<<

Clin Chem Lab Med 2017; 55(5): 667–686

Review

Véronique Desgagné, Luigi Bouchard and Renée Guérin* in lipoprotein and lipid metabolism: from biological function to clinical application

DOI 10.1515/cclm-2016-0575 Received June 29, 2016; accepted October 31, 2016; previously Introduction ­published online December 17, 2016 Since their first discovery in worms in 1993 [1], microRNAs Abstract: microRNAs (miRNAs) are short (~ 22 nucleo‑ (miRNAs) have been described as important regulators tides), non‑coding, single-stranded RNA molecules that of several biological processes from cellular differentia‑ regulate the expression of target by partial sequence- tion and development to metabolic function regulation, specific base-pairing to the targeted mRNA 3′UTR, block‑ and their deregulation in diseased states has repeatedly ing its , and promoting its degradation or its been reported. The role of miRNAs as central regulators sequestration into processing bodies. miRNAs are impor‑ of lipoprotein metabolism and cholesterol homeosta‑ tant regulators of several physiological processes includ‑ sis is now well established in the literature (reviewed in ing developmental and metabolic functions, but their [2–5]). However, the role of lipoprotein-bounded miRNAs concentration in circulation has also been reported to be as endocrine regulators has recently emerged. Although altered in many pathological conditions such as familial studies on lipoprotein-transported miRNAs suggest dis‑ hypercholesterolemia, cardiovascular diseases, obesity, tinct miRNA profiles in the two major lipoproteins found in type 2 diabetes, and . In this review, we focus on circulation, the data support that miRNAs have a stronger the role of miRNAs in lipoprotein and lipid metabolism, role in high-density lipoprotein (HDL) than in low-density with special attention to the well-characterized miR-33a/b, lipoprotein (LDL) in regulating their metabolic functions and on the huge potential of miRNAs for clinical applica‑ [6–8]. The discovery of miRNAs transported by lipopro‑ tion as biomarkers and therapeutics in the context of car‑ teins thus opens up the way for two main clinical appli‑ diometabolic diseases. cations: biomarkers of diseases and targeted therapies. In this review, we describe the basics of miRNA biology Keywords: blood miRNA; cholesterol; high-density lipo‑ and focus on the important role of miRNAs in lipoprotein protein (HDL). and lipid metabolism. We also describe where potential clinical applications currently stand and discuss future research perspectives and their challenges.

miRNA biogenesis and biological *Corresponding author: Renée Guérin, PhD, CSPQ, FCACB, Département de biochimie, Université de Sherbrooke, Sherbrooke, function Québec, Canada; and Département de biologie médicale, CIUSSS du Saguenay–Lac-St-Jean, Hôpital de Chicoutimi, Saguenay, Québec, miRNAs are evolutionarily-conserved short [~ 22 nucleo‑ Canada, Phone: (418) 541-1000-3403, tides (nt)], non‑coding, single-stranded involved in E-mail: [email protected] the post-transcriptional regulation of potentially > 60% of Véronique Desgagné: Département de biochimie, Université de Sherbrooke, Sherbrooke, Québec, Canada; and Laboratoire the human genes [9]. The miRNAs biogenesis pathway is ECOGENE-21, CIUSSS du Saguenay–Lac-St-Jean, Hôpital de detailed in Figure 1. Briefly, miRNAs are encoded in the Chicoutimi, Saguenay, Québec, Canada genome and are transcribed, mostly by the RNA polymer‑ Luigi Bouchard: Département de biochimie, Université de ase II, as a ~ 1000-nt capped and polyadenylated primary Sherbrooke, Sherbrooke, Québec, Canada; Laboratoire ECOGENE-21, miRNA (pri-miRNA) from their own promoter (intergenic) CIUSSS du Saguenay–Lac-St-Jean, Hôpital de Chicoutimi, Saguenay, Québec, Canada; and Département de biologie médicale, CIUSSS du or from co-expression with their host protein-coding Saguenay–Lac-St-Jean, Hôpital de Chicoutimi, Saguenay, Québec, (intragenic) [10]. The pri-miRNA is then cleaved by the Canada (/DGCR8) to generate a 668 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

Cytoplasm miRNA gene Nucleus Intergenic Intragenic

Mirtron Pol II Spliceosome debranching

Drosha RISC DGCR8 Exportin 5-Ran-GTP TRBP RISC m G- -AAAAA miRNA Mature Pri-miRNA Pre-miRNA duplex miRNA

7 m G- -AAAAA Target mRNA

Sequestrationintoprocessing Repression of protein bodies or degradation of mRNA translation RISC RISC

7 7 m G- -AAAAA m G- -AAAAA Ribosome

Figure 1: Canonical miRNA biogenesis pathway. miRNA genes (intragenic or intergenic) are transcribed, mostly by the RNA polymerase II (Pol II), as a primary miRNA (pri-miRNA) [10]. The pri-miRNA is cleaved the microprocessor complex (Drosha/DGCR8) to generate a hairpin-structured precursor miRNA (pre-miRNA) which is then exported to the cytoplasm by the Exportin5-RanGTP system [10]. The pre-miRNA is subjected to a second cleavage by the Dicer/TRBP complex, releasing a ~ 22 nt miRNA duplex (miRNA-3p/miRNA-5p), which is then loaded onto an protein (AGO1-4, in humans) to form the pre-RNA-induced silencing complex (RISC) [10]. Based on relative thermodynamic stability of the ends of the two miRNA strands, one strand (guide) is selected to form the mature RISC complex, while the other strand (passenger) is quickly removed and degraded [10]. Non-canonical miRNA biogenesis pathways [e.g. bypassing the processing by either Drosha (e.g. mirtrons) or Dicer (e.g. miR-451)] have also been characterized for a few specific miRNAs (reviewed in [10]). Through (mostly partial) base-pairing to the 3′ UTR of its target mRNA, miRNA regulates the expression of target genes by repressing mRNA translation and promoting mRNA sequestration into processing bodies as well as mRNA destabilization and degradation (mRNA decay, accelerated by mRNA deadenylation and decapping) [10].

~ 70-nt hairpin-structured precursor miRNA (pre-miRNA), synthesis through their (mostly partial) base-pairing to which is exported to the cytoplasm by the Exportin5- the 3′ untranslated region (UTR) of their mRNA target, RanGTP system [10]. The pre-miRNA is subjected to a which is mainly dependent of the complementarity at second cleavage by the Dicer/TRBP complex, releasing nucleotides 2–8 of the miRNA (i.e. miRNA seed region; at a ~ 22 nt miRNA duplex (miRNA-3p/miRNA-5p) [10]. The the 5′-end of the miRNA), although the nucleotides 13–16 miRNA duplex is subsequently loaded onto an Argo‑ also contribute to mRNA targeting specificity (i.e. miRNA naute protein (AGO1-4, in humans), by an ATP-dependent 3′ supplementary site) [12]. This interaction consequently mechanism involving the heat shock cognate 70 (HSC70)– inhibits mRNA translation and promotes mRNA seques‑ heat shock protein 90 (HSP90) chaperone complex, to tration into processing bodies as well as its degradation form the pre-RNA-induced silencing complex (RISC) [10]. (mRNA destabilization and decay, accelerated by mRNA Based on the relative thermodynamic stability of the two deadenylation and decapping; Figure 1), the latter pro‑ miRNA strands, the one with the weakest binding at its 5′ ducing greater repressive effect at steady state [10, 13–15]. end is more likely to become the strand (guide) selected These miRNA-mediated gene-silencing mechanisms have to form the mature RISC complex, while the other strand been reviewed in [15] and [16]. However, it has been shown (passenger) is quickly removed and degraded [10, 11]. This that miRNAs could bind functional sites in the 5′UTR as selection, however, is flexible depending on types, well as in the protein-coding region of mRNAs, promoting developmental stages and disease states [11]. This means gene translation in certain circumstances [17–19]. While a that either the miRNA-3p or the miRNA-5p may be selected single miRNA may target several (often, but not always, to form the mature RISC complex (reviewed in [11]). functionally related) genes, a single mRNA may harbor The biological function of miRNAs is usually, but many binding sites for the same or different miRNAs [20]. not exclusively, attributed to the repression of protein Moreover, two miRNA-binding sites closely located on a Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 669 mRNA could act synergistically to post-transcriptionally urine, as well as feces [38–42], demonstrating their remark‑ regulate [21]. able stability and supporting their great potential as non- Although the central role of miRNAs is to negatively invasive biomarkers. In plasma, miRNAs are transported regulate protein synthesis, other functions of miRNAs are and protected against RNAse-dependent degradation­ by emerging (reviewed in [22] and [23]). Particularly, miRNAs various carriers, such as vesicles (exosomes, microvesi‑ have been found to regulate the stability of nuclear tran‑ cles, apoptotic bodies), RNA-binding proteins [AGO2 and scripts (e.g. mRNAs, long non-coding RNAs (lncRNAs) nucleophosmin 1 (NPM1)], and lipoproteins [HDL, inter‑ or pri-miRNAs), induce epigenetic alterations at specific mediate density lipoprotein (IDL) and LDL] [37, 43]. The gene promoters, and likely modulate alternative splicing next section focuses on the role of miRNAs in lipoprotein events in the nuclear compartment [22–26]. Moreover, metabolism and function regulation. two studies have reported few specific miRNAs as ligands for toll-like receptor (TLRs; TLR7 in mice and TLR8 in humans) activation [27, 28]. Overall, miRNAs are impor‑ tant gene expression regulators that, cooperatively and at Role of miRNAs in lipoprotein and various steps of the gene expression process, orchestrate the fine-tuning of gene expression at diverse development lipid metabolism stages, for different cell types and disease states [20]. miRNAs as regulators of lipid metabolism and function miRNA expression regulation and Cholesterol is an essential molecule for animal cell physi‑ stability ology. It is involved in the regulation of the plasma mem‑ brane fluidity and integrity, as well as in membrane protein The steady-state concentration of mature miRNAs in cells functionality (e.g. cell signaling, intracellular transport). depends on their tightly regulated transcription, matura‑ It is the precursor for steroid hormones, bile acids, and tion, and decay (reviewed in [10] and [29]). It has been pro‑ vitamin D biosynthesis [44, 45]. As altered cholesterol posed that approximately ≈ 100 copies of a mature miRNA levels have been repeatedly associated with cardiometa‑ per cell is required for biologically relevant repression of bolic diseases (e.g. atherosclerosis), a strict regulation of gene translation [30, 31]. The regulation of the miRNA con‑ the lipoprotein metabolism (Figure 2; reviewed in [62]) is centration therefore plays an important part in the control essential to maintain cholesterol homeostasis and thus of the miRNA regulatory function. Briefly, miRNA tran‑ health. Since their discovery [1], miRNAs have been pro‑ scription is regulated by specific transcription factors and posed as important regulators of metabolism homeostasis epigenetic marks (e.g. DNA methylation, histone modifi‑ in humans [63–65]. The number of miRNAs with validated cations) [13]. Like protein-coding genes, certain miRNAs target genes involved in lipid and lipoprotein metabolism display a tissue-specific expression pattern [32], support‑ has increased significantly in the last decade (Figure 2 and ing their roles in cellular differentiation and develop‑ Table 1). So far, the best characterized miRNA that regu‑ ment [33]. Moreover, the miRNA-processing efficiency is lates lipid and lipoprotein metabolism is miR-33a/b. regulated through various post-translational modifica‑ tions of the maturation machinery (Drosha, DGCR8, Dicer, TRBP, AGO) [10, 13]. Also, the miRNA itself is subjected to miR-33a/b post-transcriptional modifications such as RNA editing, methylation, uridylation and adenylation [13]. These The miR-33 family member miR-33a is evolutionarily con‑ modifications, in addition with protein complex forma‑ served across animal species, whereas miR-33b is con‑ tion (e.g. AGO) and exposure to [e.g. 5′–3′ exori‑ served only in certain mammals [81, 127]. In humans, bonuclease 2 (XRN-2)], modulate the miRNA stability and miR-33a and miR-33b are encoded within intronic regions decay [29]. of the sterol regulatory element-binding transcription miRNAs show high stability (> 24 h) in cells and factor (SREBF) genes SREBF2 and SREBF1, respectively plasma, with miRNA persistence varying from one miRNA [82]. SREBFs encode key membrane-bound transcription to another [34–37]. Interestingly, miRNAs have also been factors involved in the activation of numerous genes impli‑ found in virtually all biological fluids, including cerebro‑ cated in biosynthesis and uptake of cholesterol and in the spinal fluid, tears, saliva, breast milk, amniotic fluid, synthesis of fatty acids [e.g. 3-hydroxy-3-methyglutaryl-CoA 670 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

Peripheral cell

Intestine Circulation LPL* Liver B48 MTP* B48 B48 LRP1 B48 E E E LDLR* A5 A5 * Syn-1 Exogenous pathway CM A5 mRNA CMR CMR miR-27a/b miR-30c CM miR-128 miR-130b miR-148a LPL* B100 miR-185 B100 miR-301b MTP* B100 E LDLR* E Size VLDL Density Syn-1 VLDL IDL LPL*, HL B100 LDLR* Endogenous pathway E LDL

LCAT A1 SR-BI* A1

* A1 mRNA ecto-F1-ATPase/P2Y miR-33a/b miR-145 HDLd HDL 13 miR-26 miR-148a miR-27a/b miR-223 ABCA1*

miR-19b miR-301b A1 miR-10b miR-302a ABCA1 * miR-101 miR-378 ABCG1 * miR-106b miR-455 miR-128 miR-613 Macrophage Reverse transpor t miR-130b miR-758 miR-144

Figure 2: Lipoprotein metabolism. Mature lipoproteins are spherical structures composed (roughly) of phospholipids (in white), cholesterol (in yellow), triglycerides (in red) and apolipoproteins, which allow the transport of lipids in the bloodstream [44, 46]. Lipoproteins present diverse sizes (7–1200 nm of diameter) and can be classified based on their respective density or electrophoretic mobility [47]. The exogenous lipoprotein pathway, which comprises the apoB48-containing lipoproteins, consists in dietary lipid transport from the intestine to energy-demanding tissues such as muscle, adipose tissue and the liver [46, 48–53]. The endogenous lipoprotein pathway, which comprises the apoB100-containing lipoproteins, allows the transport of lipids from the liver to peripheral tissues [46, 49–55]. The reverse cholesterol pathway consists in the efflux of excess cholesterol from extrahepatic tissues to HDLs and its transport to the liver for metabolization and excretion into the bile in the intestine [56–60]. Importantly, circulating lipoproteins undergo constant remodeling by interacting with various partners (e.g. LCAT, CETP, PLTP, HL, LPL), which implicates cargo exchanges between different lipoproteins (reviewed in [57] and [61]). All these pathways are well described with more details in the references mentioned above. Several miRNAs with validated target genes (*) involved in these lipoprotein metabolism pathways (in blue) have been identified so far (detailed in Table 1). miR-33a/b and miR-223 (in bold) have been extensively described in this review. A1, apolipoprotein-A1; A5, apolipoprotein-A5; ABCA1, ATP-binding cassette subfamily A member 1; ABCG1, ATP- binding cassette subfamily G member 1; B48, apolipoprotein-B48; B100, Apolipoprotein-B100; CETP, cholesterol ester transfer protein; CM, chylomicron; CMR, chylomicron remnant; E, apolipoprotein-E; HDL, high-density lipoprotein HDLd, nascent discoidal HDL; HL, hepatic ; IDL, intermediate density lipoprotein; LCAT, lecithin/cholesterol acyltransferase; LDL, low-density lipoprotein LDLR, low-density lipoprotein receptor; LPL, ; LRP1, low-density lipoprotein receptor-related protein 1; MTP, microsomal triglyceride transfer protein; PLTP, phospholipid transfer protein; SR-BI, scavenger receptor class B type I; Syn-1, syndecan-1 receptor; TG, triglyceride; VLDL, very low-density lipoprotein.

reductase (HMGCR), low-density lipoprotein receptor (SREBP1), repressing fatty acid oxidation, and increasing (LDLR), fatty acid synthase (FASN), stearoyl-CoA desatu‑ intracellular cholesterol levels (miR-33b) [83, 128, 130]. rase (SCD), and acetyl-CoA carboxylase alpha (ACACA)] Several studies performed using miRNA mimics/inhibi‑ [128, 129]. Under low intracellular cholesterol levels, tors in cultured cells and animal models have suggested an MIR33A is transcribed concomitantly with SREBF2 important role of miR-33a/b in HDL and lipid metabolism and increases the cholesterol biosynthesis and uptake regulation (Table 1; reviewed in [130] and [2]). miR-33a/b (SREBP2) as well as reduces cholesterol efflux and excre‑ have been shown to regulate cholesterol efflux by tar‑ tion (miR-33a) [83, 128, 130]. Alternatively, MIR33B is co- geting the cholesterol transporter gene ABCA1 in human transcribed with SREBF1 under insulin and liver x receptor hepatocytes and human and mice macrophages, and to (LXRs) activation [5], promoting fatty acid synthesis regulate ABCG1 in mice macrophages [84, 127]. miR-33a/b Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 671

Table 1: miRNAs implicated in the regulation of lipid and lipoprotein metabolism. miRNA Regulation of miRNA Target Effect/pathway involved References expression genes miR-1 Not known LXRα Transcriptional regulation of lipid metabolism genes [66] miR-9 CREB ACAT1 Intracellular cholesterol homeostasis [67] miR-10b Protocatechuic acid (gut ABCA1 HDL biogenesis and cholesterol efflux (reverse cholesterol [68, 69] microbiota metabolite), TWIST ABCG1 transport) PPAR-α Transcriptional regulation of lipid metabolism genes miR-19b Not known ABCA1 Cholesterol efflux (reverse cholesterol transport) [70] miR-21 Dietary lipids HMGCR Cholesterol biosynthesis [71] miR-26 LXR ABCA1 Cholesterol efflux (reverse cholesterol transport) [72] ARL7 miR-27a/b Hyperlipidemia, C/EBPα ABCA1 Cholesterol trafficking and efflux (reverse cholesterol [73–77] OSBPL6 transport) ACAT1 Intracellular cholesterol homeostasis SR-BI HDL-Cholesterol uptake LDLR ApoB100-containing lipoproteins (VLDL, IDL, LDL) LDLRAP1 clearance RXRα Transcriptional regulation of lipid metabolism genes PPARγ GPAM Triglyceride-rich lipoprotein metabolism NDST1 ANGPTL3 LPL miR-29 Insulin resistance, FOXA2 HMGCR Fatty acids and cholesterol metabolism [78, 79] HMGCS2 AHR SIRT1 FOXO3 miR-30c C/EBPα MTP ApoB100-containing lipoprotein assembly and secretion [80] LPGAT1 miR-33a/b Sterols, LXR, insulin ABCA1 HDL biogenesis, cholesterol trafficking and efflux (reverse [77, 81–91] ABCG1a cholesterol transport) NCP1 OSBPL6 CROT Fatty acid oxidation CPT1A HADHB CYP7A1 Bile acid synthesis and secretion ABCB11 ATP8B1 IRS2 Insulin signaling G6PC PCK1 Glucose metabolism PDK4 Mitochondrial biogenesis and ATP production SLC25A25 PPARGC1A NSF VLDL vesicular trafficking/global apolipoprotein secretion SRC3 Transcriptional regulation of lipid metabolism genes RIP140 NFYC SREBP1 SIRT6 Post-translational regulation of metabolism-associated genes miR-34a Sterols, FFAs, FXR/SHP/p53 SIRT1 Post-translational regulation of metabolism-associated [92, 93] HNF4α genes, hepatic lipid storage and secretion miR-96 SREBP2 SR-BI HDL-Cholesterol uptake [94, 95] INSIG-2 Regulation of SREBPs activity 672 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

Table 1 (continued) miRNA Regulation of miRNA Target Effect/pathway involved References expression genes miR-101 IL-6, TNFα ABCA1 Cholesterol efflux (reverse cholesterol transport) [96] miR-106b Not known ABCA1 Cholesterol efflux (reverse cholesterol transport) [97] miR-107 Dietary lipids FASN Triglyceride biosynthesis and storage [98, 99] CLOCK Circadian rhythm, metabolic homeostasis miR-122 HNF-4α, REV-ERBα, Circadian AGPAT1 TG biosynthesis and lipid storage [100–102] rhythm CIDEC PPARβ/δ Transcriptional regulation of lipid metabolism genes miR-125a Trophic hormones, cAMP, oxLDL SR-BI HDL-Cholesterol uptake, steroidogenesis [103, 104] ORP9 Lipid uptake (macrophage) miR-128 SREBP2 ABCA1 HDL biogenesis and cholesterol efflux (reverse cholesterol [105, 106] ABCG1 transport) RXRα Transcriptional regulation of lipid metabolism genes SREBP2 LDLR ApoB100-containing lipoproteins (VLDL, IDL, LDL) clearance INSR, IRS1 Insulin signaling SIRT1 Post-translational regulation of metabolism-associated genes miR-130b Dietary lipids, TNFα, NFκB ABCA1 Cholesterol efflux (reverse cholesterol transport) [105, 107–109] LDLR ApoB100-containing lipoproteins (VLDL, IDL, LDL) clearance PPARγ Transcriptional regulation of lipid metabolism genes, PPARGC1A adipogenesis miR-144 Sterols, LXR, FXR ABCA1 HDL biogenesis and cholesterol efflux (reverse cholesterol [110, 111] transport) miR-145 PPARγ ABCA1 HDL biogenesis and cholesterol efflux (reverse cholesterol [112–114] transport) miR-148a Dietary lipids, SREBP1c ABCA1 HDL biogenesis and cholesterol efflux (reverse cholesterol [105, 115] transport) LDLR ApoB100-containing lipoproteins (VLDL, IDL, LDL) clearance SIK1 Post-translational regulation of lipogenesis genes CPT1A Fatty acid oxidation miR-181a TGF-β, Wnt/β-catenin, STAT3 IDH1 Regulation of lipid biosynthesis and β-oxidation [116] miR-182 SREBP2 FBXW7 Regulation of SREBPs activity [95] miR-185 Sterols, Dietary lipids, SREBP1c SR-BI HDL-Cholesterol uptake [94, 117, 118] SREBP2 Transcriptional regulation of lipid metabolism genes LDLR ApoB100-containing lipoproteins (VLDL, IDL, LDL) clearance miR-206 Not known LXRα Transcriptional regulation of lipid metabolism genes [66] miR-223 Sterols, C/EBPα SR-BI HDL-Cholesterol uptake [94, 119] SP3 HDL biogenesis and cholesterol efflux (reverse cholesterol transport) HMGCS1 Cholesterol biosynthesis SC4MOL miR-301b Not known ABCA1 Cholesterol efflux (reverse cholesterol transport) [105] LDLR ApoB100-containing lipoprotein (VLDL, IDL, LDL) clearance miR-302a Sterols ABCA1 HDL biogenesis and cholesterol efflux (reverse cholesterol [120] transport) miR-378 Coenzyme Q10, Dietary lipids ABCG1 Cholesterol efflux (reverse cholesterol transport) [121–123] p110α Insulin signaling CRAT Energy metabolism homeostasis MED13 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 673

Table 1 (continued) miRNA Regulation of miRNA Target Effect/pathway involved References expression genes miR-455 Trophic hormones, cAMP SR-BI HDL-Cholesterol uptake, steroidogenesis [103] miR-613 SREBP1c, Insulin, PPARγ ABCA1 Cholesterol efflux (reverse cholesterol transport) [124, 125] LXRα Transcriptional regulation of lipid metabolism genes miR-758 Sterols ABCA1 HDL biogenesis and cholesterol efflux (reverse cholesterol [126] transport) aThe miR-33a/b in the 3′UTR of ABCG1 gene is present in rodents, but is not conserved in humans. have been found to target other genes associated with cho‑ in terms of both structure and functionality (reviewed lesterol metabolism, including the Niemann-Pick disease in [132]). Indeed, HDLs are composed of a large variety type C1 (NPC1) [84], cholesterol 7α-hydroxylase (CYP7A1), of proteins (> 80), lipid species (> 200), and miRNAs [6, ATPase aminophospholipid transporter class 1 type 8B 133]. The pioneering work of Vickers et al. first suggested member 1 (ATP8B1), and ATP-binding cassette subfamily that HDLs transport and deliver functional miRNAs to B member 11 (ABCB11) genes [85, 86]. In addition to their recipient cells with gene regulation capabilities [6]. role in cholesterol metabolism homeostasis, miR-33a/b Interestingly, the HDL-carried miRNA profile was consid‑ have been found to regulate the cellular fatty acid metabo‑ ered biologically distinct from the one observed in puri‑ lism by repressing many genes contributing to fatty acid fied LDLs and exosomes, these latter being more closely oxidation in hepatocytes [e.g. carnitine palmitoyltrans‑ related to each other [6]. One of the most abundant ferase 1A (CPT1a), carnitine O-octanoyltransferase(CROT)] and best characterized miRNAs carried by HDLs is miR- [83, 87] and to regulate insulin signaling by targeting the 223-3p [6, 7, 134]. HDL-transported miR-223-3p concentra‑ insulin receptor substrate 2 (IRS2) and sirtuin 6 (SIRT6) tions have been reported to be significantly increased genes [83]. Finally, the in vivo administration of miR-33a/b in participants with homozygous familial hypercholes‑ inhibitor oligonucleotides in mice and non-human pri‑ terolemia (FH) [135], and in athero-prone mice [Apolipo‑ mates has sustainably raised plasma HDL-C levels and protein E (ApoE)−/− or LDLR−/− mouse models on high-fat decreased very low-density lipoprotein (VLDL)-associated diet (HFD)] (Table 2) [6]. Moreover, hypercholesterolemia triglyceride levels [81, 82, 84, 88, 89], two factors associ‑ has also been associated with increased hepatic cho‑ ated with the metabolic syndrome [131]. Overall, miR- lesterol and miR-223-3p levels in ApoE−/− mice on HFD, 33a/b are important regulators of metabolic homeostasis, suggesting that liver miR-223-3p expression is associ‑ including cholesterol, lipid and glucose/insulin metabo‑ ated with cholesterol levels in vivo [119]. Accordingly, lism and represent a potential novel therapeutic target in miR‑223‑3p concentration in cultured human hepato‑ atherosclerosis and the metabolic syndrome [88, 89]. cytes and mice macrophages was found positively asso‑ ciated with intracellular cholesterol concentrations, and the MIR223 promoter activity was demonstrated to be cholesterol-sensitive (downregulated in low-cholesterol miRNAs carried by lipoproteins state) [119]. Importantly, miR-223-3p was found to target several genes involved in the cholesterol metabolism. High-density lipoprotein At high intracellular cholesterol levels, miR-223-3p is upregulated and represses cholesterol biosynthesis [tar‑ HDL-carried miRNA concentration is altered in diseased geting the 3-hydroxy-3-methylglutaryl-CoA synthase 1 state (HMGCS1) and methylsterol monooxygenase 1 (SC4MOL) mRNAs] as well as HDL-C uptake [targeting the scaven‑ HDLs exert their anti-atherogenic action primarily ger receptor class B type I (SR-BI) mRNA], and indirectly through their important role in promoting the reverse enhances ABCA1-mediated cholesterol efflux to HDLs cholesterol transport [56], but they also mediate several [targeting the transcription factor SP3 (SP3) mRNA] [119]. other less well-characterized cardioprotective functions, Under low intracellular cholesterol levels, miR-223-3p such as anti-inflammatory, antioxidant, anti-thrombotic, expression is downregulated and its targets repres‑ anti-apoptotic, and insulin-secretagogue actions [132]. sion is relieved; increasing cholesterol biosynthesis and HDLs comprise an heterogeneous population of particles uptake [119]. Consequently, miR-223-3p appears as an 674 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

Table 2: Lipoprotein-transported miRNAs altered in cardiovascular disease compared to healthy controls. miRNA Disease Effect References

HDL let-7f-5p Familial hypercholesterolemia + 12.3-folds [6]a miR-24-3p Familial hypercholesterolemia + 65.3-folds [6]a miR-30c-5p Acute coronary syndrome − 1.7-folds [7]b miR-92a-3p Acute coronary syndrome − 3.0-folds [7]b c Stable angina + 2.3-folds (HDL2) [8]

+ 3.8-folds (HDL3) c Unstable angina + 2.3-folds (HDL2) [8]

+ 10.0-folds (HDL3) c Myocardial infarction + 2.8-folds (HDL2) [8]

+ 7.5-folds (HDL3) miR-106a-5p Familial hypercholesterolemia + 7.9-folds [6]a c miR-122-5p Stable angina + 7.5-folds (HDL3) [8] c Unstable angina + 8.0-folds (HDL3) [8] miR-135a-3p Familial hypercholesterolemia − 2.4-folds [6]a miR-138-1-3p Familial hypercholesterolemia − 2.0-folds [6]a miR-146a-5p Acute coronary syndrome − 1.3-folds [7]b c Unstable angina + 4.7-folds (HDL3) [8] c Myocardial infarction + 4.7-folds (HDL3) [8] miR-188-5p Familial hypercholesterolemia − 2.7-folds [6]a miR-191-5p Familial hypercholesterolemia + 4.7-folds [6]a miR-218-5p Familial hypercholesterolemia + 7.8-folds [6]a miR-222-3p Familial hypercholesterolemia + 8.2-folds [6]a miR-223-3p Familial hypercholesterolemia + 3780.6-folds [6]a miR-323-3p Familial hypercholesterolemia − 2.5-folds [6]a miR-342-3p Familial hypercholesterolemia + 30.4-folds [6]a miR-412-5p Familial hypercholesterolemia + 9.5-folds [6]a c miR-486-5p Stable angina + 1.8-folds (HDL2) [8] c Unstable angina + 3.6-folds (HDL2) [8] c Myocardial infarction + 5.0-folds (HDL2) [8] miR-509-3p Familial hypercholesterolemia − 3.7-folds [6]a miR-520c-3p Familial hypercholesterolemia − 5.4-folds [6]a miR-572 Familial hypercholesterolemia − 2.5-folds [6]a miR-573 Familial hypercholesterolemia − 3.6-folds [6]a miR-625-3p Familial hypercholesterolemia − 11.0-folds [6]a miR-632 Familial hypercholesterolemia − 3.3-folds [6]a miR-877-5p Familial hypercholesterolemia − 2.3-folds [6]a LDL None yet IDL None yet

M, men; W, women. amiRNA differently concentrated in HDLs from patients with familial hypercholesterolemia (n = 5; 3 M/2 W) compared to healthy subjects (n = 6; 6 M/0 W). bmiRNAs showing different concentration in HDLs from patients with acute coronary syndrome (n = 10; 6 M/4 W) or coronary artery disease (CAD; n = 10; 6 M/4 W) compared to healthy subjects (n = 10; 6 M/4 W). No statistically significant dif- ference was observed for LDL-carried miRNAs (n = 5 for each group). cmiRNA differently concentrated in lipoproteins from 95 patients with CAD as compared to 16 healthy subjects (3 M/13 W). Patients with CAD were separated into three groups: stable angina (n = 30; 20 M/10 W), unstable angina (n = 39; 28 M/11 W) and myocardial infarction (n = 26; 22 M/4 W). miRNA analyses were performed on pooled samples (three pools per group).

important mediator of intracellular and systemic choles‑ Wagner et al. were the first to assess the contribu‑ terol homeostasis. Other HDL-carried miRNAs have been tion of HDL-carried miRNAs to the total plasmatic miRNA associated with hypercholesterolemia, inflammation, pool [7]. The percentages of HDL-bound miRNAs related and acute coronary syndrome (ACS) when compared to to those detected in plasma associated to other carriers healthy subjects (Table 2) [6, 7]. were found to be relatively low (< 1% to ~ 8%) in healthy Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 675 subjects, with HDL-carried miR-223-3p contributing HDL-carried miRNA concentration is affected by the diet the most to the plasmatic pool of circulating miRNAs (~ 8%) [7]. A contribution level varying from one miRNA More recently, studies from our and other groups tested to another suggests that over the relatively large variety whether HDL-transported miRNA concentrations were of plasmatic miRNAs carried by HDLs, a specific subset sensitive to diet-induced metabolic changes. Tabet et al. of plasmatic miRNAs might be enriched in HDLs. These showed a mild decrease in HDL-carried miR-223-3p con‑ miRNAs would be of particular biological relevance, centration following a 12-week diet rich in proteins and potentially contributing to HDL functionality (e.g. miR- that this change in HDL-miR-223-3p level was associated 223-3p, which is abundantly transported in HDLs and with a diet-induced weight loss in overweight and obese have regulatory functions in cholesterol metabolism [119] men (Table 3) [137]. Our group reported an increased con‑ and in inflammation [136]). centration of HDL-carried miR-223-3p and miR-135a-3p, Very recently, Niculescu et al. showed that miR- which was associated with an overall worsening of the car‑ 486-5p and miR-92a-3p were upregulated in the serum of diovascular disease (CVD) risk profile (e.g. lowered HDL-C patients with CAD [including stable angina (SA), unstable and increased high-sensitivity C-reactive protein (hsCRP) angina (UA) and myocardial infarction (MI)] compared and TG concentrations; Table 3) following a 4-week dou‑ to healthy subjects (Table 2), and that these differ‑ ble-blinded, randomized, crossover, controlled dietary ences were also found significant in the HDL2 and HDL3 intervention rich in trans fatty acids (TFA) from industrial subpopulations[8]. Most importantly, the authors con‑ (iTFA) or ruminant (rTFA) sources [134]. cluded that these differences in HDL-carried miR-486-5p and miR-92a-3p concentrations may discriminate between stable and vulnerable CAD patients [8]. Interestingly, miRNA transfer to HDLs miR-92a-3p has been identified as one of the most con‑ centrated miRNAs in HDLs by Wagner et al. [7]. However, The HDL-carried miRNA profile is distinct but also shares miR-92a-3p was not preferentially associated to HDLs similarities with that of plasma, LDLs, and (to a lesser relatively to other plasmatic carriers (~ 2% contribution extent) exosomes [6, 7]. This was expected considering to the plasmatic miRNA pool) [7]. This miRNA might well that HDLs are an integral part of plasma (like other car‑ remain an excellent biomarker of CAD, but our enthusi‑ riers), that a complex interplay exists between different asm is diminished regarding its specific role in the regula‑ lipoprotein pathways, and that circulating miRNAs (inde‑ tion of the many HDL functions. pendently of the carrier) may be of common cellular origin

Table 3: Lipoprotein-carried miRNAs associated with diet-induced changes in cardiometabolic risk markers. miRNA Cardiometabolic risk marker Diet Correlation direction References

HDL miR-135a-3p LDL-triglyceride Rich in rTFA (3.7% energy) + [134]b b HDL2-cholesterol Rich in iTFA (3.7% energy) − [134] HDL-ApoA1 Rich in iTFA (3.7% energy) − [134]b Total triglyceride Rich in iTFA (3.7% energy) + [134]b miR-223-3p Weight High in proteins (30% energy) + [137]a C-reactive protein Rich in rTFA (3.7% energy) + [134]b HDL-cholesterol Rich in iTFA (3.7% energy) − [134]b Total ApoA1 Rich in iTFA (3.7% energy) − [134]b HDL-ApoA1 Rich in iTFA (3.7% energy − [134]b

ApoA1, apolipoprotein A1; iTFA, trans fatty acids from industrial source; rTFA, trans fatty acids from ruminants. aObese and overweight men followed either a diet rich in proteins (HP; 30% daily energy; n = 20) or a control diet with normal protein amount (NP; 20% daily energy; n = 27), for 12 weeks. Relatively to pre-diet baseline, mean HDL-carried miR-223-3p concentration was lowered (~ –1.3-folds) following the HP diet only, and this change in HDL-miR-223-3p level was associated with diet-induced weight loss. b Following a double-blind, randomized, crossover, controlled design, nine healthy men were fed each of the three isoenergetic diets for 4 weeks each, with a washout period of ≥ 3 weeks between diets: (1) rich in iTFA (3.7% daily energy); (2) rich in rTFA (3.7% daily energy); (3) low in TFAs (control; 0.8% daily energy). The variations in HDL-carried miR-223-3p and miR-135a-3p concentration following a diet rich in TFAs compared to the control diet were strongly correlated with diet-induced changes in cardiometabolic risk markers. 676 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

(e.g. hepatocytes, macrophages, endothelial cells). Never‑ that Huh7 cells treated with HDLs from FH patients theless, certain miRNAs (e.g. miR-223-3p) were more impor‑ (abnormally abundant in miR-105-5p) showed increased tantly transported by HDLs in plasma [7], suggesting that intracellular miR-105-5p levels, whereas no change was a subset of miRNAs might potentially be more specifically observed when cells were incubated with HDLs from (actively) loaded in HDLs rather than in other carriers. healthy patients [6]. Interestingly, 60% of the concomi‑ This process could be supported by either cell type-speci‑ tantly downregulated mRNAs in Huh7 cells treated with ficity of membrane transporters and miRNA expression or HDLs from FH subjects harbored a putative target site of post-transcriptional modifications of miRNAs that would miR-105-5p [6]. This work proposed for the first time that increase their affinity for HDLs. endogenous HDL-carried miRNAs may be transferred to The exact mechanism by which miRNAs are trans‑ recipient cells with functional capabilities, although the ferred to HDLs and delivered to recipient cells is still possible contribution of other constituents of HDLs to the poorly described [138]. An obvious transporter is ABCA1, observed transcriptomic changes in cultures hepatocytes since it mediates the cholesterol and phospholipid could not be excluded. efflux to lipid-poor ApoA1 [84], but its overexpression In a more recent publication, the same group ele‑ in cultured mice macrophage resulted only in a modest gantly demonstrated that native HDL efficiently trans‑ increase in miR-223-3p levels (1.8-folds) in reconstituted fers miR-223-3p to human coronary artery endothelial HDLs (rHDL) [6]. Interestingly, sphingomyelin (SM) binds cells (HCAEC), into which miR-223-3p was not naturally RNA and prevents its degradation by RNAses [139, 140]. transcribed nor processed [136]. Importantly, Tabet et al. Moreover, a few groups have reported that small RNAs showed that change in intracellular miR-223-3p levels could bind to phospholipid membranes using vesicular following incubation with HDLs was transcription-inde‑ models [141–144]. Whether the plasma membrane SM, pendent in cells pretreated with actinomycin D, suggest‑ enriched into lipid rafts, could also bind miRNA and ing that this change in miR-223-3p levels might likely be contribute to its export to HDLs has not been published due to HDL miRNA transfer to the cell [136]. Moreover, they so far. However, the chemical inhibition of the neutral demonstrated that HDL-transferred miR‑223-3p efficiently sphingomyelinase 2 (nSMase2; the rate-limiting repressed ICAM-1 using luciferase assays [136]. This sug‑ in ceramide biosynthesis from SM) was shown to reduce gested for the first time that HDL-carried miR-223-3p likely the cellular miRNA export by exosomes [145–147]. Using contribute to the atheroprotective HDL anti-inflammatory inhibition of the nSMase2 in cultured mice macrophage, action in endothelial cells, by limiting monocyte adhe‑ Vickers et al. reported that the miR-223-3p export to rHDLs sion to endothelial cells [136]. This is concordant with the was significantly increased (35-folds), suggesting distinct previously identified anti-inflammatory function of miR- and possibly opposing mechanisms for miRNA export to 223-3p in macrophages [149]. HDLs and exosomes through the SM-ceramide pathway Another group has measured the HDL-carried miRNA [6]. This pathway may nevertheless constitute an impor‑ transfer to human umbilical venous endothelial cells tant regulatory mechanism that will have to be further (HUVEC), vascular smooth muscle cells (SMC) and periph‑ explored. eral blood mononuclear cells (PBMC) [7]. The authors first showed that HDL-introduced exogenous Caenorhabditis elegans (cel)-miR-39 can be transferred to HUVECs, but in Delivery of functional HDL-carried miRNAs to recipient low levels only (< 10 copies/cell) [7]. This group did not cells show a significant miRNA transfer from native HDLs to HUVECs, SMCs and PBMCs, but reported a slight decrease Circulating miRNAs have been suggested to play a role in in intracellular miR-223-3p, miR-92a-3p and miR-126-3p endocrine-like intercellular communication [6, 148]. The levels, especially in the PBMCs [7]. This was observed first evidence of HDL-carried miRNA biological function‑ after a relatively short incubation with native HDLs from ality was provided in 2011 by Vickers et al. [6]. As a proof healthy subjects [7]. Interestingly, the intracellular miRNA of concept, the authors showed that native HDLs loaded levels were decreased in PBMCs after incubation with with miR‑375 or miR-223-3p miRNA mimics efficiently deliv‑ HDLs from healthy subjects, whereas they were increased ered these miRNAs to cultured human hepatocytes, with after incubation with HDLs from CAD and ACS patients a concomitant decrease in mRNA levels of two putative [7]. This differential cellular response to HDLs from target genes of miR‑223‑3p [6]. Moreover, the miRNA trans‑ healthy and diseased subjects may reflect physiological fer to recipient cells was demonstrated to be mostly SR-BI- changes in HDL functions [150]. The analysis of intra­ dependent [6]. Importantly, Vickers’ group demonstrated cellular pri-miRNA and pre-miRNA levels revealed that Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 677 the observed changes in PBMCs might be due to transcrip‑ plaque and thus vascular inflammation [152]. Noteworthy, tional changes in miR-126-3p, but not miR-223-3p and miR- the miR-155-5p concentration in macrophages was found 92a-3p [7], raising the interesting hypothesis that PBMCs to be increased in presence of native and mildly oxidized may export miRNAs to HDLs. The direction of the miRNA LDLs, whereas profoundly oxidized LDLs lowered the miR- exchange between HDL and cells (import or export) could 155-5p concentration in macrophages [152]. Although the possibly be influenced by cell culture conditions and con‑ functional potential of LDL-carried miRNAs has not been tribute to explain, at least partially, the observed discrep‑ investigated yet, this observation may suggest a possible ancies between these studies. transfer of LDL-carried miR-155-5p to macrophages or, Although the transfer (and possibly exchange) of alternatively, a change in cellular miR-155-5p expression functional HDL-carried miRNAs to recipient cells has been induced by LDLs, which is likely affected by the degree demonstrated in different cultured cells [6, 7, 119], the of LDL oxidation [7]. Taken together, these pioneering HDL’s capacity to deliver (exchange) functional miRNAs studies suggest that LDL- and IDL-carried miRNA profiles in vivo has not been investigated so far. This matter will be may be altered in the diseased state, and more particularly of major importance in the future. in CAD (Table 2), and support the assessment of the role of these lipoproteins miRNAs profiles in larger studies [7, 8].

Low-density lipoprotein and other ­ApoB-containing lipoproteins Biomarkers As for HDLs, IDL and LDL particles were also found to transport miRNAs in circulation [6–8]. Despite the recog‑ miRNA as potential biomarkers of CVDs nized implication of LDLs in atherosclerosis development and the strong association between high LDL-C levels and Due to their gene regulatory function in diverse biological increased risk of CVDs [4], the LDLs’ miRNA cargo has processes and metabolic functions [63–65, 155], as well as been less studied than that of HDLs. Initial work from their high stability in biological fluids [34–37], circulat‑ Vickers et al. allowed the identification of several miRNAs ing miRNAs have been pointed out as an exciting avenue particularly abundant in LDLs including, among others, for the identification of new, non-invasive biomarkers miR-223-3p, miR-150-5p, miR-19b-3p, miR-24-3p, miR-92a-3p of diseases. Compared with the usual protein biomark‑ and miR-146a-5p [6]. From the nine miRNAs quantified by ers, miRNAs could provide early disease markers (there Wagner’s group, miR-223-3p was also reported as the most usually needs to be tissue damage for protein biomark‑ concentrated in LDLs from healthy subjects [7]. Moreover, ers to be liberated) or greater specificity. Indeed, miRNAs miR-92a-3p has been repeatedly identified as one of the can be detected quantitatively by the real-time quantita‑ most abundant miRNA carried by LDLs and IDLs [6–8]. tive polymerase chain reaction (RT-qPCR) technique with Although most miRNAs found by Wagner et al. in high sensitivity, specificity and multiplexing potential LDLs and IDLs from healthy subjects were more abundant [43, 156]. However, RT-qPCR remains a relatively time- in HDLs [7, 8], which may partially explain the greater consuming technique compared to many analyses cur‑ enthusiasm toward the HDL-carried miRNA transport as rently used in clinical laboratories, and no consensus on compared to LDLs, Wagner’s group identified one miRNA, a data normalization method is currently established (see miR-155-5p, that was more concentrated in LDLs than in Kappel et al. in this special edition for details on miRNA HDLs [7]. miR-155-5p is implicated in the regulation of detection) [156, 157]. Also, the amount of miRNA in easily various physiological processes including hematopoie‑ accessible biological matrixes (e.g. plasma, urine) is gen‑ sis, immunity, inflammation and adipogenesis. Its over‑ erally small, making a reliable miRNA quantification and expression has been previously associated with several results reproducibility more difficult [156, 157]. Some inflammation-related pathologies such as cancers, ath‑ miRNAs are modulated in more than one pathology, lim‑ erosclerosis, CVD and obesity [151–154]. miR-155-5p plays iting the specificity and thus the applicability of these a central role in the regulation of macrophage polariza‑ miRNAs as biomarkers [156]. tion toward the M1 pro-inflammatory phenotype, and Despite these challenges, several studies have inves‑ was found to play a role in the atherogenic programming tigated the potential usefulness of miRNAs based on of macrophages by indirectly upregulating the mono‑ reported differences in miRNA signature in health and cyte chemotactic protein 1 (MCP1), a chemokine known disease [43, 156, 157]. To date, most of the literature in the to promote monocyte recruitment to the atherosclerotic field focuses on cancers and CVDs [158–160]. Numerous 678 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism reviews describe miRNAs as potential biomarkers for related cluster with CVD is much more abundant and gen‑ CVDs, individually or in a panel signature, for single or erally agrees with Niculescu’s study [173, 174]. Very inter‑ mixed use (diagnostic, prognostic, and staging) [159, estingly, miR-92a-3p has been identified as an atheromiR 161]. Some studies have compared these miRNAs to the in a large scale screen in human umbilical vein endothe‑ current reference biomarkers such as troponin (I and T) lial cells (HUVECs) searching for upregulated miRNAs in [159, 161]. Interestingly, Zampetaki et al. challenged the response to the presence of oxidized LDL and low shear use of miRNAs as predicators of cardiovascular events stress [175]. This could help in refining the CAD status and showed that circulating miR-126, miR-197 and miR-223 prediction (see Zeller et al. in this special edition for more are strongly associated with the risk of future MI, and that details on miRNAs in CVD). adding the circulating levels of these three miRNAs to the The use of circulating miRNAs in a non-invasive usual Framingham risk score (FRS) improves by ~ 15% the screening, diagnostic or prognostic of disease is an excit‑ risk classification [162]. This improvement is better than ing and awaited clinical application of miRNAs. Based on that observed when adding the hsCRP levels to the risk the current knowledge, plasmatic and lipoprotein-associ‑ score calculation [163], supporting the high potential of ated miRNAs (individually or as a set of related miRNAs) circulating miRNAs. have great potential to become clinically used biomark‑ ers in the future. However, larger well-designed studies with standardized methodologies (e.g. sampling, miRNA Lipoprotein-bound miRNAs as a biomarker extraction and quantification techniques, data normaliza‑ tion method) are needed to achieve this important goal. Lipoproteins are widely used as biomarkers in the evalua‑ tion of the CVD risk. Although these biomarkers are mainly based on lipoprotein-cholesterol levels, more recent studies suggested that the lipoprotein characteristics (e.g. Therapy composition, functionality) were more strongly associ‑ ated to the CVD risk than solely its cholesterol content Nowadays, statins are one of the most prescribed classes [164–168]. The discovery that miRNAs are transported by of drugs to treat dyslipidemia and for the prevention and lipoproteins opens up our minds to new possibilities [6]. management of CVDs [176]. Statins decrease the de novo Although many plasmatic miRNAs have been identified cholesterol synthesis through HMGCR inhibition, and as potential CVD biomarkers, most of them have not been lower the cholesterol levels in plasma through indirect correlated yet with established CVD biomarkers [161]. An upregulation of LDLR via SREBP2 activation (in response interesting hypothesis would be that the carrier-specific to decreasing cellular cholesterol levels) [176–179]. Up to miRNA signature is a more accurate biomarker of a disease now, statins have been the only lipid lowering drug con‑ state than that of plasma. The association of the specific ferring a significant (~ 25%) reduction of the risk of CVD HDL-carried miRNA profile with various diseases and [176–178]. Others lipid lowering drugs, such as fibrates metabolic status (Tables 2 and 3), as well as these miRNAs’ [180, 181], niacin [178] and ezetimibe [178], are also biological relevance illustrate the potential of HDL-trans‑ available, but their effects on CVD risk prevention are ported miRNAs as biomarkers (and therapeutics; see next limited (alone or in combination with statins). Numerous section). No study has yet evaluated the clinical benefits others molecules are currently in development targeting of HDL-associated miRNAs over that of standard biomark‑ various lipoprotein biosynthesis steps, including inhibi‑ ers. As reported above, Niculescu et al. have showed that tors of ApoB biosynthesis, MTP activity, and ApoC-III among the six miRNAs upregulated in plasma from CAD synthesis, as well as the very promising proprotein con‑ compared to healthy subjects, miR-486-5p and miR-92a-3p vertase subtilisin/kexin type 9 (PCSK9) inhibitors [178, could discriminate between CAD stages in regard to their 182, 183]. In contrast, the HDL-raising therapies devel‑ levels in HDLs [8]. MiR-486-5p has been identified in oped over the past years, including over-expression of various studies [169], and its serum concentration APOA1 gene, infusion of reconstituted HDL particles or has been reported negatively correlated with systemic ven‑ recombinant lecithin-cholesterol acyltransferase (LCAT) tricular contractility following a cardiac bypass surgery and CETP inhibitors [178, 184], have failed to prevent [170]. More importantly, miR-486-5p has been associated CVD occurrence despite a huge increase (up to 180%) with the angiogenesis process and was recently shown in HDL levels [177, 178, 185–187]. This clearly shows that to regulate the cholesterol efflux from macrophages [171, the HDL particle is much more complex than previously 172]. Literature about the association of miR-92a-3p and its thought. Accordingly, our understanding of different Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 679

HDL functions and the molecular mechanisms involved still a challenge, but one that needs to be addressed in needs to be refined. order to limit the undesirable side-effects of the treat‑ To this end, novel classes of molecules are currently ment [32]. Pharmacodynamics is also difficult to assess in development, targeting HDLs’ metabolism, parti‑ since miRNAs regulate numerous genes and pathways cle structure and function [178, 185]. One such class is and the summation of these effects requires sophisticated miRNAs, which have emerged as potential candidates analytical models. Anti-miR and miR-mimetic treatments because they are important regulators of lipid metabolism may induce toxicity related to the chemical structure of and can target several mRNAs implicated in a given the molecule and also toxicity related to the hybridiza‑ pathway. As for conventional drugs, the development of tion potential of the molecule to other undesirable target a miRNA-based drug is a challenging multi-step process mRNAs (off-target) or tissue. (recently reviewed in [188] and [189]). It begins with the Even though a good number of miRNAs have been profiling of miRNAs in health and disease states [188]. described in the literature in various pathologies and Either downregulated or upregulated miRNAs (i.e. biologi‑ are candidates to become potential miRNA-based treat‑ cally relevant in disease development or progression) are ments, only a few are now under serious development then selected and tested in vitro and in vivo for gain and [188, 189]. Two miRNA mimics (miR-34 and let-7) targeting loss of function [188]. Since miRNA levels may be either oncogenes implicated in the development of solid tumors increased or decreased, two classes of miRNA-based treat‑ are presently in preclinical development [195]. miRNA ments have been described [188–190]. miRNA antagonists, antagonists are also of interest, with candidates under also known as antagomiR or anti-miR, are antisense oli‑ development: three miRNAs (miR-195 et miR-208/499) in gonucleotides which complementary bind to the targeted preclinical targeting CVD and one miRNA (miR-122) in mature miRNA, causing its loss of function by creating phase II clinical trial targeting hepatitis C viral infection a duplex unable to bind its target mRNA and inhibit its [196, 197]. Importantly, a miR-33 antagonist is currently translation [189]. Since free-synthetic and exogenous under preclinical development [129, 189]. As reported miRNAs are rapidly degraded in biological fluids, these above, miR-33 is central in cholesterol homeostasis, and antagomiRs need to be structurally stabilized in order its inhibition raises HDL-C levels, increases the choles‑ to increase their half-life and cellular uptake [189]. The terol reverse transport from macrophages and is athero‑ antagomiR structural stabilization can be achieved by the protective. Other miRNAs are also good candidates for incorporation of 2′-O-methyl modified oligonucleotides as the treatment of dyslipidemias, including miR-30c-5p and well as locked nucleic acid (LNA) modified nucleotides miR-148a-3p. The overexpression of miR-30c-5p has been or by modifying linkages between nucleotides [191, 192]. showed to reduce plaque formation in an atherosclerotic Also, adding a cholesterol molecule to the original antago‑ mouse model and, alternatively, its inhibition has induced miR structure has already been used to increase its cel‑ hyperlipidemia and increased atherosclerotic plaque for‑ lular uptake (more specifically in the cardiac tissue) [191]. mation [80]. The action of miR-30c-5p was mediated by the Another strategy to downregulate a miRNA function is the inhibition of MTP, which regulates the secretion of VLDL use of miRNA sponges, which harbor multiple comple‑ by the liver, even though miR-30c-5p can also mediate its mentary sites to the targeted miRNA [193]. action in a MTP-independent way. miR-148a-3p is also an Furthermore, the levels of miRNAs downregulated in interesting target for a miRNA-based treatment, because disease may be normalized using miRNA mimics, which this miRNA has been found to downregulate two genes are synthetic double-stranded oligonucleotides that important in lipoprotein metabolism: LDLR and ABCA1 mimic the miRNA function they replace [188, 189]. Similar [115]. The expression of miR-148a-3p was increased in the to antagomiRs, miRNA mimics are chemically modified to liver of high-fat diet mice and rhesus monkeys [105], and improve their stability and cellular uptake [188]. However, its inhibition in huh7 cells and mice models was shown only the “passenger strand” is usually modified, while the to significantly increase hepatic LDLR and ABCA1 expres‑ “guide strand” remains identical to the mature miRNA sion, concomitant with increased HDL-C and decreased of interest [189]. Alternatively, the expression of a down‑ LDL-C levels in circulation [105, 115]. Moreover, miR- regulated target miRNA may be stably normalized by the 148a-3p was also able to repress ABCA1 and the cholesterol use of adenovirus-associated vectors and tissue-specific efflux capacity in mice macrophages [105]. Also, a large ­promoters [194]. genome-wide association study (GWAS) meta-analysis Various methods of miRNA-based treatment deliv‑ (> 188,000 individuals) identified two polymorphisms ery have been described [188]. It is clear that the specific within the MIR148a promoter region that were associ‑ delivery of the molecule to a particular site of action is ated with plasma levels of total cholesterol, LDL-C and 680 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism triglycerides, suggesting that this miRNA is involved in an exciting research avenue to explore in regard of lipo‑ lipid homeostasis in humans [105]. These miRNAs have protein functionality and potential miRNA-based clinical been linked to the lipid metabolism and atherosclerosis applications for the prevention and treatment of cardio‑ and are thus very promising novel therapeutic targets. metabolic diseases. The demonstration that HDLs could transport a spe‑ cific and metabolically sensitive set of functional miRNAs Acknowledgments: We express our gratitude to Céline and deliver them to recipient cells led us to an endocrine- Bélanger (CIUSSS du Saguenay–Lac-St-Jean, Hôpital de like signaling pathway which still needs to be validated in Chicoutimi) for her thoughtful revision of the manuscript. vivo [6]. This discovery could improve our understanding Author contributions: All the authors have accepted of the functionality of HDLs in cardioprotection. It could responsibility for the entire content of this submitted support the interesting possibility of using HDL particles manuscript and approved submission. to deliver specific miRNA therapies. Infusion of synthetic Research funding: VD was recipient of a doctoral research HDL particles is presently in phase II trial [184]. These award from the Fonds de recherche du Québec en santé particles are composed of ApoA1 and phospholipids, (FRQS). LB is junior research scholar from the FRQS and mimicking nascent HDLs. Loading these synthetic HDLs member of the FRQS-funded Centre de recherche du with functional miRNAs is an exciting avenue to improve CHUS (affiliated to the Centre hospitalier universitaire de HDL-directed therapy, by which HDLs could stabilize and Sherbrooke). deliver functional antagonist or mimetic miRNAs. Employment or leadership: None declared. Honorarium: None declared. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and Conclusions interpretation of data; in the writing of the report; or in the decision to submit the report for publication. The involvement of miRNAs as central regulators of the lipoprotein metabolism and cholesterol homeostasis is now well established. The role of lipoprotein-carried References miRNAs as functional regulators or useful biomarkers is emerging. HDLs have been shown to transport and 1. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic deliver functional and metabolically-sensitive miRNAs to gene lin-4 encodes small RNAs with antisense complementarity recipient cells in vitro (e.g. hepatocytes and endothelial to lin-14. Cell 1993;75:843–54. cells), raising the interesting hypothesis of an endocrine- 2. Rayner KJ, Moore KJ. MicroRNA control of high-density lipoprotein metabolism and function. Circ Res 2014;114:183–92. like intercellular communication mediated by HDLs 3. Rotllan N, Price N, Pati P, Goedeke L, Fernandez-Hernando C. (and possibly other lipoproteins). This may contribute microRNAs in lipoprotein metabolism and cardiometabolic disor- to mediate the HDL-cardioprotective functions. Despite ders. Atherosclerosis 2016;246:352–60. the enthusiasm raised by the lipoprotein (mostly HDLs)- 4. Goedeke L, Wagschal A, Fernandez-Hernando C, Naar AM. miRNA carried miRNAs and the increasing number of publica‑ regulation of LDL-cholesterol metabolism. Biochim Biophys Acta 2016;1861(12 Pt B):2047–52. tions in this area, the current lack of data still limits our 5. DiMarco DM, Fernandez ML. The regulation of reverse cholesterol understanding of the biological relevance and functions transport and cellular cholesterol homeostasis by microRNAs. of these miRNAs in vivo. More studies are needed to elu‑ Biology (Basel) 2015;4:494–511. cidate the molecular mechanism by which miRNAs are 6. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. exchanged between lipoproteins and cells (e.g. cellular MicroRNAs are transported in plasma and delivered to recipient origin and destination, miRNA ligand, receptors, speci‑ cells by high-density lipoproteins. Nat Cell Biol 2011;13:423–33. 7. Wagner J, Riwanto M, Besler C, Knau A, Fichtlscherer S, Roxe T, ficity), as well as their functional impact in intercellu‑ et al. Characterization of levels and cellular transfer of circulat- lar communication in vivo. Moreover, the physiological ing lipoprotein-bound microRNAs. Arterioscler Thromb Vasc Biol relevance of the plasmatic miRNAs preferential location 2013;33:1392–400. (e.g. HDLs, LDLs, exosomes, RNA-binding proteins) in 8. Niculescu LS, Simionescu N, Sanda GM, Carnuta MG, Stancu CS, regard to cardiometabolic homeostasis has also not been Popescu AC, et al. MiR-486 and miR-92a identified in circulating determined yet. Larger studies will also be necessary to HDL discriminate between stable and vulnerable coronary artery disease patients. PLoS One 2015;10:e0140958. fully investigate the biomarker (refinement) and thera‑ 9. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian peutic potential of lipoprotein-carried miRNAs in dis‑ mRNAs are conserved targets of microRNAs. Genome Res eases. In conclusion, lipoprotein transport of miRNAs is 2009;19:92–105. Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 681

10. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol 31. Sarasin-Filipowicz M, Krol J, Markiewicz I, Heim MH, Filipowicz Cell Biol 2014;15:509–24. W. Decreased levels of microRNA miR-122 in individuals with 11. Meijer HA, Smith EM, Bushell M. Regulation of miRNA hepatitis C responding poorly to interferon therapy. Nat Med strand selection: follow the leader? Biochem Soc Trans 2009;15:31–3. 2014;42:1135–40. 32. Guo Z, Maki M, Ding R, Yang Y, Zhang B, Xiong L. Genome-wide 12. Witkos TM, Koscianska E, Krzyzosiak WJ. Practical aspects of survey of tissue-specific microRNA and transcription factor microRNA target prediction. Curr Mol Med 2011;11:93–109. regulatory networks in 12 tissues. Sci Rep 2014;4:5150. 13. Davis BN, Hata A. Regulation of microRNA biogenesis: a miRiad 33. Shivdasani RA. MicroRNAs: regulators of gene expression and of mechanisms. Cell Commun Signal 2009;7:18. cell differentiation. Blood 2006;108:3646–53. 14. Bratkovic T, Glavan G, Strukelj B, Zivin M, Rogelj B. Exploiting 34. Gantier MP, McCoy CE, Rusinova I, Saulep D, Wang D, Xu D, et al. microRNAs for cell engineering and therapy. Biotechnol Adv Analysis of microRNA turnover in mammalian cells following 2012;30:753–65. Dicer1 ablation. Nucleic Acids Res 2011;39:5692–703. 15. Jonas S, Izaurralde E. Towards a molecular ­understanding 35. Marzi MJ, Ghini F, Cerruti B, de Pretis S, Bonetti P, Giacomelli C, of microRNA-mediated . Nat Rev Genet et al. Degradation dynamics of microRNAs revealed by a novel 2015;16:421–33. pulse-chase approach. Genome Res 2016;26:554–65. 16. Fabian MR, Sonenberg N. The mechanics of miRNA-mediated 36. Guo Y, Liu J, Elfenbein SJ, Ma Y, Zhong M, Qiu C, et al. gene silencing: a look under the hood of miRISC. Nat Struct Mol Characterization of the mammalian miRNA turnover landscape. Biol 2012;19:586–93. Nucleic Acids Res 2015;43:2326–41. 17. Lytle JR, Yario TA, Steitz JA. Target mRNAs are repressed as effi- 37. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, ciently by microRNA-binding sites in the 5' UTR as in the 3′ UTR. Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable Proc Natl Acad Sci USA 2007;104:9667–72. blood-based markers for cancer detection. Proc Natl Acad Sci 18. Forman JJ, Legesse-Miller A, Coller HA. A search for conserved USA 2008;105:10513–8. sequences in coding regions reveals that the let-7 microRNA 38. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, targets Dicer within its coding sequence. Proc Natl Acad Sci USA et al. The microRNA spectrum in 12 body fluids. Clin Chem 2008;105:14879–84. 2010;56:1733–41. 19. Orom UA, Nielsen FC, Lund AH. MicroRNA-10a binds the 5′UTR of 39. Igaz I, Igaz P. Diagnostic relevance of microRNAs in Other body ribosomal protein mRNAs and enhances their translation. Mol fluids including urine, feces, and saliva. EXS 2015;106:245–52. Cell 2008;30:460–71. 40. Salido-Guadarrama AI, Morales-Montor JG, Rangel-Escareno C, 20. Sayed D, Abdellatif M. MicroRNAs in development and disease. Langley E, Peralta-Zaragoza O, Cruz Colin JL, et al. Urinary Physiol Rev 2011;91:827–87. microRNA-based signature improves accuracy of detection of 21. Saetrom P, Heale BS, Snove O, Jr, Aagaard L, Alluin J, Rossi JJ. clinically relevant prostate cancer within the prostate-specific Distance constraints between microRNA target sites dictate antigen grey zone. Mol Med Rep 2016;13:4549–60. efficacy and . Nucleic Acids Res 2007;35:2333–42. 41. Burgos K, Malenica I, Metpally R, Courtright A, Rakela B, 22. Cipolla GA. A non-canonical landscape of the microRNA system. Beach T, et al. Profiles of extracellular miRNA in cerebrospinal Front Genet 2014;5:337. fluid and serum from patients with Alzheimer’s and Parkinson’s 23. Roberts TC. The microRNA biology of the mammalian Nucleus. diseases correlate with disease status and features of pathol- Mol Ther Nucleic Acids 2014;3:e188. ogy. PLoS One 2014;9:e94839. 24. Chen X, Liang H, Zhang CY, Zen K. miRNA regulates noncod- 42. Wang C, Zhao K, Rong Q. Diagnostic value of fecal microRNAs for ing RNA: a noncanonical function model. Trends Biochem Sci colorectal cancer: a meta-analysis. Clin Lab 2015;61:1845–53. 2012;37:457–9. 43. Creemers EE, Tijsen AJ, Pinto YM. Circulating microRNAs: novel 25. Kim DH, Saetrom P, Snove O, Jr, Rossi JJ. MicroRNA-directed tran- biomarkers and extracellular communicators in cardiovascular scriptional gene silencing in mammalian cells. Proc Natl Acad disease? Circ Res 2012;110:483–95. Sci USA 2008;105:16230–5. 44. Gagné CG, Gaudet D. Les Dyslipoprotéinémies: L’approche 26. Liu J, Hu J, Corey DR. Expanding the action of duplex RNAs into ­Clinique. Chapitre 3: Les lipides et les lipoproténes. 3rd ed. the nucleus: redirecting alternative splicing. Nucleic Acids Res Québec, Canada: Claude Gagné, 2007. 2012;40:1240–50. 45. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. 27. Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, Nature 1990;343:425–30. et al. MicroRNAs bind to Toll-like receptors to induce pro- 46. Feingold KR, Grunfeld C. Introduction to lipids and lipoproteins. metastatic inflammatory response. Proc Natl Acad Sci USA In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, 2012;109:E2110–6. Hershman JM, et al., editors. Endotext. South Dartmouth, MA: 28. Lehmann SM, Kruger C, Park B, Derkow K, Rosenberger K, MDText.com, Inc., 2000. Baumgart J, et al. An unconventional role for miRNA: let-7 47. Mills GL, Lane PA, Weech PK. A guidebook to lipoprotein activates Toll-like receptor 7 and causes neurodegeneration. Nat ­techniques. Amsterdam, The Netherlands: Elsevier Science, 2000. Neurosci 2012;15:827–35. 48. Dash S, Xiao C, Morgantini C, Lewis GF. New insights into 29. Kai ZS, Pasquinelli AE. MicroRNA assassins: factors that regulate the regulation of chylomicron production. Annu Rev Nutr the disappearance of miRNAs. Nat Struct Mol Biol 2010;17:5–10. 2015;35:265–94. 30. Brown BD, Gentner B, Cantore A, Colleoni S, Amendola M, 49. Wilsie LC, Gonzales AM, Orlando RA. Syndecan-1 mediates Zingale A, et al. Endogenous microRNA can be broadly exploited internalization of apoE-VLDL through a low density lipoprotein to regulate transgene expression according to tissue, lineage receptor-related protein (LRP)-independent, non-clathrin- and differentiation state. Nat Biotechnol 2007;25:1457–67. mediated pathway. Lipids Health Dis 2006;5:23. 682 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

50. Mahley RW, Huang Y. Atherogenic remnant lipoproteins: role for ­transport in mice via repressing miRNA-10b. Circ Res proteoglycans in trapping, transferring, and internalizing. J Clin 2012;111:967–81. Invest 2007;117:94–8. 69. Zheng L, Lv GC, Sheng J, Yang YD. Effect of miRNA-10b in regulat- 51. Foley EM, Gordts PL, Stanford KI, Gonzales JC, Lawrence R, ing cellular steatosis level by targeting PPAR-alpha expression, a Stoddard N, et al. Hepatic remnant lipoprotein clearance by novel mechanism for the pathogenesis of NAFLD. J Gastroenterol heparan sulfate proteoglycans and low-density lipoprotein Hepatol 2010;25:156–63. receptors depend on dietary conditions in mice. Arterioscler 70. Lv YC, Tang YY, Peng J, Zhao GJ, Yang J, Yao F, et al. MicroRNA- Thromb Vasc Biol 2013;33:2065–74. 19b promotes macrophage cholesterol accumulation and aortic 52. Mahley RW, Ji ZS. Remnant lipoprotein metabolism: key path- atherosclerosis by targeting ATP-binding cassette transporter ways involving cell-surface heparan sulfate proteoglycans and A1. Atherosclerosis 2014;236:215–26. apolipoprotein E. J Lipid Res 1999;40:1–16. 71. Sun C, Huang F, Liu X, Xiao X, Yang M, Hu G, et al. miR-21 53. Stanford KI, Bishop JR, Foley EM, Gonzales JC, Niesman IR, regulates triglyceride and cholesterol metabolism in non- Witztum JL, et al. Syndecan-1 is the primary heparan sulfate alcoholic fatty liver disease by targeting HMGCR. Int J Mol Med proteoglycan mediating hepatic clearance of triglyceride-rich 2015;35:847–53. lipoproteins in mice. J Clin Invest 2009;119:3236–45. 72. Sun D, Zhang J, Xie J, Wei W, Chen M, Zhao X. MiR-26 controls 54. Hossain T, Riad A, Siddiqi S, Parthasarathy S, Siddiqi SA. Mature LXR-dependent cholesterol efflux by targeting ABCA1 and ARL7. VLDL triggers the biogenesis of a distinct vesicle from the trans- FEBS Lett 2012;586:1472–9. Golgi network for its export to the plasma membrane. Biochem J 73. Zhang M, Wu JF, Chen WJ, Tang SL, Mo ZC, Tang YY, et al. 2014;459:47–58. MicroRNA-27a/b regulates cellular cholesterol efflux, influx and 55. Mendivil CO, Zheng C, Furtado J, Lel J, Sacks FM. Metabolism of esterification/hydrolysis in THP-1 macrophages. Atherosclerosis very-low-density lipoprotein and low-density lipoprotein con- 2014;234:54–64. taining apolipoprotein C-III and not other small apolipoproteins. 74. Goedeke L, Rotllan N, Ramirez CM, Aranda JF, Canfran-Duque Arterioscler Thromb Vasc Biol 2010;30:239–45. A, Araldi E, et al. miR-27b inhibits LDLR and ABCA1 expression 56. Badimon JJ, Ibanez B. Increasing high–density lipoprotein as a but does not influence plasma and hepatic lipid levels in mice. therapeutic target in atherothrombotic disease. Rev Esp Cardiol Atherosclerosis 2015;243:499–509. 2010;63:323–33. 75. Shirasaki T, Honda M, Shimakami T, Horii R, Yamashita T, 57. Zannis VI, Fotakis P, Koukos G, Kardassis D, Ehnholm C, Sakai Y, et al. MicroRNA-27a regulates lipid metabolism and Jauhiainen M, et al. HDL biogenesis, remodeling, and inhibits hepatitis C replication in human hepatoma cells. catabolism. Handb Exp Pharmacol 2015;224:53–111. J Virol 2013;87:5270–86. 58. Phillips MC. Molecular mechanisms of cellular cholesterol efflux. 76. Vickers KC, Shoucri BM, Levin MG, Wu H, Pearson DS, J Biol Chem 2014;289:24020–9. Osei-Hwedieh D, et al. MicroRNA-27b is a regulatory hub in 59. Tanigawa H, Billheimer JT, Tohyama J, Zhang Y, Rothblat G, Rader lipid metabolism and is altered in dyslipidemia. Hepatology DJ. Expression of cholesteryl ester transfer protein in mice 2013;57:533–42. promotes macrophage reverse cholesterol transport. Circulation 77. Ouimet M, Hennessy EJ, van Solingen C, Koelwyn GJ, 2007;116:1267–73. Hussein MA, Ramkhelawon B, et al. miRNA targeting of 60. Martinez LO, Najib S, Perret B, Cabou C, Lichtenstein L. Ecto- oxysterol-binding protein-like 6 regulates cholesterol trafficking F1-ATPase/P2Y pathways in metabolic and vascular functions of and efflux. Arterioscler Thromb Vasc Biol 2016;36:942–51. high density lipoproteins. Atherosclerosis 2015;238:89–100. 78. Kurtz CL, Fannin EE, Toth CL, Pearson DS, Vickers KC, 61. Brunham LR, Hayden MR. Human genetics of HDL: Insight Sethupathy P. Inhibition of miR-29 has a significant lipid- into particle metabolism and function. Prog Lipid Res lowering benefit through suppression of lipogenic programs 2015;58:14–25. in liver. Sci Rep 2015;5:12911. 62. Ramasamy I. Recent advances in physiological lipoprotein 79. Kurtz CL, Peck BC, Fannin EE, Beysen C, Miao J, Landstreet SR, metabolism. Clin Chem Lab Med 2014;52:1695–727. et al. MicroRNA-29 fine-tunes the expression of key FOXA2- 63. Fernandez-Hernando C, Ramirez CM, Goedeke L, Suarez Y. activated lipid metabolism genes and is dysregulated in MicroRNAs in metabolic disease. Arterioscler Thromb Vasc Biol animal models of insulin resistance and diabetes. Diabetes 2013;33:178–85. 2014;63:3141–8. 64. Fernandez-Hernando C, Suarez Y, Rayner KJ, Moore KJ. Micro­ 80. Soh J, Iqbal J, Queiroz J, Fernandez-Hernando C, Hussain MM. RNAs in lipid metabolism. Curr Opin Lipidol 2011;22:86–92. MicroRNA-30c reduces hyperlipidemia and atherosclerosis in 65. Moore KJ, Rayner KJ, Suarez Y, Fernandez-Hernando C. micro- mice by decreasing lipid synthesis and lipoprotein secretion. RNAs and cholesterol metabolism. Trends Endocrinol Metab Nat Med 2013;19:892–900. 2010;21:699–706. 81. Marquart TJ, Allen RM, Ory DS, Baldan A. miR-33 links SREBP-2 66. Zhong D, Huang G, Zhang Y, Zeng Y, Xu Z, Zhao Y, et al. Micro- induction to repression of sterol transporters. Proc Natl Acad Sci RNA-1 and microRNA-206 suppress LXRalpha-induced lipogen- USA 2010;107:12228–32. esis in hepatocytes. Cell Signal 2013;25:1429–37. 82. Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, Gerszten 67. Xu J, Hu G, Lu M, Xiong Y, Li Q, Chang CC, et al. MiR-9 reduces RE, et al. MicroRNA-33 and the SREBP host genes cooperate to human acyl-coenzyme A: cholesterol acyltransferase-1 to control cholesterol homeostasis. Science 2010;328:1566–9. decrease THP-1 macrophage-derived foam cell formation. Acta 83. Davalos A, Goedeke L, Smibert P, Ramirez CM, Warrier NP, Biochim Biophys Sin (Shanghai) 2013;45:953–62. Andreo U, et al. miR-33a/b contribute to the regulation of fatty 68. Wang D, Xia M, Yan X, Li D, Wang L, Xu Y, et al. Gut ­microbiota acid metabolism and insulin signaling. Proc Natl Acad Sci USA metabolism of anthocyanin promotes reverse ­cholesterol 2011;108:9232–7. Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 683

84. Rayner KJ, Suarez Y, Davalos A, Parathath S, Fitzgerald ML, 99. Bhatia H, Verma G, Datta M. miR-107 orchestrates ER stress Tamehiro N, et al. MiR-33 contributes to the regulation of induction and lipid accumulation by post-transcriptional cholesterol homeostasis. Science 2010;328:1570–3. regulation of fatty acid synthase in hepatocytes. Biochim 85. Allen RM, Marquart TJ, Albert CJ, Suchy FJ, Wang DQ, Biophys Acta 2014;1839:334–43. Ananthanarayanan M, et al. miR-33 controls the expression 100. Hsu SH, Wang B, Kota J, Yu J, Costinean S, Kutay H, et al. of biliary transporters, and mediates statin- and diet-induced Essential metabolic, anti-inflammatory, and anti-tumorigenic hepatotoxicity. EMBO Mol Med 2012;4:882–95. functions of miR-122 in liver. J Clin Invest 2012;122:2871–83. 86. Li T, Francl JM, Boehme S, Chiang JY. Regulation of 101. Gatfield D, Le Martelot G, Vejnar CE, Gerlach D, Schaad O, cholesterol and bile acid homeostasis by the cholesterol Fleury-Olela F, et al. Integration of microRNA miR-122 in hepatic 7alpha-hydroxylase/steroid response element-binding circadian gene expression. Genes Dev 2009;23:1313–26. protein 2/microRNA-33a axis in mice. Hepatology 102. Jopling C. Liver-specific microRNA-122: Biogenesis and func- 2013;58:1111–21. tion. RNA Biol 2012;9:137–42. 87. Gerin I, Clerbaux LA, Haumont O, Lanthier N, Das AK, Burant 103. Hu Z, Shen WJ, Kraemer FB, Azhar S. MicroRNAs 125a and 455 CF, et al. Expression of miR-33 from an SREBP2 intron inhib- repress lipoprotein-supported steroidogenesis by targeting its cholesterol export and fatty acid oxidation. J Biol Chem scavenger receptor class B type I in steroidogenic cells. Mol 2010;285:33652–61. Cell Biol 2012;32:5035–45. 88. Rayner KJ, Sheedy FJ, Esau CC, Hussain FN, Temel RE, 104. Chen T, Huang Z, Wang L, Wang Y, Wu F, Meng S, et al. Micro- Parathath S, et al. Antagonism of miR-33 in mice promotes RNA-125a-5p partly regulates the inflammatory response, lipid reverse cholesterol transport and regression of atherosclerosis. uptake, and ORP9 expression in oxLDL-stimulated monocyte/ J Clin Invest 2011;121:2921–31. macrophages. Cardiovasc Res 2009;83:131–9. 89. Rayner KJ, Esau CC, Hussain FN, McDaniel AL, Marshall SM, van 105. Wagschal A, Najafi-Shoushtari SH, Wang L, Goedeke L, Sinha S, Gils JM, et al. Inhibition of miR-33a/b in non-human primates deLemos AS, et al. Genome-wide identification of microRNAs raises plasma HDL and lowers VLDL triglycerides. Nature regulating cholesterol and triglyceride homeostasis. Nat Med 2011;478:404–7. 2015;21:1290–7. 90. Horie T, Ono K, Horiguchi M, Nishi H, Nakamura T, Nagao K, et al. 106. Adlakha YK, Khanna S, Singh R, Singh VP, Agrawal A, Saini MicroRNA-33 encoded by an intron of sterol regulatory element- N. Pro-apoptotic miRNA-128-2 modulates ABCA1, ABCG1 and binding protein 2 (Srebp2) regulates HDL in vivo. Proc Natl Acad RXRalpha expression and cholesterol homeostasis. Cell Death Sci USA 2010;107:17321–6. Dis 2013;4:e780. 91. Karunakaran D, Thrush AB, Nguyen MA, Richards L, Geoffrion M, 107. Lee EK, Lee MJ, Abdelmohsen K, Kim W, Kim MM, Srikantan S, Singaravelu R, et al. Macrophage mitochondrial energy status et al. miR-130 suppresses adipogenesis by inhibiting peroxi- regulates cholesterol efflux and is enhanced by anti-miR33 in some proliferator-activated receptor gamma expression. Mol atherosclerosis. Circ Res 2015;117:266–78. Cell Biol 2011;31:626-38. 92. Lee J, Padhye A, Sharma A, Song G, Miao J, Mo YY, et al. 108. Wang YC, Li Y, Wang XY, Zhang D, Zhang H, Wu Q, et al. A ­pathway involving farnesoid X receptor and small Circulating miR-130b mediates metabolic crosstalk between heterodimer partner positively regulates hepatic sirtuin 1 fat and muscle in overweight/obesity. Diabetologia levels via microRNA-34a inhibition. J Biol Chem 2013;56:2275–85. 2010;285:12604–11. 109. Kim C, Lee H, Cho YM, Kwon OJ, Kim W, Lee EK. TNFalpha- 93. Xu Y, Zalzala M, Xu J, Li Y, Yin L, Zhang Y. A metabolic stress- induced miR-130 resulted in adipocyte dysfunction during inducible miR-34a-HNF4alpha pathway regulates lipid and obesity-related inflammation. FEBS Lett 2013;587:3853–8. lipoprotein metabolism. Nat Commun 2015;6:7466. 110. Ramirez CM, Rotllan N, Vlassov AV, Davalos A, Li M, Goedeke 94. Wang L, Jia XJ, Jiang HJ, Du Y, Yang F, Si SY, et al. MicroRNAs 185, L, et al. Control of cholesterol metabolism and plasma 96, and 223 repress selective high-density lipoprotein choles- high-density lipoprotein levels by microRNA-144. Circ Res terol uptake through posttranscriptional inhibition. Mol Cell Biol 2013;112:1592–601. 2013;33:1956–64. 111. de Aguiar Vallim TQ, Tarling EJ, Kim T, Civelek M, Baldan A, 95. Jeon TI, Esquejo RM, Roqueta-Rivera M, Phelan PE, Moon YA, Esau C, et al. MicroRNA-144 regulates hepatic ATP binding Govindarajan SS, et al. An SREBP-responsive microRNA cassette transporter A1 and plasma high-density lipoprotein operon contributes to a regulatory loop for intracellular lipid after activation of the nuclear receptor farnesoid X receptor. homeostasis. Cell Metab 2013;18:51–61. Circ Res 2013;112:1602–12. 96. Zhang N, Lei J, Lei H, Ruan X, Liu Q, Chen Y, et al. MicroRNA-101 112. Kang MH, Zhang LH, Wijesekara N, de Haan W, Butland S, overexpression by IL-6 and TNF-alpha inhibits cholesterol efflux Bhattacharjee A, et al. Regulation of ABCA1 protein expression by suppressing ATP-binding cassette transporter A1 expression. and function in hepatic and pancreatic islet cells by miR-145. Exp Cell Res 2015;336:33–42. Arterioscler Thromb Vasc Biol 2013;33:2724–32. 97. Kim J, Yoon H, Ramirez CM, Lee SM, Hoe HS, Fernandez- 113. Sala F, Aranda JF, Rotllan N, Ramirez CM, Aryal B, Elia L, et al. Hernando C, et al. MiR-106b impairs cholesterol efflux and MiR-143/145 deficiency attenuates the progression of athero- increases Abeta levels by repressing ABCA1 expression. Exp sclerosis in Ldlr-/-mice. Thromb Haemost 2014;112:796–802. Neurol 2012;235:476–83. 114. Dharap A, Pokrzywa C, Murali S, Kaimal B, Vemuganti R. Mutual 98. Daimiel-Ruiz L, Klett-Mingo M, Konstantinidou V, Mico V, induction of transcription factor PPARgamma and microRNAs Aranda JF, Garcia B, et al. Dietary lipids modulate the expression miR-145 and miR-329. J Neurochem. 2015;135:139–46. of miR-107, a miRNA that regulates the circadian system. Mol 115. Goedeke L, Rotllan N, Canfran-Duque A, Aranda JF, Ramirez Nutr Food Res 2015;59:1865–78. CM, Araldi E, et al. MicroRNA-148a regulates LDL receptor and 684 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

ABCA1 expression to control circulating lipoprotein levels. Nat International Association for the Study of Obesity. Circulation Med 2015;21:1280–9. 2009;120:1640–5. 116. Chu B, Wu T, Miao L, Mei Y, Wu M. MiR-181a regulates lipid 132. Asztalos BF, Tani M, Schaefer EJ. Metabolic and functional metabolism via IDH1. Sci Rep 2015;5:8801. ­relevance of HDL subspecies. Curr Opin Lipidol 117. Yang M, Liu W, Pellicane C, Sahyoun C, Joseph BK, Gallo-Ebert 2011;22:176–85. C, et al. Identification of miR-185 as a regulator of de novo 133. Annema W, von Eckardstein A. High-density lipoproteins. Multi- cholesterol biosynthesis and low density lipoprotein uptake. J functional but vulnerable protections from atherosclerosis. Circ Lipid Res 2014;55:226–38. J 2013;77:2432–48. 118. Jiang H, Zhang J, Du Y, Jia X, Yang F, Si S, et al. micro- 134. Desgagne V, Guay SP, Guerin R, Corbin F, Couture P, RNA-185 modulates low density lipoprotein receptor expres- Lamarche B, et al. Variations in HDL-carried miR-223 and sion as a key posttranscriptional regulator. Atherosclerosis miR-135a concentrations after consumption of dietary trans fat 2015;243:523–32. are associated with changes in blood lipid and inflammatory 119. Vickers KC, Landstreet SR, Levin MG, Shoucri BM, Toth CL, markers in healthy men – an exploratory study. Taylor RC, et al. MicroRNA-223 coordinates cholesterol 2016;11:438–48. homeostasis. Proc Natl Acad Sci USA 2014;111:14518–23. 135. Rader DJ, Cohen J, Hobbs HH. Monogenic hypercholester- 120. Meiler S, Baumer Y, Toulmin E, Seng K, Boisvert WA. MicroRNA olemia: new insights in pathogenesis and treatment. J Clin 302a is a novel modulator of cholesterol homeostasis and Invest 2003;111:1795–803. atherosclerosis. Arterioscler Thromb Vasc Biol 2015;35:323–31. 136. Tabet F, Vickers KC, Cuesta Torres LF, Wiese CB, Shoucri 121. Wang D, Yan X, Xia M, Yang Y, Li D, Li X, et al. Coenzyme Q10 BM, Lambert G, et al. HDL-transferred microRNA-223 regu- promotes macrophage cholesterol efflux by regulation of the lates ICAM-1 expression in endothelial cells. Nat Commun activator protein-1/miR-378/ATP-binding cassette trans- 2014;5:3292. porter G1-signaling pathway. Arterioscler Thromb Vasc Biol 137. Tabet F, Cuesta Torres LF, Ong KL, Shrestha S, Choteau SA, 2014;34:1860–70. Barter PJ, et al. High-density lipoprotein-associated miR-223 is 122. Liu W, Cao H, Ye C, Chang C, Lu M, Jing Y, et al. Hepatic miR-378 altered after diet-induced weight loss in overweight and obese targets p110alpha and controls glucose and lipid homeosta- males. PLoS One 2016;11:e0151061. sis by modulating hepatic insulin signalling. Nat Commun 138. Boon RA, Vickers KC. Intercellular transport of microRNAs. 2014;5:5684. Arterioscler Thromb Vasc Biol 2013;33:186–92. 123. Carrer M, Liu N, Grueter CE, Williams AH, Frisard MI, Hulver MW, 139. Micheli M, Albi E, Leray C, Magni MV. Nuclear sphingomyelin et al. Control of mitochondrial metabolism and systemic energy protects RNA from RNase action. FEBS Lett 1998;431:443–7. homeostasis by microRNAs 378 and 378*. Proc Natl Acad Sci 140. Rossi G, Magni MV, Albi E. Sphingomyelin-cholesterol and USA 2012;109:15330–5. double stranded RNA relationship in the intranuclear complex. 124. Ou Z, Wada T, Gramignoli R, Li S, Strom SC, Huang M, et al. Arch Biochem Biophys 2007;459:27–32. MicroRNA hsa-miR-613 targets the human LXRalpha gene and 141. Khvorova A, Kwak YG, Tamkun M, Majerfeld I, Yarus M. RNAs mediates a feedback loop of LXRalpha autoregulation. Mol that bind and change the permeability of phospholipid mem- Endocrinol 2011;25:584–96. branes. Proc Natl Acad Sci USA 1999;96:10649–54. 125. Zhao R, Feng J, He G. miR-613 regulates cholesterol efflux 142. Janas T, Yarus M. Visualization of membrane RNAs. RNA by targeting LXRalpha and ABCA1 in PPARgamma acti- 2003;9:1353–61. vated THP-1 macrophages. Biochem Biophys Res Commun 143. Janas T, Janas T, Yarus M. Specific RNA binding to ordered phos- 2014;448:329–34. pholipid bilayers. Nucleic Acids Res 2006;34:2128–36. 126. Ramirez CM, Davalos A, Goedeke L, Salerno AG, Warrier N, 144. Michanek A, Kristen N, Hook F, Nylander T, Sparr E. RNA and Cirera-Salinas D, et al. MicroRNA-758 regulates cholesterol DNA interactions with zwitterionic and charged lipid mem- efflux through posttranscriptional repression of ATP-binding branes – a DSC and QCM-D study. Biochim Biophys Acta cassette transporter A1. Arterioscler Thromb Vasc Biol 2010;1798:829–38. 2011;31:2707–14. 145. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland 127. Fernandez-Hernando C, Moore KJ. MicroRNA modulation F, et al. Ceramide triggers budding of exosome vesicles into of cholesterol homeostasis. Arterioscler Thromb Vasc Biol multivesicular endosomes. Science 2008;319:1244–7. 2011;31:2378–82. 146. Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya 128. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the T. Secretory mechanisms and intercellular transfer of microR- complete program of cholesterol and fatty acid synthesis in the NAs in living cells. J Biol Chem 2010;285:17442–52. liver. J Clin Invest 2002;109:1125–31. 147. Rayner KJ, Hennessy EJ. Extracellular communication via 129. Ono K. Functions of microRNA-33a/b and microRNA therapeu- microRNA: lipid particles have a new message. J Lipid Res tics. J Cardiol 2016;67:28–33. 2013;54:1174–81. 130. Rottiers V, Naar AM. MicroRNAs in metabolism and metabolic 148. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. disorders. Nat Rev Mol Cell Biol 2012;13:239–50. Exosome-mediated transfer of mRNAs and microRNAs is a novel 131. Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, mechanism of genetic exchange between cells. Nat Cell Biol Donato KA, et al. Harmonizing the metabolic syndrome: a joint 2007;9:654–9. interim statement of the International Diabetes Federation 149. Zhuang G, Meng C, Guo X, Cheruku PS, Shi L, Xu H, et al. Task Force on Epidemiology and Prevention; National Heart, A novel regulator of macrophage activation: miR-223 in Lung, and Blood Institute; American Heart Association; World obesity-associated adipose tissue inflammation. Circulation Heart Federation; International Atherosclerosis Society; and 2012;125:2892–903. Desgagné et al.: microRNAs in lipoprotein and lipid metabolism 685

150. Rosenson RS, Brewer HB, Jr, Ansell BJ, Barter P, Chapman MJ, 168. Mackey RH, Greenland P, Goff DC, Jr, Lloyd-Jones D, Sibley Heinecke JW, et al. Dysfunctional HDL and atherosclerotic CT, Mora S. High-density lipoprotein cholesterol and particle cardiovascular disease. Nat Rev Cardiol 2016;13:48–60. concentrations, carotid atherosclerosis, and coronary events: 151. Elton TS, Selemon H, Elton SM, Parinandi NL. Regulation of the MESA (multi-ethnic study of atherosclerosis). J Am Coll Cardiol MIR155 host gene in physiological and pathological processes. 2012;60:508–16. Gene 2013;532:1–12. 169. Wang J, Zhang KY, Liu SM, Sen S. Tumor-associated 152. Nazari-Jahantigh M, Wei Y, Noels H, Akhtar S, Zhou Z, Koenen circulating microRNAs as biomarkers of cancer. Molecules RR, et al. MicroRNA-155 promotes atherosclerosis by repressing 2014;19:1912–38. Bcl6 in macrophages. J Clin Invest 2012;122:4190–202. 170. Lai CT, Ng EK, Chow PC, Kwong A, Cheung YF. Circulating micro- 153. Karkeni E, Astier J, Tourniaire F, El Abed M, Romier B, RNA expression profile and systemic right ventricular function Gouranton E, et al. Obesity-associated Inflammation Induces in adults after atrial switch operation for complete transposi- microRNA-155 expression in adipocytes and adipose tissue: tion of the great arteries. BMC Cardiovasc Disord 2013;13:73. outcome on adipocyte function. J Clin Endocrinol Metab 171. Liu D, Zhang M, Xie W, Lan G, Cheng HP, Gong D, et al. MiR-486 2016;101:1615–26. regulates cholesterol efflux by targeting HAT1. Biochem Bio- 154. Liu G, Abraham E. MicroRNAs in immune response and phys Res Commun 2016;472:418–24. macrophage polarization. Arterioscler Thromb Vasc Biol 172. Shi XF, Wang H, Xiao FJ, Yin Y, Xu QQ, Ge RL, et al. MiRNA-486 2013;33:170–7. regulates angiogenic activity and survival of mesenchymal 155. Zhang X, Dong H, Tian Y. miRNA biology in pathological pro- stem cells under hypoxia through modulating Akt signal. cesses. MicroRNA detection and pathological functions, 1 ed. Biochem Biophys Res Commun 2016;470:670–7. Springer-Verlag Berlin Heidelberg, 2015:7–22. 173. Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, 156. Moldovan L, Batte KE, Trgovcich J, Wisler J, Marsh CB, Piper M. Liebetrau C, et al. Circulating microRNAs in patients with Methodological challenges in utilizing miRNAs as circulating coronary artery disease. Circ Res 2010;107:677–84. biomarkers. J Cell Mol Med 2014;18:371–90. 174. Ren J, Zhang J, Xu N, Han G, Geng Q, Song J, et al. Signature of 157. Engelhardt S. Small RNA biomarkers come of age. J Am Coll circulating microRNAs as potential biomarkers in vulnerable Cardiol 2012;60:300–3. coronary artery disease. PLoS One 2013;8:e80738. 158. Hayes J, Peruzzi PP, Lawler S. MicroRNAs in cancer: biomarkers, 175. Loyer X, Potteaux S, Vion AC, Guerin CL, Boulkroun S, functions and therapy. Trends Mol Med 2014;20:460–9. Rautou PE, et al. Inhibition of microRNA-92a prevents 159. Zampetaki A, Willeit P, Drozdov I, Kiechl S, Mayr M. Profiling endothelial dysfunction and atherosclerosis in mice. Circ Res of circulating microRNAs: from single biomarkers to re-wired 2014;114:434–43. networks. Cardiovasc Res 2012;93:555–62. 176. Hennessy DA, Tanuseputro P, Tuna M, Bennett C, Perez R, 160. Santovito D, Egea V, Weber C. Small but smart: MicroRNAs Shields M, et al. Population health impact of statin treatment orchestrate atherosclerosis development and progression. in Canada. Health Rep 2016;27:20–8. Biochim Biophys Acta 2015. 177. Pahan K. Lipid-lowering drugs. Cell Mol Life Sci 161. Schulte C, Zeller T. microRNA-based diagnostics and therapy 2006;63:1165–78. in cardiovascular disease-Summing up the facts. Cardiovasc 178. Barter PJ, Rye KA. New era of lipid-lowering drugs. Pharmacol Diagn Ther 2015;5:17–36. Rev 2016;68:458–75. 162. Zampetaki A, Willeit P, Tilling L, Drozdov I, Prokopi M, Renard 179. Brown MS, Goldstein JL. Biomedicine. Lowering LDL–not only JM, et al. Prospective study on circulating MicroRNAs and risk how low, but how long? Science 2006;311:1721–3. of myocardial infarction. J Am Coll Cardiol 2012;60:290–9. 180. Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, et al. Effects of 163. Anderson TJ, Gregoire J, Hegele RA, Couture P, Mancini GB, fibrates on cardiovascular outcomes: a systematic review and McPherson R, et al. 2012 update of the Canadian Cardiovas- meta-analysis. Lancet 2010;375:1875–84. cular Society guidelines for the diagnosis and treatment of 181. Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leitersdorf E, dyslipidemia for the prevention of cardiovascular disease in Fruchart JC. Mechanism of action of fibrates on lipid and the adult. Can J Cardiol 2013;29:151–67. lipoprotein metabolism. Circulation 1998;98:2088–93. 164. Barter PJ, Ballantyne CM, Carmena R, Castro Cabezas M, Chap- 182. Dadu RT, Ballantyne CM. Lipid lowering with PCSK9 inhibitors. man MJ, Couture P, et al. Apo B versus cholesterol in estimating Nat Rev Cardiol 2014;11:563–75. cardiovascular risk and in guiding therapy: report of the thirty- 183. Zhang XL, Zhu QQ, Zhu L, Chen JZ, Chen QH, Li GN, et al. Safety person/ten-country panel. J Intern Med 2006;259:247–58. and efficacy of anti-PCSK9 : a meta-analysis of 25 165. Sniderman AD, Furberg CD, Keech A, Roeters van Lennep JE, randomized, controlled trials. BMC Med 2015;13:123. Frohlich J, Jungner I, et al. Apolipoproteins versus lipids as indi- 184. Chyu KY, Shah PK. HDL/ApoA-1 infusion and ApoA-1 gene ces of coronary risk and as targets for statin treatment. Lancet therapy in atherosclerosis. Front Pharmacol 2015;6:187. 2003;361:777–80. 185. Kingwell BA, Chapman MJ, Kontush A, Miller NE. HDL-targeted 166. Cromwell WC, Otvos JD, Keyes MJ, Pencina MJ, Sullivan L, Vasan therapies: progress, failures and future. Nat Rev Drug Discov RS, et al. LDL Particle Number and Risk of Future Cardiovascu- 2014;13:445–64. lar Disease in the Framingham Offspring Study – Implications 186. Schwartz GG, Olsson AG, Abt M, Ballantyne CM, Barter PJ, for LDL Management. J Clin Lipidol 2007;1:583–92. Brumm J, et al. Effects of dalcetrapib in patients with a recent 167. Khera AV, Cuchel M, de la Llera-Moya M, Rodrigues A, Burke acute coronary syndrome. N Engl J Med 2012;367:2089–99. MF, Jafri K, et al. Cholesterol efflux capacity, high-density 187. Barter PJ, Caulfield M, Eriksson M, Grundy SM, Kastelein JJ, lipoprotein function, and atherosclerosis. N Engl J Med Komajda M, et al. Effects of torcetrapib in patients at high risk 2011;364:127–35. for coronary events. N Engl J Med 2007;357:2109–22. 686 Desgagné et al.: microRNAs in lipoprotein and lipid metabolism

188. Christopher AF, Kaur RP, Kaur G, Kaur A, Gupta V, Bansal P. toward microRNA-based therapeutics for hepatocellular MicroRNA therapeutics: Discovering novel targets and develop- ­carcinoma. Gastroenterol Hepatol Bed Bench ing specific therapy. Perspect Clin Res 2016;7:68–74. 2014;7:43–54. 189. van Rooij E, Purcell AL, Levin AA. Developing microRNA thera- 194. Hinkel R, Trenkwalder T, Kupatt C. Gene therapy for ischemic peutics. Circ Res 2012;110:496–507. heart disease. Expert Opin Biol Ther 2011;11:723–37. 190. Thum T. MicroRNA therapeutics in cardiovascular medicine. 195. Kasinski AL, Kelnar K, Stahlhut C, Orellana E, Zhao J, Shimer E, EMBO Mol Med 2012;4:3–14. et al. A combinatorial microRNA therapeutics approach 191. Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, to suppressing non-small cell lung cancer. Oncogene Manoharan M, et al. Silencing of microRNAs in vivo with 2015;34:3547–55. ‘antagomirs’. Nature 2005;438:685–9. 196. Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres 192. Krutzfeldt J, Kuwajima S, Braich R, Rajeev KG, Pena J, Tuschl T, M, Patel K, et al. Treatment of HCV infection by targeting micro- et al. Specificity, duplex degradation and subcellular localiza- RNA. N Engl J Med 2013;368:1685–94. tion of antagomirs. Nucleic Acids Res 2007;35:2885–92. 197. Romaine SP, Tomaszewski M, Condorelli G, Samani NJ. Micro- 193. Moshiri F, Callegari E, D’Abundo L, Corra F, Lupini L, Sabbioni S, RNAs in cardiovascular disease: an introduction for clinicians. et al. Inhibiting the oncogenic mir-221 by microRNA sponge: Heart 2015;101:921–8.