standardisation against housekeeping protein β-tubulin in five independent experiments. *P > 0.05 using t-test. C, G. Concentration of IL-18 in cell supernatant was determined using ELISA assay. Abbreviations: GSDMD — gasdermin D; HUVECs — human umbilical vein endothelial cells; SD — standard deviation; VSMCs — vascular smooth muscle cells.

These results indicated that in both studied cell lines, NEXN exerted the regulatory effects of pyroptosis through the canonical inflammasome pathway, which was consistent with the effects of lncRNA NEXN-AS1.

LncRNA NEXN-AS1 regulated NEXN expression, but NEXN did not affect lncRNA NEXN-AS1 expression

We further examined lncRNA levels of NEXN-AS1 in VSMCs and HUVECs treated with both LV-NEXN-AS1 and siNEXN to discover whether lncRNA NEXN-AS1 could be regulated by NEXN. Compared to the negative control, our results showed that the experimental group treated with both LV-NEXN-AS1 and siNEXN showed significant upregulation of NEXN-AS1 at RNA levels similar to the LV-NEXN-AS1 group (Fig. 5A). However, both the mRNA (Figure 5B) and protein levels (Figs. 5C, D) of NEXN recovered to normal.

These findings showed that the upregulation effects of LV-NEXN-AS1 on NEXN expression could be neutralised by knockdown of NEXN. They also indicated that lncRNA NEXN-AS1 could regulate the expression levels of NEXN, but NEXN could not influence the expression level of lncRNA NEXN-AS1.

Figure 5. Effects of LV-NEXN-AS1 on expression of lncRNA NEXN-AS1 and NEXN can be reversed by siNEXN. AE. HUVECs and VSMCs were treated with LV-mock vs. siNC, LV-NEXN-AS1 vs. siNC, LV-mock vs. siNEXN, LV-NEXN-AS1 vs. siNEXN, respectively. Then qRT-PCR and Western blot analysis were performed. A. Relative lncRNA levels of NEXN-AS1. B. Relative mRNA levels of NEXN. C. Representative Western blot images of NEXN protein levels. D. Band intensity after standardisation against housekeeping protein β-tubulin. All results were obtained from five independent experiments, each performed in triplicate. Column chart shows fold differences in mean (± SD). Relative RNA levels of NEXN-AS1 and NEXN standardised against housekeeping genes U6 and GAPDH, respectively. Relative protein levels of NEXN standardised against housekeeping protein β-tubulin. *P < 0.05, **P < 0.01 using one-way analysis of variance analysis. Abbreviations: HUVECs — human umbilical vein endothelial cells; SD — standard deviation; VSMCs — vascular smooth muscle cells.
LncRNA NEXN-AS1 inhibited pyroptosis via NEXN

We found that lncRNA NEXN-AS1 and NEXN regulated the expression levels of the canonical inflammasome pathway molecules of pyroptosis, but whether lncRNA NEXN-AS1 inhibited pyroptosis via NEXN remains unclear. In this study, we transfected HUVECs and VSMCs with an LV-mock or an LV-NEXN-AS1 and then incubated them with siNC or siNEXN. We found that the expression of genes encoding the molecules of pyroptosis (NLRP3, caspase-1, GSDMD, IL-1β, and IL-18) was significantly suppressed by LV-NEXN-AS1, and they could be reversed at the levels of mRNA by treatment with siNEXN (Figs. 6A, E).

Figure 6. Knockdown of NEXN can reverse effects of overexpression of lncRNA NEXN-AS1 on canonical inflammasome pathway biomarkers of pyroptosis. AF. LV-mock vs. siNC, LV-NEXN-AS1 vs. siNC, LV-mock vs. siNEXN, and LV-NEXN-AS1 vs. siNEXN were tested on HUVECs and VSMCs, respectively. A, E. Biomarkers of canonical inflammasome pathway, NLRP3, caspase-1, GSDMD, IL-1β, and IL-18 in HUVECs (A) and VSMCs (E), measured by real-time PCR in HUVECs and VSMCs. After standardisation of results against Ct value of housekeeping gene GAPDH, five independent experiments, carried out in triplicate, are shown in column chart, *P < 0.05 using one-way analysis of variance analysis. B, D, E. Measurement of NLRP3, caspase-1p45, caspase-1p20, and GSDMD-C protein levels using Western blot technique. D. Representative Western blot images. B, E. In five independent experiments, mean (± SD) values of NLRP3, caspase-1p45, caspase-1p20, GSDMD full length, GSDMD-C, IL-1β p31, IL-1β p17, and IL-18 band intensity in HUVECs and VSMCs were compared after standardising against band intensity of housekeeping protein β-tubulin, *P < 0.05, **P < 0.05 using one-way analysis of variance analysis. C, G. Concentration of IL-18 in supernatant was measured using ELISA assay. Abbreviations: GSDMD — gasdermin D; HUVECs — human umbilical vein endothelial cells; SD — standard deviation; VSMCs — vascular smooth muscle cells.

Consistent with the mRNA expression, Western blot analysis showed that the protein levels of the molecules of pyroptosis (NLPR3, cleavage caspase-1 p20, cleavage IL-1β p17, IL-18, and GSDMD-C) were downregulated by LV-NEXN-AS1, upregulated by siNEXN, and recovered to normal levels by the synergistic effects of LV-NEXN-AS1 and siNEXN. There were no significant differences in the mRNA (Figs. 6B, E, F) and protein (Figs. 6C, G) levels of the precursors of caspase-1, IL-1β, and GSDMD in two cell types. In addition, our results showed that after transfection with LV-NEXN-AS1, the level of IL-18 produced by HUVECs and VSMCs and secreted into the supernatant decreased, but after transfection with siNEXN, the level of IL-18 increased. Nevertheless, after co-transfection with LV-NEXN-AS1 and siNEXN, the level of IL-18 returned to the normal level (Figs. 6C, G).

All these results indicated that lncRNA NEXN-AS1 could suppress the protein levels of pyroptosis-associated molecules via the regulation of its downstream gene, NEXN.

DISCUSSION

In this study, we have found that in HUVECs and VSMCs, overexpression of lncRNA NEXN-AS1 upregulated the expression of its cognate gene NEXN and downregulated both mRNA and protein levels of pyroptosis-associated molecules in the canonical inflammation pathway (NLRP3, caspase-1, GSDMD, IL-1β, and IL-18). Knockdown of NEXN upregulated the expression of NLRP3, caspase-1, GSDMD, IL-, and IL-18 genes. Furthermore, the downregulation effects on NLRP3, caspase-1, GSDMD, IL-, and IL-18 induced by the overexpression of lncRNA NEXN-AS1 could be reversed by knocking down NEXN.

Atherosclerosis is a chronic, progressive vascular pathology in which cell death plays a critical role. Pyroptosis is a relatively novel form of programmed cell death characterised by flattening-like rupture, the release of inflammatory factors (such as IL-1β and IL-18), and severe inflammatory reaction [34]. Pyroptosis has been implicated in the atherosclerotic process in several cell type models, including vascular wall cells such as endothelial and smooth muscle cells, and immune cells (such as T-lymphocytes and macrophages) [32–34].

During pyroptosis, a chain reaction occurs in which many molecules and cytokines, including NLRP3, inflammatory caspases (caspase-1 and -11), gasdermin D, and proinflammatory interleukins (IL-1β, IL-18) are involved in a sequence. There are two signals leading to pyroptosis, each with a different initiation mechanism, i.e. the canonical inflammasome pathway and the non-canonical inflammasome pathway. The former is usually stimulated by danger-associated molecular patterns (DAMPs), while the latter is usually stimulated by pathogen-associated molecular patterns (PAMPs) [35]. Cholesterol crystals and hyperlipidemia serve as DAMPs to induce cell pyroptosis via the canonical inflammasome pathway to promote atherosclerosis [36].

NLRP3 organising inflammasomes are recruited, oligomerised, and activated by DAMPs. NLRP3’s N-terminal pyrin segment (PYD) recruits pro-caspase 1 to the inflammasome [37]. A caspase-1 p20 or p10 fragment is then produced following caspase-1 oligomerisation and autoproteolytic cleavage. Caspase-1 that has been activated performs a catalytic function acting as cysteine-dependent protease, causing the cleavage of pro-IL-1β and pro-IL-18 into the biologically active cytokines IL-1β and IL-18, respectively [38, 39]. Caspases also cleave gasdermin D into C-terminal and N-terminal fragments (GSDMD-N), the latter of which initiates pyroptosis by forming non-selective membranes. The GSDMD N-terminal fragment is released from membrane pores when active caspase-1 cleaves GSDMD [38].

Endothelial cells are neatly arranged on the inner wall of blood vessels, forming a physical barrier to maintain blood vessel integrity. Endothelial injury has been shown to be involved in many vascular pathological processes, including thrombosis and atherosclerosis, resulting from the physical barrier destruction and altered secretory profile of ECs [40]. Activated or injured HUVECs secrete large amounts of inflammatory molecules (caspase-1, IL-1β, and IL-18) and adhesion molecules [P-selectin, intercellular adhesion molecule-1 (ICAM-1), as well as vascular cell adhesion molecule-1 (VCAM-1)] [41].

Inflammatory molecules promote the expression of NLRP3, caspase-1, and GSDMD-C by activating the inflammatory reaction, while adhesion molecules recruit monocytes and other inflammatory cells to infiltrate the endothelium and expose it to increased levels of lipids, the main characteristics of hyperlipidemia. NLRP3 and GSDMD expression levels are promoted by caspase-1 in the endothelial injury, which are all the main components of the canonical inflammasome pathway of pyroptosis [42]. They are related to the pyroptosis of HUVECs and could greatly affect the progression of atherosclerosis. Furthermore, the pyroptosis of HUVECs promotes the production of adhesion molecules, the accumulation of inflammatory factors, and the infiltration of multiple leukocytes into the intima. Consequently, these factors lead to the further deterioration of atherosclerosis. Therefore, control of pyroptosis in HUVECs may be a potential strategy to mitigate atherosclerosis, and in this respect the findings from our current study are consistent with previous research.

Our results showed that lncRNAs NEXN-AS1 and NEXN could regulate the expression of NLRP3, caspase-1, GSDMD, IL-, and IL-18, and particularly activate the caspase-1 p20 fragment, GSDMD-N, and cleavage of IL-1β p17. These findings suggest that lncRNA NEXN-AS1 and NEXN could regulate cell pyroptosis through the canonical inflammasome pathway because key molecules of this pathway were all affected. As a result, lncRNA NEXN-AS1 and NEXN have for the first time been reported in this current study as molecular targets for regulating pyroptosis in HUVECs.

VSMCs are important components of the medium layer of the arterial wall that play a vital role in the progression of atherosclerosis. On the one hand, migration of VSMCs from the media to the intima of the arteries promotes atherosclerosis [43]. On the other hand, VSMCs keep atherosclerotic plaques stable by producing extracellular matrix molecules such as collagen and elastic fibres [44]. However, VSMC pyroptosis can lead to the loss of collagen fibres and trigger a severe inflammatory response, causing the fibres and plaques to become thinner and unstable. Many pro-atherosclerotic factors, such as a high concentration of oxidized low density lipoprotein cholesterin, can induce VSMC pyroptosis. Pyroptosis could further increase the levels of inflammatory factors by activating in the inflammasomes GSDMD-C, NLRP3, and caspase-1. As a result, the plaques become prone to rupture promoting thrombus formation, which can result in cardiovascular events [11].

Therefore, it is particularly important to identify targets to inhibit pyroptosis of VSMC. In our study, we are the first to find that lncRNA NEXN-AS1 inhibits the expression level of pyroptosis-related molecules in VSMCs via the regulation of its downstream gene, NEXN.

In addition, emerging research is confirming that an increasing number of lncRNAs are involved in pyroptosis. For example, lncRNA MALAT1 has been found to affect the occurrence and development of experimental diabetic nephropathy by interacting with miR-23c to inhibit the expression of closely related pyroptosis molecules such as ELAVL1, NLRP3, caspase-1, and IL-1β in HK-2 cells [45]. LncRNA LINC00339 promotes renal tubular epithelial pyroptosis by regulating the miR-22-3p/NLRP3 pathway [46], and lncRNA MEG3 mediates EC pyroptosis via the miR-223/NLRP3 pathway [47]. These results demonstrate the vital role played by some lncRNAs in regulating pyroptosis.

Consistent with the above research, we found that a previously unknown lncRNA, NEXN-AS1, involved in inflammation and atherosclerosis, could regulate pyroptosis in both HUVECs and VSMCs. Interestingly, both inflammation and pyroptosis were found to be closely related to atherosclerosis [48–50]. Therefore, precise regulation of cell pyroptosis and the release of inflammatory molecules could serve as a new direction for the treatment and prevention of atherosclerosis. Our findings have demonstrated that lncRNA NEXN-AS1 could regulate the expression levels of pyroptosis-related molecules, which should offer new insights into the therapeutic and preventive potential of lncRNA in atherosclerosis. However, we have only demonstrated the expression of pyroptosis molecule alterations. The specific processes of activation of such molecules remain to be identified.

In summary, we have found that lncRNA NEXN-AS1 inhibited pyroptosis via the canonical inflammasome pathway in HUVECs and VSMCs by regulating the expression of its adjacent gene NEXN, suggesting that lncRNAs NEXN-AS1 and NEXN could have anti-inflammatory and anti-atherosclerotic effects by regulating pyroptosis.

These findings have provided a new direction for determining the mechanisms of lncRNAs action in the regulation of atherosclerotic process, as well as new insights into the potential of lncRNAs in the prevention and treatment of atherosclerosis.

Article information and declarations

Data availability statement

The datasets used and/or analysed during the current study are available from the corresponding author upon reasonable request.

Ethics statement

Not applicable.

Authors’ contributions

FC: Acquisition of data, writing of manuscript, obtaining financing; YYL: acquisition, analysis and interpretation ofdata; RC: analysis and interpretation of data, statistical analysis; WJL: acquisition of data, critical revision of manuscript for intellectual content; YWW: statistical analysis, critical revision of manuscript for intellectual content; SQ: conceptualisation and design of research, critical revision of manuscript for intellectual content; LW: conceptualisation and design of research, acquisition of data, obtaining financing.

All authors read and approved the final draft.

Funding

This study was supported by: (1) the Science and Tech­nology Programme of Guangzhou city (grant numbers: 2023A03J0492 and 202102010049); and (2) Research project of Traditional Chinese Medicine Bureau of Guangdong Province (grant number: 20231235).

Acknowledgements

We would like to acknowledge the hard and dedicated work of all the staff who implemented the intervention and evaluation components of this study.

Conflicts of interest

The authors declare that they have no competing interests.

References

  1. Tabas I. Heart disease: death-defying plaque cells. Nature. 2016; 536(7614): 32–33, doi: 10.1038/nature18916, indexed in Pubmed: 27437578.
  2. Kim BS, Chung PW, Park KY, et al. Burden of intracranial atherosclerosis is associated with long-term vascular outcome in patients with ischemic stroke. Stroke. 2017; 48(10): 2819–2826, doi: 10.1161/STROKEAHA.117.017806, indexed in Pubmed: 28798260.
  3. Zhao Y, Yang Y, Xing R, et al. Hyperlipidemia induces typical atherosclerosis development in Ldlr and Apoe deficient rats. Atherosclerosis. 2018; 271: 26–35, doi: 10.1016/j.atherosclerosis.2018.02.015, indexed in Pubmed: 29459263.
  4. Liu S, Hou W, Qin H, et al. Oxidized LDL stimulates lipid peroxidation-derived DNA and protein adducts in human vascular endothelial and smooth muscle cells. J Huazhong Univ Sci Technolog Med Sci. 2015; 35(2): 200–205, doi: 10.1007/s11596-015-1411-8, indexed in Pubmed: 25877352.
  5. McAteer MA, Akhtar AM, von Zur Muhlen C, et al. An approach to molecular imaging of atherosclerosis, thrombosis, and vascular inflammation using microparticles of iron oxide. Atherosclerosis. 2010; 209(1): 18–27, doi: 10.1016/j.atherosclerosis.2009.10.009, indexed in Pubmed: 19883911.
  6. Garrido AM, Bennett M. Assessment and consequences of cell senescence in atherosclerosis. Curr Opin Lipidol. 2016; 27(5): 431– –438, doi: 10.1097/MOL.0000000000000327, indexed in Pubmed: 27261931.
  7. Yang Q, Yuan H, Chen M, et al. Metformin ameliorates the progression of atherosclerosis via suppressing macrophage infiltration and inflammatory responses in rabbits. Life Sci. 2018; 198: 56–64, doi: 10.1016/j.lfs.2018.02.017, indexed in Pubmed: 29452166.
  8. Bäck M, Yurdagul A, Tabas I, et al. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat Rev Cardiol. 2019; 16(7): 389–406, doi: 10.1038/s41569-019-0169-2, indexed in Pubmed: 30846875.
  9. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005; 352(16): 1685–1695, doi: 10.1056/NEJMra043430, indexed in Pubmed: 15843671.
  10. Chen X, He WT, Hu L, et al. Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel- -mediated necroptosis. Cell Res. 2016; 26(9): 1007–1020, doi: 10.1038/cr.2016.100, indexed in Pubmed: 27573174.
  11. Pan J, Han Li, Guo J, et al. AIM2 accelerates the atherosclerotic plaque progressions in ApoE–/– mice. Biochem Biophys Res Commun. 2018; 498(3): 487–494, doi: 10.1016/j.bbrc.2018.03.005, indexed in Pubmed: 29510138.
  12. Zhaolin Z, Jiaojiao C, Peng Wu, et al. OxLDL induces vascular endothelial cell pyroptosis through miR-125a-5p/TET2 pathway. J Cell Physiol. 2019; 234(5): 7475–7491, doi: 10.1002/jcp.27509, indexed in Pubmed: 30370524.
  13. Xu YJ, Zheng L, Hu YW, et al. Pyroptosis and its relationship to atherosclerosis. Clin Chim Acta. 2018; 476: 28–37, doi: 10.1016/j.cca.2017.11.005, indexed in Pubmed: 29129476.
  14. Tan YT, Lin JF, Li T, et al. LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond). 2021; 41(2): 109–120, doi: 10.1002/cac2.12108, indexed in Pubmed: 33119215.
  15. Martins-Taylor K, Chamberlain S. Roles of long non-coding RNAs in genomic imprinting. Molecular Biology of Long Non-coding RNAs. 2013: 95–114, doi: 10.1007/978-1-4614-8621-3_4.
  16. Cajigas I, Leib DE, Cochrane J, et al. Evf2 lncRNA/BRG1/DLX1 interactions reveal RNA-dependent inhibition of chromatin remodeling. Development. 2015; 142(15): 2641–2652, doi: 10.1242/dev.126318, indexed in Pubmed: 26138476.
  17. Kim T, Jeon YJ, Cui Ri, et al. Role of MYC-regulated long noncoding RNAs in cell cycle regulation and tumorigenesis. J Natl Cancer Inst. 2015; 107(4), doi: 10.1093/jnci/dju505, indexed in Pubmed: 25663692.
  18. Liu X, Li D, Zhang W, et al. Long non-coding RNA gadd7 interacts with TDP-43 and regulates Cdk6 mRNA decay. EMBO J. 2012; 31(23): 4415–4427, doi: 10.1038/emboj.2012.292, indexed in Pubmed: 23103768.
  19. Caballero J, Gilbert I, Fournier E, et al. Exploring the function of long non-coding RNA in the development of bovine early embryos. Reprod Fertil Dev. 2014; 27(1): 40–52, doi: 10.1071/RD14338, indexed in Pubmed: 25472043.
  20. Huarte M. The emerging role of lncRNAs in cancer. Nat Med. 2015; 21(11): 1253–1261, doi: 10.1038/nm.3981, indexed in Pubmed: 26540387.
  21. Yang Y, Deng X, Li Q, et al. Emerging roles of long noncoding RNAs in cholangiocarcinoma: Advances and challenges. Cancer Commun (Lond). 2020; 40(12): 655–680, doi: 10.1002/cac2.12109, indexed in Pubmed: 33142045.
  22. Shan K, Jiang Q, Wang XQ, et al. Role of long non-coding RNA-RNCR3 in atherosclerosis-related vascular dysfunction. Cell Death Dis. 2016; 7(6): e2248, doi: 10.1038/cddis.2016.145, indexed in Pubmed: 27253412.
  23. Meng F, Yan J, Ma Q, et al. Expression status and clinical significance of lncRNA in the progression of atherosclerosis. PeerJ. 2018; 6: e4246, doi: 10.7717/peerj.4246, indexed in Pubmed: 29372117.
  24. Bitarafan S, Yari M, Broumand MA, et al. Association of increased levels of lncRNA H19 in PBMCs with risk of coronary artery disease. Cell J. 2019; 20(4): 564–568, doi: 10.22074/cellj.2019.5544, indexed in Pubmed: 30124004.
  25. Chi JS, Li JZ, Jia JJ, et al. Long non-coding RNA ANRIL in gene regulation and its duality in atherosclerosis. J Huazhong Univ Sci Technolog Med Sci. 2017; 37(6): 816–822, doi: 10.1007/s11596-017-1812-y, indexed in Pubmed: 29270737.
  26. Li H, Han S, Sun Q, et al. Long non-coding RNA CDKN2B-AS1 reduces inflammatory response and promotes cholesterol efflux in atherosclerosis by inhibiting ADAM10 expression. Aging (Albany NY). 2019; 11(6): 1695–1715, doi: 10.18632/aging.101863, indexed in Pubmed: 30926762.
  27. Guo FX, Wu Q, Li P, et al. The role of the LncRNA-FA2H-2-MLKL pathway in atherosclerosis by regulation of autophagy flux and inflammation through mTOR-dependent signaling. Cell Death Differ. 2019; 26(9): 1670–1687, doi: 10.1038/s41418-018-0235-z, indexed in Pubmed: 30683918.
  28. Hu YW, Guo FX, Xu YJ, et al. Long noncoding RNA NEXN-AS1 mitigates atherosclerosis by regulating the actin-binding protein NEXN. J Clin Invest. 2019; 129(3): 1115–1128, doi: 10.1172/JCI98230, indexed in Pubmed: 30589415.
  29. Wu LM, Wu SG, Chen F, et al. Atorvastatin inhibits pyroptosis through the lncRNA NEXN-AS1/NEXN pathway in human vascular endothelial cells. Atherosclerosis. 2020; 293: 26–34, doi: 10.1016/j.atherosclerosis.2019.11.033, indexed in Pubmed: 31830726.
  30. Al-Hassnan Z, Almesned A, Tulbah S, et al. Identification of a novel homozygous NEXN gene mutation in recessively inherited dilated cardiomyopathy. J Saudi Heart Assoc. 2013; 25(2): 171–172, doi: 10.1016/j.jsha.2013.03.180.
  31. Wu C, Yan H, Sun J, et al. NEXN is a novel susceptibility gene for coronary artery disease in Han Chinese. PLoS One. 2013; 8(12): e82135, doi: 10.1371/journal.pone.0082135, indexed in Pubmed: 24349201.
  32. Chen X, He WT, Hu L, et al. Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis. Cell Res. 2016; 26(9): 1007–1020, doi: 10.1038/cr.2016.100, indexed in Pubmed: 27573174.
  33. Wu X, Zhang H, Qi W, et al. Nicotine promotes atherosclerosis via ROS-NLRP3-mediated endothelial cell pyroptosis. Cell Death Dis. 2018; 9(2): 171, doi: 10.1038/s41419-017-0257-3, indexed in Pubmed: 29416034.
  34. Chang W, Lin J, Dong J, et al. Pyroptosis: an inflammatory cell death implicates in atherosclerosis. Med Hypotheses. 2013; 81(3): 484–486, doi: 10.1016/j.mehy.2013.06.016, indexed in Pubmed: 23831306.
  35. Man SiM, Kanneganti TD. Gasdermin D: the long-awaited executioner of pyroptosis. Cell Res. 2015; 25(11): 1183–1184, doi: 10.1038/cr.2015.124, indexed in Pubmed: 26482951.
  36. Duewell P, Kono H, Rayner KJ, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010; 464(7293): 1357–1361, doi: 10.1038/nature08938, indexed in Pubmed: 20428172.
  37. Cai X, Chen J, Xu H, et al. Prion-like polymerization underlies signal transduction in antiviral immune defense and inflammasome activation. Cell. 2014; 156(6): 1207–1222, doi: 10.1016/j.cell.2014.01.063, indexed in Pubmed: 24630723.
  38. He Wt, Wan H, Hu L, et al. Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res. 2015; 25(12): 1285–1298, doi: 10.1038/cr.2015.139, indexed in Pubmed: 26611636.
  39. Man SiM, Kanneganti TD. Converging roles of caspases in inflammasome activation, cell death and innate immunity. Nat Rev Immunol. 2016; 16(1): 7–21, doi: 10.1038/nri.2015.7, indexed in Pubmed: 26655628.
  40. Teng Yh, Lu Dz, Fan Cl, et al. Proteomic study on the protective effect of tanshinone IIA against oxidative injury to human umbilical vein endothelial cells by hydrogen peroxide. Current Proteomics. 2014; 10(4): 312–321, doi: 10.2174/1570164610999131125102832.
  41. Le Hiress M, Tu Ly, Ricard N, et al. Proinflammatory signature of the dysfunctional endothelium in pulmonary hypertension. Role of the macrophage migration inhibitory factor/cd74 complex. Am J Respir Crit Care Med. 2015; 192(8): 983–997, doi: 10.1164/rccm.201402-0322OC, indexed in Pubmed: 26203495.
  42. Li JY, Wang YY, Shao T, et al. The zebrafish NLRP3 inflammasome has functional roles in ASC-dependent interleukin-1β maturation and gasdermin E-mediated pyroptosis. J Biol Chem. 2020; 295(4): 1120–1141, doi: 10.1074/jbc.RA119.011751, indexed in Pubmed: 31852739.
  43. Guo Y, Zhao Y, Li L, et al. Concentrationdependent effects of paeoniflorin on proliferation and apoptosis of vascular smooth muscle cells. Mol Med Rep. 2017; 16(6): 9567–9572, doi: 10.3892/mmr.2017.7776, indexed in Pubmed: 29039520.
  44. Heidari M, Mandato C, Lehoux S. Vascular smooth muscle cell phenotypic modulation and the extracellular matrix. Artery Res. 2015; 9(C): 14, doi: 10.1016/j.artres.2014.12.002.
  45. Li X, Zeng Li, Cao C, 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; 350(2): 327–335, doi: 10.1016/j.yexcr.2016.12.006, indexed in Pubmed: 27964927.
  46. Song Z, Zhang Y, Gong B, et al. Long noncoding RNA LINC00339 promotes renal tubular epithelial pyroptosis by regulating the miR-22--3p/NLRP3 axis in calcium oxalate-induced kidney stone. J Cell Biochem. 2019; 120(6): 10452–10462, doi: 10.1002/jcb.28330, indexed in Pubmed: 30614043.
  47. Zhang Y, Liu X, Bai X, et al. Melatonin prevents endothelial cell pyroptosis via regulation of long noncoding RNA MEG3/miR-223/NLRP3 axis. J Pineal Res. 2018; 64(2), doi: 10.1111/jpi.12449, indexed in Pubmed: 29024030.
  48. Puylaert P, Van Praet M, Vaes F, et al. Gasdermin D deficiency limits the transition of atherosclerotic plaques to an inflammatory phenotype in ApoE knock-out mice. Biomedicines. 2022; 10(5), doi: 10.3390/biomedicines10051171, indexed in Pubmed: 35625908.
  49. Opoku E, Traughber CA, Zhang D, et al. Gasdermin D mediates inflammation-induced defects in reverse cholesterol transport and promotes atherosclerosis. Front Cell Dev Biol. 2021; 9: 715211, doi: 10.3389/fcell.2021.715211, indexed in Pubmed: 34395445.
  50. Rayner KJ. Cell death in the vessel wall: the good, the bad, the ugly. Arterioscler Thromb Vasc Biol. 2017; 37(7): e75–e81, doi: 10.1161/ATVBAHA.117.309229, indexed in Pubmed: 28637702.

This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, allowing to download articles and share them with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially.