Articles in PresS. Physiol Genomics (October 29, 2013). doi:10.1152/physiolgenomics.00106.2013

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23

MicroRNA Associated with Dyslipidemia and Coronary Disease in Humans Elena Flowers,1 Bradley E. Aouizerat1, 2 1 2

Department of Physiological Nursing, School of Nursing, University of California, San Francisco Institute for Human Genetics, University of California, San Francisco

Running Head: MicroRNA and Lipids in Humans Contact Information: Elena Flowers Department of Physiological Nursing 2 Koret Way, #605L San Francisco, CA 94143-0610 415-476-0983 [email protected] Author Contributions: Disclosures/conflicts of interest: none

24 25

1

Copyright © 2013 by the American Physiological Society.

26 27 28

ABSTRACT

29

messenger RNA translation. Recently, there is significant interest in the application of microRNA as a

30

blood-based biomarker of underlying physiologic conditions. Dyslipidemia is a complex, heterogeneous

31

condition conferring substantially increased risk for cardiovascular disease. The purpose of this review is

32

to describe the current body of knowledge on the role of microRNA regulation of lipoprotein metabolism

33

in humans, and to discuss relevant methodologic and study design considerations. We highlight the

34

potential roles for microRNA in gene-environment interactions.

35 36 37

KEY WORDS

38

microRNA

39

lipids

MicroRNAs are structural components of an epigenetic mechanism of post-transcriptional regulation of

40 41

Figure 1 Legend

42 43 44 45 46 47

CAD: coronary artery disease TC: total cholesterol LDL: low-density lipoprotein cholesterol HDL: high-density lipoprotein cholesterol PBMCs: peripheral blood mononuclear cells

2

48 49

Introduction Dyslipidemia is a common risk factor for cardiovascular disease, and reductions in the levels of

50

atherogenic lipoproteins are associated with substantially decreased risk. (39) The two primary

51

approaches to treatment for dyslipidemia are pharmacologic agents and lifestyle change. (39) However,

52

even in combination these interventions are not completely effective at achieving adequate risk reduction

53

in all individuals. One important reason that risk reduction efforts fail to achieve full success is

54

incomplete understanding of inter-individual differences in the underlying pathophysiology. While single

55

genes and discrete environmental exposures that increase risk have been identified, for most individuals

56

the etiology is complex (i.e., multifactorial). (9)

57

A recent paradigm shift in biomarker research has resulted from the development of methods to

58

measure adaptive changes of the genome and its expression, termed epigenetics. Epigenetic events are

59

particularly attractive for clinical research because they represent an aggregate response to many

60

upstream events. In other words, epigenetic events are the sum of genetic risk factors and sustained

61

environmental exposures that drive an organism towards adaptive responses to maintain homeostasis. To

62

date, three primary epigenetic mechanisms have been identified and described in detail elsewhere: histone

63

modification (16), DNA methylation (22), and microRNA (54).

64

MicroRNAs (miRs) are an epigenetic mechanism of post-transcriptional regulation of messenger

65

RNA (mRNA). MiRs function by binding to a complementary 18-24 nucleotide precursor region on

66

mRNA known as the “seed” sequence, thereby preventing initiation of translation of mRNA to amino

67

acids. (28) MiR regulation is dynamic; the function can be temporary, when the microRNA release the

68

mRNA, or permanent, causing degradation of the mRNA strand. (46) Currently, there are 2,042 discrete

69

miR species identified in humans, and approximately 300 are detectable in blood. (27) MiR are the

70

subject of numerous research studies because they currently have three potential clinical implications: (1)

71

biomarkers of underlying disease; (2) therapeutic treatments (i.e., synthetic miRs or miR antagonists); and

72

(3) as markers for response to therapeutic interventions to prevent and treat disease. The subset of miR

73

detectable in blood are of particular interest because their measurement is minimally invasive and blood3

74

based diagnostic testing is common in the clinical setting. Additional studies are needed in order to

75

determine the origins of miRs found in blood to fully understand the pathophysiological underpinnings of

76

dyslipidemia.

77 78

Non-Human Studies

79

Numerous in vitro and animal model studies have examined the regulatory role of microRNA on

80

lipid metabolism (Table 1). (7, 8, 12, 36, 37, 42, 45) MiR-122 is the most prevalent miR in animal model

81

hepatocytes, targeting numerous genes involved in lipid metabolism in the liver (i.e., diacylglycerol O-

82

acyltransferase 2, glycogen synthase 1, solute carrier family 7, acetyl-coA carboxylase α, nuclear receptor

83

subfamily 1, group H, member 3/liver X receptor α, fatty acid synthase, sterol regulatory element binding

84

transcription factor-1c , cholesterol 7α-hydroxylase), and adipocytes (i.e., adenosine triphosphate binding

85

cassette transporter-1, diacylglycerol O-acyltransferase 2) with associated changes in lipid parameters.

86

Greater detail on the effects of miR-122 on lipid metabolism has previously been published. (12) The

87

aggregate effects of miR-122 in non-human studies are decreased total cholesterol and triglycerides and

88

decreased production of the LDL receptor. Further research on miR-122 in humans is needed to determine

89

whether this miR has the same effects as those seen in in vitro and animal model studies, as well as

90

whether this miR might be useful as a therapeutic agent for modification of lipoprotein levels.

91

Recently, miR-33 has become a potential therapeutic target for dyslipidemia in animal models. In

92

African Green Monkeys, inhibition of miR-33 results in decreased serum triglycerides and increased

93

HDL. (41) Additional studies in the same primate model and also in mouse models demonstrated miR-33

94

is located in an intron of the sterol-regulatory binding element transcription factor gene and regulates this

95

mRNA as well as adenosine triphosphate binding cassette transporter and others. (5, 12, 18, 38, 43)

96

Although the findings from in vitro and animal model studies established the role of miR-33 in

97

modulating biological pathways underlying dyslipidemia, the findings have limited generalizability to

98

humans. For example, the miR-33a isoform, which is readily detectable in mice, is not expressed in

4

99 100 101

humans; thus initial findings from studies of mouse models had limited implications for potential translation to clinical practice. Although there is growing evidence to support miR regulation of lipoprotein metabolism, the

102

translational implications of studies from in vitro and animal model studies are limited. Currently

103

published reviews in this area have not concentrated on findings from studies in humans with more

104

imminent translational potential (i.e., miRs as surrogate biomarkers for underlying pathophysiology). The

105

primary focus of this review is the synthesis of findings from studies of miR regulation of lipid

106

metabolism in humans.

107 108

Human Studies

109

Cardiovascular Risk Factors

110

There is a small but growing body of literature about miR expression and dyslipidemia in

111

humans, and particularly blood-based expression of miR associated with underlying perturbations in lipid

112

metabolism and/or sequelae (e.g., coronary artery disease (CAD)) (Table 1). Karolina, et. al.,

113

characterized miRs in whole blood and exosomes in 265 Singaporean patients with and without the

114

metabolic syndrome and its component risk factors (i.e., dyslipidemia, hypertension, and type-2 diabetes).

115

(26) Using microarray-based screening of known human miRs followed by validation of a subset of

116

targets by quantitative polymerase chain reaction (qPCR), unique miR profiles characterizing three risk

117

factor profiles associated with metabolic syndrome were identified. (26) Eight miR species (miR-103,

118

miR-17, miR-183, miR-197, miR-23a, miR-509-5p, miR-584, miR-652) were unique to the metabolic

119

syndrome and dyslipidemia cluster (n = 41) compared to the controls (n = 17) by microarray. (26) Three

120

of these (miR-197, miR-23a, miR-509-5p) were also significantly correlated with body mass index. (26)

121

Only a subset (miR-17, miR-197, miR-509-5p) were detectable by qPCR in exosomes. (26) Flowers,

122

et.al., (in review) similarly used microarray methods to screen for the presence of 85 known miR species

123

in South Asian men with (n=22) and without (n=22) atherogenic dyslipidemia. Fifteen miRs (miR-100,

124

miR-106b, miR-125b, miR-143, miR-148a, miR-17, miR-17*, miR-18a, miR-20a, miR-21, miR-221, 5

125

miR-374a, miR-7, miR-93, miR-96) were found to be differentially expressed at a magnitude of at least

126

two-fold. Of note, miR-17 was observed to be differentially expressed by microarray-based measurement

127

in both studies, and has previously been observed to target and suppress angiogenic pathways in in vitro

128

studies. (6, 24) Karolina, et.al., observed increased expression in dyslipidemic individuals compared to

129

healthy controls (fold change 2.77), whereas Flowers, et.al., observed decreased expression (fold change -

130

2.27). One possible explanation for these apparently discrepant findings is the specific dyslipidemic

131

phenotype selected for case status was primarily focused on hypercholesterolemia (i.e., LDL > 3.4

132

mmol/L or total cholesterol > 5.0 mmol/L (26) as compared to hypertrigclyeridemia (i.e., HDL < 1.03

133

mmol/L and triglycerides >1.7 mmol/L (Flowers, et.al.)) and the differing racial background of each

134

sample.

135

Coronary Artery Disease

136

Two studies have reported blood-based differential expression of miRs in individuals with and

137

without CAD, with associated alterations in lipoprotein profiles. Gao, et.al. studied 355 Chinese patients

138

who underwent coronary angioagraphy, 255 of whom had dyslipidemia (total cholesterol ≥ 5.72mmol/L

139

and/or triglycerides ≥ 1.7mmol/L). (15) QPCR was used to measure blood plasma expression of miR-122,

140

miR-370, miR-33a, and miR-33b, miRs frequently examined in in vitro and animal model studies of lipid

141

metabolism. (15) Hyperlipidemic patients displayed statistically significantly greater expression of both

142

miR-122 and miR-370, and this effect was attenuated in those treated with statins. (15) Both miRs were

143

positively associated with total cholesterol, triglycerides, and LDL cholesterol, but no relationship was

144

observed for HDL cholesterol. (15) Patients with CAD also displayed greater expression of both miRs,

145

and this relationship remained significant after adjusting for covariates, including lipoprotein levels. (15)

146

Of note, the two miR-33 isoforms were undetectable in plasma. (15) In a similar study, Tabuchi, et.al.

147

studied miR-34 in 70 Japanese patients undergoing coronary angiography and 48 controls with no

148

symptoms of CAD. (49) The patients with CAD were randomized to receive one of two statins following

149

angiography. (49) This study also used qPCR but quantified miR expression from peripheral blood

150

mononuclear cells, which are a subset of the cell types found in whole blood. (49) MiR-34 was chosen 6

151

based on prior data from in vitro studies indicating that this miR regulates silent information regulator 1

152

(SIRT1), which has anti-atherosclerotic effects by influencing vascular endothelial cell homeostasis and is

153

a proposed therapeutic target of statins. MiR-34 showed statistically significantly greater expression in

154

patients with CAD compared to the controls, and an inverse association with SIRT1 protein levels. (49)

155

Post-treatment, no differences between the two statin groups for miR-34 or SIRT1 levels were observed.

156

(49) Comparisons of pre-post statin treatment were not reported, thus no determination of the effects of

157

statins on miR-34 or SIRT1 levels can be made. Both of these studies provide evidence that individual

158

circulating miRs are associated with underlying pathophysiology (i.e., dyslipidemia and CAD). Although

159

the samples, covariates (i.e., statin therapy), and miR measurement methods were similar between these

160

two studies, there was no overlap in the miR targets tested. Further, the studies evaluated different tissue

161

sources (i.e., plasma versus peripheral blood mononuclear cells), and used different reference genes to

162

calculate normalized miR expression. Although comparison of the miR results is not possible, this

163

aggregate set of miR species with a plausible role in lipid metabolism warrant further investigation.

164 165 166 167

Methodological Considerations

168

expression associated with dyslipidemia in humans. (11) The first is the tissue source used for detection

169

of miRs. Similar to mRNA, miR expression varies among tissues. In most cases, peripheral blood is the

170

most readily accessible tissue source in routine clinical care, which is a fundamental driver of widespread

171

interest in circulating miRs as a biomarker(s) for underlying disease processes and risk. MiRs are readily

172

detectable in the circulation, and are differentially expressed in numerous disease states and/or

173

pathophysiological processes compared to healthy controls. (3, 35) MiRs are detectable in several blood

174

compartments: plasma, serum, exosomes, microparticles, platelets, erythrocytes, and leukocytes. (14, 19,

175

51, 56) Prior studies of miR and lipid metabolism examined several discrete blood compartments (Figure

176

1). Comparison of blood-based expression of miRs must consider the specific source from which miR

177

were isolated. For example, in the studies of miR expression and CAD, Gao, et.al. quantified miRs in

There are several important methodological considerations for interpretation of studies of miR

7

178

blood plasma, whereas Tabuchi, et.al. studied peripheral blood mononuclear cells. Even plasma and

179

serum show some differences in the specific species and quantity of miRs detectable. (56) To date, it is

180

unclear whether detection of blood-based miRs detects causal pathways to diseaseor whether miRs

181

originate from apoptotic cells in ischemic or damaged solid organs that are sloughed into the circulation.

182

Depending on the origin of the miRs and underlying pathophysiology (i.e., blood-based versus solid

183

organ-based), some miRs may need compartment-specific measures and others will not. There are

184

examples of both types of biomarkers in routine clinical use at present. Cardiac troponins, which are

185

released into the circulation by ischemic cardiomyocytes, reflect a causal relationship and are widely used

186

in clinical care to diagnose myocardial infarction. By contrast, elevated LDL cholesterol is used to

187

identify individuals who are at high risk for atherosclerosis, but the biomarker itself is not diagnostic for

188

CAD. Additional studies are needed to determine the origins of circulating miR in order to understand the

189

primary physiology. Regardless of the underlying causal relationship, blood-based miR expression has

190

the potential to be a useful marker for disease.

191

The second consideration is how miRs are measured. Until recently, the gold standard for

192

measurement was qPCR, and for practical reasons (i.e., cost, access to resources, data analysis), it remains

193

the primary method reported in the literature. Whole transcriptome sequencing is the next generation of

194

measurement for RNA, including miR, improving measurement of miR considerably through more

195

accurate quantitation of the miR species and number. Another option, microarray, uses hybridization to

196

detect the type and number of miRs present, and offers higher throughput and greater sensitivity and

197

specificity compared to qPCR. (11) There are several commercially available platforms for both

198

microarray and qPCR-based quantitation of miRs. Comparison studies show a high level of variability in

199

the discrete miR species detected and correlation in the magnitude of expression between commercial

200

miR measurement products (2, 56), suggesting that some caution should be taken in comparing results of

201

studies using differing measurement platforms and commercial products.

202 203

Commonly, studies of gene expression report normalized expression, in which expression of each individual sample is standardized to the expression of a reference gene or combination of genes that is 8

204

ubiquitously expressed at constant levels in most tissue types across individuals. (1, 50, 52) Such

205

normalization controls for variation in the starting quantity of biologic material that could otherwise skew

206

comparisons of relative expression between groups (i.e., diseased versus non-diseased). However for

207

miR, a universally accepted normalizer control has not been identified. A commonly used normalizer is

208

the small nuclear RNU6b. However, recent studies have reported considerable variation of expression of

209

this RNA. (40, 57) Normalization to the average expression of all measured miRs in all samples in a

210

single experiment, normalization to the least variable miR in each experiment, and alternative approaches

211

have been proposed. (4, 33, 34, 58) To date, there is no universally accepted method for normalization of

212

expression, and different strategies used between studies are an important consideration for interpreting

213

conclusions.

214 215 216

Gene-Environment Interactions A potential implication of miR activity is modulation of the effects of environmental exposures.

217

Gene-environment interactions are widely known to effect risk factors for cardiovascular disease,

218

including dyslipidemia. (10) MiRs may be the structural cause of gene-environment interactions, as is the

219

case when a DNA polymorphism lies in the miR coding site or within the seed sequence in the promoter

220

region of the mRNA target and secondary exposure to an environmental insult triggers deleterious

221

consequences. For example, a single nucleotide polymorphism in the lipoprotein lipase (LPL) gene

222

(rs13702) appears to lie within the miR-410 seed sequence. (44) This polymorphism is also linked to a

223

gene-environment interaction, as the rare C allele is associated with an additive inverse relationship

224

between polyunsaturated fat intake and triglycerides. (44) Genome-wide association data indicate that the

225

rare C allele is associated with lower triglycerides and higher HDL, and in vitro transfection of the

226

genomic region containing the rare C allele along with a miR-410 mimic results in gain of function of

227

LPL compared to transfection with the common T allele. (44) Further, there is a dose-dependent response

228

for the miR-410 mimic in the major allele transfected cells. (44)

9

229

Alternatively, miR may be a mechanism for gene-environment interactions through its inherent

230

regulatory function on gene expression, resulting in increased or decreased expression of an mRNA target

231

as an adaptive response to environmental exposure. For example, workers exposed to perflouroaklyl

232

chemicals (PFCs) in their workplace show an inverse correlation between blood levels of PFCs and HDL,

233

and array-based differential expression of 9 miRs (miR-127, miR-601, miR-106b, miR-24, miR-199a-3p,

234

miR-92a, miR-26b, miR-30c, miR-30b) compared to nearby residents of in the same community who

235

were not exposed to PFCs. (55) These differences were validated by qPCR for miR-26b and miR-199a-

236

3p, and a linear association was seen between fold-change miR expression and blood levels of PFCs. (55)

237

Another example of how miR may function to allow an organism to adapt to environmental changes is

238

through diet-induced miR activity. We were unable to find any published studies describing the effect of

239

dietary factors on miR activity and dyslipidemia performed in humans, but several in vitro and animal

240

model studies of other conditions indicate that this relationship is plausible.(23, 25, 29, 31, 47, 48, 53, 60)

241

Increasing interest in the implications of miRs in the area of gene-environment interactions is evidenced

242

by the recent creation of the miREnvironment Database, which cross-references miR species with

243

environmental risk factors and phenotypes. (59)

244 245 246

Racial Ancestry and MiR Expression Although the currently published studies reporting miR expression associated with dyslipidemia

247

do not lend themselves to direct comparison because of differences in study design, sample, and methods,

248

we can anticipate that future studies will incorporate comprehensive screening of miR targets detectable

249

in blood in samples with well-characterized phenotypes, creating the possibility of direct comparison. The

250

trajectory of miR-based research, although relatively new, closely follows the arc of both genetic (i.e.,

251

DNA polymorphisms) and gene-expression (i.e., mRNA) studies. Particularly in genetic studies, there

252

exists variability among studies in the observed associations between gene polymorphisms and risk

253

phenotypes. (21) This is particularly true for multi-factorial phenotypes like dyslipidemia (17) and CAD

254

(30). Proposed alternative explanations for discrepant findings in genetic studies include unknown and 10

255

unmeasured gene-gene and gene-environment interactions. (10, 30) Differences in the underlying

256

prevalence of gene polymorphisms among racial groups may add another layer of complexity to the

257

aforementioned interactions. (13, 32) While the myriad gene-gene and gene-environment interactions for

258

dyslipidemia have yet to be fully characterized, methods to quantify population substructure (i.e., racial

259

ancestry), have been developed – namely measurement of ancestry informative markers (AIMs) that

260

distinguish five racial groups on the basis of genotype prevalence. (20) To date, no studies investigating

261

the corollary for miRs have been reported. We posit that differential expression of miRs might be

262

influenced in part by differences in underlying genetic makeup between ancestry groups. For example,

263

four of the studies on miRs and lipid metabolism cited above had samples of Asian ancestry (i.e.,

264

Singaporean, South Asian, Chinese, Japanese) – racial groups which, although categorized as Asian, may

265

not share equal genetic commonalities. Thus, confounding may occur in studies of miRs in humans when

266

genetic background is not properly accounted for in data analyses. Further investigation of possible miR

267

AIMs will bring to light the prospect of controlling for differences in ancestry that may be confounding

268

observed associations or lack of associations between miR expression and disease states.

269 270

Conclusion

271

Dyslipidemia is a complex, multi-factorial risk factor for cardiovascular disease that arises in the

272

setting of both genetic and environmental exposures. MiRs are an epigenetic mechanism of regulation of

273

gene expression that facilitate dynamic adaptation of organisms to their environment. As such, miRs are

274

an appealing possible biomarker with two important implications: (1) earlier detection of impending

275

cardiovascular risk than is currently available through routine screening of lipoprotein levels; and (2)

276

elucidation of the underlying genetic mechanisms at play in a given individual. As blood is readily

277

accessible in clinical care, there is particular interest in detection of both causal and non-causal blood-

278

based miR expression associated with underlying pathophysiology. In addition to the biomarker

279

implications, the possibility exists that miRs may be a therapeutic agent, through either administration of

280

synthetic miR or miR antagonists. To date, a number of in vitro and animal model studies describing miR 11

281

regulation of lipoprotein metabolism have been published, but less is known about these mechanisms in

282

humans, with virtually no data to describe the molecular functions of miR in humans. The few human

283

studies consistently show differential expression of miRs in blood compartments of individuals with

284

dyslipidemia and/or CAD. Methodologic differences in the design of these studies (i.e., miR tissue

285

source, specific miR targets evaluated, measurement method) limit the possibility for making meaningful

286

conclusions about which miR are of potential clinical relevance for detection and treatment of

287

dyslipidemia as well as comparison of results between studies. Additional possible methodologic and

288

study design issues relevant to studies of miRs and dyslipidemia include gene-environment interactions

289

and racial ancestry. Further work is needed to evaluate the full cadre of blood-based miRs associated with

290

dyslipidemia in humans, and to delve into possible racial differences in the prevalence and variability of

291

individual miR species and the presence of gene-environment interactions exerting pressure on miR

292

regulation of gene expression.

12

293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342

References 1. Bas A, Forsberg G, Hammarstrom S, and Hammarstrom ML. Utility of the housekeeping genes 18S rRNA, beta-actin and glyceraldehyde-3-phosphate-dehydrogenase for normalization in realtime quantitative reverse transcriptase-polymerase chain reaction analysis of gene expression in human T lymphocytes. Scandinavian journal of immunology 59: 566-573, 2004. 2. Callari M, Dugo M, Musella V, Marchesi E, Chiorino G, Grand MM, Pierotti MA, Daidone MG, Canevari S, and De Cecco L. Comparison of microarray platforms for measuring differential microRNA expression in paired normal/cancer colon tissues. PLoS One 7: e45105, 2012. 3. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, Li Q, Li X, Wang W, Wang J, Jiang X, Xiang Y, Xu C, Zheng P, Zhang J, Li R, Zhang H, Shang X, Gong T, Ning G, Zen K, and Zhang CY. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 18: 997-1006, 2008. 4. D'Haene B, Mestdagh P, Hellemans J, and Vandesompele J. miRNA expression profiling: from reference genes to global mean normalization. Methods Mol Biol 822: 261-272, 2012. 5. 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, and Fernandez-Hernando C. miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc Natl Acad Sci U S A 108: 9232-9237, 2011. 6. Doebele C, Bonauer A, Fischer A, Scholz A, Reiss Y, Urbich C, Hofmann WK, Zeiher AM, and Dimmeler S. Members of the microRNA-17-92 cluster exhibit a cell-intrinsic antiangiogenic function in endothelial cells. Blood 115: 4944-4950, 2010. 7. Fernandez-Hernando C, and Moore KJ. MicroRNA modulation of cholesterol homeostasis. Arterioscler Thromb Vasc Biol 31: 2378-2382, 2011. 8. Fernandez-Hernando C, Ramirez CM, Goedeke L, and Suarez Y. MicroRNAs in metabolic disease. Arterioscler Thromb Vasc Biol 33: 178-185, 2013. 9. Flowers E, Froelicher ES, and Aouizerat BE. Gene-environment interactions in cardiovascular disease. European journal of cardiovascular nursing : journal of the Working Group on Cardiovascular Nursing of the European Society of Cardiology 2011. 10. Flowers E, Froelicher ES, and Aouizerat BE. Gene-environment interactions in cardiovascular disease. European journal of cardiovascular nursing : journal of the Working Group on Cardiovascular Nursing of the European Society of Cardiology 11: 472-478, 2012. 11. Flowers E, Froelicher ES, and Aouizerat BE. Measurement of MicroRNA: A Regulator of Gene Expression. Biological research for nursing 15: 167-178, 2013. 12. Flowers E, Froelicher ES, and Aouizerat BE. MicroRNA regulation of lipid metabolism. Metabolism 2012. 13. Freedman ML, Reich D, Penney KL, McDonald GJ, Mignault AA, Patterson N, Gabriel SB, Topol EJ, Smoller JW, Pato CN, Pato MT, Petryshen TL, Kolonel LN, Lander ES, Sklar P, Henderson B, Hirschhorn JN, and Altshuler D. Assessing the impact of population stratification on genetic association studies. Nat Genet 36: 388-393, 2004. 14. Gallo A, Tandon M, Alevizos I, and Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One 7: e30679, 2012. 15. Gao W, He HW, Wang ZM, Zhao H, Lian XQ, Wang YS, Zhu J, Yan JJ, Zhang DG, Yang ZJ, and Wang LS. Plasma levels of lipometabolism-related miR-122 and miR-370 are increased in patients with hyperlipidemia and associated with coronary artery disease. Lipids in health and disease 11: 55, 2012. 16. Greer EL, and Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet 13: 343-357, 2012. 17. Hegele RA. Plasma lipoproteins: genetic influences and clinical implications. Nat Rev Genet 10: 109-121, 2009. 13

343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393

18. 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, and Kita T. MicroRNA-33 encoded by an intron of sterol regulatory element-binding protein 2 (Srebp2) regulates HDL in vivo. Proc Natl Acad Sci U S A 107: 17321-17326, 2010. 19. Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, Xiao T, Schafer J, Lee ML, Schmittgen TD, Nana-Sinkam SP, Jarjoura D, and Marsh CB. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One 3: e3694, 2008. 20. International HapMap C, Altshuler DM, Gibbs RA, Peltonen L, Altshuler DM, Gibbs RA, Peltonen L, Dermitzakis E, Schaffner SF, Yu F, Peltonen L, Dermitzakis E, Bonnen PE, Altshuler DM, Gibbs RA, de Bakker PI, Deloukas P, Gabriel SB, Gwilliam R, Hunt S, Inouye M, Jia X, Palotie A, Parkin M, Whittaker P, Yu F, Chang K, Hawes A, Lewis LR, Ren Y, Wheeler D, Gibbs RA, Muzny DM, Barnes C, Darvishi K, Hurles M, Korn JM, Kristiansson K, Lee C, McCarrol SA, Nemesh J, Dermitzakis E, Keinan A, Montgomery SB, Pollack S, Price AL, Soranzo N, Bonnen PE, Gibbs RA, Gonzaga-Jauregui C, Keinan A, Price AL, Yu F, Anttila V, Brodeur W, Daly MJ, Leslie S, McVean G, Moutsianas L, Nguyen H, Schaffner SF, Zhang Q, Ghori MJ, McGinnis R, McLaren W, Pollack S, Price AL, Schaffner SF, Takeuchi F, Grossman SR, Shlyakhter I, Hostetter EB, Sabeti PC, Adebamowo CA, Foster MW, Gordon DR, Licinio J, Manca MC, Marshall PA, Matsuda I, Ngare D, Wang VO, Reddy D, Rotimi CN, Royal CD, Sharp RR, Zeng C, Brooks LD, and McEwen JE. Integrating common and rare genetic variation in diverse human populations. Nature 467: 52-58, 2010. 21. Ioannidis JP, Ntzani EE, Trikalinos TA, and Contopoulos-Ioannidis DG. Replication validity of genetic association studies. Nat Genet 29: 306-309, 2001. 22. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13: 484-492, 2012. 23. Joven J, Espinel E, Rull A, Aragones G, Rodriguez-Gallego E, Camps J, Micol V, HerranzLopez M, Menendez JA, Borras I, Segura-Carretero A, Alonso-Villaverde C, and Beltran-Debon R. Plant-derived polyphenols regulate expression of miRNA paralogs miR-103/107 and miR-122 and prevent diet-induced fatty liver disease in hyperlipidemic mice. Biochim Biophys Acta 1820: 894-899, 2012. 24. Kaluza D, Kroll J, Gesierich S, Manavski Y, Boeckel JN, Doebele C, Zelent A, Rossig L, Zeiher AM, Augustin HG, Urbich C, and Dimmeler S. Histone Deacetylase 9 Promotes Angiogenesis by Targeting the Antiangiogenic MicroRNA-17-92 Cluster in Endothelial Cells. Arterioscler Thromb Vasc Biol 33: 533-543, 2013. 25. Karere GM, Glenn JP, VandeBerg JL, and Cox LA. Differential microRNA response to a high-cholesterol, high-fat diet in livers of low and high LDL-C baboons. BMC Genomics 13: 320, 2012. 26. Karolina DS, Tavintharan S, Armugam A, Sepramaniam S, Pek SL, Wong MT, Lim SC, Sum CF, and Jeyaseelan K. Circulating miRNA profiles in patients with metabolic syndrome. The Journal of clinical endocrinology and metabolism 97: E2271-2276, 2012. 27. Kozomara A, and Griffiths-Jones S. miRBase: integrating microRNA annotation and deepsequencing data. Nucleic Acids Res 39: D152-157, 2011. 28. Lai EC. Micro RNAs are complementary to 3' UTR sequence motifs that mediate negative posttranscriptional regulation. Nat Genet 30: 363-364, 2002. 29. Lam TK, Shao S, Zhao Y, Marincola F, Pesatori A, Bertazzi PA, Caporaso NE, Wang E, and Landi MT. Influence of quercetin-rich food intake on microRNA expression in lung cancer tissues. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 21: 2176-2184, 2012. 30. Lanktree MB, and Hegele RA. Gene-gene and gene-environment interactions: new insights into the prevention, detection and management of coronary artery disease. Genome medicine 1: 28, 2009. 31. Mandal CC, Ghosh-Choudhury T, Dey N, Choudhury GG, and Ghosh-Choudhury N. miR21 is targeted by omega-3 polyunsaturated fatty acid to regulate breast tumor CSF-1 expression. Carcinogenesis 33: 1897-1908, 2012. 14

394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444

32. Marchini J, Cardon LR, Phillips MS, and Donnelly P. The effects of human population structure on large genetic association studies. Nat Genet 36: 512-517, 2004. 33. Mestdagh P, Van Vlierberghe P, De Weer A, Muth D, Westermann F, Speleman F, and Vandesompele J. A novel and universal method for microRNA RT-qPCR data normalization. Genome Biol 10: R64, 2009. 34. Meyer SU, Kaiser S, Wagner C, Thirion C, and Pfaffl MW. Profound effect of profiling platform and normalization strategy on detection of differentially expressed microRNAs--a comparative study. PLoS One 7: e38946, 2012. 35. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, and Tewari M. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A 105: 1051310518, 2008. 36. Moore KJ, Rayner KJ, Suarez Y, and Fernandez-Hernando C. The role of microRNAs in cholesterol efflux and hepatic lipid metabolism. Annual review of nutrition 31: 49-63, 2011. 37. Najafi-Shoushtari SH. MicroRNAs in cardiometabolic disease. Current atherosclerosis reports 13: 202-207, 2011. 38. Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, Gerszten RE, and Naar AM. MicroRNA-33 and the SREBP host genes cooperate to control cholesterol homeostasis. Science 328: 1566-1569, 2010. 39. NCEP. Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation 106: 3143-3421, 2002. 40. Peltier HJ, and Latham GJ. Normalization of microRNA expression levels in quantitative RTPCR assays: identification of suitable reference RNA targets in normal and cancerous human solid tissues. RNA 14: 844-852, 2008. 41. 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, and Moore KJ. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature 478: 404-407. 42. Rayner KJ, Fernandez-Hernando C, and Moore KJ. MicroRNAs regulating lipid metabolism in atherogenesis. Thrombosis and haemostasis 107: 642-647, 2012. 43. 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, and Moore KJ. Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J Clin Invest 121: 2921-2931, 2011. 44. Richardson K, Nettleton JA, Rotllan N, Tanaka T, Smith CE, Lai CQ, Parnell LD, Lee YC, Lahti J, Lemaitre RN, Manichaikul A, Keller M, Mikkila V, Ngwa J, van Rooij FJ, Ballentyne CM, Borecki IB, Cupples LA, Garcia M, Hofman A, Ferrucci L, Mozaffarian D, Perala MM, Raitakari O, Tracy RP, Arnett DK, Bandinelli S, Boerwinkle E, Eriksson JG, Franco OH, Kahonen M, Nalls M, Siscovick DS, Houston DK, Psaty BM, Viikari J, Witteman JC, Goodarzi MO, Lehtimaki T, Liu Y, Zillikens MC, Chen YD, Uitterlinden AG, Rotter JI, Fernandez-Hernando C, and Ordovas JM. Gain-of-function lipoprotein lipase variant rs13702 modulates lipid traits through disruption of a microRNA-410 seed site. Am J Hum Genet 92: 5-14, 2013. 45. Rottiers V, and Naar AM. MicroRNAs in metabolism and metabolic disorders. Nature reviews Molecular cell biology 13: 239-250, 2012. 46. Saxena S, Jonsson ZO, and Dutta A. Small RNAs with imperfect match to endogenous mRNA repress translation. Implications for off-target activity of small inhibitory RNA in mammalian cells. J Biol Chem 278: 44312-44319, 2003. 47. Shankar S, Kumar D, and Srivastava RK. Epigenetic modifications by dietary phytochemicals: implications for personalized nutrition. Pharmacology & therapeutics 138: 1-17, 2013. 15

445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484

48. Sun C, Alkhoury K, Wang YI, Foster GA, Radecke CE, Tam K, Edwards CM, Facciotti MT, Armstrong EJ, Knowlton AA, Newman JW, Passerini AG, and Simon SI. IRF-1 and miRNA126 modulate VCAM-1 expression in response to a high-fat meal. Circ Res 111: 1054-1064, 2012. 49. Tabuchi T, Satoh M, Itoh T, and Nakamura M. MicroRNA-34a regulates the longevityassociated protein SIRT1 in coronary artery disease: effect of statins on SIRT1 and microRNA-34a expression. Clinical science 123: 161-171, 2012. 50. Tricarico C, Pinzani P, Bianchi S, Paglierani M, Distante V, Pazzagli M, Bustin SA, and Orlando C. Quantitative real-time reverse transcription polymerase chain reaction: normalization to rRNA or single housekeeping genes is inappropriate for human tissue biopsies. Anal Biochem 309: 293300, 2002. 51. Turchinovich A, Weiz L, Langheinz A, and Burwinkel B. Characterization of extracellular circulating microRNA. Nucleic Acids Res 39: 7223-7233, 2011. 52. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, and Speleman F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 3: RESEARCH0034, 2002. 53. Varma SD, and Kovtun S. Protective effect of caffeine against high sugar-induced transcription of microRNAs and consequent gene silencing: a study using lenses of galactosemic mice. Molecular vision 19: 493-500, 2013. 54. Wahid F, Shehzad A, Khan T, and Kim YY. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta 1803: 1231-1243, 2010. 55. Wang J, Zhang Y, Zhang W, Jin Y, and Dai J. Association of perfluorooctanoic acid with HDL cholesterol and circulating miR-26b and miR-199-3p in workers of a fluorochemical plant and nearby residents. Environmental science & technology 46: 9274-9281, 2012. 56. Wang K, Yuan Y, Cho JH, McClarty S, Baxter D, and Galas DJ. Comparing the MicroRNA Spectrum between Serum and Plasma. PLoS One 7: e41561, 2012. 57. Wotschofsky Z, Meyer HA, Jung M, Fendler A, Wagner I, Stephan C, Busch J, Erbersdobler A, Disch AC, Mollenkopf HJ, and Jung K. Reference genes for the relative quantification of microRNAs in renal cell carcinomas and their metastases. Anal Biochem 417: 233-241, 2011. 58. Wylie D, Shelton J, Choudhary A, and Adai AT. A novel mean-centering method for normalizing microRNA expression from high-throughput RT-qPCR data. BMC research notes 4: 555, 2011. 59. Yang Q, Qiu C, Yang J, Wu Q, and Cui Q. miREnvironment database: providing a bridge for microRNAs, environmental factors and phenotypes. Bioinformatics 27: 3329-3330, 2011. 60. Yang YM, Seo SY, Kim TH, and Kim SG. Decrease of microRNA-122 causes hepatic insulin resistance by inducing protein tyrosine phosphatase 1B, which is reversed by licorice flavonoid. Hepatology 56: 2209-2220, 2012.

16

485

Table 1. Prior studies reporting microRNA expression associated with dyslipidemia. NON-HUMAN STUDIES – PRIOR REVIEWS MicroRNA

Cell/tissue source

Citation

miR-33a/b

Human (hsa) cell lines, mouse (mmu) models

Fernandez-Hernando, et.al. (7)

miR-27, miR-33a/b, miR-3a, miR-103/107, miR-122, miR-124, miR-143, miR-148, miR-182, miR-370, miR378/378*

Human (hsa) cell lines

Fernandez-Hernando, et.al. (8)

miR-let7, miR-9, miR-24, miR-26, miR-29, miR-33a/b, miR-3a, miR-103/107, miR-122, miR-208, miR-375

Mouse (mmu) models

miR-9, miR-96

Rat (rno) cell lines

miR-122 miR-370 miR-758 miR-106b

Human (hsa) cell lines, mouse (mmu) models Human (hsa) cell lines Human (hsa) cell lines, mouse (mmu) models Human (hsa) cell lines

Flowers et.al. (12)

miR-122

Human (hsa) cell lines, mouse (mmu) models

Najafi-Shoushtari, et.al. (33)

miR-370

Human (hsa) cell lines

miR-33

Human (hsa) cell lines, mouse (mmu) models

miR-33a/b

Human (hsa) cell l lines, mouse (mmu) models, primate (sae) models

Rayner, et.al. (38)

miR-33a/b

Human (hsa) cell lines, mouse (mmu) models, primate (sae) models

Rottiers et.al. (41)

miR-122

Mouse (mmu) models, primate (sae, ptr) models

Moore, et.al. (32)

miR-370

Human (hsa) cell lines

miR-378/378*

Human (hsa) cell lines

miR-33

Human (hsa) cell l lines, mouse (mmu) models HUMAN STUDIES Whole Blood 17

MicroRNA identified

Tissue source

Racial characteristics

Phenotype

Citation

Dyslipidemia: miR-103, miR-17, miR183, miR-197, miR-23a, miR-509-5p, miR-584, miR-652 BMI: miR-197, miR-23a, miR-509-5p

Whole blood

miR-17, miR-197, miR-509-5p, miR92a, miR-320a

Exosomes

Singaporean

Metabolic syndrome components

miR-100, miR-106b, miR-125b, miR143, miR-148a, miR-17/17*, miR-18a, miR-20a, miR-21, miR-221, miR374a, miR-7, miR-93, miR-96

Whole blood

South Asian

High triglycerides/low HDL

Flowers et.al.

Singaporean

Metabolic syndrome components Karolina et.al. (24)

Blood Components

486 487 488 489 490 491

Dyslipidemia: miR-103, miR-17, miR183, miR-197, miR-23a, miR-509-5p, miR-584, miR-652 BMI: miR-197, miR-23a, miR-509-5p

Exosomes

Singaporean

Metabolic syndrome components

Karolina et.al. (24)

miR-17, miR-197, miR-509-5p, miR92a, miR-320a

Blood plasma

Chinese

Coronary artery disease

Gao et.al. (15)

miR-34

Peripheral blood mononuclear cells

Japanese

Coronary artery disease

Tabuchi et.al. (43)

qPCR: quantitative polymerase chain reaction BMI: body mass index HDL: high-density lipoprotein cholesterol

18

Figure 1. Perturbations in microRNA expression associated with dyslipidemia and coronary artery disease on whole blood and blood components.

Legend CAD: coronary artery disease TC: total cholesterol LDL: low-density lipoprotein cholesterol HDL: high-density lipoprotein cholesterol PBMCs: peripheral blood mononuclear cells

MicroRNA associated with dyslipidemia and coronary disease in humans.

MicroRNAs are structural components of an epigenetic mechanism of posttranscriptional regulation of messenger RNA translation. Recently, there has bee...
230KB Sizes 0 Downloads 0 Views