Cardiovascular Research Advance Access published June 3, 2014

1 microRNAs and HDL life cycle

Alberto Canfrán-Duque1,2, Cristina M. Ramírez1,2, Leigh Goedeke1,2, Chin-Sheng Lin3 and Carlos Fernández-Hernando1,2,4 1

Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven,

Connecticut, USA Integrative Cell Signalling and Neurobiology of Metabolism Program, Section of Comparative

Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 3

Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense

Medical Center, No. 325, Sec. 2, Chen-Kung Rd., Neihu 114, Taipei, Taiwan 4

Corresponding author: Carlos Fernández-Hernando, PhD, 10 Amistad Street, Amistad Research

Building, Room 337C, New Haven, CT 06510, Yale University School of Medicine, Tel: (203) 737-4615,

Accepted Manuscript

Fax: (203) 737-2290, Email: [email protected]

Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2014. For permissions please email: [email protected].

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

2

2 ABSTRACT miRNAs have emerged as important regulators of lipoprotein metabolism. Work over the past few years has demonstrated that miRNAs control the expression of most of the genes associated with high-density lipoprotein (HDL) metabolism, including the ATP transporters, ABCA1 and ABCG1, and the scavenger receptor SRB1. These findings strongly suggest that miRNAs regulate HDL biogenesis, cellular cholesterol efflux and HDL cholesterol (HDL-C) uptake in the liver, thereby controlling all of

manipulating miRNA levels including miR-33 can increase circulating HDL-C. Importantly, antagonizing miR-33 in vivo enhances the regression and reduces the progression of atherosclerosis. These findings support the idea of developing miRNA inhibitors for the treatment of dyslipidaemia and related cardiovascular disorders such as atherosclerosis. This review article focuses on how HDL metabolism is regulated by miRNAs and how antagonizing miRNA expression could be a potential

Accepted Manuscript

therapy for treating cardiometabolic diseases.

Keywords: miRNAs, cholesterol metabolism, ABCA1 and SRB1.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

the steps of reverse cholesterol transport. Recent work in animal models has demonstrated that

3 INTRODUCTION Cholesterol is a major component of the plasma membrane in mammalian cells. In addition to its structural requirement, cholesterol is important for other cell functions such as cell proliferation and bile acid and hormone biosynthesis1, 2. Despite its pivotal role in controlling multiple physiological processes, abnormal levels of cholesterol can trigger a number of cardiometabolic diseases including atherosclerosis and type-II diabetes3. Because mammalian cells cannot degrade cholesterol,

cholesterol must be removed and transported from the peripheral tissues to the liver for reutilization and excretion into feces in a physiological process traditionally known as reverse cholesterol transport (RCT)4. During RCT, plasma high-density lipoprotein (HDL) is thought to function as a sterol transporter that facilitates the movement of sterols from the peripheral cells to the liver. HDL integrates a heterogeneous class of lipoproteins with a density > 1.063 g/ml5. HDL particles contain

Accepted Manuscript

various apolipoproteins of which ApoA1 and ApoA2 are quantitatively the most abundant6, 7. HDL formation occurs in the liver and intestine. The interaction between lipid poor ApoA1 with the ATP binding cassette (ABC) A1 mediates this first step in HDL formation6. ABCA1 is a member of the ABC family of membrane transporters that promotes phospholipid and cholesterol transfer from cells to poorly-lipidated ApoA1. Even though the mechanism by which ABCA1 regulates this process is not fully characterized, it is thought that poorly-lipidated ApoA1 binds to ABCA1 in the membrane surface leading to increased stability and activity of the transporter in the plasma membrane. In response to ATP hydrolysis, ABCA1 promotes the trans-bilayer transport of phospholipids from the inner to outer leaflet of the plasma membrane. The uneven phospholipid packing in the plasma membrane bilayer leads to the formation of extravesiculated lipid domains. Lipid poor ApoA1 binds to this phospholipid and cholesterol-rich domain and promotes the spontaneous solubilization to form pre β-HDL particles8. The efflux of cholesterol and HDL formation through the ABCA1 pathway remains predominant. This finding was brought to light when the mutation in Tangier disease, a condition characterized by low plasma HDL levels, was attributed to mutations in the ABCA1 gene9-11. However other proteins also

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

cholesterol removal is indispensable in order to prevent cholesterol accumulation in cells. Excess

4 play a role in HDL metabolism, including ABCG1 and the scavenger receptor class B type 1 (SRB1), which is involved in the maturation of HDL particles. Studies in vitro have shown that ABCA1 and ABCG1 synergistically mediate cholesterol efflux to HDL12. These models propose that the lipidated ApoA1 formed after the ApoA1/ABCA1 interaction serves as an acceptor for cholesterol that is effluxed from cells in an ABCG1-dependent process12. The importance of ABCG1 in regulating cholesterol efflux is well established, however the mechanism of ABCG1 action is controversial. In

facilitate the distribution of specific intracellular sterols away from the endoplasmatic reticulum13. Absence of ABCA1 and ABCG1 in mice results in massive accumulation of macrophage foam cells in various tissues such as in the spleen, heart, thymus, liver and lung14, 15. However, there are conflicting data regarding the role of both transporters during the progression of atherosclerosis. While transplantation of bone marrow from Abca1-/- mice into Ldlr-/- or ApoE-/- recipients caused an increase

Accepted Manuscript

in atherosclerosis16, deficiency of ABCG1 in bone marrow cells resulted in either a modest increase, or decrease of atherosclerosis17, 18. Despite the well-established role of ABCA1 in HDL formation, its effect on atherogenesis is less clear. Even though the increase in the incidence of atherosclerosis has been reported in people affected with Tangier disease, not all the subjects develop atherosclerosis. Moreover, mice lacking ABCA1 in the liver develop similar atherosclerotic lesions than Ldlr-/- mice19. Interestingly, absence of ABCA1 in the liver markedly diminish plasma HDL-C levels (less than 50%) but cause also a marked reduction in circulating VLDL and LDL. In addition to ABCA1/ABCG1-mediated cholesterol efflux, excess intracellular cholesterol can also be eliminated by aqueous diffusion. This process consists of desorption of free cholesterol molecules from the plasma cell membrane into the surrounding aqueous phase. Collision of desorbed cholesterol molecules with HDL particles diffusing in the extracellular aqueous space leads to their rapid uptake into the lipoprotein acceptor SRB1 mediates bidirectional flux of cholesterol between cells and HDL, modulating changes in the composition and structure of HDL particles20, 21. SRB1 facilitates the delivery of cholesterol to steroidogenic tissues and liver. Mouse studies have revealed the importance of this receptor in

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

contrast to ABCA1, it has been recently reported that ABCG1 is localized in endocytic vesicles where

5 controlling lipoprotein metabolism and the progression of atherosclerosis22, 23. Remarkably, absence of SRB1 results in a dramatic increase of atherosclerosis at a very young age. Srb1-/-ApoE-/- mice develop atherosclerotic plaques in the coronary arteries, a rare phenotype observed in mouse models of atherosclerosis22. HDL particles are constantly remodelled by a number of plasma enzymes and proteins. The lecithincholesterol acyltransferase (LCAT), a liver synthesized glycoprotein, mediates the transfer of fatty

Glomset proposed that esterification of cholesterol would drive the net efflux from cells because esterification would prevent the back-exchange of cholesterol from HDL to cells4, 26. The cholesteryl ester generated by LCAT in HDL could be transferred to other lipoproteins, including very-low density lipoproteins (VLDL). This process is catalyzed by the cholesteryl ester transfer protein (CETP), an enzyme that facilitates the transport of cholesteryl esters and triglycerides between the lipoproteins27, 28

Accepted Manuscript

. This neutral lipid transfer process results in a net gain of triglycerides and net loss of cholesteryl

ester in HDL28. In addition to CETP, human plasma also contains a second lipid transfer protein, designated phospholipid transfer protein (PLTP), which mediates transfer of phospholipids from ApoB-containing lipoproteins to HDL. Moreover, two members of the triglyceride lipase family, the hepatic lipase (HL) and the endothelial lipase (EL) also participate in HDL metabolism. HL is a glycoprotein synthesized primarily by the liver and has both triglyceride lipase and phospholipase and hydrolyzes HDL, generating smaller subspecies, including pre β-HDL29. EL acts primarily as a phospholipase and hydrolyzes HDL phospholipids and is primarily located in vascular endothelial cells30. HDL particles deliver cholesterol and cholesterol esters to the liver and steroidogenic tissues through the scavenger receptor SRB1. Within the liver, a portion of cholesterol is enzymatically converted in bile salt molecules. This process is unique to the liver because only hepatocytes express high levels of the enzyme cholesterol 7a-hydroxilase (CYP7A1), which initiates and rate-limits the multi-step conversion process31. Cholesterol and bile acid molecules have different physical properties. While cholesterol is insoluble in water, bile acid salts are biological amphiphiles and highly soluble allowing

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

acids from phospholipids to free cholesterol present in the pre β-HDL to form cholesteryl ester24, 25.

6 the transport of cholesterol in the digestive system by forming micelles. Biliary lipids are secreted across the apical (canalicular) membrane of hepatocytes by three different transmembrane transporters: ABCB11 (aka BESP), ABCG5/ABCG8 (an obligate heterodimer that facilitates cholesterol efflux) and ABCB4 (aka MDR3; which pumps phospholipids)32. Another transporter, ATP8B1 maintains the asymmetry of phospholipids to promote the required lipid packing of the canalicular membrane for resistance to hydrophobic bile salts and canalicular membrane transport33, . All these transporters play an essential role in the removal of cholesterol from the peripheral

tissues towards the liver for excretion35, 36. This process represents the last step of RCT, a protective mechanism against the development of atherosclerosis.

TRANSCRIPTIONAL REGULATION OF ABCA1 AND ABCG1 EXPRESSION As mentioned above, ABCA1 regulates cellular cholesterol efflux and HDL biogenesis. Importantly,

Accepted Manuscript

ABCA1 mRNA and protein half-life is very short (1-2 hours), suggesting that de novo transcription and translation are critical for controlling its expression in response to environmental changes, such as cholesterol loading. ABCA1 and ABCG1 are oxysterol-regulated genes and both are transcriptionally regulated by the liver X receptors (LXRs)12, 37, 38. These nuclear hormone receptors [(LXRα (NR1H3) and LXRβ (NR1H2)] form heterodimers with the retinoic-X-receptor (RXR)39 leading to the transcriptional activation of both ABC transporters. Genetic deletion of LXRα and LXRβ results in a massive accumulation of cholesterol in peripheral tissues suggesting the critical role of both transcription factors in controlling cholesterol removal from cells40, 41. Moreover, absence of LXRα and LXRβ markedly increase the progression of atherosclerosis in mice40. In addition to the transcriptional regulation of ABCA1 and ABCG1 by LXRs, several groups have recently identified that the expression of both transporters are significantly post-transcriptionally regulated by microRNAs (miRNAs)42-44 (Table).

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

34

7 microRNAs AND LIPID METABOLISM MicroRNAs (miRNAs) are small (18–25 nucleotides), evolutionarily conserved, non-coding RNAs that have an important function in gene regulation, acting predominantly at the post-transcriptional level45, 46

. Since miRNAs have been described in Caenorhabditis elegans, hundreds of miRNAs have been

identified in animals, plants and viruses45, 46. They have been shown to participate in almost every cellular process investigated, including cholesterol homeostasis and lipoprotein metabolism42-46.

hundreds or thousands of nucleotides through sequential processing by the ribonuclease DROSHA47, 48

. This ultimately produces a nuclear hairpin precursor called the pre-miRNA, which is then exported

to the cytoplasm where it is processed by DICER to produce the mature miRNA47, 48. miRNAs typically control the expression of their target genes by imperfect base pairing to the 3′-untranslated region (3’UTR) of mRNAs. miRNAs are preferentially incorporated into the RISC complex where they

Accepted Manuscript

associate with Argonaute proteins directing the binding of the RISC complex to the 3’UTR of their target mRNAs. This association produces mRNA repression either by transcript destabilization, translational inhibition or both47, 48. One miRNA often regulates multiple genes that are involved in a specific signalling cascade or cellular mechanism, thus making miRNAs potent biological regulators. In the past few years, several groups have demonstrated the important role of miRNAs including miR33, miR-122 and miR-30c in controlling lipoprotein metabolism49-54. In the following sections of this review article we will discuss the most recent findings regarding the importance of miRNAs in regulating HDL metabolism.

miR-33 REGULATION OF HDL METABOLISM AND ATHEROGENESIS The miR-33 family consists of two intronic miRNAs, miR-33a and miR-33b, which are encoded within the introns of the sterol regulatory element-binding proteins (SREBP) 2 and 1 genes, respectively51-53, 55

. The SREBPs are a family of membrane-bound transcription factors that regulate cellular lipid

synthesis and clearance of pro-atherogenic lipoproteins56, 57. SREBP2 regulates the expression of genes involved in the cholesterol biosynthetic pathway as well as the low-density lipoprotein receptor

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

Mature miRNA products are generated from precursors called pri-miRNAs, which are composed of

8 (LDLR). The function of SREBP1 is more complex because the same gene encodes two different SREBP isoforms (SREBP1c and SREBP1a). SREBP1c increases the expression of genes that regulate fatty acid synthesis and SREBP1a regulates genes that control cholesterol metabolism and fatty acid synthesis. SREBP1c is the predominant SREBP1 isoform in adult liver and it’s activated in response to insulin. The fact that miR-33a and miR-33b are co-transcribed with their respective host genes, suggests that miR-33a/b regulate related physiological processes controlled by SREBP2 and

cholesterol and fatty acid levels during times of need. Under conditions that stimulate SREBP transcription, miR-33a/b are co-expressed with their host genes and reciprocally regulate genes involved in cellular cholesterol efflux/HDL biogenesis (ABCA1 and ABCG1) and fatty acid degradation (CPT1A, CROT, HADHB, AMPK1A)51-53, 55. These findings illustrate an elegant genetic regulatory mechanism by which miR-33a/b and their host genes cooperate to tightly regulate intracellular

Accepted Manuscript

cholesterol and fatty acid levels. In addition to the genes mentioned above, Baldan and colleagues have reported that miR-33 also regulates the expression of a number of bile acid transporters, including ABCB11 and ATP8B1 that control bile secretion58. Moreover, a recent study also identified CYP7A1 as a miR-33 target gene59. Altogether, these findings support the hypothesis that miR-33 controls whole-body cholesterol homeostasis by affecting HDL biogenesis (via ABCA1), cellular cholesterol efflux from peripheral tissues (via ABCA1 and ABCG1) and bile acid synthesis (via CYP7A1) and secretion (via ATP8B1 and ABCB11). Given that miR-33 levels markedly regulate ABCA1 expression, several groups assessed the efficacy of anti-miR-33 therapy for increasing circulating HDL-C. Three independent studies demonstrated that silencing of miR-33 in mice using modified anti-sense oligonucleotides, or viral delivery of hairpin inhibitors, increased hepatic ABCA1 expression and plasma HDL levels by 25-35%51-53. In addition to the elevated circulating HDL-C levels observed in mice treated with anti-miR-33 oligonucleotides, antagonism of miR-33 in vivo also enhanced RCT53, 58. The effect of miR-33 inhibitors on plasma HDL-C levels was later confirmed in miR-33 deficient mice, which showed a significant increase in hepatic ABCA1 expression and a 25% increase in serum HDL-C compared to wild-type mice60. Most

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

SREBP151-53, 55. Indeed, it has been demonstrated that miR-33a and miR-33b help to boost cellular

9 importantly, anti-miR-33 therapy also resulted in increased plasma HDL-C levels in non-human primates61, 62. A number of observational studies, including the Framingham Heart Study, have shown a strong inverse correlation of plasma HDL-C levels with coronary heart disease (CHD). To demonstrate whether anti-miR-33 therapy reduces the progression and enhances the regression of atherosclerosis in atherosclerosis-prone mouse models, several groups inhibited miR-33 using chemically modified

fluoro/methoxylethyl (2’F/MOE) anti-miR-33 oligonucleotides of Ldlr null mice fed previously a western diet (WD) for 14-weeks increased circulating HDL-C and enhanced the regression of atherosclerosis64. The results of the atherosclerosis progression studies, however, are somehow conflicting. While Baldan’s group showed that prolonged anti-miR-33 therapy failed to raise plasma HDL-C and did not prevent the progression of atherosclerosis63, our group demonstrated that

Accepted Manuscript

antagonism of miR-33 reduced atherogenesis despite the fact that HDL-C levels were not affected65. The different outcomes observed in the last two studies might be explained by different cholesterol content in the Western diets (0.3% and 1.25%), oligonucleotide chemical modifications and length of treatment. Indeed, while we demonstrated that the (2’F/MOE) anti-miR-33 oligonucleotides enhances ABCA1 expression in the artery wall, Baldan’s study did not assess the efficacy of anti-miR-33 therapy in increasing ABCA1 expression in atherosclerotic plaques. Finally, the fact that miR-33 null mice have significant protection against the progression of atherosclerosis strongly suggests that inhibiting miR-33 in vivo might be useful for treating atherosclerotic vascular disease66.

miR-33 REGULATION OF FATTY ACID AND GLUCOSE METABOLISM Besides the main role of miR-33 in regulating cholesterol metabolism, miR-33 also contributes to the regulation of additional metabolic pathways such as fatty acid metabolism and insulin signalling55, 67. miR-33 regulates the expression of carnitine O-octanyl transferase (CROT), carnitine palmitoyltransferase 1A (CPT1A), and hydroxyacyl-coenzyme A dehydrogenase-3-ketoacylcoenzyme A thiolase-enoyl-coenzyme A hydratase (trifunctional protein) β-subunit (HADHB), thereby

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

antisense oligonucleotides63-65. The first study reported that a 4-week treatment with 2’-

10 controlling fatty acid β-oxidation55, 67. CROT and CPT1A regulate the transport of fatty acids to the mitochondria for their degradation and HADHB is required for the last steps of the mitochondria βoxidation pathway. Inhibition of miR-33 in human hepatic cells increases the degradation of fatty acids, suggesting that anti–miR-33 therapy may be useful for treating hepatic steatosis by increasing the degradation rate of fatty acids in the liver55, 67. miR-33 also regulates the post-transcriptional expression of the AMPK-activated protein kinase

glucose metabolism55. This observation suggests that anti-miR-33 therapy could increase insulin sensitivity. In addition to the regulation of insulin signalling in human hepatic cells, it has recently been shown that miR-33 also modulates the expression of ABC transporters and insulin secretion in human and mouse pancreatic islets. Of note, inhibition of miR-33 in pancreatic islets increases ABCA1 expression and enhances insulin secretion while overexpression of miR-33 has the opposite effects68.

Accepted Manuscript

Altogether, these findings indicate that antagonism of miR-33 might increase plasma HDL-C and insulin sensitivity and reduce hepatic lipid accumulation and plasma triglyceride levels. However, it has been recently shown that miR-33 deficient mice develop obesity, hepatic steatosis and insulin resistance69. Mechanistically, Horie and colleagues found that miR-33 inhibits SREBP1 expression, thereby increasing fatty acid synthesis69. Moreover, we have also reported that miR-33 regulates gluconeogenesis, suggesting that derepression of miR-33 might increase hepatic glucose production70. Together, these observations indicate that miR-33 regulates multiple metabolic processes and that further experiments are warranted to fully understand the molecular mechanism by which miR-33 controls lipid and glucose metabolism. Although most of the studies have focused on the role of miR-33a-5p (guide strand), we have also recently discovered that the guide strand (miR-33-3p; aka miR-33*) accumulates in a number of tissues and targets similar genes as miR-3371. These findings suggest that both strands of the miR-33 locus may work together to control cellular lipid metabolism. Collectively, these studies have illuminated the key role of miR-33 in regulating lipid and glucose metabolism and how targeting miR33 might be a useful therapy for treating cardiometabolic disorders.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

(AMPK), sirtuin 6 (SIRT6) and insulin receptor substrate 2 (IRS2), thus controlling fatty acid and

11

OTHER miRNAs THAT REGULATE ABCA1 EXPRESSION AND HDL METABOLISM miR-33 was the first miRNA described to regulate hepatic ABCA1 expression and plasma HDL-C levels in vivo. However in the last years it has become clear that ABCA1 expression is highly regulated at the post-transcriptional level by multiple miRNAs, including miR-758, miR-26, miR-106b, miR-27, miR-145, miR-10b and miR-14471-78. These miRNAs can regulate ABCA1 expression and

hepatocytes. The relative importance of these miRNAs in controlling ABCA1 expression will most likely be dictated by their relative abundance and the expression of other miRNA targets in specific cells or tissues. Additionally, the expression of these miRNAs and mRNA targets can also be regulated by physiological stimuli that alter miRNA expression levels. Importantly, several reports have demonstrated that cellular lipid metabolism influences miRNA expression, representing positive

Accepted Manuscript

or negative feedback models that contribute to the complex regulation of ABCA1 expression. This is the case of miR-758, an intergenic miRNA that, similarly to miR-33, is downregulated after cholesterol loading in macrophages and in the liver of mice fed a high-fat diet75. Additionally, two reports have recently demonstrated the role of miR-144 in regulating cholesterol metabolism79, 80. miR-144 is synthesized as a polycistronic transcript together with miR-451. In vertebrates, this conserved miRNA cluster plays an important role in eritropoiesis and cancer and was first described to bypass the classic Dicer processing step during miRNA biogenesis81-83. We identified miR-144 using an unbiased genome-wide screen of miRNAs modulated by LXR ligands in combination with bioinformatic tools for miRNA target predictions. We found that miR-144 directly targets ABCA1 and its overexpression markedly reduces ABCA1 protein levels in human and mouse macrophages and hepatic cell lines. Importantly, our in vivo results indicated that delivery of miR-144 mimics to mice inhibits hepatic ABCA1 expression levels and reduces circulating HDL-C76. More importantly, inhibition of endogenous miR-144 levels using anti-miR-144 conjugated particles in mice increases hepatic ABCA1 expression and raises plasma HDL levels. In a second report, de Aguiar Vallim and colleagues identified miR-144 using a genome-wide screening aimed at identifying miRNAs regulated

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

function in a variety of cell types such as macrophages, neurons, pancreatic β-cells, enterocytes and

12 by farnesoid X receptor (FXR), a nuclear receptor that controls hepatic sterol and bile acid levels80. Similar to our findings, gain and loss-of-function experiments also showed that changes in hepatic miR-144 levels influences hepatic ABCA1 expression and circulating HDL-C. These results suggest a novel model by which miR-144 contributes to the FXR effect by inhibiting hepatic ABCA1 and promoting the redirection of hepatic cholesterol to biliary excretion80. The role of miR-144 in regulating plasma HDL levels has also recently been confirmed in a mouse model of

impairing RCT and promoting pro-inflammatory cytokine production in ApoE-/- mice. A recent study has shown that the intestinal microbiota can regulate RCT by modulating the expression of miR-10b expression. The authors found that protocatechuic acid (PCA), a metabolite produced by the gut microbiota from cyaniding-3 to O-β-glucoside (Cy-3-G) inhibits ABCA1 and ABCG1 expression. Importantly, PCA accelerates macrophage cholesterol efflux and Cy-3-G

Accepted Manuscript

consumption promotes RCT and regresses atherosclerosis in ApoE-/- mice. miR-145 also regulates ABCA1 expression in HepG2 cells and in murine pancreatic islets. Overexpression of miR-145 inhibits cholesterol efflux in HepG2 cells and causes cholesterol accumulation in pancreatic islets resulting in a marked decrease in glucose-stimulated insulin secretion. Finally, miR-26 and miR-27 inhibit ABCA1 expression and cholesterol efflux in mouse and human macrophage cell lines, respectively73, 77, 85. miRNAs also regulate the expression of LXR, thereby controlling the transcriptional activation of ABCA1. LXR is directly targeted by miR-1, miR-206, miR-613 and miR-15586-88. miR-1, miR-206 and miR-613 suppress lipogenesis by inhibiting LXRα and its target genes including SREBP1, acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS)86-88. miR-155 also inhibits LXR expression and its absence confers protection against hepatic steatosis in mice86. LXR activation also regulates the expression of miRNAs that control the expression levels of some LXR-induced genes such as ABCA1 and the ADP-ribosylation factor-like-7 (ARL7)77. Interestingly, miR-26 expression is inhibited in cells treated with LXR agonists, suggesting that the downregulation of miR-26 might cooperate with the LXR transcriptional activation to increase ABCA1 expression77. RXRα is also regulated by miRNAs including miR-128-289. This miRNA targets RXRα, thus inhibiting

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

atherosclerosis84. Overexpression of miR-144 accelerates the progression of atherosclerosis by

13 LXR-induced ABCA1 expression. Interestingly, miR-128-2 increases SREBP2 expression and decreases SREBP1, ABCA1, ABCG1 and RXRα expression. Overall, these data suggest the existence of a complex miRNA network operating under different physiological conditions in a number of different cell types and tissues to orchestrate post-transcriptional regulation of lipid metabolism.

POST-TRANSCRIPTIONAL REGULATION OF ABCA1 BY HuR

demonstrated in a number of cells and tissues, recent findings suggest that miRNA action might occur in conjunction with RNA-binding proteins (RBPs)90-92. RBPs bind to AU-rich elements (AREs) in the 3’UTR of genes, thereby modulating their expression by increasing or decreasing translation and/or mRNA stability. Around 20 RBPs, including the well-known members of the ELAV family (HuR, HuB,

Accepted Manuscript

HuC and HuD) have been identified93. Interestingly, we have recently found that HuR binds to the 3’UTR of ABCA1 and increases its expression by enhancing protein translation94. A number of studies have shown that RBP compete or cooperate with miRNAs to control gene expression95, 96. As such, the miRNA-mediated regulation of ABCA1 expression may be influenced by HuR binding to the ARE motifs in the 3’UTR (Figure 2). Further studies would be important to determine whether HuR might compete or cooperate with miRNAs in the regulation of ABCA1 expression.

miRNA REGULATION OF SRB1 EXPRESSION SRB1 regulates the cholesterol transport from HDL to the liver for excretion35, 36. Absence of SRB1 impairs RCT and causes massive atherosclerosis in mice22. Indeed, SRB1 null mice develop coronary atherosclerosis, a rare phenotype observed in few mouse models of atherosclerosis22, 97, 98. SRB1 expression is regulated by different transcription factors including LXR, SREBP, liver receptor homologue 1 (LRH-1) and peroxisome proliferator-activated receptors (PPAR)99. Moreover, SRB1 expression is controlled at the post-transcriptional level by alternative splicing and its interaction with PDX domain containing 1 (PDZK1), a scaffolding protein that regulates SRB1 cellular localization and function100, 101. In addition to this mechanism of regulation, SRB1 expression is also controlled by

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

Although the importance of miRNAs in the regulation of ABCA1 and ABCG1 expression has been

14 miRNAs. In this regard, a number of recent studies have uncovered several miRNAs including miR455, miR-125a, miR-185, miR-96 and miR-223 that bind directly to the 3’UTR of SRB1 and suppress its expression102, 103. Overexpression of miR-455, miR-125a, miR-185, miR-96 and miR-223 reduces SRB1 protein levels and HDL-C uptake. Conversely, antagonism of these miRNAs enhances SRB1 expression and increases HDL-C uptake. Interestingly, the levels of miR-96 and miR-185 inversely correlate with the increased expression of SRB1 in the livers of ApoE knockout mice fed a high-fat

transcriptional level by alternative splicing, protein localization and miRNAs.

HDL TRANSPORTS ENDOGENOUS miRNAS AND DELIVERS THEM TO RECIPIENT CELLS In addition to the classic view of HDL as sterol transporter that facilitates the movement if sterols from peripheral cells to the liver for its reutilization or excretion, it is now recognize that HDL function is

Accepted Manuscript

much more complex in terms of differential lipids and proteins that it transports. Surprisingly, it has been recently reported that HDL can transport miRNAs and deliver them to the receiving cells influencing their gene expression104. Amongst of them, miR-223 is one of the most abundant miRNAs (10000 copies/µg of HDL). Notably, HDL-derived miR-223 is transferred to human hepatic cells (Huh7) via SRB1 receptor leading to a significant reduction of miR-223 target genes expression. Similarly, HDL-derived miR-223 is also transferred to endothelial cells and inhibits the intracellular adhesion molecule 1 (ICAM1), thereby reducing monocyte adhesion and inflammation105 (Figure 3). This finding might explain in part the well-known anti-inflammatory effects of HDL. However, additional studies are necessary to address some important questions including the mechanism of miRNA loading in HDL particles and the receptors that mediate the transfer of HDL-derived miRNAs and the recipient cells within the atherosclerotic plaques (endothelial cells, macrophages and smooth muscle cells).

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

diet103. Overall, these observations suggest that SRB1 expression is regulated at the post-

15 CONCLUSIONS miRNAs have emerged as critical regulators of almost all biological processes including lipoprotein metabolism. Work over the last years has demonstrated that miRNAs play an important role in regulating HDL metabolism (Figure 1). A number of genes associated with HDL biogenesis, cellular cholesterol efflux and biliary secretion are post-trancriptionally regulated by miRNAs. Most of the studies have identified miRNAs that regulate ABCA1 and SRB1 expression (Table), however, it is not

CETP and LCAT, are modulated by miRNAs. This possibility is unlikely as both genes have a very short 3’UTR (less than 200 nt) and no conserved miRNA binding sites within their 3’UTR across species. Antagonizing the expression of some miRNAs, including miR-33, has shown to markedly increase circulating HDL-C in mice and non-human primates. Moreover, anti-miR-33 therapy reduces the

Accepted Manuscript

progression and enhances the regression of atherosclerosis in mice. Work over the last year has identified additional miRNAs that regulate HDL metabolism by increasing hepatic SRB1 expression. These findings suggest that antagonizing a set of miRNAs in the liver to increase ABCA1 and SRB1 expression might enhance RCT. Most of the therapies aimed to increase plasma HDL-C levels, such us CETP inhibitors, fail to protect against coronary artery disease. Moreover, recent results from Mendelian randomization studies also fail to demonstrate an association between circulating HDL-C levels and cardiovascular risk106. Even thought, these studies argue about the benefit of HDL-C to protect against myocardial infarction, it is clear that HDL-C levels are not necessarily reflective of the broad antiatherogenic properties of HDL particles including RCT. Indeed, Rader and colleagues have shown that the cholesterol efflux capacity from macrophages, a metric of HDL function, has a strong inverse association with both carotid intima-media thickness and the likelihood of angiographic coronary artery disease, independently of HDL-C107. It may be possible that anti-miR-33 therapies, which influence HDL metabolism by controlling HDL biogenesis, cellular cholesterol efflux and bile excretion might be useful for treating dyslipidaemias and cardiovascular related disorders.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

known whether or not the expression of other key players that control HDL metabolism, such as

16

FUNDING CF-H laboratory is supported by the National Institutes of Health (R01HL107953 and R01HL106063) and The Leducq Foundation.

REFERENCES Chen HW, Heiniger HJ, Kandutsch AA. Stimulation of sterol and DNA synthesis in leukemic blood cells by low concentrations of phytohemagglutinin. Exp Cell Res 1977;109:253-262. 2.

Fernandez C, Lobo Md Mdel V, Gomez-Coronado D, Lasuncion MA. Cholesterol is essential for mitosis progression and its deficiency induces polyploid cell formation. Exp Cell Res 2004;300:109-120.

3.

Ikonen E. Mechanisms for cellular cholesterol transport: defects and human disease. Physiol

Accepted Manuscript

Rev 2006;86:1237-1261. 4.

Glomset JA, Norum KR. The metabolic role of lecithin: cholesterol acyltransferase: perspectives form pathology. Adv Lipid Res 1973;11:1-65.

5.

Eisenberg S. High density lipoprotein metabolism. Journal of lipid research 1984;25:10171058.

6.

Duong PT, Collins HL, Nickel M, Lund-Katz S, Rothblat GH, Phillips MC. Characterization of nascent HDL particles and microparticles formed by ABCA1-mediated efflux of cellular lipids to apoA-I. Journal of lipid research 2006;47:832-843.

7.

Karlsson H, Leanderson P, Tagesson C, Lindahl M. Lipoproteomics II: mapping of proteins in high-density lipoprotein using two-dimensional gel electrophoresis and mass spectrometry. Proteomics 2005;5:1431-1445.

8.

Vedhachalam C, Duong PT, Nickel M, Nguyen D, Dhanasekaran P, Saito H, Rothblat GH, Lund-Katz S, Phillips MC. Mechanism of ATP-binding cassette transporter A1-mediated cellular lipid efflux to apolipoprotein A-I and formation of high density lipoprotein particles. The Journal of biological chemistry 2007;282:25123-25130.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

1.

17 9.

Bodzioch M, Orso E, Klucken J, Langmann T, Bottcher A, Diederich W, Drobnik W, Barlage S, Buchler C, Porsch-Ozcurumez M, Kaminski WE, Hahmann HW, Oette K, Rothe G, Aslanidis C, Lackner KJ, Schmitz G. The gene encoding ATP-binding cassette transporter 1 is mutated in Tangier disease. Nat Genet 1999;22:347-351.

10.

Brooks-Wilson A, Marcil M, Clee SM, Zhang LH, Roomp K, van Dam M, Yu L, Brewer C, Collins JA, Molhuizen HO, Loubser O, Ouelette BF, Fichter K, Ashbourne-Excoffon KJ,

Pimstone S, Kastelein JJ, Genest J, Jr., Hayden MR. Mutations in ABC1 in Tangier disease and familial high-density lipoprotein deficiency. Nat Genet 1999;22:336-345. 11.

Rust S, Rosier M, Funke H, Real J, Amoura Z, Piette JC, Deleuze JF, Brewer HB, Duverger N, Denefle P, Assmann G. Tangier disease is caused by mutations in the gene encoding ATPbinding cassette transporter 1. Nat Genet 1999;22:352-355.

Accepted Manuscript

12.

Gelissen IC, Harris M, Rye KA, Quinn C, Brown AJ, Kockx M, Cartland S, Packianathan M, Kritharides L, Jessup W. ABCA1 and ABCG1 synergize to mediate cholesterol export to apoAI. Arteriosclerosis, thrombosis, and vascular biology 2006;26:534-540.

13.

Tarling EJ, Edwards PA. ATP binding cassette transporter G1 (ABCG1) is an intracellular sterol transporter. Proceedings of the National Academy of Sciences of the United States of America 2011;108:19719-19724.

14.

Baldan A, Tarr P, Vales CS, Frank J, Shimotake TK, Hawgood S, Edwards PA. Deletion of the transmembrane transporter ABCG1 results in progressive pulmonary lipidosis. The Journal of biological chemistry 2006;281:29401-29410.

15.

Westerterp M, Murphy AJ, Wang M, Pagler TA, Vengrenyuk Y, Kappus MS, Gorman DJ, Nagareddy PR, Zhu X, Abramowicz S, Parks JS, Welch C, Fisher EA, Wang N, Yvan-Charvet L, Tall AR. Deficiency of ATP-binding cassette transporters A1 and G1 in macrophages increases inflammation and accelerates atherosclerosis in mice. Circulation research 2013;112:1456-1465.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

Sensen CW, Scherer S, Mott S, Denis M, Martindale D, Frohlich J, Morgan K, Koop B,

18 16.

Aiello RJ, Brees D, Bourassa PA, Royer L, Lindsey S, Coskran T, Haghpassand M, Francone OL. Increased atherosclerosis in hyperlipidemic mice with inactivation of ABCA1 in macrophages. Arteriosclerosis, thrombosis, and vascular biology 2002;22:630-637.

17.

Out R, Hoekstra M, Hildebrand RB, Kruit JK, Meurs I, Li Z, Kuipers F, Van Berkel TJ, Van Eck M. Macrophage ABCG1 deletion disrupts lipid homeostasis in alveolar macrophages and moderately influences atherosclerotic lesion development in LDL receptor-deficient mice.

18.

Ranalletta M, Wang N, Han S, Yvan-Charvet L, Welch C, Tall AR. Decreased atherosclerosis in low-density lipoprotein receptor knockout mice transplanted with Abcg1-/- bone marrow. Arteriosclerosis, thrombosis, and vascular biology 2006;26:2308-2315.

19.

Bi X, Zhu X, Duong M, Boudyguina EY, Wilson MD, Gebre AK, Parks JS. Liver ABCA1 deletion in LDLrKO mice does not impair macrophage reverse cholesterol transport or

Accepted Manuscript

exacerbate atherogenesis. Arteriosclerosis, thrombosis, and vascular biology 2013;33:22882296. 20.

Ji Y, Jian B, Wang N, Sun Y, Moya ML, Phillips MC, Rothblat GH, Swaney JB, Tall AR. Scavenger receptor BI promotes high density lipoprotein-mediated cellular cholesterol efflux. The Journal of biological chemistry 1997;272:20982-20985.

21.

Jian B, de la Llera-Moya M, Ji Y, Wang N, Phillips MC, Swaney JB, Tall AR, Rothblat GH. Scavenger receptor class B type I as a mediator of cellular cholesterol efflux to lipoproteins and phospholipid acceptors. The Journal of biological chemistry 1998;273:5599-5606.

22.

Braun A, Trigatti BL, Post MJ, Sato K, Simons M, Edelberg JM, Rosenberg RD, Schrenzel M, Krieger M. Loss of SR-BI expression leads to the early onset of occlusive atherosclerotic coronary artery disease, spontaneous myocardial infarctions, severe cardiac dysfunction, and premature death in apolipoprotein E-deficient mice. Circulation research 2002;90:270-276.

23.

Rigotti A, Trigatti BL, Penman M, Rayburn H, Herz J, Krieger M. A targeted mutation in the murine gene encoding the high density lipoprotein (HDL) receptor scavenger receptor class B

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

Arteriosclerosis, thrombosis, and vascular biology 2006;26:2295-2300.

19 type I reveals its key role in HDL metabolism. Proceedings of the National Academy of Sciences of the United States of America 1997;94:12610-12615. 24.

Fielding CJ, Shore VG, Fielding PE. A protein cofactor of lecithin:cholesterol acyltransferase. Biochem Biophys Res Commun 1972;46:1493-1498.

25.

Jonas A. Lecithin-cholesterol acyltransferase in the metabolism of high-density lipoproteins. Biochim Biophys Acta 1991;1084:205-220. Glomset JA. The plasma lecithins:cholesterol acyltransferase reaction. Journal of lipid research 1968;9:155-167.

27.

Fielding CJ, Fielding PE. Molecular physiology of reverse cholesterol transport. Journal of lipid research 1995;36:211-228.

28.

Tall AR. Plasma cholesteryl ester transfer protein. Journal of lipid research 1993;34:12551274.

Accepted Manuscript

29.

Connelly PW. The role of hepatic lipase in lipoprotein metabolism. Clin Chim Acta 1999;286:243-255.

30.

Shimizu M, Kanazawa K, Hirata K, Ishida T, Hiraoka E, Matsuda Y, Iwai C, Miyamoto Y, Hashimoto M, Kajiya T, Akita H, Yokoyama M. Endothelial lipase gene polymorphism is associated with acute myocardial infarction, independently of high-density lipoproteincholesterol levels. Circ J 2007;71:842-846.

31.

de Aguiar Vallim TQ, Tarling EJ, Edwards PA. Pleiotropic roles of bile acids in metabolism. Cell metabolism 2013;17:657-669.

32.

Esteller A. Physiology of bile secretion. World journal of gastroenterology : WJG 2008;14:5641-5649.

33.

Paulusma CC, Folmer DE, Ho-Mok KS, de Waart DR, Hilarius PM, Verhoeven AJ, Oude Elferink RP. ATP8B1 requires an accessory protein for endoplasmic reticulum exit and plasma membrane lipid flippase activity. Hepatology 2008;47:268-278.

34.

Paulusma CC, Groen A, Kunne C, Ho-Mok KS, Spijkerboer AL, Rudi de Waart D, Hoek FJ, Vreeling H, Hoeben KA, van Marle J, Pawlikowska L, Bull LN, Hofmann AF, Knisely AS, Oude

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

26.

20 Elferink RP. Atp8b1 deficiency in mice reduces resistance of the canalicular membrane to hydrophobic bile salts and impairs bile salt transport. Hepatology 2006;44:195-204. 35.

Acton S, Rigotti A, Landschulz KT, Xu S, Hobbs HH, Krieger M. Identification of scavenger receptor SR-BI as a high density lipoprotein receptor. Science 1996;271:518-520.

36.

Kozarsky KF, Donahee MH, Rigotti A, Iqbal SN, Edelman ER, Krieger M. Overexpression of the HDL receptor SR-BI alters plasma HDL and bile cholesterol levels. Nature 1997;387:414-

37.

Tontonoz P. Transcriptional and posttranscriptional control of cholesterol homeostasis by liver X receptors. Cold Spring Harb Symp Quant Biol 2011;76:129-137.

38.

Venkateswaran A, Laffitte BA, Joseph SB, Mak PA, Wilpitz DC, Edwards PA, Tontonoz P. Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXR alpha. Proceedings of the National Academy of Sciences of the United States of America 2000;97:12097-12102.

Accepted Manuscript

39.

Willy PJ, Umesono K, Ong ES, Evans RM, Heyman RA, Mangelsdorf DJ. LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes Dev 1995;9:1033-1045.

40.

Bradley MN, Hong C, Chen M, Joseph SB, Wilpitz DC, Wang X, Lusis AJ, Collins A, Hseuh WA, Collins JL, Tangirala RK, Tontonoz P. Ligand activation of LXR beta reverses atherosclerosis and cellular cholesterol overload in mice lacking LXR alpha and apoE. The Journal of clinical investigation 2007;117:2337-2346.

41.

Peet DJ, Turley SD, Ma W, Janowski BA, Lobaccaro JM, Hammer RE, Mangelsdorf DJ. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell 1998;93:693-704.

42.

Fernandez-Hernando C, Ramirez CM, Goedeke L, Suarez Y. MicroRNAs in metabolic disease. Arteriosclerosis, thrombosis, and vascular biology 2013;33:178-185.

43.

Fernandez-Hernando C, Suarez Y, Rayner KJ, Moore KJ. MicroRNAs in lipid metabolism. Curr Opin Lipidol 2011;22:86-92.

44.

Moore KJ, Rayner KJ, Suarez Y, Fernandez-Hernando C. microRNAs and cholesterol metabolism. Trends Endocrinol Metab 2010;21:699-706.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

417.

21 45.

Ambros V. The functions of animal microRNAs. Nature 2004;431:350-355.

46.

Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281297.

47.

Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009;136:215-233.

48.

Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nature reviews Genetics 2008;9:102-114. Elmen J, Lindow M, Silahtaroglu A, Bak M, Christensen M, Lind-Thomsen A, Hedtjarn M, Hansen JB, Hansen HF, Straarup EM, McCullagh K, Kearney P, Kauppinen S. Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver. Nucleic Acids Res 2008;36:1153-1162.

50.

Esau C, Davis S, Murray SF, Yu XX, Pandey SK, Pear M, Watts L, Booten SL, Graham M, McKay R, Subramaniam A, Propp S, Lollo BA, Freier S, Bennett CF, Bhanot S, Monia BP.

Accepted Manuscript

miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell metabolism 2006;3:87-98. 51.

Marquart TJ, Allen RM, Ory DS, Baldan A. miR-33 links SREBP-2 induction to repression of sterol transporters. Proceedings of the National Academy of Sciences of the United States of America 2010;107:12228-12232.

52.

Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, Gerszten RE, Naar AM. MicroRNA33 and the SREBP host genes cooperate to control cholesterol homeostasis. Science 2010;328:1566-1569.

53.

Rayner KJ, Suarez Y, Davalos A, Parathath S, Fitzgerald ML, Tamehiro N, Fisher EA, Moore KJ, Fernandez-Hernando C. MiR-33 contributes to the regulation of cholesterol homeostasis. Science 2010;328:1570-1573.

54.

Soh J, Iqbal J, Queiroz J, Fernandez-Hernando C, Hussain MM. MicroRNA-30c reduces hyperlipidemia and atherosclerosis in mice by decreasing lipid synthesis and lipoprotein secretion. Nature medicine 2013;19:892-900.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

49.

22 55.

Davalos A, Goedeke L, Smibert P, Ramirez CM, Warrier NP, Andreo U, Cirera-Salinas D, Rayner K, Suresh U, Pastor-Pareja JC, Esplugues E, Fisher EA, Penalva LO, Moore KJ, Suarez Y, Lai EC, Fernandez-Hernando C. miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proceedings of the National Academy of Sciences of the United States of America 2011;108:9232-9237.

56.

Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of

2002;109:1125-1131. 57.

Yokoyama C, Wang X, Briggs MR, Admon A, Wu J, Hua X, Goldstein JL, Brown MS. SREBP1, a basic-helix-loop-helix-leucine zipper protein that controls transcription of the low density lipoprotein receptor gene. Cell 1993;75:187-197.

58.

Allen RM, Marquart TJ, Albert CJ, Suchy FJ, Wang DQ, Ananthanarayanan M, Ford DA,

Accepted Manuscript

Baldan A. miR-33 controls the expression of biliary transporters, and mediates statin- and dietinduced hepatotoxicity. EMBO Mol Med 2012;4:882-895. 59.

Li T, Francl JM, Boehme S, Chiang JY. Regulation of cholesterol and bile acid homeostasis by the cholesterol 7alpha-hydroxylase/steroid response element-binding protein 2/microRNA-33a axis in mice. Hepatology 2013;58:1111-1121.

60.

Horie T, Ono K, Horiguchi M, Nishi H, Nakamura T, Nagao K, Kinoshita M, Kuwabara Y, Marusawa H, Iwanaga Y, Hasegawa K, Yokode M, Kimura T, Kita T. MicroRNA-33 encoded by an intron of sterol regulatory element-binding protein 2 (Srebp2) regulates HDL in vivo. Proceedings of the National Academy of Sciences of the United States of America 2010;107:17321-17326.

61.

Rayner KJ, Esau CC, Hussain FN, McDaniel AL, Marshall SM, van Gils JM, Ray TD, Sheedy FJ, Goedeke L, Liu X, Khatsenko OG, Kaimal V, Lees CJ, Fernandez-Hernando C, Fisher EA, Temel RE, Moore KJ. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature 2011;478:404-407.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

cholesterol and fatty acid synthesis in the liver. The Journal of clinical investigation

23 62.

Rottiers V, Obad S, Petri A, McGarrah R, Lindholm MW, Black JC, Sinha S, Goody RJ, Lawrence MS, deLemos AS, Hansen HF, Whittaker S, Henry S, Brookes R, Najafi-Shoushtari SH, Chung RT, Whetstine JR, Gerszten RE, Kauppinen S, Naar AM. Pharmacological inhibition of a microRNA family in nonhuman primates by a seed-targeting 8-mer antimiR. Sci Transl Med 2013;5:212ra162.

63.

Marquart TJ, Wu J, Lusis AJ, Baldan A. Anti-miR-33 therapy does not alter the progression of

and vascular biology 2013;33:455-458. 64.

Rayner KJ, Sheedy FJ, Esau CC, Hussain FN, Temel RE, Parathath S, van Gils JM, Rayner AJ, Chang AN, Suarez Y, Fernandez-Hernando C, Fisher EA, Moore KJ. Antagonism of miR33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. The Journal of clinical investigation 2011;121:2921-2931.

Accepted Manuscript

65.

Rotllan N, Ramirez CM, Aryal B, Esau CC, Fernandez-Hernando C. Therapeutic silencing of microRNA-33 inhibits the progression of atherosclerosis in Ldlr-/- mice--brief report. Arteriosclerosis, thrombosis, and vascular biology 2013;33:1973-1977.

66.

Horie T, Baba O, Kuwabara Y, Chujo Y, Watanabe S, Kinoshita M, Horiguchi M, Nakamura T, Chonabayashi K, Hishizawa M, Hasegawa K, Kume N, Yokode M, Kita T, Kimura T, Ono K. MicroRNA-33 deficiency reduces the progression of atherosclerotic plaque in ApoE-/- mice. J Am Heart Assoc 2012;1:e003376.

67.

Gerin I, Clerbaux LA, Haumont O, Lanthier N, Das AK, Burant CF, Leclercq IA, MacDougald OA, Bommer GT. Expression of miR-33 from an SREBP2 intron inhibits cholesterol export and fatty acid oxidation. The Journal of biological chemistry 2010;285:33652-33661.

68.

Wijesekara N, Zhang LH, Kang MH, Abraham T, Bhattacharjee A, Warnock GL, Verchere CB, Hayden MR. miR-33a modulates ABCA1 expression, cholesterol accumulation, and insulin secretion in pancreatic islets. Diabetes 2012;61:653-658.

69.

Horie T, Nishino T, Baba O, Kuwabara Y, Nakao T, Nishiga M, Usami S, Izuhara M, Sowa N, Yahagi N, Shimano H, Matsumura S, Inoue K, Marusawa H, Nakamura T, Hasegawa K, Kume

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

atherosclerosis in low-density lipoprotein receptor-deficient mice. Arteriosclerosis, thrombosis,

24 N, Yokode M, Kita T, Kimura T, Ono K. MicroRNA-33 regulates sterol regulatory elementbinding protein 1 expression in mice. Nature communications 2013;4:2883. 70.

Ramirez CM, Goedeke L, Rotllan N, Yoon JH, Cirera-Salinas D, Mattison JA, Suarez Y, de Cabo R, Gorospe M, Fernandez-Hernando C. MicroRNA 33 regulates glucose metabolism. Mol Cell Biol 2013;33:2891-2902.

71.

Goedeke L, Vales-Lara FM, Fenstermaker M, Cirera-Salinas D, Chamorro-Jorganes A,

microRNA 33* in controlling lipid metabolism gene expression. Mol Cell Biol 2013;33:23392352. 72.

de Aguiar Vallim T, Tarling E, Kim T, Civelek M, Baldan A, Esau C, Edwards P. MicroRNA-144 Regulates Hepatic ABCA1 and Plasma HDL Following Activation of the Nuclear Receptor FXR. Circulation research 2013.

Accepted Manuscript

73.

Kang MH, Zhang LH, Wijesekara N, de Haan W, Butland S, Bhattacharjee A, Hayden MR. Regulation of ABCA1 protein expression and function in hepatic and pancreatic islet cells by miR-145. Arteriosclerosis, thrombosis, and vascular biology 2013;33:2724-2732.

74.

Kim J, Yoon H, Ramirez CM, Lee SM, Hoe HS, Fernandez-Hernando C. MiR-106b impairs cholesterol efflux and increases Abeta levels by repressing ABCA1 expression. Exp Neurol 2012;235:476-483.

75.

Ramirez CM, Davalos A, Goedeke L, Salerno AG, Warrier N, Cirera-Salinas D, Suarez Y, Fernandez-Hernando C. MicroRNA-758 regulates cholesterol efflux through posttranscriptional repression of ATP-binding cassette transporter A1. Arteriosclerosis, thrombosis, and vascular biology 2011;31:2707-2714.

76.

Ramirez CM, Rotllan N, Vlassov AV, Davalos A, Li M, Goedeke L, Aranda JF, Cirera-Salinas D, Araldi E, Salerno A, Wanschel AC, Zavadil J, Castrillo A, Jungsu K, Suarez Y, FernandezHernando C. Control of Cholesterol Metabolism and Plasma HDL Levels by miRNA-144. Circulation research 2013.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

Ramirez CM, Mattison JA, de Cabo R, Suarez Y, Fernandez-Hernando C. A regulatory role for

25 77.

Sun D, Zhang J, Xie J, Wei W, Chen M, Zhao X. MiR-26 controls LXR-dependent cholesterol efflux by targeting ABCA1 and ARL7. FEBS letters 2012;586:1472-1479.

78.

Wang D, Xia M, Yan X, Li D, Wang L, Xu Y, Jin T, Ling W. Gut microbiota metabolism of anthocyanin promotes reverse cholesterol transport in mice via repressing miRNA-10b. Circulation research 2012;111:967-981.

79.

Ramirez CM, Rotllan N, Vlassov AV, Davalos A, Li M, Goedeke L, Aranda JF, Cirera-Salinas

Hernando C. Control of cholesterol metabolism and plasma high-density lipoprotein levels by microRNA-144. Circulation research 2013;112:1592-1601. 80.

de Aguiar Vallim TQ, Tarling EJ, Kim T, Civelek M, Baldan A, Esau C, Edwards PA. MicroRNA-144 regulates hepatic ATP binding cassette transporter A1 and plasma highdensity lipoprotein after activation of the nuclear receptor farnesoid X receptor. Circulation

Accepted Manuscript

research 2013;112:1602-1612. 81.

Cheloufi S, Dos Santos CO, Chong MM, Hannon GJ. A dicer-independent miRNA biogenesis pathway that requires Ago catalysis. Nature 2010;465:584-589.

82.

Cifuentes D, Xue H, Taylor DW, Patnode H, Mishima Y, Cheloufi S, Ma E, Mane S, Hannon GJ, Lawson ND, Wolfe SA, Giraldez AJ. A novel miRNA processing pathway independent of Dicer requires Argonaute2 catalytic activity. Science 2010;328:1694-1698.

83.

Yang JS, Maurin T, Robine N, Rasmussen KD, Jeffrey KL, Chandwani R, Papapetrou EP, Sadelain M, O'Carroll D, Lai EC. Conserved vertebrate mir-451 provides a platform for Dicerindependent, Ago2-mediated microRNA biogenesis. Proceedings of the National Academy of Sciences of the United States of America 2010;107:15163-15168.

84.

Hu YW, Hu YR, Zhao JY, Li SF, Ma X, Wu SG, Lu JB, Qiu YR, Sha YH, Wang YC, Gao JJ, Zheng L, Wang Q. An Agomir of miR-144-3p Accelerates Plaque Formation through Impairing Reverse Cholesterol Transport and Promoting Pro-Inflammatory Cytokine Production. PloS one 2014;9:e94997.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

D, Araldi E, Salerno A, Wanschel A, Zavadil J, Castrillo A, Kim J, Suarez Y, Fernandez-

26 85.

Zhang M, Wu JF, Chen WJ, Tang SL, Mo ZC, Tang YY, Li Y, Wang JL, Liu XY, Peng J, Chen K, He PP, Lv YC, Ouyang XP, Yao F, Tang DP, Cayabyab FS, Zhang DW, Zheng XL, Tian GP, Tang CK. MicroRNA-27a/b regulates cellular cholesterol efflux, influx and esterification/hydrolysis in THP-1 macrophages. Atherosclerosis 2014;234:54-64.

86.

Miller AM, Gilchrist DS, Nijjar J, Araldi E, Ramirez CM, Lavery CA, Fernandez-Hernando C, McInnes IB, Kurowska-Stolarska M. MiR-155 has a protective role in the development of non-

87.

Ou Z, Wada T, Gramignoli R, Li S, Strom SC, Huang M, Xie W. MicroRNA hsa-miR-613 targets the human LXRalpha gene and mediates a feedback loop of LXRalpha autoregulation. Mol Endocrinol 2011;25:584-596.

88.

Zhong D, Huang G, Zhang Y, Zeng Y, Xu Z, Zhao Y, He X, He F. MicroRNA-1 and microRNA206 suppress LXRalpha-induced lipogenesis in hepatocytes. Cell Signal 2013;25:1429-1437.

Accepted Manuscript

89.

Adlakha YK, Khanna S, Singh R, Singh VP, Agrawal A, Saini N. Pro-apoptotic miRNA-128-2 modulates ABCA1, ABCG1 and RXRalpha expression and cholesterol homeostasis. Cell Death Dis 2013;4:e780.

90.

Ciafre SA, Galardi S. microRNAs and RNA-binding proteins: a complex network of interactions and reciprocal regulations in cancer. RNA biology 2013;10:935-942.

91.

Ho JJ, Marsden PA. Competition and collaboration between RNA-binding proteins and microRNAs. Wiley interdisciplinary reviews RNA 2014;5:69-86.

92.

Srikantan S, Tominaga K, Gorospe M. Functional interplay between RNA-binding protein HuR and microRNAs. Current protein & peptide science 2012;13:372-379.

93.

Srikantan S, Gorospe M. HuR function in disease. Frontiers in bioscience 2012;17:189-205.

94.

Ramirez CM, Lin CS, Abdelmohsen K, Goedeke L, Yoon JH, Madrigal-Matute J, MartinVentura JL, Vo DT, Uren PJ, Penalva LO, Gorospe M, Fernandez-Hernando C. RNA-binding protein HuR regulates expression of ABCA1. Journal of lipid research 2014.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

alcoholic hepatosteatosis in mice. PloS one 2013;8:e72324.

27 95.

Bhattacharyya SN, Habermacher R, Martine U, Closs EI, Filipowicz W. Relief of microRNAmediated translational repression in human cells subjected to stress. Cell 2006;125:11111124.

96.

Eiring AM, Harb JG, Neviani P, Garton C, Oaks JJ, Spizzo R, Liu S, Schwind S, Santhanam R, Hickey CJ, Becker H, Chandler JC, Andino R, Cortes J, Hokland P, Huettner CS, Bhatia R, Roy DC, Liebhaber SA, Caligiuri MA, Marcucci G, Garzon R, Croce CM, Calin GA, Perrotti D.

leukemic blasts. Cell 2010;140:652-665. 97.

Fernandez-Hernando C, Ackah E, Yu J, Suarez Y, Murata T, Iwakiri Y, Prendergast J, Miao RQ, Birnbaum MJ, Sessa WC. Loss of Akt1 leads to severe atherosclerosis and occlusive coronary artery disease. Cell metabolism 2007;6:446-457.

98.

Kuhlencordt PJ, Gyurko R, Han F, Scherrer-Crosbie M, Aretz TH, Hajjar R, Picard MH, Huang

Accepted Manuscript

PL. Accelerated atherosclerosis, aortic aneurysm formation, and ischemic heart disease in apolipoprotein E/endothelial nitric oxide synthase double-knockout mice. Circulation 2001;104:448-454. 99.

Rigotti A, Miettinen HE, Krieger M. The role of the high-density lipoprotein receptor SR-BI in the lipid metabolism of endocrine and other tissues. Endocr Rev 2003;24:357-387.

100.

Kocher O, Krieger M. Role of the adaptor protein PDZK1 in controlling the HDL receptor SRBI. Curr Opin Lipidol 2009;20:236-241.

101.

Kocher O, Yesilaltay A, Cirovic C, Pal R, Rigotti A, Krieger M. Targeted disruption of the PDZK1 gene in mice causes tissue-specific depletion of the high density lipoprotein receptor scavenger receptor class B type I and altered lipoprotein metabolism. The Journal of biological chemistry 2003;278:52820-52825.

102.

Hu Z, Shen WJ, Kraemer FB, Azhar S. MicroRNAs 125a and 455 repress lipoproteinsupported steroidogenesis by targeting scavenger receptor class B type I in steroidogenic cells. Mol Cell Biol 2012;32:5035-5045.

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

miR-328 functions as an RNA decoy to modulate hnRNP E2 regulation of mRNA translation in

28 103.

Wang L, Jia XJ, Jiang HJ, Du Y, Yang F, Si SY, Hong B. MicroRNAs 185, 96, and 223 repress selective high-density lipoprotein cholesterol uptake through posttranscriptional inhibition. Mol Cell Biol 2013;33:1956-1964.

104.

Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nature cell biology 2011;13:423-433. Tabet F, Vickers KC, Cuesta Torres LF, Wiese CB, Shoucri BM, Lambert G, Catherinet C, Prado-Lourenco L, Levin MG, Thacker S, Sethupathy P, Barter PJ, Remaley AT, Rye KA. HDL-transferred microRNA-223 regulates ICAM-1 expression in endothelial cells. Nature communications 2014;5:3292.

106.

Voight BF, Peloso GM, Orho-Melander M, Frikke-Schmidt R, Barbalic M, Jensen MK, Hindy G, Holm H, Ding EL, Johnson T, Schunkert H, Samani NJ, Clarke R, Hopewell JC, Thompson JF,

Accepted Manuscript

Li M, Thorleifsson G, Newton-Cheh C, Musunuru K, Pirruccello JP, Saleheen D, Chen L, Stewart A, Schillert A, Thorsteinsdottir U, Thorgeirsson G, Anand S, Engert JC, Morgan T, Spertus J, Stoll M, Berger K, Martinelli N, Girelli D, McKeown PP, Patterson CC, Epstein SE, Devaney J, Burnett MS, Mooser V, Ripatti S, Surakka I, Nieminen MS, Sinisalo J, Lokki ML, Perola M, Havulinna A, de Faire U, Gigante B, Ingelsson E, Zeller T, Wild P, de Bakker PI, Klungel OH, Maitland-van der Zee AH, Peters BJ, de Boer A, Grobbee DE, Kamphuisen PW, Deneer VH, Elbers CC, Onland-Moret NC, Hofker MH, Wijmenga C, Verschuren WM, Boer JM, van der Schouw YT, Rasheed A, Frossard P, Demissie S, Willer C, Do R, Ordovas JM, Abecasis GR, Boehnke M, Mohlke KL, Daly MJ, Guiducci C, Burtt NP, Surti A, Gonzalez E, Purcell S, Gabriel S, Marrugat J, Peden J, Erdmann J, Diemert P, Willenborg C, Konig IR, Fischer M, Hengstenberg C, Ziegler A, Buysschaert I, Lambrechts D, Van de Werf F, Fox KA, El Mokhtari NE, Rubin D, Schrezenmeir J, Schreiber S, Schafer A, Danesh J, Blankenberg S, Roberts R, McPherson R, Watkins H, Hall AS, Overvad K, Rimm E, Boerwinkle E, TybjaergHansen A, Cupples LA, Reilly MP, Melander O, Mannucci PM, Ardissino D, Siscovick D, Elosua R, Stefansson K, O'Donnell CJ, Salomaa V, Rader DJ, Peltonen L, Schwartz SM,

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

105.

29 Altshuler D, Kathiresan S. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet 2012;380:572-580. 107.

Khera AV, Cuchel M, de la Llera-Moya M, Rodrigues A, Burke MF, Jafri K, French BC, Phillips JA, Mucksavage ML, Wilensky RL, Mohler ER, Rothblat GH, Rader DJ. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. The New England journal of medicine 2011;364:127-135.

Figure 1. miRNAs regulate reverse cholesterol transport (RCT). ABCA1 is regulated by a number of miRNAs that reduce the cholesterol efflux to lipid-poor ApoA1 that originates nascent HDL particles. miR-33 also inhibits the expression of bile acid transporters (ABCB11 and ATP8B1) in the liver, thereby regulating the last step of the RCT. ABCA1 is also

Accepted Manuscript

regulated by miRNAs in the intestine and in the macrophages accumulated in atherosclerotic plaques. Free cholesterol in the nascent HDL is further esterified to cholesteryl esters by lecithin-cholesterol acyltransferase (LCAT) leading to the formation of mature HDL particles. HDL particles deliver cholesterol to the liver via SRB1 receptor, which is also regulated by several miRNAs including miR185, miR-223, miR-96 and miR-185. This figure was performed using the Servier Medical Art illustration resources (http://www.servier.com).

Figure 2. Interplay between miRNAs and HuR in the post-transcriptional regulation of ABCA1 expression. HuR reguales ABCA1 expression at post-transcriptional level. HuR might cooperate with miRNAs to enhance translational repression or activation and ARE-mediated mRNA decay (A) or compete by counteracting the miRNA binding to the 3’UTR (B).

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

FIGURE LEGEND

30 Figure 3. HDL-derived miR-223 is transferred to endothelial cells and reduces inflammation. HDL transports miRNAs including miR-92a, miR-126 and miR-223. miR-223 can be transferred to endothelial cells (ECs). The receptor that facilitates the transfer in ECs is unknown but previous studies demonstrate that SRB1 regulates the transport in human hepatic cells (Huh7). miR-223

Downloaded from http://cardiovascres.oxfordjournals.org/ at Memorial University of Newfoundland on July 6, 2014

Accepted Manuscript

inhibit ICAM-1 expression in ECs thereby reducing monocyte adhesion and inflammation.

INTESTINE Biliary Excretion

ABCG1

miR-10b MACROPHAGE

ABCA1

miR-26

ATP8B1

miR-144 CYP7A1

miR-185

LXR

miR-758 miR-33

miR-145 miR-33

epted Manuscript

miR-96

miR-223

miR-758

HDL Biogenesis

miR-27

miR-125a Lipid poor apoA1

LDLR

nascent HDL HL, EL

LCAT

Cholesterol Efflux

miR-26

LIVER

Figure 1

mature HDL CETP

VLDL/LDL

ATHEROSCLEROTIC PLAQUE

A ABCA1 5’UTR

ORP

3’UTR miR-33

miR-144

AUUUA

AUUUA

HuR

HuR

miR-758

miR-145

epted Manuscript

Ÿ  TRANSLATIONAL REPRESSION COOPERATIVE

B

Ÿ  TRANSLATIONAL ACTIVATION Ÿ  ARE-MEDIATED mRNA DECAY

ABCA1 5’UTR

ORP

3’UTR

miR-33

COMPETITIVE

miR-144

AUUUA

AUUUA

HuR

HuR

miR-758 miR-145

Ÿ  COUNTERACT Figure 2

HDL

MONOCYTES

miR-126

miR-92a miR-223

✖  

epted Manuscript

SRB1 ?

ENDOTHELIUM

ICAM1

miR-223 ICAM1

ATHEROSCLEROTIC PLAQUE

Figure 3

Target genes

miR-33a/b miR-33* miR-758

ABCA1, ABCG1, NPC1, CPT1A, SIRT6, AMPK, HADHB, CROT, CYP7A1, ABCB11, ATP8B1, NSF, SRC3, PCK1, G6PC, IRS2, RIP140, NFYC, SREBP1 NPC1, RIP140, SRC3, NFYC, IRS2, CROT ABCA1

miR-26 miR-145

ABCA1, ARL7 ABCA1

miR-106b

ABCA1

miR-10b

ABCA1, ABCG1

miR-144

ABCA1

ccepted Manuscript

miRNA

miR-27 miR-206 miR-613 miR-96 miR-223 miR-185 miR-125a

ABCA1

miR-455

SRB1

LXRα LXRα SRB1 SRB1 SRB1 SRB1

  Table 1. miRNA regulation of HDL metabolism

Cell type /Tissue

Ref

Macrophages (primary mouse peritoneal macs, THP- 51, 52, 53, 1), Endothelial cells (EAhy926), hepatic cells (HepG2, 55, 58, 59, HEPA, Fu5aH, Hep3B) and mouse liver. 60, 67 Hepatic cells (Huh-7), macrophages (THP-1) 71 Macrophages (primary mouse peritoneal macs, J774, 75 THP-1), hepatic cells (HepG2, Huh-7, HEPA) and neuroglyomal cells (H4). Macrophages (Raw264.7 and THP-1). 77 73 Hepatic cells (HepG2) and pancreatic β cells (MIN6 and primary mouse β cells). Mouse primary hippocampal neurons (DIV14) and 74 mouse neuroblastoma cells (Neuro2a). Macrophages (primary mouse peritoneal macs, 78 J774,THP-1). Macrophages (mouse peritoneal macrophages, J774, 79, 80, 84 THP-1), hepatic cells (HepG2, Huh-7, Hepa), endothelial cells (EAhy926), mouse liver, human hepatic cells (Hep3B) and mouse primary hepatocytes. Macrophages (THP-1) 85 Macrophages (THP-1) 88 Hepatic cells (HepG2) 87 Hepatic cells (HepG2) 103 Carried on HDL 103 Hepatic cells (HepG2) 103 Hepatic cells (Hepa), steroidogenic cell lines (MLTC102 1, granulosa cells) Hepatic cells (Hepa), steroidogenic cell lines (MLTC1, granulosa cells)

102

microRNAs and HDL life cycle.

miRNAs have emerged as important regulators of lipoprotein metabolism. Work over the past few years has demonstrated that miRNAs control the expressio...
2MB Sizes 2 Downloads 3 Views