Journal List > J Lipid Atheroscler > v.9(1) > 1141427

Enchill, Lantz, and Thorp: Select Macrophage Noncoding RNAs of Interest in Cardiovascular Disease

Abstract

Cardiovascular disease remains a leading cause of morbidity and mortality worldwide. Aspects of disease severity that are associated with heightened inflammation, such as during atherosclerosis or after myocardial infarction, are correlated with macrophage activation and macrophage polarization of the transcriptome and secretome. In this setting, non-coding RNAs (ncRNAs) may be as abundant as protein-coding genes and are increasingly recognized as significant modulators of macrophage gene expression and cytokine secretion, although the functions of most ncRNAs—and in particular, long non-coding RNAs—remain unknown. Herein, we discuss a subset of specific ncRNAs of interest in macrophages in atherosclerosis and during myocardial inflammation.

INTRODUCTION TO CARDIOVASCULAR DISEASE-ASSOCIATED MACROPHAGE INFLAMMATION

Cardiovascular disease (CVD) remains the leading cause of death globally and in the United States.1 CVD encompasses a wide range of diseases that affect the heart and blood vessels, including but not limited to conditions such as coronary heart disease, heart failure, and hypertensive heart disease. While the scope of CVD is broad, there is a gradient in terms of the burden of various types of CVD, with atherosclerotic vascular disease currently dominating worldwide.2 Atherosclerosis is marked by immune activation and lipid accumulation in the arterial wall. It is also associated with risk factors such as high blood pressure, smoking, obesity, and high cholesterol.3 The pathogenesis of atherosclerosis involves a complex interplay of multiple cell types.4 In particular, macrophages are central protagonists in the genesis and progression of atherosclerotic CVD. During atherogenesis, infiltration of macrophage precursor monocytes into atherosclerotic lesions and in response to the retention of sub-endothelial lipoprotein are important components of the progression from pathologic intimal thickening to late fibroatheroma.5 Macrophages also play a critical role in cardiac repair and recovery following atherothrombotic myocardial infarction.6 In particular, heterogeneous macrophage populations carry out distinct functions during cardiac repair following cardiac insult.78 Numerous regulatory mechanisms govern macrophages and their inflammatory functions. Non-coding RNAs (ncRNAs), historically considered unnecessary products of so-called “junk” DNA, have emerged as key contributors to macrophage regulation.

ncRNAs AND lncRNAs

Modern genomic and transcriptomic approaches have opened our eyes to the potential role of ncRNAs in the nuanced regulation of gene expression, cell function,9 and disease pathogenesis.10 Initially characterized in bacteria,11 the field quickly expanded to identify roles played by ncRNAs in mitochondrial12 and macrophage13 regulation. ncRNAs are categorized based on size. Small ncRNAs are less than 200 nucleotides in length, while long non-coding RNAs (lncRNAs) are greater than 200 nucleotides. The size of ncRNAs also generally correlates with their function. For example, microRNAs (miRNAs) are 20–24 nucleotides in length and bind to the 3' untranslated region of messenger RNA (mRNA), thereby sterically blocking translation and increasing mRNA degradation.14 On the longer side of the spectrum, lncRNAs are able to interact with DNA, RNA, and proteins to exert cellular regulation at multiple levels, including chromatin remodeling,15 mRNA splicing,16 mRNA translation,17 and multi-protein complex assembly.18 The enhancer RNA subset of lncRNAs are less than 2,000 nucleotide-long transcripts that are produced from enhancer domains to regulate the transcription of target genes19 or nearby genes through diverse mechanisms.20 Importantly, many lncRNAs fall within intergenic regions of the genome (lincRNAs), which are under less selective pressure than genic sequences. These lincRNAs may thus have opportunities for functional diversification compared to their genic counterparts, and could encode species-specific functions.21 In humans, ncRNAs are implicated in cardiogenesis,22 ventricular remodeling after myocardial infarction,23 and wound healing.24
In this brief review, we discuss the role of select ncRNAs, including their potential role in myeloid cell metabolism (immunometabolism). Given the relatively early state of the field, we chose to focus on specific ncRNAs that either have been extensively validated by independent laboratories or have significant potential for future study. We focused on the aspects of these ncRNAs that affect macrophage inflammatory function in the context of atherosclerosis and myocardial infarction. It is also important to note that ncRNAs are expressed in non-myeloid cells; however, the focus of this review is myeloid cells and macrophages. For example, the complexity and heterogeneity of macrophages require dynamic metabolic reprogramming.25 In recent years, novel experimental approaches have enabled researchers to identify specific metabolic variations and their direct connections to immune cell activation. In lipopolysaccharide-activated macrophages, dendritic cells, and activated B and T cells, there is often evidence of enhanced glycolysis following activation.25 In atherosclerosis and related cardiovascular diseases, the integration of excess lipid metabolism and myeloid metabolic processes provides a unique set of circumstances that are likely under the control of lncRNAs.

SPECIFIC ncRNAs OF INTEREST FOR MACROPHAGES AND CARDIOVASCULAR DISEASE

Below we highlight specific ncRNAs of interest, as also shown in Fig. 1. The lncRNA ANRIL (CDKN2BAS) is remarkable in its capacity to regulate genes that have been extensively linked to glucose and fatty acid metabolism.26 Like protein-coding genes, ANRIL is alternatively spliced into multiple linear and circular forms in a tissue-specific manner. In macrophages, ANRIL expression triggers increased glycolysis and induces apoptosis.27 ANRIL is induced by glucose uptake28 and proinflammatory factors such as interferon gamma29 and tumor necrosis factor alpha (TNF-α).30 ANRIL may work through numerous mechanisms, including as an epigenetic modifier or miRNA sponge.31 ANRIL has the capacity to regulate interleukin (IL)-6 cytokine production, for example through the transcription factor Yin Yang 1 (YY1), a component of the nuclear factor-κB pathway.30 Moreover, ANRIL is associated with hematopoietic stem cell proliferation, a key component of the myeloid response after myocardial infarction.32 To maintain homeostasis, excess hematopoietic stem cell proliferation is partially controlled by the tumor suppressor genes cyclin-dependent kinase 2A (CDKN2A) and 2B (CDKN2B).33 Located within a single topological domain, ANRIL negatively regulates CDKN2A/B expression via chromatin remodeling.26 In THP1 macrophage-derived foam cell lines, ANRIL suppressed atherosclerotic-like inflammatory responses and promoted cholesterol efflux.34 ANRIL was shown to regulate ADAM10, a disintegrin and metalloproteinase domain-containing protein, that has been shown to be involved in neuroinflammation35 and expressed at high levels in atherosclerosis.36 The overexpression of ANRIL was found to result in methylation of the ADAM10 gene, which has been shown to inhibit atherosclerotic inflammation.34 Importantly, single-nucleotide polymorphisms within the ANRIL sequence are correlated with an increased risk of coronary artery disease and type 2 diabetes.3738 It will be interesting to determine whether ANRIL expression alters myeloid function through metabolic manipulation after ischemic insults.
Fig. 1

Select ncRNAs of potential significance in the regulation of macrophage function during cardiovascular disease. ncRNAs enact multiple regulatory functions in macrophages. Depicted in the schematic are the potential roles of the ncRNAs NEAT1, GAS5, DAPK-IT1, miR-33, ANRIL, and MeXis. NEAT1 and GAS5 acts as sponges of miR-342-3p and miR-135a, respectively, leading to the upregulation of inflammatory cytokines. DAPK-IT1 and miR-33 both inhibit expression of the ABCA1 gene, leading to reduced expression of ABCA1 cholesterol transporters. ANRIL and MeXis increase macrophage cholesterol efflux, with Mexis working specifically at the ABCA1 gene locus, resulting in increased ABCA1 expression.

ncRNA, non-coding RNA.
jla-9-153-g001
Another important lncRNA involved in cholesterol metabolism in macrophages is MeXis, which lies in close proximity to the locus of the cholesterol-efflux gene ABCA1.3940 MeXis enhances binding of the transcriptional coactivator DDX17 to enhancer regions in the ABCA1 gene locus, thereby increasing nuclear receptor LXR-mediated gene expression of ABCA1 in macrophages. Single-nucleotide polymorphism variants within the MeXis locus are correlated with an increased risk of coronary artery disease in humans.39 Importantly, MeXis is preferentially expressed in macrophages in the heart and kidney, whereas a distinct lncRNA, LeXis, is preferentially expressed in the liver.39 MeXis and LeXis are interesting in that they reveal a mechanism by which cells respond uniquely to the same stimulus, resulting in tissue-specific changes in metabolic regulation. Consequently, MeXis expression leads to increased levels of intracellular cholesterol in cardiac macrophages, in turn inducing inflammatory pathways that are implicated in myocardial infarction.
The lncRNA NEAT1 is involved in cholesterol metabolism and atherosclerosis development. Since its discovery, NEAT1 expression has been discovered in a number of diseases, including lupus41 and several disparate cancers.4243 A lncRNA screen of peripheral blood mononuclear cells revealed differential expression of NEAT1 in patients following myocardial infarction.44 NEAT1 was the most highly expressed and subsequently deregulated lncRNA identified. Multivariate statistical analysis revealed that NEAT1 levels were correlated with post-myocardial infarction status, independent of statin intake, left ventricular ejection fraction, low-density lipoprotein (LDL) or high-density lipoprotein cholesterol, age, diabetes, and smoking.44 NEAT1 has also been shown to be upregulated in THP1 macrophages stimulated by oxidized LDLs (ox-LDLs).45 Knockdown of NEAT1 in these THP1 cells repressed the formation of foam cells triggered by ox-LDLs.45 NEAT1 functions partly by acting as a sponge for miR-342-3p, as depicted in Fig. 1. Repression of NEAT1 and overexpression of miR-342-3p inhibited lipid uptake in THP1 cells. Therefore, it appears that NEAT1 promotes inflammation and lipid uptake through miR-342-3p repression.45 NEAT1 has been experimentally shown to inhibit another miRNA, miR-128. Analysis of RAW264.7 cells stimulated by ox-LDL showed increased NEAT1 expression in combination with decreased miR-128 expression.46 Knockdown of NEAT1 in the same cells repressed foam cell formation and downregulated IL-6, IL-1β, and TNF-α.46 As with miR-342-3p, overexpression of miR-128 inhibited atherosclerotic development triggered by NEAT1.46 In mice, Neat1 enhanced activation of the NLRP3, NLRC4, and AIM2 inflammasomes by stabilizing inflammasome caspase tetramers.47 With all of these considerations in mind, further research into NEAT1 could lead to the identification of possible therapeutic targets in atherosclerotic development.
In recent years, miR-33 has emerged as a significant regulator of macrophage function and cholesterol efflux.48 In humans, miR-33 is co-expressed with the SREBF1/2 genes, which code for transcription factors that regulate fatty acid synthesis and uptake.4950 miR-33 expression inhibits the expression of ABCA1 and ABCG1, two key transporters involved in cholesterol efflux in macrophages, while SREBF1/2 regulates cholesterol biosynthesis and cellular uptake.51 Therefore, miR-33 and SREBF1/2 co-expression is dependent on intracellular cholesterol concentrations, with low-sterol conditions leading to the upregulation of cholesterol biosynthesis through SREBF1/2, and reduced cholesterol efflux through miR-33. Moreover, miR-33 can regulate mitochondrial respiration and metabolism by inhibiting expression of the PGC-1α, PDK4, and SLC25A25 proteins.49 As oxidative phosphorylation is a downstream function of PGC-1α expression, anti-miR-33 treatment was found to lead to increased mitochondrial biogenesis and efficient ATP production. miR-33 has also been shown to regulate autophagy and lipid metabolism, further showcasing this miRNA's ability to regulate cholesterol homeostasis. Targeting these metabolic pathways through anti-mi-R33 resulted in improved cholesterol efflux, decreased foam cell formation, and reduced atherosclerotic lesions in atherosclerosis mouse models. The full therapeutic potential of manipulating ncRNAs in the regulation of cardiovascular disease, including cardiac inflammation, remains unclear; however, ncRNAs can be targeted to improve metabolic dysfunction in non-human primates.51 Therefore, ncRNAs could modulate the immunometabolic rewiring of macrophages after myocardial infarction by controlling the expression of metabolic genes. Consequently, manipulation of miR-33 and other relevant ncRNAs may fine-tune the inflammatory response after cardiac injury. ncRNAs may both adjust the regulation of gene expression, which is important during cardiac repair, and further encode human-specific forms not found in experimental rodents.
In macrophage-derived foam cells, both the ncRNA DAPK-IT1 and lipoprotein lipase (LPL) were upregulated after increased LDL exposure.52 In a separate co-expression gene network analysis, DAPK-IT1 was identified as a differentially expressed lncRNA during the progression of atherosclerosis.53 Increased levels of DAPK-IT1 in THP1 macrophages were also associated with reduced expression of miR-590-3p. In this context, in a subsequent analysis of ApoE-deficient mice, DAPK-IT1 was shown to promote LPL expression through the suppression of miR-590-30 in foam cells. These data are consistent with the proposal that the DAPK-IT1/miR-590-3p/LPL axis plays a regulatory role in cholesterol homeostasis and inflammation. Like miR-33, one of the mechanisms through which the DAPK-IT1/miR-590-3p/LPL axis promotes atherogenic effects is through downregulation of the key reverse cholesterol transporters, ABCA1 and ABCG1 (Fig. 1).
The lncRNA growth arrest-specific transcript 5 (GAS5) encodes several functional small nucleolar RNAs that have been shown to act as regulators of apoptosis, as well as a number of other inflammatory conditions such as hepatocellular carcinoma and osteoarthritis. GAS5 has also been identified as a possible biomarker of coronary artery disease54 and cardiac fibrosis.55 In patients with coronary artery disease, GAS5 expression was lower than in healthy controls.54 Like other lncRNAs described in this review, GAS5 also has the capacity to act as a miRNA sponge. In active cardiac fibroblast tissue, GAS5-induced suppression of miR-21 resulted in inhibition of fibrosis.55 These studies suggest that GAS5 may play a protective role in cardiovascular disease. Interestingly, GAS5 appears to have opposite effects in the aggravation of atherosclerosis. In THP1 macrophages, increased expression of GAS5 further increased the ox-LDL-induced release of the proinflammatory cytokines IL-6, IL-1β, and TNF-α.56 Increased expression of GAS5 was also associated with the upregulation of matrix metalloproteinases (MMPs), leading to degradation of the extracellular matrix and atherosclerotic plaque rupture.56 Specifically, GAS5 expression suppressed miR-221, which has been shown to decrease MMP expression and to stabilize atherosclerotic plaques through various anti-inflammatory pathways.56 In a separate study, GAS5 was shown to act as a sponge of miR-135a, another anti-atherosclerotic miRNA.57 Silencing GAS5 led to decreased expression of IL-6, IL-1β, and TNF-α, as depicted in Fig. 1.57 Taken together, these studies suggest that GAS5 plays a pro-inflammatory role in atherosclerosis. It would be interesting to further characterize the role of GAS5 in the development and progression of various CVDs. Research into the role of GAS5 in CVD currently remains limited, and further studies are needed to uncover its full functionality and possible therapeutic potential.

CONCLUSION

Taken together, lncRNAs are a fascinating area of research with implications for many disease processes. In the context of CVD and macrophages, lncRNAs appear to encode a myriad of functions, ranging from transcriptional activation to regulation of immunometabolism and macrophage polarization. This also includes the potential to regulate the clearance of dying cells by the lncRNA MIAT.58 Efferocytosis is critical to the resolution of atherosclerosis and myocardial infarction.59 Significant insights have been gleaned connecting lncRNAs with the regulation of cholesterol homeostasis and miRNAs in macrophages. Future research is necessary to shed light on the full therapeutic potential of targeting ncRNAs relative to other standards of care.

Notes

Funding None.

Conflict of Interest The authors have no conflicts of interest to disclose.

Author Contributions

  • Conceptualization: Thorp EB.

  • Writing - review & editing: Enchill Z, Lantz C, Thorp EB.

References

1. Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, et al. Heart disease and stroke statistics-2019 update: a report from the American Heart Association. Circulation. 2019; 139:e56–e528.
2. Roth GA, Johnson C, Abajobir A, Abd-Allah F, Abera SF, Abyu G, et al. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J Am Coll Cardiol. 2017; 70:1–25.
pmid pmc
3. Kinoshita M, Yokote K, Arai H, Iida M, Ishigaki Y, Ishibashi S, et al. Japan Atherosclerosis Society (JAS) guidelines for prevention of atherosclerotic cardiovascular diseases 2017. J Atheroscler Thromb. 2018; 25:846–984.
crossref pmid pmc
4. Bobryshev YV, Ivanova EA, Chistiakov DA, Nikiforov NG, Orekhov AN. Macrophages and their role in atherosclerosis: pathophysiology and transcriptome analysis. BioMed Res Int. 2016; 2016:9582430.
crossref
5. Otsuka F, Kramer MC, Woudstra P, Yahagi K, Ladich E, Finn AV, et al. Natural progression of atherosclerosis from pathologic intimal thickening to late fibroatheroma in human coronary arteries: a pathology study. Atherosclerosis. 2015; 241:772–782.
crossref pmid pmc
6. Dick SA, Macklin JA, Nejat S, Momen A, Clemente-Casares X, Althagafi MG, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. 2019; 20:29–39.
crossref pmid
7. Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling JD, et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci U S A. 2014; 111:16029–16034.
crossref pmid pmc
8. Wang L, Zhang YL, Lin QY, Liu Y, Guan XM, Ma XL, et al. CXCL1-CXCR2 axis mediates angiotensin II-induced cardiac hypertrophy and remodelling through regulation of monocyte infiltration. Eur Heart J. 2018; 39:1818–1831.
crossref pmid
9. Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014; 15:7–21.
crossref
10. Batista PJ, Chang HY. Long noncoding RNAs: cellular address codes in development and disease. Cell. 2013; 152:1298–1307.
crossref pmid pmc
11. Mizuno T, Chou MY, Inouye M. A unique mechanism regulating gene expression: translational inhibition by a complementary RNA transcript (micRNA). Proc Natl Acad Sci U S A. 1984; 81:1966–1970.
crossref pmid pmc
12. De Paepe B, Lefever S, Mestdagh P. How long noncoding RNAs enforce their will on mitochondrial activity: regulation of mitochondrial respiration, reactive oxygen species production, apoptosis, and metabolic reprogramming in cancer. Curr Genet. 2018; 64:163–172.
crossref pmid
13. Das S, Reddy MA, Senapati P, Stapleton K, Lanting L, Wang M, et al. Diabetes mellitus-induced long noncoding RNA Dnm3os regulates macrophage functions and inflammation via nuclear mechanisms. Arterioscler Thromb Vasc Biol. 2018; 38:1806–1820.
crossref pmid pmc
14. Engels BM, Hutvagner G. Principles and effects of microRNA-mediated post-transcriptional gene regulation. Oncogene. 2006; 25:6163–6169.
crossref pmid
15. Zhu Y, Rowley MJ, Böhmdorfer G, Wierzbicki AT. A SWI/SNF chromatin-remodeling complex acts in noncoding RNA-mediated transcriptional silencing. Mol Cell. 2013; 49:298–309.
crossref pmid pmc
16. Moseley ML, Zu T, Ikeda Y, Gao W, Mosemiller AK, Daughters RS, et al. Bidirectional expression of CUG and CAG expansion transcripts and intranuclear polyglutamine inclusions in spinocerebellar ataxia type 8. Nat Genet. 2006; 38:758–769.
crossref pmid
17. Carrieri C, Cimatti L, Biagioli M, Beugnet A, Zucchelli S, Fedele S, et al. Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature. 2012; 491:454–457.
crossref pmid
18. Lorenzen JM, Thum T. Long noncoding RNAs in kidney and cardiovascular diseases. Nat Rev Nephrol. 2016; 12:360–373.
crossref pmid
19. Kim TK, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, et al. Widespread transcription at neuronal activity-regulated enhancers. Nature. 2010; 465:182–187.
crossref
20. Ding M, Liu Y, Liao X, Zhan H, Liu Y, Huang W. Enhancer RNAs (eRNAs): new insights into gene transcription and disease treatment. J Cancer. 2018; 9:2334–2340.
crossref pmid pmc
21. Ransohoff JD, Wei Y, Khavari PA. The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol. 2018; 19:143–157.
crossref pmid
22. Klattenhoff CA, Scheuermann JC, Surface LE, Bradley RK, Fields PA, Steinhauser ML, et al. Braveheart, a long noncoding RNA required for cardiovascular lineage commitment. Cell. 2013; 152:570–583.
crossref pmid pmc
23. Zangrando J, Zhang L, Vausort M, Maskali F, Marie PY, Wagner DR, et al. Identification of candidate long non-coding RNAs in response to myocardial infarction. BMC Genomics. 2014; 15:460.
crossref pmid pmc
24. Roy S, Sen CK. MiRNA in innate immune responses: novel players in wound inflammation. Physiol Genomics. 2011; 43:557–565.
crossref pmid
25. O'Neill LA, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol. 2016; 16:553–565.
26. Bochenek G, Häsler R, El Mokhtari NE, König IR, Loos BG, Jepsen S, et al. The large non-coding RNA ANRIL, which is associated with atherosclerosis, periodontitis and several forms of cancer, regulates ADIPOR1, VAMP3 and C11ORF10 . Hum Mol Genet. 2013; 22:4516–4527.
crossref pmid
27. Holdt LM, Stahringer A, Sass K, Pichler G, Kulak NA, Wilfert W, et al. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat Commun. 2016; 7:12429.
crossref
28. Thomas AA, Feng B, Chakrabarti S. ANRIL: a regulator of VEGF in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2017; 58:470–480.
crossref pmid
29. Harismendy O, Notani D, Song X, Rahim NG, Tanasa B, Heintzman N, et al. 9p21 DNA variants associated with coronary artery disease impair interferon-γ signalling response. Nature. 2011; 470:264–268.
crossref pmid pmc
30. Zhou X, Han X, Wittfeldt A, Sun J, Liu C, Wang X, et al. Long non-coding RNA ANRIL regulates inflammatory responses as a novel component of NF-κB pathway. RNA Biol. 2016; 13:98–108.
crossref pmid
31. Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M, et al. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15INK4B tumor suppressor gene. Oncogene. 2011; 30:1956–1962.
crossref pmid
32. Dutta P, Sager HB, Stengel KR, Naxerova K, Courties G, Saez B, et al. Myocardial infarction activates CCR2+ hematopoietic stem and progenitor cells. Cell Stem Cell. 2015; 16:477–487.
crossref pmid pmc
33. Janzen V, Forkert R, Fleming HE, Saito Y, Waring MT, Dombkowski DM, et al. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a . Nature. 2006; 443:421–426.
crossref pmid
34. Li H, Han S, Sun Q, Yao Y, Li S, Yuan C, 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; 11:1695–1715.
crossref pmid pmc
35. Zhou X, Tao H, Cai Y, Cui L, Zhao B, Li K. Stage-dependent involvement of ADAM10 and its significance in epileptic seizures. J Cell Mol Med. 2019; 23:4494–4504.
crossref pmid pmc
36. van der Vorst EP, Weber C, Donners MM. A disintegrin and metalloproteases (ADAMs) in cardiovascular, metabolic and inflammatory diseases: aspects for theranostic approaches. Thromb Haemost. 2018; 118:1167–1175.
crossref pmid
37. Pasmant E, Sabbagh A, Vidaud M, Bièche I. ANRIL, a long, noncoding RNA, is an unexpected major hotspot in GWAS. FASEB J. 2011; 25:444–448.
38. Visel A, Zhu Y, May D, Afzal V, Gong E, Attanasio C, et al. Targeted deletion of the 9p21 non-coding coronary artery disease risk interval in mice. Nature. 2010; 464:409–412.
crossref pmid pmc
39. Sallam T, Jones M, Thomas BJ, Wu X, Gilliland T, Qian K, et al. Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA. Nat Med. 2018; 24:304–312.
crossref pmid pmc
40. Sallam T, Jones MC, Gilliland T, Zhang L, Wu X, Eskin A, et al. Feedback modulation of cholesterol metabolism by the lipid-responsive non-coding RNA LeXis . Nature. 2016; 534:124–128.
crossref pmid pmc
41. Zhang F, Wu L, Qian J, Qu B, Xia S, La T, et al. Identification of the long noncoding RNA NEAT1 as a novel inflammatory regulator acting through MAPK pathway in human lupus. J Autoimmun. 2016; 75:96–104.
crossref
42. Liu X, Liang Y, Song R, Yang G, Han J, Lan Y, et al. Long non-coding RNA NEAT1-modulated abnormal lipolysis via ATGL drives hepatocellular carcinoma proliferation. Mol Cancer. 2018; 17:90.
crossref pmid pmc
43. Yong W, Yu D, Jun Z, Yachen D, Weiwei W, Midie X, et al. Long noncoding RNA NEAT1, regulated by LIN28B, promotes cell proliferation and migration through sponging miR-506 in high-grade serous ovarian cancer. Cell Death Dis. 2018; 9:861.
crossref pmid pmc
44. Gast M, Rauch BH, Haghikia A, Nakagawa S, Haas J, Stroux A, 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.
crossref pmid
45. Wang L, Xia JW, Ke ZP, Zhang BH. 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.
crossref pmid
46. Chen DD, Hui LL, Zhang XC, 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.
crossref
47. Zhang P, Cao L, Zhou R, Yang X, Wu M. The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun. 2019; 10:1495.
crossref pmid pmc
48. Rayner KJ, Suárez Y, Dávalos A, Parathath S, Fitzgerald ML, Tamehiro N, et al. MiR-33 contributes to the regulation of cholesterol homeostasis. Science. 2010; 328:1570–1573.
crossref pmid pmc
49. Karunakaran D, Thrush AB, Nguyen MA, Richards L, Geoffrion M, Singaravelu R, et al. Macrophage mitochondrial energy status regulates cholesterol efflux and is enhanced by anti-miR33 in atherosclerosis. Circ Res. 2015; 117:266–278.
crossref pmid pmc
50. Ouimet M, Ediriweera H, Afonso MS, Ramkhelawon B, Singaravelu R, Liao X, et al. microRNA-33 regulates macrophage autophagy in atherosclerosis. Arterioscler Thromb Vasc Biol. 2017; 37:1058–1067.
crossref pmid pmc
51. Rayner KJ, Esau CC, Hussain FN, McDaniel AL, Marshall SM, van Gils JM, et al. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature. 2011; 478:404–407.
crossref pmid pmc
52. Zhen Z, Ren S, Ji H, Ding X, Zou P, Lu J. The lncRNA DAPK-IT1 regulates cholesterol metabolism and inflammatory response in macrophages and promotes atherogenesis. Biochem Biophys Res Commun. 2019; 516:1234–1241.
crossref pmid
53. Wang CH, Shi HH, Chen LH, Li XL, Cao GL, Hu XF. Identification of key lncRNAs associated with atherosclerosis progression based on public datasets. Front Genet. 2019; 10:123.
crossref pmid pmc
54. Yin Q, Wu A, Liu M. Plasma long non-coding RNA (lncRNA) GAS5 is a new biomarker for coronary artery disease. Med Sci Monit. 2017; 23:6042–6048.
crossref pmid pmc
55. Tao H, Zhang JG, Qin RH, Dai C, Shi P, Yang JJ, et al. LncRNA GAS5 controls cardiac fibroblast activation and fibrosis by targeting miR-21 via PTEN/MMP-2 signaling pathway. Toxicology. 2017; 386:11–18.
crossref pmid
56. Ye J, Wang C, Wang D, Yuan H. LncRBA GSA5, up-regulated by ox-LDL, aggravates inflammatory response and MMP expression in THP-1 macrophages by acting like a sponge for miR-221. Exp Cell Res. 2018; 369:348–355.
crossref pmid
57. Shen S, Zheng X, Zhu Z, Zhao S, Zhou Q, Song Z, et al. Silencing of GAS5 represses the malignant progression of atherosclerosis through upregulation of miR-135a. Biomed Pharmacother. 2019; 118:109302.
crossref
58. Ye ZM, Yang S, Xia YP, Hu RT, Chen S, Li BW, et al. LncRNA MIAT sponges miR-149-5p to inhibit efferocytosis in advanced atherosclerosis through CD47 upregulation. Cell Death Dis. 2019; 10:138.
crossref pmid pmc
59. Thorp EB. Contrasting inflammation resolution during atherosclerosis and post myocardial infarction at the level of monocyte/macrophage phagocytic clearance. Front Immunol. 2012; 3:39.
crossref pmid pmc
TOOLS
ORCID iDs

Zenaida Enchill
https://orcid.org/0000-0003-3431-1379

Connor Lantz
https://orcid.org/0000-0003-3317-6486

Edward B. Thorp
https://orcid.org/0000-0003-1387-7058

Similar articles