Next Article in Journal
Validating Prediction Tools for Autoimmune Encephalitis in Adult Taiwanese Patients: A Retrospective Study
Next Article in Special Issue
A History and Atlas of the Human CD4+ T Helper Cell
Previous Article in Journal
COVID-19 and HIV: Clinical Outcomes among Hospitalized Patients in the United States
Previous Article in Special Issue
Dysfunctions of Neutrophils in the Peripheral Blood of Children with Cystic Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Macrophage lncRNAs in Mediating Inflammatory Processes in Atherosclerosis and Sepsis

1
BK21 Plus KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
2
Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(7), 1905; https://doi.org/10.3390/biomedicines11071905
Submission received: 26 May 2023 / Revised: 25 June 2023 / Accepted: 3 July 2023 / Published: 5 July 2023
(This article belongs to the Special Issue Pathophysiological Mechanisms of Leukocyte Activation and Recruitment)

Abstract

:
Long noncoding RNAs (lncRNAs) are molecules >200 bases in length without protein-coding functions implicated in signal transduction and gene expression regulation via interaction with proteins or RNAs, exhibiting various functions. The expression of lncRNAs has been detected in many cell types, including macrophages, a type of immune cell involved in acute and chronic inflammation, removal of dead or damaged cells, and tissue repair. Increasing evidence indicates that lncRNAs play essential roles in macrophage functions and disease development. Additionally, many animal studies have reported that blockage or modulation of lncRNA functions alleviates disease severity or morbidity rate. The present review summarizes the current knowledge regarding lncRNAs expressed in macrophages, focusing on their molecular targets and the biological processes regulated by them during the development of inflammatory diseases such as atherosclerosis and sepsis. Possible application of this information to lncRNA-targeting therapy is also discussed. The studies regarding macrophage lncRNAs described in this review can help provide valuable information for developing treatments for various pathological conditions involving macrophages.

Graphical Abstract

1. Introduction

Recent advances in molecular biology have revealed that although noncoding RNAs (ncRNAs) are not translated into proteins, they play various roles in cellular processes and disease pathogeneses. Long ncRNAs (lncRNAs) are >200 nucleotides in length and have been extensively researched in various fields of biology [1]. Nuclear lncRNAs have been implicated in regulating chromatin organization, gene transcription, RNA splicing, and epigenetic modifications [2,3,4,5]. Certain lncRNAs that exhibit structural features similar to those of mRNAs can be transported to the cytoplasm to modulate signaling pathways and post-transcriptional gene expression regulation by affecting mRNA stability and translation or sponging microRNAs (miRNAs) to block their function [6,7,8,9,10].
Macrophages exhibit immunoregulatory functions during acute and chronic inflammation, pathogenesis of various diseases, and cancer development. They are categorized into M1 and M2 functional groups. M1 macrophages (also known as “killer” or classically activated macrophages) phagocytose pathogens and foreign substances and promote inflammation, whereas M2 macrophages (also known as “repair” or alternatively activated macrophages) mediate tissue repair and inflammation resolution [11,12]. M1 macrophages metabolize arginine to nitric oxide and synthesize ATP via glycolysis. Furthermore, the mitochondrial citric acid cycle is shut down in these cells. Conversely, M2 macrophages metabolize arginine into ornithine or proline and mostly synthesize ATPs via the citric acid cycle [13]. The bacterial endotoxin lipopolysaccharide (LPS) and interferon (IFN)-γ induce the differentiation of undifferentiated (M0) macrophages into M1 macrophages, which produce a range of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-12. These cytokines and M1 macrophages have been associated with acute inflammation and tissue damage. Conversely, M2 differentiation, induced by IL-4 and IL-13, is characterized by the production of anti-inflammatory cytokines, such as IL-10 and transforming growth factor (TGF)-β. These cytokines and M2 macrophages help regulate immune responses and promote tissue repair [13,14]. Given that the activation and differentiation pathways and immunological roles of M1 and M2 macrophages differ, a detailed understanding of the factors that maintain or regulate the balance between them is important to effectively treat various diseases, such as autoimmune diseases, inflammatory bowel diseases, diabetes, obesity, rheumatoid arthritis (RA), and systemic sclerosis [13,15].
With the identification of an increasing number of lncRNAs involved in regulating macrophage activities and maintaining M1 and M2 polarization balance, lncRNAs are expected to become effective therapeutic targets for various diseases. Previous studies have extensively summarized the implications of lncRNAs regulating inflammatory diseases and macrophage M1 and M2 polarization [16,17]. Herein, we investigated the roles of recently identified and/or salient macrophage lncRNAs in the pathogenesis of atherosclerosis and sepsis, two diseases in which macrophage-mediated inflammation plays an important role.

2. Atherosclerosis

Atherosclerosis is a chronic inflammatory disease characterized by the narrowing and hardening of arteries due to the accumulation of lipid-laden plaques on their inner walls. Macrophages engulf modified low-density lipoprotein (LDL) particles, such as oxidized LDL (oxLDL), and differentiate into foam cells, thereby aggravating chronic inflammatory conditions, stimulating plaque growth, and destabilizing plaques [18,19]. Atherosclerosis has been associated with various health problems, including coronary artery diseases (CADs) and stroke. M1 and M2 macrophages have been implicated in atherogenesis. M1 macrophages promote inflammation and plaque rupture by producing proinflammatory cytokines, chemokines, reactive oxygen species (ROS), and extracellular matrix-degrading enzymes. Conversely, M2 macrophages resolve inflammation by releasing anti-inflammatory cytokines, such as IL-10 [13,20,21]. Most studies on the role of macrophage lncRNAs in atherosclerosis have focused on the regulation of proinflammatory activities, oxLDL-mediated lipid accumulation, cholesterol efflux, and foam cell formation, as described below.
Numerous lncRNAs have been linked to atherogenesis, and these lncRNAs are categorized and representative ones are described in the text. Table 1 lists the macrophage lncRNAs that regulate atherogenesis, their targets, and the cellular functions affected by them. Figure 1 summarizes the functions performed by these lncRNAs, and Figure 2 illustrates the action mechanisms of representative lncRNAs.

2.1. LncRNAs That Promote Inflammation and Foam Cell Formation

Many macrophage lncRNAs involved in atherosclerosis development are induced by oxLDL and promote foam cell formation through enhancing inflammatory changes (Figure 2). Previous studies found that lncRNA H19 expression was upregulated after oxLDL treatment in peripheral blood mononuclear cells of patients with CAD, plaque macrophages of an atherosclerotic mouse model, and RAW264.7 murine macrophage-like cells [22,23]. Transfecting H19-specific short hairpin RNA (shRNA) into RAW264.7 cells decreased oxLDL-induced lipid accumulation and proinflammatory mediator expression by regulating miR-130b activity [24]. Additionally, H19 facilitated lipid accumulation in macrophages via sponging miR-146a-5p and, consequently, protecting angiopoietin-like 4 (ANGPTL4) (miR-146a-5p/ANGPTL4 axis) [23]. ANGPTL4 is a released glycoprotein that regulates lipid metabolism and insulin sensitivity; thus, alterations in ANGPTL4 expression affect the risk of developing atherosclerosis and type 2 diabetes mellitus [25].
NEAT1 (LINC00084, shortened from either nuclear paraspeckle assembly transcript 1 or nuclear enriched abundant transcript 1) is a nucleus-restricted lncRNA involved in the formation of paraspeckles, which are subnuclear structures implicated in antiviral responses [26]. NEAT1 expression is upregulated in oxLDL-treated THP-1 cells, a human monocytic leukemia cell line with macrophage-like properties. Furthermore, NEAT1 participates in the formation of paraspeckles and the development of subsequent proinflammatory responses by regulating p65 phosphorylation. Additionally, NEAT1 modulates lipid uptake by regulating the expression of a scavenger receptor, CD36 [27]. The treatment of RAW264.7 cells with oxLDL increases NEAT1 expression, which in turn stimulates proinflammatory cytokine and ROS production, subsequently promoting foam cell formation by sponging miR-128 [28]. Similar results were observed in oxLDL-treated THP-1 cells; however, the target of NEAT1 action was miR-342-3p [29]. Treating BMDMs with titanium particles induces the expression of NEAT1, which then sponges miR-188-5p to stabilize Bruton’s tyrosine kinase (BTK) mRNA. This NEAT1/miR-188-5p/BTK axis eventually promotes the activation of NF-κB, formation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome, and M1 polarization [30]. Thus, NEAT1 expression in activated macrophages enhances proinflammatory changes. However, one contradicting study reported decreased NEAT1 levels in peripheral blood mononuclear cells (PBMCs) of post-myocardial infarction (MI) patients and demonstrated enhanced inflammatory activation of macrophages in NEAT1-knockout mice [31].
In THP-1 cells, oxLDL treatment upregulated lncRNA urothelial cancer-associated 1 (UCA1) expression levels, which further exacerbated atherosclerotic events, such as CD36 expression, foam cell formation, and ROS generation via sponging miR-206 [32]. In atherosclerotic animal models, lncRNA dynamin 3 opposite strand (Dnm3os) expression was increased in atherosclerotic plaques. Dnm3os regulated macrophage proinflammatory activities via the miR-27b-3p/signaling lymphocytic activation molecule 7 (SLAMF7) axis [33]. SLAMF7 is a membrane protein whose expression is upregulated in macrophages during phagocytosis and macrophage differentiation in atherosclerotic plaques [34].

2.2. LncRNAs That Regulate Cholesterol Efflux and Foam Cell Formation

Numerous clinical and animal studies have demonstrated that defects in reverse cholesterol transport and cholesterol efflux are associated with an increased risk of cardiovascular diseases and atherosclerosis [18,19]. ATP-binding cassette subfamily A member 1 (ABCA1)-mediated cholesterol efflux reduces the formation of lipid-laden foam cells in atherosclerotic plaques. Because regulation of cholesterol efflux is crucial for the prevention of atherosclerosis, ABCA1 has been one of the primary targets in lncRNA research. Interestingly, most of these lncRNAs are involved in regulating chromatin activity and the transcription of the ABCA1 gene, as described below.
The lncRNA macrophage-expressed liver X receptor (LXR)-induced sequence (MeXis) is reportedly involved in LXR-dependent transcriptional activation of Abca1 by guiding the promoter binding of the transcription coactivator DEAD-box helicase 17 (DDX17). Furthermore, bone marrow cells from MeXis-deficient mice exhibited altered chromosome architecture at the Abca1 locus, impaired cholesterol efflux, and accelerated atherosclerosis development. Notably, the genes encoding ABCA1 and MeXis are located near one another to ensure tissue-selective activation of this regulatory circuit [35]. Prostate cancer antigen 3 (PCA3), another lncRNA, also promoted ABCA1 expression by activating the transcription activator RFX7 through sponging miR-140-5p [36]. In the case of HAND2 antisense RNA 1 (HAND2-AS1), ABCA1 expression was promoted by sponging miR-128 and subsequently activating the class II histone deacetylase, sirtuin 1 (SIRT1) [37].
In contrast, the lncRNA growth arrest-specific 5 (GAS5) exerts inhibitory effects on ABCA1 function through its interaction with and stabilization of the enhancer of zeste homolog 2 (EZH2), a chromatin-repressive complex known to promote trimethylation of lysine 27 (H3K27) at the Abca1 promoter [38]. Notably, a significant elevation in GAS5 levels was detected in the serum of patients with coronary heart disease, exhibiting a correlation with heightened proinflammatory markers [39]. Similarly, the lncRNA Kcnq1 overlapping transcript 1 (KCNQ1OT1) impedes ABCA1-mediated cholesterol efflux by suppressing chromatin activity at the Abca1 locus via the miR-452-3p/histone deacetylase 3 (HDAC3) axis [40]. Intriguingly, lncRNA AI662270 directly binds to ABCA1, exerting a limiting effect on its function and thereby promoting lipid accumulation and foam cell formation [41]. Consistent with expectations, animal models have demonstrated that lncRNAs promoting ABCA1 expression alleviate atherosclerosis, whereas those suppressing ABCA1 expression exacerbate its progression [36,38,40].

2.3. LncRNAs That Regulate Macrophage Apoptosis, Pyroptosis, or Autophagy in Atherosclerosis

Cellular processes such as apoptosis, pyroptosis, and autophagy need to be balanced with macrophage proliferation. Disruption of the balance may destabilize atherosclerotic plaques. Apoptotic cell death, especially in overstimulated or exhausted foam cells, can enhance inflammation and trigger blood clot formation, which may lead to heart attack or stroke [18,19].
Table 1. Macrophage lncRNAs involved in atherosclerosis development. The grouping is arbitrary and lncRNA in one group may have roles that are relevant to other categories.
Table 1. Macrophage lncRNAs involved in atherosclerosis development. The grouping is arbitrary and lncRNA in one group may have roles that are relevant to other categories.
LncRNACell TypeTargetAffected FunctionRef.
Group 1. LncRNAs that regulate inflammation and foam cell formation
H19-miR-130bModulates cell survival, lipid accumulation, and inflammatory response[24]
H19Foam cellmiR-146a-5p/ANGPTL4ox-LDL-induced, regulates lipid accumulation, accelerates foam cell formation[23]
Dnm3os-miR-27b-3p/SLAMF7Streptosotocin-induced, intermedin-repressed, participates in ox-LDL absorption [33]
KCNQ10T1-miR-145-5p/PPARαox-LDL-induced, participates in lipid metabolism and inflammation[42]
MALAT1Foam cellmiR-330-5p/IκBαActivates NF-κB pathway, enhances lipid accumulation and inflammation[43]
MALAT1--MALAT1-deficient apoE−/− mice exhibit enhanced inflammation and atherosclerosis [44]
MALAT1Foam cellβ-catenin/CD36ox-LDL-induced, induces lipid uptake[45]
MALAT1Foam cellSIRT1/MAPK/NF-κBox-LDL-induced, inhibits activation of NF-κB pathway[46]
NEAT1-NONO and p65 ox-LDL-induced, mediates paraspeckle formation, regulates lipid uptake[27]
NEAT1M1miR-188-5p or KLF4/BTKActivates NLRP3 inflammasome and NF-κB pathway, induces M1 polarization[30]
NEAT1Foam cellmiR-128Facilitates inflammation, oxidative stress responses and foam cell formation[28]
NEAT1-miR-342-3pox-LDL-induced, regulates inflammation and lipid uptake[29]
NEXN-AS1-BAZ1A/NEXNAttenuates adhesion activity of macrophage, suppresses inflammatory gene expression[47]
SNHG16Foam cellmiR-17-5p/NF-κBox-LDL-induced, enhances cell proliferation and inflammatory responses[48]
UCA1Foam cellmiR-206ox-LDL-induced, facilitates foam cell formation, regulates ROS levels[32]
Group 2. LncRNAs that regulate cholesterol efflux and foam cell formation
AI662270Foam cellABCA1ox-LDL-induced, suppresses cholesterol efflux, enhances lipid accumulation[41]
CDKN2B-AS1Foam cellDNMT1/ADAM10Promotes cholesterol efflux, reduces inflammatory responses[49]
GAS5Foam cellEZH2/ABCA1Inhibits cholesterol efflux, enhances lipid accumulation and atherogenesis[38]
HAND2-AS1Foam cellmiR-1208/SIRT1/ABCA1Inhibits foam cell formation, accelerates cholesterol efflux, attenuates atherosclerosis [37]
KCNQ1OT1-miR-452-3p/HDAC3/ABCA1ox-LDL-induced, inhibits cholesterol efflux, facilitates foam cell formation[40]
MALAT1Foam cellmiR-17-5p/ABCA1Decreases in ox-LDL induced THP-1 derived macrophage, inhibits cholesterol efflux[50]
MeXisFoam cellDDX17/ABCA1LXR-induced, remodels chromatin at the target locus, induces cholesterol efflux[35]
PCA3Foam cellmiR-140-5p/RFX7ABCA1promotes cholesterol efflux, inhibits atherosclerosis progression[36]
TUG1Foam cellmiR-92a/FXR1Regulates apolipoprotein M, downregulates cholesterol efflux, aggravates atherosclerosis[51]
ZFAS1Foam cellmiR-654-3p/ADAM10
miR-654-3p/RAB22A
ox-LDL-induced, activates inflammation, inhibits cholesterol efflux[52]
Group 3. LncRNAs that regulate apoptosis, pyroptosis, and autophagy
ANRIL-Alu repeats etc.Increases proliferation, enhances metabolic activity, decreases apoptosis[53]
GAS5Foam cellmiR-145-5p/Plin2Facilitates oxLDL uptake and autophagy, accelerates foam cell formation[54]
MIAT-miR-149-5p/CD47ox-LDL-induced, inhibits efferocytosis of macrophage, accelerates atherogenic process[55]
MALAT1-miR-23c/ELAVL1ox-LDL-induced, activates NLRP3 inflammasome mediate pyroptosis[56]
MAARS-ELAVL1(HuR)Activates HuR target gene expression, promotes apoptosis, decreases efferocytosis[57]
MALAT1-miR-23-3p/LAMP1Rapa induced, facilitates autophagy activity[58]
p21-Mdm2Inhibits cell proliferation, induces apoptosis, regulates p300/p53 interaction[59]
TUG1-miR-133a/FGF1Facilitates cell proliferation, activates inflammation, inhibits apoptosis[60]
XIST-miR-599/TLR4ox-LDL-induced, inhibits apoptosis, aggravates atherosclerosis progression[61]
RAPIA-miR-183-5p/ITGB1Induced by the action of FoxO1, activates cell proliferation, inhibits apoptosis[62]
SMILR-miR-10b-3p/KLF5ox-LDL-induced, activates cell proliferation, inhibits apoptosis[63]
Group 4. LncRNAs that acts through exosomes
GAS5-p53Produced by macrophages, induces apoptosis in macrophages and vascular endothelial cells[64]
MALAT1M2-Exported via ox-LDL-induced HUVEC-derived exosome, induces M2 polarization[65]
MALAT1M1miR-25-3p/CDC42Affects endothelial cells. Inhibits angiogenesis and myocardial regeneration, aggravates MI[66]
MRGPRF-6:1M1TLR4/MyD88/MAPKEnhances foam cell formation and M1 polarization. Detected in the plasma exosomes [67]
LOC100129516Foam cellPPARγ/LXRα/ABCA1MSC-derived exosomes, inhibits foam cell cholesterol efflux, aggravates atherosclerosis[68]
LncRNAs that inhibit apoptosis tend to aggravate atherogenesis [53,60,61,62,63]. By suppressing apoptosis, macrophages are allowed to proliferate and promote plaque formation. In addition to inhibiting apoptosis, lncRNAs such as taurine-upregulated gene 1 (TUG1) and X-inactive specific transcript (XIST) enhance inflammation via fibroblast growth factor 1 and Toll-like receptor 4 (TLR4), respectively [60,61]. LncRNAs associated with the progression and intervention of atherosclerosis (RAPIA) and smooth-muscle-induced lncRNA (SMILR) also inhibit apoptosis and enhance atherosclerosis by regulating cellular receptor integrin beta 1 (ITGB1) and a transcription factor, Krueppel-like factor 5 (KLF5), respectively [62,63]. In contrast, apoptosis-inducing lncRNAs such as lncRNA-p21 have an atheroprotective role by reducing macrophage proliferation and consequent inflammation [59]. These effects of apoptosis-regulating lncRNAs on atherogenesis have been confirmed in animal models in the cases of p21 and TUG1 [59,60].
The progression of atherosclerosis may also be aggravated by defects in a process known as efferocytosis, the clearance of apoptotic cells by macrophages. Simion et al., detected high-level expression of a macrophage-associated atherosclerotic lncRNA sequence (MAARS) in the aortic intima of atherogenic animal models, and it decreased with the regression of atherosclerosis. Knockdown experiments indicated that MAARS promotes macrophage apoptosis, thereby inhibiting efferocytosis, through its interaction with HuR (ELAVL1), an RNA-binding protein with an apoptosis regulator function [57]. Another lncRNA, MI-associated transcript (MIAT), directly affects efferocytosis [55]. Its expression has been detected in the serum of patients with advanced atherosclerosis and necrotic core macrophages. In RAW264.7 cells, treatment with oxLDL increases MIAT levels. MIAT prevents efferocytosis by sponging miR-149-5p and, consequently, increasing the expression of the antiphagocytic molecule CD47. Suppressing MIAT expression in experimental animal models attenuated atherosclerosis progression by reducing the necrotic core size and increasing plaque stability.
Autophagy and inflammasome-mediated pyroptotic cell death in macrophages may also affect plaque stability, and GAS5 and MALAT1 have been reported to be involved in these processes (Table 1).

2.4. LncRNAs Functioning via Exosomes in Atherogenesis

Many studies have indicated that exosomes can be used as carriers of lncRNA to regulate cellular activities of neighboring cells. The upregulation of lnc-MRGPRF-6:1 expression and as its correlation with levels of proinflammatory mediators have been detected in the plasma exosomes of patients with CAD [67]. The expression level of lnc-MRGPRF-6:1 following M1 induction was higher than that following M2 induction in THP-1 cells. The knockout of lnc-MRGPRF-6:1 reduced ROS generation, lipid accumulation, and subsequent foam cell formation. Furthermore, lnc-MRGPRF-6:1 knockout in human monocyte-derived macrophages suppressed M1 marker and inflammatory cytokine expression and enhanced M2 marker expression by modulating the TLR4/myeloid differentiation primary response 88 (MyD88)/mitogen-activated protein kinase (MAPK) signaling pathway [67].
Notably, exosomes derived from mesenchymal stem cells (MSCs) have been developed as a method for drug delivery for therapeutic purposes [69]. Delivery of LOC100129516-specific siRNA using MSC-derived exosomes blocked cholesterol efflux in foam cells by suppressing the LOC100129516/PPARγ/LXRα/ABCA1 axis [68]. The overexpression of GAS5 in THP-1 cells enhanced oxLDL-induced apoptosis. The resulting apoptotic cells further secreted GAS5-containing exosomes, thereby enhancing apoptosis in vascular endothelial cells [64].

2.5. Multiple Function of MALAT1 in Atherogenesis

The lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) affects multiple atherosclerotic processes, such as foam cell formation and macrophage apoptosis, autophagy, and pyroptosis (Figure 2). Treating THP-1 cells with oxLDL upregulated MALAT1 expression in a nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB)-dependent manner [43,45]. MALAT1 enhanced lipid uptake by inducing CD36 expression by recruiting β-catenin to its binding sites on the CD36 promoter [45]. MALAT1 also enhanced NF-κB activation and, subsequently, foam cell formation, apoptosis, and inflammation via sponging miR-330-5p [43]. Additionally, MALAT1 promoted autophagy in RAW264.7 cells by modulating the miR-23-3p/lysosomal-associated membrane protein 1 (LAMP1) axis [58]. LAMP1 plays a key role in the fusion of autophagosomes with lysosomes. In THP-1 cells, MALAT1 mediated oxLDL-induced autophagy by inhibiting sirtuin 1, a deacetylase enzyme regulating various transcription factors and enzymes, and activating the MAPK/NF-κB pathway [46]. These effects of MALAT1 have been confirmed in an atherosclerotic mouse model, wherein MALAT1 overexpression enhanced disease severity [43]. Treating diabetic atherosclerosis rat models with sinapic acid, a derivative of cinnamic acid, improved rat body weight and reduced their blood glucose levels. Bone-marrow-derived macrophages (BMDMs) isolated from disease animal models treated with low doses of sinapic acid exhibited less inflammasome activation and pyroptotic cell death [56]. In vitro analyses revealed that these beneficial effects of sinapic acid originate from its ability to suppress the expression of MALAT1, which promotes pyroptotic cell death in macrophages and, consequently, the progression of atherosclerosis. Pyroptosis is an inflammasome-mediated programmed cell death that occurs as a defense mechanism against intracellular pathogens. MALAT1 was also found in extracellular vesicles (EVs) derived from M1 BMDMs. These EVs affect myocyte proliferation and angiogenesis in MI animal models [66]. Thus, the findings of these studies indicate that MALAT1 expression is upregulated in macrophages by stimulating agents, such as oxLDL, and MALAT1 enhances atherosclerosis by inducting lipid uptake, foam cell formation, and cell death in macrophages.
However, there are reports of an opposite role played by MALAT1 in atherosclerosis. For instance, in an apolipoprotein E (apoE)-knockout mouse model, MALAT1 deficiency accelerated inflammation and atherosclerosis. Treating MALAT1-deficient BMDMs with LPS enhanced TNF-α and inducible nitric oxide synthase expression, suppressed matrix metalloproteinase-9 expression, and impaired phagocytic activity [44]. Furthermore, decreased MALAT1 levels have been detected in the serum of patients with atherosclerosis and in oxLDL-treated THP-1 cells. MALAT1 knockdown in THP-1 cells increased oxLDL uptake, lipid accumulation, and total cholesterol levels by regulating the miR-17-5p/ABCA1 axis [50]. Notably, treating THP-1 cells with exosomal MALAT1 derived from oxLDL-treated human umbilical vein endothelial cells (HUVECs) suppressed M1 marker expression and enhanced M2 marker expression [65]. These results suggest that suppressing or downregulating MALAT1 expression accelerates atherosclerosis progression. The reason for the discrepancy in the role of MALAT1 in atherosclerosis remains unknown; thus, additional analyses are warranted.

3. Sepsis

Blood monocytes/macrophages and endothelial cells lining the blood vessels respond to gram-negative bacteria infiltration by releasing a flood of chemicals, including cytokines, into circulation to fight the infection. Macrophages can remove pathogens by phagocytosis and regulate the extent of sepsis by producing anti-inflammatory cytokines. However, the production of excess inflammatory cytokines, such as IL-6, IL-1β, and especially TNF-α, may damage the surrounding normal tissues and organs, which can be life-threatening [70,71]. As observed in other diseases, the proinflammatory activity of M1 macrophages aggravates sepsis, whereas the anti-inflammatory activity of M2 macrophages mitigates it [72]. Most studies regarding the role of macrophage lncRNAs in sepsis have focused on the proinflammatory effects and M1/M2 polarization of macrophages, as described below and summarized in Figure 3.

3.1. NEAT1 Enhances Sepsis Progression through Promoting Inflammation

Previous studies have found a considerable increase in NEAT1 levels in serum of patients with sepsis and septic mouse models [73,74,75,76]. These studies agree that NEAT1 is involved in the inflammatory activation of macrophages; however, the targets of its action differ. In THP-1 cells, LPS-induced NEAT1 expression enhances inflammatory responses by modulating the miR-17-5p/TLR4 axis [76]. LPS-stimulated Kupffer or RAW264.7 cells exhibit the expression of NEAT1, which exerts its proinflammatory activities through the Let-7q/TLR4 axis [73]. Other studies have reported that NEAT1 promotes inflammation in LPS-treated RAW264.7 cells by modulating the miR-495-3p/signal transducer and activator of transcription 3 (STAT3), miR-211/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), miR-370-3p/thrombospondin-1, or miR-31-5p/POU domain, class 2, transcription factor 1 (POU2F1) axes [74,75,77]. Wang et al., reported that this effect of NEAT1 was mediated through the miR-125a-5p/TNF receptor-associated factor 6 (TRAF6)/TGF-β-activated kinase 1 (TAK1) axis and that the downregulation of NEAT1 expression promoted M2 polarization [78]. Thus, NEAT1 expressed in activated macrophages enhances their proinflammatory activity. As most previous experiments have been performed using cell lines, additional studies involving animal models or primary macrophages isolated from patients with sepsis are warranted to confirm the role of NEAT1 in sepsis progression.

3.2. MALAT1 Promotes M1 Polarization and Inflammation in Sepsis

An increase in lncRNA MALAT1 levels was detected in the serum of late-onset sepsis patients and in activated primary macrophages and macrophage cell lines [79]. MALAT1-knockout mice exhibited reduced inflammation and death upon sepsis induction. Particularly, suppressing MALAT1 expression increased the antioxidant capacity of macrophages through the methyltransferase 16 (METTL16)/methionine adenosyltransferase 2 A (MAT2A) axis, wherein MALAT1 binds to METTL16, thereby stabilizing the METTL16 N6-methyladenosine (m6A) modification activity [79]. MAT2A regulates cellular metabolism and catalyzes S-adenosylmethionine production [80]. Intraperitoneal LPS injection in mice induces septic lung injury, substantially increasing MALAT1 expression in lung tissues. Additional intravenous MALAT1-specific small interfering RNA (siRNA) injection reduces the number of inflammatory cells and cytokine levels in the bronchoalveolar lavage fluid (BALF) of these animal models by inhibiting the p38 MAPK/p65 NF-κB signaling pathway [81]. Cui et al., reported that LPS and IL-4 treatments increased and decreased MALAT1 expression levels, respectively, in human and mouse macrophage cell lines and primary macrophages. Attenuated MALAT1 expression suppresses M1 and enhances M2 polarization in primary macrophages. Although MALAT1-knockout mice exhibit attenuated symptoms of LPS-induced acute lung injury, profibrotic macrophage differentiation and pulmonary fibrosis are promoted in these mice [82]. Thus, macrophage MALAT1 levels increase during LPS-induced macrophage activation and sepsis development. MALAT1 also mediates proinflammatory changes, further implicating MALAT1 as an attractive therapeutic target of sepsis.
However, a few studies have reported different observations regarding the role of MALAT1. For instance, Yang et al., reported a significant decrease in MALAT1 serum levels and an increase in hsa-miR-346 levels in patients with sepsis. Activated RAW264.7 cells also exhibit reduced expression of MALAT1. Additional experiments demonstrated that MALAT1 regulates macrophage proliferation through the hsa-miR-346/small mothers against decapentaplegic homolog 3 (SMAD3) axis [83]. SMAD3 is a receptor-regulated signaling adaptor activated by serine kinases. Zhao et al., demonstrated that LPS treatment upregulated MALAT1 expression in human and mouse macrophage-like cell lines and primary macrophages. This increase in MALAT1 expression levels is NF-κB dependent, and MALAT1 suppresses the expression of proinflammatory cytokines, such as TNF-α and IL-6, by interacting with NF-κB to block its DNA-binding activity [84]. The cause of this discrepancy in the role of macrophage MALAT1 in sepsis development is currently unknown, and more research on this subject will aid in reaching a definitive conclusion.

3.3. Other lncRNAs Involved in Sepsis Development

LPS-induced NF-κB activation in THP-1 cells and the subsequent release of proinflammatory cytokines were shown to be regulated by lncRNA colorectal neoplasia differentially expressed (CRNDE) via the miR-181-5p/TLR4 axis. A considerable increase in CRNDE expression levels and decrease in miR-181-5p expression levels have been detected in the peripheral blood of patients with sepsis. Furthermore, the extent of these changes correlates with the survival rate of the patients [85]. In sepsis patients, the serum levels of G-quadruplex-forming sequence-containing lncRNA (GSEC) were observed to be elevated, and GSEC was identified as a regulator of inflammation and proliferation in RAW264.7 cells through its interaction with miR-873-3p [86]. Conversely, sepsis patients exhibited a significant decrease in serum levels of lncRNA MEG3. In vitro analysis demonstrated that MEG3 overexpression suppressed macrophage apoptosis and NF-κB-mediated inflammation. Notably, in a septic mouse model, silencing of lncRNA Cox2 resulted in the amelioration of sepsis symptoms by suppressing M1 and enhancing M2 macrophage activities [87]. Moreover, elevated expression of lncRNA PVT1 was detected in heart-infiltrating macrophages of septic mice, and PVT1 was found to enhance M1 polarization through the miR-29a/HMGB1 axis [88].

4. Discussion and Conclusions

Macrophage lncRNAs are induced by causative agents, such as oxLDL, and affect atherogenesis by regulating a wide range of processes including NF-κB activation, ROS generation, cholesterol efflux, apoptosis, efferocytosis, autophagy, inflammasome formation, and pyroptosis. These processes lead to lipid accumulation, proinflammatory activation, and foam cell formation in macrophages (Figure 1). Similarly, macrophage lncRNAs play a role in sepsis development by regulating inflammation, ROS generation, and M1 polarization (Figure 3). Pyroptosis and apoptotic cell death are crucial factors affecting sepsis progression [89,90], and macrophage lncRNAs that mediate these processes in sepsis are expected to be revealed soon. While many lncRNAs have been found to be transferred by exosomes in atherosclerosis, exosomal lncRNA has not been detected in conditions associated with sepsis.
It is intriguing to observe that NEAT1 and MALAT1 serve as key regulators of macrophage functions in both atherosclerosis and sepsis. Despite their distinct target profiles, both MALAT1 and NEAT1 promote inflammation and facilitate M1 polarization or foam cell formation. When considering the roles of these two lncRNAs, it is important to acknowledge the presence of a series of tRNA-like transcripts generated from the evolutionarily conserved NEAT1-MALAT1 gene cluster. Targeted disruption of these tRNA-like transcripts has revealed their critical involvement in the innate immune responses of monocytes/macrophages [91]. Exploring the interrelationship between these lncRNAs and tRNA-like transcripts in the pathogenesis of atherosclerosis and sepsis promises to be an intriguing avenue for future research.
In summary, manipulating the expression of numerous macrophage lncRNAs in experimental animal models has shown promising effects on the severity of atherosclerosis and sepsis. For instance, the introduction of MALAT1 siRNA effectively reduced macrophage-mediated inflammation and improved lung injury in a mouse model of LPS-induced sepsis [81,82]. Conversely, MALAT1 overexpression was found to exacerbate disease severity in an atherosclerotic mouse model [43]. Furthermore, overexpression of PCA3 or knockdown of KCNQ1OT1 in apoE−/− mice demonstrated favorable outcomes by promoting reverse cholesterol transport and impeding atherosclerosis progression, whereas overexpression of GAS5 exerted contrasting effects [36,38,40]. However, it is important to exercise caution when drawing conclusions solely based on lncRNA overexpression studies, as many lncRNAs may not be expressed at sufficiently high levels to function as effective miRNA sponges. Although clinical trial results are currently unavailable, the compelling findings from these animal studies underscore the potential of lncRNA-based therapy as a promising avenue for the treatment of atherosclerosis and sepsis. Further research and clinical investigations are warranted to explore the therapeutic implications and translate these findings into clinical practice.

Author Contributions

Conceptualization, W.-H.L. and K.S.; writing—original draft preparation, H.-S.S., J.P., J.-J.S. and I.A.; writing—review and editing, W.-H.L. and K.S.; supervision, W.-H.L.; project administration, W.-H.L.; funding acquisition, W.-H.L. and K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (Ministry of Science and ICT) (No. 2022R1A2C1010005 and 2017R1A5A2015391).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABCA1, ATP binding cassette subfamily a member 1; ADAM10, A disintegrin and metalloproteinase domain-containing protein 10; AI662270, Mus musculus expressed sequence AI662270; AKT, Protein kinase B; ANGPTL4, Angiopoietin-like 4; ANRIL, Antisense non-coding RNA in the INK4 locus; ATP, Adenosine triphosphate; BALF, Bronchoalveolar lavage fluid; BAZ1A, Bromodomain adjacent to Zinc finger domain 1A; BMDM, Bone marrow-derived macrophage; BTK, Bruton tyrosine kinase; C/EBPβ, CCAAT/enhancer binding protein beta; CAD, Coronary artery disease; cAMP, cyclic adenosine monophosphate; CD36, Cluster of differentiation 36; CD42, Cluster of differentiation 42; CD47, Cluster of differentiation 47; CDKN2B, Cyclin dependent kinase inhibitor 2B; CDKN2B-AS1, CDKN2B antisense RNA 1; COX-2, Cycloxygenase-2; CPT1, Camitine palmitoyltransferase I; CREB, cAMP response element-binding protein; CRNDE, Colorectal neoplasia differentially expressed; DDX17, DEAD-box helicase 17; Dnm3os, Dynamin 3 opposite strand; DNMT1, DNA(cytosine-5)-methyltransferase 1; ELAVL1, Embryonic lethal abnormal vision like RNA binding protein 1; EV, Extracellular vesicle; EZH2, Enhancer of zeste 2 polycomb repressive complex 2 subunit; FGF1, Fibroblast growth factor 1; FXR1, Fragile-X mental retardation autosomal homolog 1; GAS5, Growth arrest-specific 5; GPR, G-protein coupled receptor; GSEC, G-quadruplex-forming sequence containing lncRNA; H3K27, Trimethylation of lysine 27; HAND2, Heart and neural crest derivatives expressed 2; HAND2-AS1, HAND2 antisense RNA 1; HDAC3, Histone deacetylase 3; HMGB1, High mobility group box 1; HUVEC, Human umbilical vein endothelial cell; IFN-γ, Interferon gamma; IL-1, Interleukin 1; IL-1β, Interleukin 1 beta; IL-4, Interleukin 4; IL-6, Interleukin 6; IL-8, Interleukin 8; IL-10, Interleukin 10; IL-12, Interleukin 12; IL-13, Interleukin 13; INK4, Inhibitor of cyclin-dependent kinase 4; iNOS, inducible nitric oxide synthase; ITGB1, Integrin beta 1; KCNQ1, Potassium voltage-gated channel subfamily Q member 1; KCNQ1OT1, Kcnq1 overlapping transcript 1; KLF4, Kruppel-like factor 4; KLF5, Kruepeel-like factor 5; LAMP1, Lysosomal-associated membrane protein 1; LDL, Low-density lipoprotein; LncRNA H19, LncRNA encoded by the H19 gene; lncRNA, Long noncoding RNA; LncRNA-COX2, LncRNA located about 50kb upstream of the protein-coding gene COX-2; LPS, Lipopolysaccharide; LXR, Liver X receptor; m6A, N6-methyladenosine; MAARS, Macrophage-associated atherosclerotic lncRNA sequence; MALAT1, Metastasis-associated lung adenocarcinoma transcript 1; MAPK, Mitogen-activated protein kinase; MAS, Marker assisted selection or marker aided selection; MAT2A, Methionine adenosyltransferase 2A; Mdm2, Mouse double minute 2 homolog; MEG3, Maternally expressed 3; METTL16, Methyltransferase 16; MeXis, Macrophage-expressed LXR-induced sequence; MI, Myocardial infarction; MIAT, MI-associated transcript; miRNA, MicroRNA; MMP-9, Matrix metalloproteinase 9; MRGPRF, MAS related GPR family member F; MSC, mesenchymal stem cell; MyD88, Myeloid diffentiation primary response 88; ncRNA, Noncoding RNA; NEAT1, Shortened from either nuclear paraspeckle assembly transcript 1 or Nuclear enriched abundant transcript 1; NEXN, Nexilin F-actin binding protein; NEXN-AS1, NEXN antisense RNA 1; NFX1, Nuclear transcription factor X-box binding 1; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cell; NLR, Nucleotide-binding domain leucine-rich repeat containing; NLRP3, NLR family pyrin domain containing 3; NONO, Non-POU domain containing octamer binding; oxLDL, Oxidized LDL; PBMC, Peripheral blood mononuclear cell; PCA3, Prostate cancer associated 3; PI3K, Phosphoinositide 3-kinase; Plin2, Perilipin 2; POU2F1, POU domain, class2, transcription factor 1; PPARα, Peroxisome proliferator activated receptor alpha PPARγ, Peroxisome proliferator activated receptor gamma PVT1, Plasmacytoma variant translocation 1; Rab, Ras-associated binding protein; RAB22A, Ras-related protein Rab-22A; RAPIA, LncRNA associated with the progression and intervention of atherosclerosis; RFX7, Regulatory factor X7; ROS, Reactive oxygen species; siRNA, Small interfering RNA; SIRT1, Sirtuin 1; SLAMF7, Signaling lymphocytic activation molecule 7; SMAD3, Small mothers against decapentaplegic homolog 3; SMILR, Smooth muscle-induced lncRNA; SNHG16, Small nucleolar RNA host gene 16; STAT3, Signal transducer and activator of transcription 3; TAK1, TGF-β-activated kinase 1; TGF-β, Transforming growth factor beta; TLR4, Toll-like receptor 4; TNF, Tumor necrosis factor; TNF-α, Tumor necrosis factor alpha; TRAF6, TNF receptor-associated factor 6; TUG1, Taurine-up-regulated gene 1; UCA1, Urothelial cancer associated 1; XIST, X-inactive specific transcript; ZFAS1, ZNFX1 antisense RNA 1; ZNFX1, Zinc finger NFX1-type containing.

References

  1. Winkle, M.; El-Daly, S.M.; Fabbri, M.; Calin, G.A. Noncoding RNA therapeutics—Challenges and potential solutions. Nat. Rev. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef] [PubMed]
  2. Herman, A.B.; Tsitsipatis, D.; Gorospe, M. Integrated lncRNA function upon genomic and epigenomic regulation. Mol. Cell 2022, 82, 2252–2266. [Google Scholar] [CrossRef]
  3. Huang, W.; Li, H.; Yu, Q.; Xiao, W.; Wang, D.O. LncRNA-mediated DNA methylation: An emerging mechanism in cancer and beyond. J. Exp. Clin. Cancer Res. 2022, 41, 100. [Google Scholar] [CrossRef]
  4. Senmatsu, S.; Hirota, K. Roles of lncRNA transcription as a novel regulator of chromosomal function. Genes Genet. Syst. 2021, 95, 213–223. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, C.; Wang, L.; Ding, Y.; Lu, X.; Zhang, G.; Yang, J.; Zheng, H.; Wang, H.; Jiang, Y.; Xu, L. LncRNA Structural Characteristics in Epigenetic Regulation. Int. J. Mol. Sci. 2017, 18, 2659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Zhao, Z.; Sun, W.; Guo, Z.; Zhang, J.; Yu, H.; Liu, B. Mechanisms of lncRNA/microRNA interactions in angiogenesis. Life Sci. 2020, 254, 116900. [Google Scholar] [CrossRef] [PubMed]
  7. Ferrè, F.; Colantoni, A.; Helmer-Citterich, M. Revealing protein-lncRNA interaction. Brief. Bioinform. 2016, 17, 106–116. [Google Scholar] [CrossRef] [Green Version]
  8. Khorkova, O.; Hsiao, J.; Wahlestedt, C. Basic biology and therapeutic implications of lncRNA. Adv. Drug Deliv. Rev. 2015, 87, 15–24. [Google Scholar] [CrossRef] [Green Version]
  9. Mathieu, E.L.; Belhocine, M.; Dao, L.T.; Puthier, D.; Spicuglia, S. Functions of lncRNA in development and diseases. Med. Sci. 2014, 30, 790–796. [Google Scholar] [CrossRef] [Green Version]
  10. Peng, W.X.; Koirala, P.; Mo, Y.Y. LncRNA-mediated regulation of cell signaling in cancer. Oncogene 2017, 36, 5661–5667. [Google Scholar] [CrossRef]
  11. Orecchioni, M.; Ghosheh, Y.; Pramod, A.B.; Ley, K. Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front. Immunol. 2019, 10, 1084. [Google Scholar] [CrossRef] [Green Version]
  12. Tardito, S.; Martinelli, G.; Soldano, S.; Paolino, S.; Pacini, G.; Patane, M.; Alessandri, E.; Smith, V.; Cutolo, M. Macrophage M1/M2 polarization and rheumatoid arthritis: A systematic review. Autoimmun. Rev. 2019, 18, 102397. [Google Scholar] [CrossRef] [Green Version]
  13. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef]
  14. Bashir, S.; Sharma, Y.; Elahi, A.; Khan, F. Macrophage polarization: The link between inflammation and related diseases. Inflamm. Res. 2016, 65, 1–11. [Google Scholar] [CrossRef]
  15. Funes, S.C.; Rios, M.; Escobar-Vera, J.; Kalergis, A.M. Implications of macrophage polarization in autoimmunity. Immunology 2018, 154, 186–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Jiang, P.; Li, X. Regulatory Mechanism of lncRNAs in M1/M2 Macrophages Polarization in the Diseases of Different Etiology. Front. Immunol. 2022, 13, 835932. [Google Scholar] [CrossRef] [PubMed]
  17. Liao, K.; Xu, J.; Yang, W.; You, X.; Zhong, Q.; Wang, X. The research progress of LncRNA involved in the regulation of inflammatory diseases. Mol. Immunol. 2018, 101, 182–188. [Google Scholar] [CrossRef] [PubMed]
  18. Tabas, I. Macrophage death and defective inflammation resolution in atherosclerosis. Nat. Rev. Immunol. 2010, 10, 36–46. [Google Scholar] [CrossRef] [Green Version]
  19. Gibson, M.S.; Domingues, N.; Vieira, O.V. Lipid and Non-lipid Factors Affecting Macrophage Dysfunction and Inflammation in Atherosclerosis. Front. Physiol. 2018, 9, 654. [Google Scholar] [CrossRef] [Green Version]
  20. Momtazi-Borojeni, A.A.; Abdollahi, E.; Nikfar, B.; Chaichian, S.; Ekhlasi-Hundrieser, M. Curcumin as a potential modulator of M1 and M2 macrophages: New insights in atherosclerosis therapy. Heart Fail. Rev. 2019, 24, 399–409. [Google Scholar] [CrossRef]
  21. Oh, J.; Riek, A.E.; Weng, S.; Petty, M.; Kim, D.; Colonna, M.; Cella, M.; Bernal-Mizrachi, C. Endoplasmic reticulum stress controls M2 macrophage differentiation and foam cell formation. J. Biol. Chem. 2012, 287, 11629–11641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Bitarafan, S.; Yari, M.; Broumand, M.A.; Ghaderian, S.M.H.; Rahimi, M.; Mirfakhraie, R.; Azizi, F.; Omrani, M.D. Association of Increased Levels of lncRNA H19 in PBMCs with Risk of Coronary Artery Disease. Cell J. 2019, 20, 564–568. [Google Scholar] [CrossRef]
  23. Huang, S.F.; Zhao, G.; Peng, X.F.; Ye, W.C. The Pathogenic Role of Long Non-coding RNA H19 in Atherosclerosis via the miR-146a-5p/ANGPTL4 Pathway. Front. Cardiovasc. Med. 2021, 8, 770163. [Google Scholar] [CrossRef] [PubMed]
  24. Han, Y.; Ma, J.; Wang, J.; Wang, L. Silencing of H19 inhibits the adipogenesis and inflammation response in ox-LDL-treated Raw264.7 cells by up-regulating miR-130b. Mol. Immunol. 2018, 93, 107–114. [Google Scholar] [CrossRef] [PubMed]
  25. Mehta, N.; Qamar, A.; Qu, L.; Qasim, A.N.; Mehta, N.N.; Reilly, M.P.; Rader, D.J. Differential association of plasma angiopoietin-like proteins 3 and 4 with lipid and metabolic traits. Arter. Thromb. Vasc. Biol. 2014, 34, 1057–1063. [Google Scholar] [CrossRef] [Green Version]
  26. Naganuma, T.; Nakagawa, S.; Tanigawa, A.; Sasaki, Y.F.; Goshima, N.; Hirose, T. Alternative 3′-end processing of long noncoding RNA initiates construction of nuclear paraspeckles. EMBO J. 2012, 31, 4020–4034. [Google Scholar] [CrossRef] [Green Version]
  27. Huang-Fu, N.; Cheng, J.S.; Wang, Y.; Li, Z.W.; Wang, S.H. Neat1 regulates oxidized low-density lipoprotein-induced inflammation and lipid uptake in macrophages via paraspeckle formation. Mol. Med. Rep. 2018, 17, 3092–3098. [Google Scholar] [CrossRef]
  28. Chen, D.D.; Hui, L.L.; Zhang, X.C.; Chang, Q. NEAT1 contributes to ox-LDL-induced inflammation and oxidative stress in macrophages through inhibiting miR-128. J. Cell. Biochem. 2018, 120, 2493–2501. [Google Scholar] [CrossRef]
  29. Wang, L.; Xia, J.W.; Ke, Z.P.; Zhang, B.H. Blockade of NEAT1 represses inflammation response and lipid uptake via modulating miR-342-3p in human macrophages THP-1 cells. J. Cell. Physiol. 2019, 234, 5319–5326. [Google Scholar] [CrossRef]
  30. Lin, S.; Wen, Z.; Li, S.; Chen, Z.; Li, C.; Ouyang, Z.; Lin, C.; Kuang, M.; Xue, C.; Ding, Y. LncRNA Neat1 promotes the macrophage inflammatory response and acts as a therapeutic target in titanium particle-induced osteolysis. Acta Biomater. 2022, 142, 345–360. [Google Scholar] [CrossRef]
  31. Gast, M.; Rauch, B.H.; Haghikia, A.; Nakagawa, S.; Haas, J.; Stroux, A.; Schmidt, D.; Schumann, P.; Weiss, S.; Jensen, L.; et al. Long noncoding RNA NEAT1 modulates immune cell functions and is suppressed in early onset myocardial infarction patients. Cardiovasc. Res. 2019, 115, 1886–1906. [Google Scholar] [CrossRef] [PubMed]
  32. Hu, X.; Ma, R.; Fu, W.; Zhang, C.; Du, X. LncRNA UCA1 sponges miR-206 to exacerbate oxidative stress and apoptosis induced by ox-LDL in human macrophages. J. Cell. Physiol. 2019, 234, 14154–14160. [Google Scholar] [CrossRef] [PubMed]
  33. Su, Y.; Guan, P.; Li, D.; Hang, Y.; Ye, X.; Han, L.; Lu, Y.; Bai, X.; Zhang, P.; Hu, W. Intermedin attenuates macrophage phagocytosis via regulation of the long noncoding RNA Dnm3os/miR-27b-3p/SLAMF7 axis in a mouse model of atherosclerosis in diabetes. Biochem. Biophys. Res. Commun. 2021, 583, 35–42. [Google Scholar] [CrossRef] [PubMed]
  34. Xia, Z.; Gu, M.; Jia, X.; Wang, X.; Wu, C.; Guo, J.; Zhang, L.; Du, Y.; Wang, J. Integrated DNA methylation and gene expression analysis identifies SLAMF7 as a key regulator of atherosclerosis. Aging 2018, 10, 1324–1337. [Google Scholar] [CrossRef] [PubMed]
  35. Sallam, T.; Jones, M.; Thomas, B.J.; Wu, X.; Gilliland, T.; Qian, K.; Eskin, A.; Casero, D.; Zhang, Z.; Sandhu, J.; et al. Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA. Nat. Med. 2018, 24, 304–312. [Google Scholar] [CrossRef]
  36. Zhao, Z.W.; Zhang, M.; Liao, L.X.; Zou, J.; Wang, G.; Wan, X.J.; Zhou, L.; Li, H.; Qin, Y.S.; Yu, X.H.; et al. Long non-coding RNA PCA3 inhibits lipid accumulation and atherosclerosis through the miR-140-5p/RFX7/ABCA1 axis. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158904. [Google Scholar] [CrossRef]
  37. Ma, L.; He, S.; Li, H.; Zhang, S.; Yin, Y. HAND2-AS1 targeting miR-1208/SIRT1 axis alleviates foam cell formation in atherosclerosis. Int. J. Cardiol. 2022, 346, 53–61. [Google Scholar] [CrossRef]
  38. Meng, X.D.; Yao, H.H.; Wang, L.M.; Yu, M.; Shi, S.; Yuan, Z.X.; Liu, J. Knockdown of GAS5 Inhibits Atherosclerosis Progression via Reducing EZH2-Mediated ABCA1 Transcription in ApoE−/− Mice. Mol. Ther. Nucleic Acids 2020, 19, 84–96. [Google Scholar] [CrossRef]
  39. Jiang, Y.; Du, T. Relation of circulating lncRNA GAS5 and miR-21 with biochemical indexes, stenosis severity, and inflammatory cytokines in coronary heart disease patients. J. Clin. Lab. Anal. 2022, 36, e24202. [Google Scholar] [CrossRef]
  40. Yu, X.H.; Deng, W.Y.; Chen, J.J.; Xu, X.D.; Liu, X.X.; Chen, L.; Shi, M.W.; Liu, Q.X.; Tao, M.; Ren, K. LncRNA kcnq1ot1 promotes lipid accumulation and accelerates atherosclerosis via functioning as a ceRNA through the miR-452-3p/HDAC3/ABCA1 axis. Cell Death Dis. 2020, 11, 1043. [Google Scholar] [CrossRef]
  41. Hong, Y.; Zhang, Y.; Chen, H.; Tang, X.; Zhao, H.; Meng, Z.; Jia, X.; Liu, W.; Li, X.; Wang, L.; et al. Genetic dissection of the impact of lncRNA AI662270 during the development of atherosclerosis. J. Transl. Med. 2023, 21, 97. [Google Scholar] [CrossRef]
  42. Wang, Y.; Liu, L.; Li, J. LncRNA KCNQ1OT1 depletion inhibits the malignant development of atherosclerosis by miR-145-5p. Microvasc. Res. 2022, 139, 104236. [Google Scholar] [CrossRef] [PubMed]
  43. Shi, Z.; Zheng, Z.; Lin, X.; Ma, H. Long Noncoding RNA MALAT1 Regulates the Progression of Atherosclerosis by miR-330-5p/NF-kappaB Signal Pathway. J. Cardiovasc. Pharmacol. 2021, 78, 235–246. [Google Scholar] [CrossRef] [PubMed]
  44. Gast, M.; Rauch, B.H.; Nakagawa, S.; Haghikia, A.; Jasina, A.; Haas, J.; Nath, N.; Jensen, L.; Stroux, A.; Bohm, A.; et al. Immune system-mediated atherosclerosis caused by deficiency of long non-coding RNA MALAT1 in ApoE−/− mice. Cardiovasc. Res. 2019, 115, 302–314. [Google Scholar] [CrossRef] [PubMed]
  45. Huangfu, N.; Xu, Z.; Zheng, W.; Wang, Y.; Cheng, J.; Chen, X. LncRNA MALAT1 regulates oxLDL-induced CD36 expression via activating beta-catenin. Biochem. Biophys. Res. Commun. 2018, 495, 2111–2117. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, J.; Lin, X.; Wang, L.; Sun, T.; Zhao, Q.; Ma, Q.; Zhou, Y. LncRNA MALAT1 Enhances ox-LDL-Induced Autophagy through the SIRT1/MAPK/NF-kappaB Pathway in Macrophages. Curr. Vasc. Pharmacol. 2020, 18, 652–662. [Google Scholar] [CrossRef] [PubMed]
  47. Hu, Y.W.; Guo, F.X.; Xu, Y.J.; Li, P.; Lu, Z.F.; McVey, D.G.; Zheng, L.; Wang, Q.; Ye, J.H.; Kang, C.M.; et al. Long noncoding RNA NEXN-AS1 mitigates atherosclerosis by regulating the actin-binding protein NEXN. J. Clin. Investig. 2019, 129, 1115–1128. [Google Scholar] [CrossRef]
  48. An, J.H.; Chen, Z.Y.; Ma, Q.L.; Wang, H.J.; Zhang, J.Q.; Shi, F.W. LncRNA SNHG16 promoted proliferation and inflammatory response of macrophages through miR-17-5p/NF-kappaB signaling pathway in patients with atherosclerosis. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8665–8677. [Google Scholar] [CrossRef]
  49. Li, H.; Han, S.; Sun, Q.; Yao, Y.; Li, S.; Yuan, C.; Zhang, B.; Jing, B.; Wu, J.; Song, Y.; et al. Long non-coding RNA CDKN2B-AS1 reduces inflammatory response and promotes cholesterol efflux in atherosclerosis by inhibiting ADAM10 expression. Aging 2019, 11, 1695–1715. [Google Scholar] [CrossRef]
  50. Liu, L.; Tan, L.; Yao, J.; Yang, L. Long non-coding RNA MALAT1 regulates cholesterol accumulation in ox-LDL-induced macrophages via the microRNA-17-5p/ABCA1 axis. Mol. Med. Rep. 2020, 21, 1761–1770. [Google Scholar] [CrossRef] [Green Version]
  51. Yang, L.; Li, T. LncRNA TUG1 regulates ApoM to promote atherosclerosis progression through miR-92a/FXR1 axis. J. Cell. Mol. Med. 2020, 24, 8836–8848. [Google Scholar] [CrossRef] [PubMed]
  52. Tang, X.; Yin, R.; Shi, H.; Wang, X.; Shen, D.; Wang, X.; Pan, C. LncRNA ZFAS1 confers inflammatory responses and reduces cholesterol efflux in atherosclerosis through regulating miR-654-3p-ADAM10/RAB22A axis. Int. J. Cardiol. 2020, 315, 72–80. [Google Scholar] [CrossRef] [PubMed]
  53. Gareev, I.; Kudriashov, V.; Sufianov, A.; Begliarzade, S.; Ilyasova, T.; Liang, Y.; Beylerli, O. The role of long non-coding RNA ANRIL in the development of atherosclerosis. Non-Coding RNA Res. 2022, 7, 212–216. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, C.; Qin, Q.; Xu, J.; Li, X.; Cong, H. Phthalate promotes atherosclerosis through interacting with long-non coding RNA and induces macrophage foam cell formation and vascular smooth muscle damage. Chemosphere 2022, 308, 136383. [Google Scholar] [CrossRef] [PubMed]
  55. Ye, Z.M.; Yang, S.; Xia, Y.P.; Hu, R.T.; Chen, S.; Li, B.W.; Chen, S.L.; Luo, X.Y.; Mao, L.; Li, Y.; et al. LncRNA MIAT sponges miR-149-5p to inhibit efferocytosis in advanced atherosclerosis through CD47 upregulation. Cell Death Dis. 2019, 10, 138. [Google Scholar] [CrossRef] [Green Version]
  56. Han, Y.; Qiu, H.; Pei, X.; Fan, Y.; Tian, H.; Geng, J. Low-dose Sinapic Acid Abates the Pyroptosis of Macrophages by Downregulation of lncRNA-MALAT1 in Rats With Diabetic Atherosclerosis. J. Cardiovasc. Pharmacol. 2018, 71, 104–112. [Google Scholar] [CrossRef]
  57. Simion, V.; Zhou, H.; Haemmig, S.; Pierce, J.B.; Mendes, S.; Tesmenitsky, Y.; Perez-Cremades, D.; Lee, J.F.; Chen, A.F.; Ronda, N.; et al. A macrophage-specific lncRNA regulates apoptosis and atherosclerosis by tethering HuR in the nucleus. Nat. Commun. 2020, 11, 6135. [Google Scholar] [CrossRef]
  58. Ma, Z.; Zhang, J.; Xu, X.; Qu, Y.; Dong, H.; Dang, J.; Huo, Z.; Xu, G. LncRNA expression profile during autophagy and Malat1 function in macrophages. PLoS ONE 2019, 14, e0221104. [Google Scholar] [CrossRef]
  59. Wu, G.; Cai, J.; Han, Y.; Chen, J.; Huang, Z.P.; Chen, C.; Cai, Y.; Huang, H.; Yang, Y.; Liu, Y.; et al. LincRNA-p21 regulates neointima formation, vascular smooth muscle cell proliferation, apoptosis, and atherosclerosis by enhancing p53 activity. Circulation 2014, 130, 1452–1465. [Google Scholar] [CrossRef] [Green Version]
  60. Zhang, L.; Cheng, H.; Yue, Y.; Li, S.; Zhang, D.; He, R. TUG1 knockdown ameliorates atherosclerosis via up-regulating the expression of miR-133a target gene FGF1. Cardiovasc. Pathol. 2018, 33, 6–15. [Google Scholar] [CrossRef]
  61. Yang, K.; Xue, Y.; Gao, X. LncRNA XIST Promotes Atherosclerosis by Regulating miR-599/TLR4 Axis. Inflammation 2021, 44, 965–973. [Google Scholar] [CrossRef] [PubMed]
  62. Sun, C.; Fu, Y.; Gu, X.; Xi, X.; Peng, X.; Wang, C.; Sun, Q.; Wang, X.; Qian, F.; Qin, Z.; et al. Macrophage-Enriched lncRNA RAPIA: A Novel Therapeutic Target for Atherosclerosis. Arter. Thromb. Vasc. Biol. 2020, 40, 1464–1478. [Google Scholar] [CrossRef]
  63. Li, H.; Pan, Z.; Chen, Q.; Yang, Z.; Zhang, D. SMILR Aggravates the Progression of Atherosclerosis by Sponging miR-10b-3p to Regulate KLF5 Expression. Inflammation 2020, 43, 1620–1633. [Google Scholar] [CrossRef]
  64. Chen, L.; Yang, W.; Guo, Y.; Chen, W.; Zheng, P.; Zeng, J.; Tong, W. Exosomal lncRNA GAS5 regulates the apoptosis of macrophages and vascular endothelial cells in atherosclerosis. PLoS ONE 2017, 12, e0185406. [Google Scholar] [CrossRef] [PubMed]
  65. Huang, C.; Han, J.; Wu, Y.; Li, S.; Wang, Q.; Lin, W.; Zhu, J. Exosomal MALAT1 derived from oxidized low-density lipoprotein-treated endothelial cells promotes M2 macrophage polarization. Mol. Med. Rep. 2018, 18, 509–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chen, B.; Luo, L.; Wei, X.; Gong, D.; Li, Z.; Li, S.; Tang, W.; Jin, L. M1 Bone Marrow-Derived Macrophage-Derived Extracellular Vesicles Inhibit Angiogenesis and Myocardial Regeneration Following Myocardial Infarction via the MALAT1/MicroRNA-25-3p/CDC42 Axis. Oxidative Med. Cell. Longev. 2021, 2021, 9959746. [Google Scholar] [CrossRef]
  67. Hu, D.; Wang, Y.; You, Z.; Lu, Y.; Liang, C. lnc-MRGPRF-6:1 Promotes M1 Polarization of Macrophage and Inflammatory Response through the TLR4-MyD88-MAPK Pathway. Mediat. Inflamm. 2022, 2022, 6979117. [Google Scholar] [CrossRef]
  68. Sun, L.; He, X.; Zhang, T.; Han, Y.; Tao, G. Knockdown of mesenchymal stem cell-derived exosomal LOC100129516 suppresses the symptoms of atherosclerosis via upregulation of the PPARgamma/LXRalpha/ABCA1 signaling pathway. Int. J. Mol. Med. 2021, 48, 1–11. [Google Scholar] [CrossRef]
  69. Huang, P.; Wang, L.; Li, Q.; Tian, X.; Xu, J.; Xu, J.; Xiong, Y.; Chen, G.; Qian, H.; Jin, C.; et al. Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding RNA H19. Cardiovasc. Res. 2020, 116, 353–367. [Google Scholar] [CrossRef]
  70. Pravda, J. Sepsis: Evidence-based pathogenesis and treatment. World J. Crit. Care Med. 2021, 10, 66–80. [Google Scholar] [CrossRef]
  71. Chousterman, B.G.; Swirski, F.K.; Weber, G.F. Cytokine storm and sepsis disease pathogenesis. Semin. Immunopathol. 2017, 39, 517–528. [Google Scholar] [CrossRef]
  72. Chen, X.; Liu, Y.; Gao, Y.; Shou, S.; Chai, Y. The roles of macrophage polarization in the host immune response to sepsis. Int. Immunopharmacol. 2021, 96, 107791. [Google Scholar] [CrossRef]
  73. Zhang, C.C.; Niu, F. LncRNA NEAT1 promotes inflammatory response in sepsis-induced liver injury via the Let-7a/TLR4 axis. Int. Immunopharmacol. 2019, 75, 105731. [Google Scholar] [CrossRef] [PubMed]
  74. Xia, D.; Yao, R.; Zhou, P.; Wang, C.; Xia, Y.; Xu, S. LncRNA NEAT1 reversed the hindering effects of miR-495-3p/STAT3 axis and miR-211/PI3K/AKT axis on sepsis-relevant inflammation. Mol. Immunol. 2020, 117, 168–179. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, X.Y.; Fang, Y.; Zheng, F.X.; Zhang, Y.Z.; Li, Q.L. LncRNA NEAT1 facilitates the progression of sepsis through up-regulating TSP-1 via sponging miR-370-3p. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 333–344. [Google Scholar] [CrossRef]
  76. Li, Y.; Guo, W.; Cai, Y. NEAT1 Promotes LPS-induced Inflammatory Injury in Macrophages by Regulating MiR-17-5p/TLR4. Open Med. 2020, 15, 38–49. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, Y.; Xue, J.; Qin, L.; Zhang, J.; Liu, J.; Yu, J. LncRNA NEAT1 Promotes Inflammatory Response in Sepsis via the miR-31-5p/POU2F1 Axis. Inflammation 2021, 44, 1518–1528. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, W.; Guo, Z.H. Downregulation of lncRNA NEAT1 Ameliorates LPS-Induced Inflammatory Responses by Promoting Macrophage M2 Polarization via miR-125a-5p/TRAF6/TAK1 Axis. Inflammation 2020, 43, 1548–1560. [Google Scholar] [CrossRef]
  79. Chen, J.; Tang, S.; Ke, S.; Cai, J.J.; Osorio, D.; Golovko, A.; Morpurgo, B.; Guo, S.; Sun, Y.; Winkle, M.; et al. Ablation of long noncoding RNA MALAT1 activates antioxidant pathway and alleviates sepsis in mice. Redox Biol. 2022, 54, 102377. [Google Scholar] [CrossRef]
  80. Bottiglieri, T. S-Adenosyl-L-methionine (SAMe): From the bench to the bedside--molecular basis of a pleiotrophic molecule. Am. J. Clin. Nutr. 2002, 76, 1151S–1157S. [Google Scholar] [CrossRef] [Green Version]
  81. Lin, L.P.; Niu, G.H.; Zhang, X.Q. Influence of lncRNA MALAT1 on septic lung injury in mice through p38 MAPK/p65 NF-kappaB pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 1296–1304. [Google Scholar] [CrossRef]
  82. Cui, H.; Banerjee, S.; Guo, S.; Xie, N.; Ge, J.; Jiang, D.; Zornig, M.; Thannickal, V.J.; Liu, G. Long noncoding RNA Malat1 regulates differential activation of macrophages and response to lung injury. JCI Insight 2019, 4, e124522. [Google Scholar] [CrossRef] [Green Version]
  83. Yang, Q.; Cao, K.; Jin, G.; Zhang, J. Hsa-miR-346 plays a role in the development of sepsis by downregulating SMAD3 expression and is negatively regulated by lncRNA MALAT1. Mol. Cell. Probes 2019, 47, 101444. [Google Scholar] [CrossRef]
  84. Zhao, G.; Su, Z.; Song, D.; Mao, Y.; Mao, X. The long noncoding RNA MALAT1 regulates the lipopolysaccharide-induced inflammatory response through its interaction with NF-kappaB. FEBS Lett. 2016, 590, 2884–2895. [Google Scholar] [CrossRef] [Green Version]
  85. Wang, Y.; Xu, Z.; Yue, D.; Zeng, Z.; Yuan, W.; Xu, K. Linkage of lncRNA CRNDE sponging miR-181a-5p with aggravated inflammation underlying sepsis. Innate Immun. 2020, 26, 152–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Xiao, J.; Wu, Y.; Paranggan, P.; Gao, W.; Gao, Z.; Liu, J.; Wu, L. Involvement of plasma lncRNA GSEC in sepsis discrimination and prognosis, and its correlation with macrophage cell inflammation and proliferation. Immunobiology 2022, 227, 152264. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, Q.; Xie, Y.; He, Q.; Geng, Y.; Xu, J. LncRNA-Cox2 regulates macrophage polarization and inflammatory response through the CREB-C/EBPbeta signaling pathway in septic mice. Int. Immunopharmacol. 2021, 101, 108347. [Google Scholar] [CrossRef] [PubMed]
  88. Luo, Y.Y.; Yang, Z.Q.; Lin, X.F.; Zhao, F.L.; Tu, H.T.; Wang, L.J.; Wen, M.Y.; Xian, S.X. Knockdown of lncRNA PVT1 attenuated macrophage M1 polarization and relieved sepsis induced myocardial injury via miR-29a/HMGB1 axis. Cytokine 2021, 143, 155509. [Google Scholar] [CrossRef]
  89. Luo, R.; Li, X.; Wang, D. Reprogramming Macrophage Metabolism and its Effect on NLRP3 Inflammasome Activation in Sepsis. Front. Mol. Biosci. 2022, 9, 917818. [Google Scholar] [CrossRef]
  90. Qiu, P.; Liu, Y.; Zhang, J. Review: The Role and Mechanisms of Macrophage Autophagy in Sepsis. Inflammation 2019, 42, 6–19. [Google Scholar] [CrossRef]
  91. Gast, M.; Nageswaran, V.; Kuss, A.W.; Tzvetkova, A.; Wang, X.; Mochmann, L.H.; Rad, P.R.; Weiss, S.; Simm, S.; Zeller, T.; et al. tRNA-like Transcripts from the NEAT1-MALAT1 Genomic Region Critically Influence Human Innate Immunity and Macrophage Functions. Cells 2022, 11, 3970. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Action mechanisms of macrophage lncRNAs involved in atherogenesis. These lncRNAs affect various cellular processes that ultimately regulate inflammation, lipid accumulation, and foam cell formation.
Figure 1. Action mechanisms of macrophage lncRNAs involved in atherogenesis. These lncRNAs affect various cellular processes that ultimately regulate inflammation, lipid accumulation, and foam cell formation.
Biomedicines 11 01905 g001
Figure 2. An overview of macrophage lncRNAs involved in atherosclerosis development. LncRNAs exert their functions through both direct and indirect mechanisms. Certain lncRNAs act as miRNA sponges, indirectly regulating protein expression by inhibiting miRNA function. In contrast, other lncRNAs directly interact with proteins to modulate their activity. Additionally, some lncRNAs can regulate gene expression through epigenetic modifications. Within the figures, arrows are employed to indicate the molecule responsible for activating or inhibiting its specific target.
Figure 2. An overview of macrophage lncRNAs involved in atherosclerosis development. LncRNAs exert their functions through both direct and indirect mechanisms. Certain lncRNAs act as miRNA sponges, indirectly regulating protein expression by inhibiting miRNA function. In contrast, other lncRNAs directly interact with proteins to modulate their activity. Additionally, some lncRNAs can regulate gene expression through epigenetic modifications. Within the figures, arrows are employed to indicate the molecule responsible for activating or inhibiting its specific target.
Biomedicines 11 01905 g002
Figure 3. Action mechanisms of macrophage lncRNAs involved in sepsis development. Most of these lncRNAs are involved in LPS-induced inflammatory activation and subsequent M1 polarization and proliferation of macrophages.
Figure 3. Action mechanisms of macrophage lncRNAs involved in sepsis development. Most of these lncRNAs are involved in LPS-induced inflammatory activation and subsequent M1 polarization and proliferation of macrophages.
Biomedicines 11 01905 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shin, H.-S.; Shin, J.-J.; Park, J.; Arab, I.; Suk, K.; Lee, W.-H. Role of Macrophage lncRNAs in Mediating Inflammatory Processes in Atherosclerosis and Sepsis. Biomedicines 2023, 11, 1905. https://doi.org/10.3390/biomedicines11071905

AMA Style

Shin H-S, Shin J-J, Park J, Arab I, Suk K, Lee W-H. Role of Macrophage lncRNAs in Mediating Inflammatory Processes in Atherosclerosis and Sepsis. Biomedicines. 2023; 11(7):1905. https://doi.org/10.3390/biomedicines11071905

Chicago/Turabian Style

Shin, Hyeung-Seob, Jae-Joon Shin, Jeongkwang Park, Imene Arab, Kyoungho Suk, and Won-Ha Lee. 2023. "Role of Macrophage lncRNAs in Mediating Inflammatory Processes in Atherosclerosis and Sepsis" Biomedicines 11, no. 7: 1905. https://doi.org/10.3390/biomedicines11071905

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop