the emerging roles of micrornas in cancer metabolism

8
1 2 Mini-review 4 Roles of microRNA in cancer metabolism 5 6 7 Brian Chan Q1 , Jacob Manley, Jae Lee, Shree Ram Singh 8 Basic Research Laboratory, National Cancer Institute, Frederick, MD 21702, USA 9 11 article info 12 Article history: 13 Received 9 October 2014 14 Accepted 9 October 2014 15 Available online xxxx 16 Keywords: 17 Cancer 18 Metabolism 19 MicroRNAs 20 Glycolysis 21 TCA cycle 22 Cancer therapy 23 24 abstract 25 The major task of cancer therapy to destroy cancer cells without harming normal cells. However, because 26 cancer cells have incredible heterogeneity and adaptability, it is difficult to target them therapeutically. 27 Metabolic reprogramming has emerged as a common feature of cancer. Ever since microRNAs (miRNAs) 28 have been found to influence metabolism, researchers have been trying to address the connection 29 between cancer cells and specific miRNAs. Many of the well-known miRNAs relate with crucial genes that 30 can impact metabolic pathways, both negatively and positively. With a better understanding of how 31 different pathways are affected, the roles of miRNAs will be more transparent, which could lead to 32 discovering new ideas about the concept of tumorigenesis and other cancer-related topics Q2 . 33 Published by Elsevier Ireland Ltd. 34 35 36 37 Introduction 38 MicroRNAs (miRNAs) are non-coding, 18–24 nucleotide-long 39 RNAs that attach to the 3 0 untranslated region (UTR) of a target 40 mRNA, resulting in altered gene expression [1,2]. The first miRNA 41 lin-4, and its target lin-14 were discovered in C. elegans [3,4]. Since 42 then a large number of miRNAs have been identified and conserved 43 from worms to mammals. miRNA biogenesis initiates with 44 transcription, followed by several processing steps to produce the 45 mature miRNA. miRNAs are originally transcribed by RNA pol II 46 in the nucleus to produce primary miRNA transcripts (pri-miRNA) 47 [5]. In the next step, a protein known as Pasha binds to Drosha, a 48 RNase III enzyme, to form a microprocessor complex [6–13]. This 49 complex is able to cleave the stem-loop portion of the pri-miRNA 50 to result in a precursor miRNA (pre-miRNA) sequence. The result- 51 ing sequence is then ready to be exported to the cytoplasm with 52 the help of exportin 5, which is a RanGTP dependent transporter 53 protein [14,15]. Once it is in the cytoplasm, the pre-miRNA is 54 converted to a miRNA: miRNA * duplex due to its interaction with 55 Dicer and TRBP [16–18]. The duplex unwinds at this point and 56 the miRNA * strand is degraded. Meanwhile, an Argonaute protein 57 is brought in and RISC assembly occurs so that the mature miRNA 58 can bind with a target mRNA [19]. Once it is bound, translational 59 repression or target mRNA cleavage can ensue, depending on the 60 complementarity of the strands. mRNAs are conserved across all 61 organisms and are involved in many physiological and pathological 62 processes including development, tissue homeostasis, cell prolifer- 63 ation, tissue growth, cell death, neurogenesis, metabolism, immu- 64 nity, cell fate determination, stem cells, aging, and cancer [20–30]. 65 Altered metabolism is a common feature of cancer. Normally, 66 non-cancerous cells catabolize glucose by oxidative phosphoryla- 67 tion in the mitochondria to produce ATP [31]. However, proliferat- 68 ing cancer cells metabolize significant amounts of glucose into 69 lactate, even in the presence of normal oxygen. These ideas stem 70 from an observation known as the ‘‘Warburg effect’’ [32]. The pref- 71 erence to utilize glycolysis offers several advantages to cancer cells 72 including adaptation in hypoxic and acidic environments (lactate 73 production), which help cancer cells in invasion, and rapid prolifer- 74 ation [33,34]. Oxidative stress and high levels of ROS (reaction 75 oxygen species) in cells play a crucial role in tumor formation. Can- 76 cer cells use pyruvate and nicotinamide adenine dinucleotide 77 phosphate (NADPH) to counter high oxidative stress [35]. 78 Since it was discovered that miRNAs are abnormally expressed 79 in cancer, accumulative studies show that miRNAs play important 80 roles in tumor growth by regulating oncogenes and tumor suppres- 81 sor genes [20,30,36,37]. In the last few years, a large number of 82 miRNAs have been identified to regulate cancer metabolism 83 [31,38–53]; Table 1. Carbohydrate, protein, lipid, and nucleic acid 84 metabolism are all affected by miRNAs that influence the growth 85 and production of cells. Many researchers seem to focus on 86 carbohydrate/glucose metabolism far more than other types of 87 metabolism (e.g. lipid) when it comes to cancer metabolism, but 88 it is beneficial to look at metabolism in a broader view since 89 miRNAs are involved in all these metabolic processes. Because of http://dx.doi.org/10.1016/j.canlet.2014.10.011 0304-3835/Published by Elsevier Ireland Ltd. Corresponding author at: Basic Research Laboratory, Stem cell and Animal Aging Section, National Cancer Institute, Frederick, MD 21702, USA. Tel.: +1 301 846 733. E-mail address: [email protected] (S.R. Singh). Cancer Letters xxx (2014) xxx–xxx Contents lists available at ScienceDirect Cancer Letters journal homepage: www.elsevier.com/locate/canlet CAN 12080 No. of Pages 8, Model 5G 23 October 2014 Please cite this article in press as: B. Chan et al., Roles of microRNA in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/ j.canlet.2014.10.011

Upload: shree-ram

Post on 02-Mar-2017

224 views

Category:

Documents


0 download

TRANSCRIPT

1

2

45

6

7 Q1

8

9

1 1

12131415

1617181920212223

2 4

Q2

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

Cancer Letters xxx (2014) xxx–xxx

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

Contents lists available at ScienceDirect

Cancer Letters

journal homepage: www.elsevier .com/locate /canlet

Mini-review

Roles of microRNA in cancer metabolism

http://dx.doi.org/10.1016/j.canlet.2014.10.0110304-3835/Published by Elsevier Ireland Ltd.

⇑ Corresponding author at: Basic Research Laboratory, Stem cell and AnimalAging Section, National Cancer Institute, Frederick, MD 21702, USA. Tel.: +1 301 846733.

E-mail address: [email protected] (S.R. Singh).

Please cite this article in press as: B. Chan et al., Roles of microRNA in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/1j.canlet.2014.10.011

Brian Chan, Jacob Manley, Jae Lee, Shree Ram Singh ⇑Basic Research Laboratory, National Cancer Institute, Frederick, MD 21702, USA

a r t i c l e i n f o a b s t r a c t

25262728293031323334

Article history:Received 9 October 2014Accepted 9 October 2014Available online xxxx

Keywords:CancerMetabolismMicroRNAsGlycolysisTCA cycleCancer therapy

The major task of cancer therapy to destroy cancer cells without harming normal cells. However, becausecancer cells have incredible heterogeneity and adaptability, it is difficult to target them therapeutically.Metabolic reprogramming has emerged as a common feature of cancer. Ever since microRNAs (miRNAs)have been found to influence metabolism, researchers have been trying to address the connectionbetween cancer cells and specific miRNAs. Many of the well-known miRNAs relate with crucial genes thatcan impact metabolic pathways, both negatively and positively. With a better understanding of howdifferent pathways are affected, the roles of miRNAs will be more transparent, which could lead todiscovering new ideas about the concept of tumorigenesis and other cancer-related topics.

Published by Elsevier Ireland Ltd.

35

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

Introduction

MicroRNAs (miRNAs) are non-coding, 18–24 nucleotide-longRNAs that attach to the 30 untranslated region (UTR) of a targetmRNA, resulting in altered gene expression [1,2]. The first miRNAlin-4, and its target lin-14 were discovered in C. elegans [3,4]. Sincethen a large number of miRNAs have been identified and conservedfrom worms to mammals. miRNA biogenesis initiates withtranscription, followed by several processing steps to produce themature miRNA. miRNAs are originally transcribed by RNA pol IIin the nucleus to produce primary miRNA transcripts (pri-miRNA)[5]. In the next step, a protein known as Pasha binds to Drosha, aRNase III enzyme, to form a microprocessor complex [6–13]. Thiscomplex is able to cleave the stem-loop portion of the pri-miRNAto result in a precursor miRNA (pre-miRNA) sequence. The result-ing sequence is then ready to be exported to the cytoplasm withthe help of exportin 5, which is a RanGTP dependent transporterprotein [14,15]. Once it is in the cytoplasm, the pre-miRNA isconverted to a miRNA: miRNA* duplex due to its interaction withDicer and TRBP [16–18]. The duplex unwinds at this point andthe miRNA* strand is degraded. Meanwhile, an Argonaute proteinis brought in and RISC assembly occurs so that the mature miRNAcan bind with a target mRNA [19]. Once it is bound, translationalrepression or target mRNA cleavage can ensue, depending on thecomplementarity of the strands. mRNAs are conserved across all

86

87

88

89

organisms and are involved in many physiological and pathologicalprocesses including development, tissue homeostasis, cell prolifer-ation, tissue growth, cell death, neurogenesis, metabolism, immu-nity, cell fate determination, stem cells, aging, and cancer [20–30].

Altered metabolism is a common feature of cancer. Normally,non-cancerous cells catabolize glucose by oxidative phosphoryla-tion in the mitochondria to produce ATP [31]. However, proliferat-ing cancer cells metabolize significant amounts of glucose intolactate, even in the presence of normal oxygen. These ideas stemfrom an observation known as the ‘‘Warburg effect’’ [32]. The pref-erence to utilize glycolysis offers several advantages to cancer cellsincluding adaptation in hypoxic and acidic environments (lactateproduction), which help cancer cells in invasion, and rapid prolifer-ation [33,34]. Oxidative stress and high levels of ROS (reactionoxygen species) in cells play a crucial role in tumor formation. Can-cer cells use pyruvate and nicotinamide adenine dinucleotidephosphate (NADPH) to counter high oxidative stress [35].

Since it was discovered that miRNAs are abnormally expressedin cancer, accumulative studies show that miRNAs play importantroles in tumor growth by regulating oncogenes and tumor suppres-sor genes [20,30,36,37]. In the last few years, a large number ofmiRNAs have been identified to regulate cancer metabolism[31,38–53]; Table 1. Carbohydrate, protein, lipid, and nucleic acidmetabolism are all affected by miRNAs that influence the growthand production of cells. Many researchers seem to focus oncarbohydrate/glucose metabolism far more than other types ofmetabolism (e.g. lipid) when it comes to cancer metabolism, butit is beneficial to look at metabolism in a broader view sincemiRNAs are involved in all these metabolic processes. Because of

0.1016/

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

2 B. Chan et al. / Cancer Letters xxx (2014) xxx–xxx

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

the importance of metabolism in cancer, studying the role of miR-NAs is crucial in understanding their effect on metabolic pathways,and their ultimate effect on cancer cells. Here we present a briefoverview on the involvement of miRNAs in cancer metabolismand their targets in cancer therapy.

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204205

206

207

208

209

210

211

212

213

214

miRNAs in cancer metabolism

miRNAs in glucose metabolism

As seen with the Warburg effect, alterations in glucose metab-olism can result in the proliferation of cancer cells. One reasonfor this effect can be attributed to specific modifications from miR-NAs in the glycolysis pathway. There are a large number of miRNAsreported to regulate gene transcription and expression of glucosetransporters (GLUTs). Of the 14 members of the GLUT familyreported, only 11 of them facilitate glucose transport. However,only three of the GLUTs (GLUT1, GLUT2, and GLUT3) are highlyexpressed in most cancer tissues [54]. Deregulation of GLUTsresults in high glucose uptake, accelerated metabolism, andincreased glucose requirements by tumorous tissues. miRNAs canpossibly control the glucose uptake by regulating the expressionof some GLUTs in cancer tissues. It has been reported that mir-195-5p directly regulates the expression of GLUT3 in bladder can-cer [55]. In addition, Yang et al. [56] also reported that miR-21 reg-ulates the expression of GLUT1 and GLUT3 in bladder cancer.Further, a study involving renal cell carcinoma showed that miR-1291 regulates the GLUT1 expression [57].

Accumulative studies suggest that miRNAs control severalessential enzymes of glycolysis including hexokinases (HKs), glyc-eraldehyde-3-phosphate dehydrogenase (GAPDH), and 6-phosp-hofructo-1-kinase (PFK1). HKs catalyze the first reversible step ofglucose metabolism and exist in four isoforms HK1 through HK4.Among them is HK2, which is frequently overexpressed in cancerand plays an important role in tumor growth, survival, and metas-tasis by repressing mitochondrial function [58]. It has been demon-strated that miR-143 down-regulates HK2 and inhibits glucosemetabolism in head and neck squamous cell carcinoma (HNSCC)[59], breast cancer [60], lung cancer [61], and colon cancer [62].In addition, Zhao et al. [63] reported that miR-143 is downregu-lated in human and rat glioblastoma and the overexpression ofmiR-143 inhibits glycolysis by downregulating HK2. Jiang et al.[64] have shown that miR-155 promotes HK2 transcription bythe activation of signal transducer and activator of transcription3 (STAT3), and represses miR-143 by targeting c/EBPb. Further,miR-125a/b reported to regulate HK2 in lung cancer [61], miR-125b in hepatocellular carcinoma cells [64], and in chronic lym-phocytic leukemia [65]. Furthermore, p53-inducible miR-34arepressed HK1 and HK2 in lung and colon cancer [66]. In addition,miR-34a and miR-21 regulate the expression of HK1 and HK2 incolon cancer [66], and bladder cancer [56].

In addition to HKs, other components in glycolysis are regulatedby miRNAs as well. Aldolase A (Aldo A), a glycolytic enzyme, whichcatalyzes the reversible conversion of fructose-1,6-bisphosphate toglyceraldehyde 3-phosphate and dihydroxyacetone phosphate, isshown to be a target of miR-122 in liver cancer [67] and miR-15a/16-1 inhibit the expression of Aldo A, aldehyde dehydrogenase6 family, member A1 (Aldo6A1) and triosephosphate isomerase 1(TPI1) in leukemia [68]. Further, TPI1 is also reported to beregulated by miR-195 in bladder cancer [69]. Further, it has beenreported that Glucose-6-phosphate isomerase (PGI), an enzy-me that catalyzes the conversion of glucose-6-phosphate into fruc-tose 6-phosphate in the second step of glycolysis, regulates theexpression of miR-200 in breast cancer cells [53]. In addition, astudy demonstrated that miR-29a regulates the expression of

Please cite this article in press as: B. Chan et al., Roles of microRNAj.canlet.2014.10.011

Phosphoglucomutase-1 (PGM1), Enolase 1 (ENO1) and Phospho-glycerate kinase 1 (PGK1) in lung cancer cell lines [70]. In the lastfew years, pyruvate kinase muscle isozyme (PKM2) has emerged asa major player in cancer development because it gives metabolicadvantage to cancer cells to utilize phosphor-metabolitesupstream of pyruvate as precursors for the synthesis of lipids,amino acids and nucleic acids [41,71,72]. There are several miRNAsregulating the expression of PKM1 and PKM2 such as miR-99, miR-133a and 133b, miR-326, miR-124, miR-137, miR-340, miR-122and miR-21 [56,72–76]; see Table 1. During aerobic glycolysis,most of the pyruvate is converted to lactate in the cytoplasm byenzyme lactate dehydrogenase (LDH). Lactate is then secreted out-side of the cells by monocarboxylate transporter 1 (MCT1), whichin humans is encoded by the SLC16A1 gene. Li et al. [77] demon-strated that miR-124 is a direct regulator of SLC16A1 in medullo-blastoma cells. Further, miR-375 is involved in regulation oflactate dehydrogenase B (LDHB), an enzyme that catalyzes thereversible conversion of lactate and pyruvate, and NAD and NADH,in the glycolytic pathway, in maxillary sinus squamous cell carci-noma [78].

miRNAs in TCA cycle, ROS and glutamine metabolism

Like glycolysis, the tricarboxylic acid (TCA) cycle has also seen avariety of miRNAs that target the steps in this pathway. Cancercells mostly utilize aerobic glycolysis (lactate production) thanthe TCA cycle. These changes in metabolism allow cancer cells toproduce crucial biosynthesis materials by maintaining high ATPlevels. It was predicted that miR-103 and miR-107 controlsacetyl-coA and lipid levels by upregulating the pantothenatekinase (PanK), the first enzyme in the Coenzyme A biosyntheticpathway that phosphorylates pantothenate to form 40-phospho-pantothenate [79]. There are several other miRNAs that are knownto downregulate or upregulate the expression of metabolic associ-ated genes. miR-183 regulates isocitrate dehydrogenase 1 (IDH2)in glioma cells [80]. miR-210 regulates the TCA cycle by targetingiron-sulfur cluster assembly proteins (ISCU1/2), cytochrome c oxi-dase10 (COX10), succinate dehydrogenase, subunit D (SDHD), ADHdehydrogenase (ubiquinone) 1 alpha subcomplex, 4 (NDUFA4) inseveral cancers [81–84], miR-1 and miR-206 regulates glycerol-3-phosphate dehydrogenase 2 (GPD2) [48], and miR-378* also reg-ulates the TCA cycle in breast cancer [85].

Production of ROS is a crucial component of cellular metabolismthat is generated by mitochondria and NADPH oxidases (NOX)[86]. A recent study demonstrates that miR-17* can suppresstumorigenicity in prostate cancer by inhibiting the importantmitochondrial antioxidant enzymes such as manganese superoxidedismutase (MnSOD), glutathione peroxidase-2 (GPX2), and thiore-doxin reductase-2 (TrxR2) [87]. Studies suggest that ROS levels arealso regulated by several other miRNAs such as miR-17-92, miR-126, miR-128a, miR-141, miR-200, miR-21, miR-34a, miR-210,miR-122, miR-335, and miR-320 [82,88–92,43,93].

Glutaminase is crucial in mitochondrial metabolism as it con-verts glutamine to glutamate, which further converts to producea-ketoglutarate. Glutaminolysis is also controlled by several miR-NAs [94–96]. Further, studies suggest that p53, which is a directtarget of miR-125b, miR-30, miR-504, plays an essential role insustaining glutamine levels by activating GLS2 [97]. In addition,it has been demonstrated that Myc downregulates mir-23a and23b in lymphoma and prostate cancer cells and increases gutamin-ase expression [96]. Since autophagy plays a crucial role in cancercell survival when there is decreased oxygen, insufficient nutrients,and metabolic stress, it has been demonstrated that miR-23b cansuppress autophagy by regulating ATG12 in pancreatic cancer cells[98].

in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/

Table 1MicroRNAs, targets, and regulation of cancer metabolism.

microRNAs Target genes Metabolic activity/Pathway Phenotypes/cancer types References

miR-210 ISCU Hypoxia, Krebs cycle,glycolysis

Breast, colon, head and neckcancer

Favaro et al. [81]

ISCU, COX10 Hypoxia, ROS, glycolysis Colon, breast, andesophageal cancer

Chen et al. [82]

SDHD, NDUFA4, ISCU1/2 HIF-1a, ETC Non-small cell lung cancer Puissegur et al. [83]ISCU1/2 Hypoxia, ROS, TCA cycle,

ETCHuman colorectaladenocarcinoma, breastcancer, promyelocyticLeukemia cell lines

Chan et al. [84]

miR-122 SMARCD1, MAP3K3, CAT-1 Mitochondrial metabolism,Krebscycle

Hepatocellular carcinoma Burchard et al. [89]

PKM2 Glycolysis Hepatocellular carcinoma Liu et al. [73]AldoA, TPI1 Glycolysis Leukemia Calin et al. [68]Agpat1, Cidec, Stard4 Lipid metabolism Liver cancer Hsu et al. [103]

Tsai et al. [104]

miR-155/miR-143 HK2 Aerobic glycolysis Breast cancer Jiang et al. [60]

miR-143 HK2 Glycolysis, Glucosemetabolism

Human lung cancer Fang et al. [61]Human and ratglioblastoma,

Zhao et al. [63]

colon cancer cell line, Gregersen et al [62]head and neck squamouscell carcinoma

Peschiaroli et al. [59]

miR-23a/miR-23b GLS2 c-Myc, glutaminemetabolism,gluconeogenesis, STAT3

Lymphoma and prostatecancer cells, hepatocellularcarcinoma

Gao et al. [94]PGC-1a, G6PC Wang et al. [95]

miR-23b ATG12 Autophagy Pancreatic cancer cells Wang et al. [98]mir-23b* POX/PRODH c-Myc, proline and

glutamine metabolismLymphoma and prostatecancer

Liu et al. [96]

miR-26a PDHX Glucose metabolism Colorectal cancer cells Chen et al. [29]

miR-34a HK1, HK2, GP1, PDK1 P53, glucose metabolism Human Non-small cell lungand colon cancer cell lines

Kim et al. [66]

NOX2 ROS Human glioma cell line Li et al. [93]

miR-21 SOD3 and TNFa ROS Human bronchial epithelialcell lines

Zhang et al. [91]

GLUT1, GLUT3, LDHA,LDHB, HK1, HK2, PKM,HIF-1a

PTEN/PI3K/AKT/mTOR pathways,aerobic glycolysis

Bladder cancer Yang et al. [56]

miR-326 PKM2 AMPK Glioblastoma Kefas et al. [75]miR-126 IRS1 Mitochondrial energy

metabolism, AKT signalingMesothelioma Tomasetti et al. [43]

miR-451 CAB39 LKB1/AMPK Glioblastoma Godlewski et al. [110]miR-185, miR-342 SREBP-1, SREBP-2, FASN,

HMGCRSREBP-lipogenesis-cholesterogenesis

Prostate cancer cells Li et al. [76]

miR-320 PFKm Glycolysis Human lungadenocarcinoma

Tang et al. [47]

miR-1, miR-206 G6PD, TKT, 6PGD, GPD2 Carbon flux toward the PPPand the TCA cycle,reprogramming glucosemetabolism

Human lung carcinoma andprostate cancer cell lines

Singh et al. [48]

miR-378* ESRRG, GABPA, ERRc Shift from aerobic, oxidativemetabolism (OXPHOS) toglycolytic metabolism

Human breast cancer Eichner et al. [85]

miR-124, miR-137, miR-340 PKM alternative splicing Inhibit the glycolysis rate,elevate the glucose flux intooxidative phosphorylation(glucose metabolism)

Colorectal cancer Sun et al. [72]Proteins (PTB1, hnRNAPA1,hnRNAPA2)

miR-124 SLC16A1 Eflux lactic acid duringaerobic glycolysis

Medulloblastoma Li et al. [77]

PKM1, HNF4a Glucose metabolism Colorectal cancer cells Sun et al. [125]miR-125 HK2 Glucose metabolism Human hepatocellular

carcinoma cellsJiang et al. [64]

miR-125b PCTP, LIPA, GSS, ACSS1, HK2,SCD1, AKT2, PDK1

Glucose, glutathione, lipid,and glycerolipidmetabolism

chronic lymphocyticleukemias

Tili et al. [65]

miR-181a PTEN Glycolysis, PTEN/AKTpathway

Colon cancer cells Wei et al. [49]

miR-183 IDH2 TCA cycle Glioma cells Tanaka et al. [80]miR-520 PFKP Glycolysis Hepatocellular carcinoma Park et al. [133]miR-205 ACSL1 Lipid metabolism Hepatocellular carcinoma Cui et al. [102]miR-1291 NMN, NNMT Nicotinamide metabolism Pancreatic cancer Bi et al. [45]

SLC2A1/GLUT1 Gucose metabolism Renal cell carcinoma Yamasaki et al. [57]

(continued on next page)

B. Chan et al. / Cancer Letters xxx (2014) xxx–xxx 3

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

Please cite this article in press as: B. Chan et al., Roles of microRNA in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/j.canlet.2014.10.011

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

Table 1 (continued)

microRNAs Target genes Metabolic activity/Pathway Phenotypes/cancer types References

miR-195-5p GLUT3 Glucose uptake (glucosemetabolism)

Human bladder cancer Fei et al. [55]

miR-106a SLC2A3 Glucose uptake (glucosemetabolism)

Gioblastoma Dai et al. [51]

miR-17* MnSOD, GPX2, TrxR2 Mitochondrial antioxidant Prostate cancer Xu et al. [87]miR-17-3p LDH-A Enhance glycolysis pathway

and inhibit mitochondrialmetabolism

Prostate cancer Velez and Xu [52]

miR-17-92 E2F1 ROS Lung cancer Ebi et al. [88]miR-200 PGI Glycolysis Breast cancer Ahmad et al. [53]miR-133a, miR-133b PKM2 Glycolysis Tongue squamous cell

carcinomaWong et al. [74]

miR-375 LDHB Anaerobic glycolysis Maxillary sinus squamouscell carcinoma

Kinoshita et al. [78]

4 B. Chan et al. / Cancer Letters xxx (2014) xxx–xxx

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

miRNAs in lipid and amino acid metabolism

Lipids are crucial biosynthesis materials for organelles and cells.Altered lipid metabolism is a unique feature of cancer metabolism.Several miRNAs are involved in regulating lipid metabolism incancer [98–102]. miR-122 was found to regulate fat and choles-terol metabolism by controlling the expression of 1-acylglycerol-3-phosphate O-acyltransferase 1 (Agpat1), cell death-inducingDFFA-like effector c (Cidec) and StAR-related lipid transfer protein4 (Stard4) in live cancer [103,104]. It has been demonstrated thatmiR-185 and miR-342 control lipogenesis and cholesterogenesisin prostate cancer cells by reducing the expression of SREBP-1and 2 and down-regulating their targeted genes, including fattyacid synthase (FASN) and 3-hydroxy-3-methylglutaryl CoA reduc-tase (HMGCR) [101]. Further, Tili et al. [100] reported that meta-bolic enzymes such as phosphatidylcholine transfer protein(PCTP), lipase A (LIPA), glutathione synthetase (GSS), argininosuc-cinate synthase 1 (ACSS1), HK2, stearoyl-CoA desaturase-1(SCD1), AKT2, and pyruvate dehydrogenase lipoamide kinase iso-zyme 1 (PDK1), are potential targets of miR-125b and miR-125b.Recently, it has been demonstrated that miR-205 deregulates lipidmetabolism in hepatocellular carcinoma by targeting ACSL1, a lipidmetabolism enzyme in liver [102]. miRNAs have also reported toregulate amino acid catabolism. Liu et al. [96] reported thatmiR-32b* regulates the proline oxidase in kidney cancer. Thesestudies indicate that miRNAs alter the lipid and amino acid metab-olism in cancer.

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

Cancer metabolism: microRNAs and signaling pathways

Dysregulation of signaling pathways and altered expression ofmiRNAs results in defective cell metabolism and carcinogenesis[39] Fig. 1. These pathways include p53, c-Myc, Ras, AMPK, AKTand HIFs. p53 is one of the well-known tumor suppressor genesthat regulate cellular metabolism by modulating glycolysis andoxidative phosphorylation. It has been demonstrated that p53 con-trols the glycolysis by inhibiting the expression of GLUT1, GLUT4,phosphoglycerate mutase (PGM) and TP53-inducible glycolysisand apoptosis regulator (TIGAR), and enhances the expression ofcytochrome c oxidase, SCO2, and p53R2 [31,39,105]. p53 regulatesseveral miRNAs to control metabolism in cancer. Some otherstudies reported that p53 controls the expression of miR-34, let-7, miR-107, which further inhibits their target genes’ expressionsuch as lactate dehydrogenase A (LDHA), MYC, sirtuin-1 (SIRT1),hypoxia inducible factor 1 (HIF1) [66,21,106].p53 also controlsthe expression of c-Myc, a proto-oncogene that is known beinvolved in cancer metabolism. p53 regulate c-Myc expression bymiR-145 [107]. c-Myc targets LDHA as well as GLUT1, HK2, PFKM,and ENO1 expression [108]. These genes are essential for the

Please cite this article in press as: B. Chan et al., Roles of microRNAj.canlet.2014.10.011

proper function of glycolysis and oxidative phosphorylation aswell. c-Myc regulates the expression of several miRNAs includingmiR-17-92, miR-23a/b and miR-23* [94,96,109].

In addition to p53 and c-Myc, other pathways such as AMP-activated protein kinase (AMPK), AKT, RAS, and hypoxia-induciblefactors (HIFs) are interact with several miRNAs and regulate metab-olism in cancer. AMPK is activated during metabolic stress toincrease energy conservation and glucose uptake. This allows cellsto survive when the energy supply is low. It has been shown thatmiR-451 controls the AMPK signaling in glioma cells [110].Recently, it has been demonstrated that miR-195 and miR-451 tar-get calcium binding protein 9 (CAB39) and regulate LKB1/AMPKsignaling [39]. Kefas et al. [75] reported that PKM2 is a target ofmiR-326 and that knocking down PKM2 decreased ATP and gluta-thione levels and resulted in activation of AMPK. The AKT pathwayis generally activated in human cancers, which is a major regulatorof apoptosis. AKT is downstream of PI3 K and elevates glycolysis byenhancing the expression of glucose transporters and phosphory-lating hexokinase. It has been shown that with the continuouselevation of the PI3/AKT/mTOR pathway in cancer, the pathwaycan control metabolism possibly by HIF-1a induction undernormoxia [38,41]. There are several miRNAs that are known to reg-ulate the PI3/AKT/mTOR pathway in cancer [41,56,111–114]. TheHIFs are crucial for the maintenance of cellular oxygen homeosta-sis and hypoxia adaptation when oxygen supplies are too low forthe cell. Hypoxia and HIFs are known to regulate different pro-cesses of metabolism in cancer cells [115]. There are severalreports that show the association between miRNAs and hypoxiain regulating cancer metabolism including miR-210 [81–84].Further, Kras also known to regulate cancer metabolism becauseit abrupt the mitochondrial function by inactivating oxidativephosphorylation. Kras also enhance tumor growth by suppressingpyruvate flux through TCA cycle and stimulates glutamine metab-olism [31,116,117]. In recent years, there have been several miR-NAs reported to be associated with Kras in regulating cancermetabolism [118,119].

miRNAs, metabolism and metastatic microenvironment

The proper condition of tumor microenvironment is vital totumor growth and development. The tumor microenvironment iscollection of cancer-associated fibroblasts (CAF), inflammatorycells, endothelial cells (ECs), extracellular matrix, and cytokines[120–123]. In addition to the genes and pathways that influencethe tumor cell metabolism, unusual tumor microenvironmentsplay crucial parts in deciding the fate of tumor cells. Tumors residein a very heterogeneous environments and mutual communicationbetween the cancer cells and tumor stromal cells (tumor microen-vironment) is crucial in determining whether a tumor cell is in a

in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/

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

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367Q3368369370371372373374375376377378379380381382383384385386387388389390391392393394395396397398399

Metabolic reprogramming in

cancer cellsOncogenes

cMyc, HIF1α, Ras, PI3K/AKT

Tumor supressorsp53, LKB1/AMPK

Tumorigenesis

Oncogenic miRNAs

Tumor suppressor

miRNAs

Fig. 1. Metabolic regulation by the tumor suppressors and oncogenes, and involvement of microRNAs in enhancing or suppressing tumorigenesis.

B. Chan et al. / Cancer Letters xxx (2014) xxx–xxx 5

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

quiescent state or whether it should become invasive and result inmetastatic cancer. miRNAs play critical roles in the activation oftumor stromal cells, from their quiescent state to cancer angiogen-esis [124]. Several microRNAs regulate cancer angiogenesis, CAFs,ECs, and tumor microenvironment [121,127]. miR-29 emerged asa master orchestrator of the tumor microenvironment (Chouet al., 2013). It has been reported that the metabolic interactionbetween mitochondria in cancer cells and catabolism in CAFsenhance tumor growth and metastasis [126]. Further, it has beensuggested that miRNAs establish a novel communication betweencancer and stromal cells through microvasicles to facilitate angio-genesis and tumor growth [127].

microRNAs as a target in cancer therapy

Metabolic reprogramming plays an essential step in tumordevelopment and metastasis. Targeting cancer metabolism holdsgreat promise in developing anti-cancer therapy. There are manytypes of compounds that have been developed to specificallyinhibit the metabolic enzymes, which are crucial in tumor develop-ment [128]. However, some of the cells have similar metabolicprogramming as cancer cells such as T lymphocytes, which use glu-tamine metabolism [128]. Studies show that miRNA deregulationdoes not affect the growth of normal cells, but it does reduce thegrowth of cancer cells, which suggest that miRNAs can be thera-peutic targets in cancer [41,129]. In addition, tumor suppressorp53 induced several miRNAs, such as miR-34, let-7, miR-107, andmiR-200, which function as regulators of LDHA, Myc and SIRT1[130]. These suggest that p53/miR-34 is the major metabolic regu-lator in tumor suppression and that targeting LDHA, Myc and SIRT1or other metabolic pathways or enzymes would be an innovativestrategy in cancer treatment [130]. Further, it has been reportedthat metformin is effective in reprogramming the metabolism incancer cells by directly modulating the metabolic genes, and miR-NAs [131]. miRNAs can be used both as targets and tools in anti-cancer therapy, however, development of engineered animal mod-els to examine cancer-associated miRNAs and the advancement ofthe effectiveness of microRNAs/anti-miRs in vivo delivery is still amajor hurdle in clinics [132]. Some recent studies, however, dem-onstrated that systematic administration of microRNAs reducesdifferent types of cancer growth in vivo without cell toxicity[41,104,133–135].

Conclusion

miRNAs are crucial regulators of cancer metabolism. miRNAscontrol the cancer metabolic reprogramming by regulating theexpression of genes/pathways/enzymes involved in glycolysis

Please cite this article in press as: B. Chan et al., Roles of microRNAj.canlet.2014.10.011

and energy metabolism. Although there are many roles of miRNAsthat are unknown at this moment, it is important to study them tounderstand how tumor cells form. Further, the specific miRNAsthat regulate glycolysis or mitochondrial metabolism in cancerwould be promising therapeutic targets in cancers. However,improving in vivo delivery of these microRNAs is crucial in exploit-ing the full potential of miRNAs. It is hopeful that scientists can usethe knowledge gained from miRNAs to develop cancer treatmentsthat will target areas or pathways of metabolic alteration.

Conflicts of Interest

The authors declare no conflict of interest.

Acknowledgement

This research was supported by the Intramural Research Pro-gram of the National Cancer Institute, National Institutes of Health.

References

[1] S. Vasudevan, Y. Tong, J.A. Steitz, Switching from repression to activation:microRNAs can up-regulate translation, Science 318 (5858) (2007) 1931–1934.

[2] A.S. Flynt, E.C. Lai, Biological principles of microRNA-mediated regulation:shared themes amid diversity, Nat. Rev. Genet. 9 (11) (2008) 831–842.

[3] R.C. Lee, R.L. Feinbaum, V. Ambros, The C. elegans heterochronic gene lin-4encodes small RNAs with antisense complementarity to lin-14, Cell 75 (1993)843–854.

[4] B. Wightman, I. Ha, G. Ruvkun, Posttranscriptional regulation of theheterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C.elegans, Cell 75 (1993) 855–862.

[5] Y. Lee, M. Kim, J. Han, K.H. Yeom, S. Lee, S.H. Baek, V.N. Kim, MicroRNA genesare transcribed by RNA polymerase II, EMBO J. 23 (20) (2004) 4051–4060.

[6] Y. Lee, C. Ahn, J. Han, H. Choi, J. Kim, J. Yim, J. Lee, P. Provost, O. Rådmark, S.Kim, V.N. Kim, The nuclear RNase III Drosha initiates microRNA processing,Nature 425 (6956) (2003) 415–419.

[7] A.M. Denli, B.B. Tops, R.H. Plasterk, R.F. Ketting, G.J. Hannon, Processing ofprimary microRNAs by the Microprocessor complex, Nature 432 (7014)(2004) 231–235.

[8] R.I. Gregory, K.P. Yan, G. Amuthan, T. Chendrimada, B. Doratotaj, N. Cooch, R.Shiekhattar, The Microprocessor complex mediates the genesis of microRNAs,Nature 432 (7014) (2004) 235–240.

[9] M. Landthaler, A. Yalcin, T. Tuschl, The human DiGeorge syndrome criticalregion gene 8 and Its D. melanogaster homolog are required for miRNAbiogenesis, Curr. Biol. 14 (23) (2004) 2162–2167.

[10] M.A. Carmell, G.J. Hannon, RNase III enzymes and the initiation of genesilencing, Nat. Struct. Mol. Biol. 11 (3) (2004) 214–218.

[11] H. Zhang, F.A. Kolb, L. Jaskiewicz, E. Westhof, W. Filipowicz, Single processingcenter models for human Dicer and bacterial RNase III, Cell 118 (1) (2004)57–68.

[12] J. Han, Y. Lee, K.H. Yeom, J.W. Nam, I. Heo, J.K. Rhee, S.Y. Sohn, Y. Cho, B.T.Zhang, V.N. Kim, Molecular basis for the recognition of primary microRNAs bythe Drosha-DGCR8 complex, Cell 125 (5) (2006) 887–901.

in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/

400401402403404405406407408409410411412413414415416417418419420421422423424425426427428429430431432433434435436437438439440441442443444445446447448449450451452453454455456457458459460461462463464465466467468469470471472473474475476477478479480481482483484

485486487488489490491492493494495496497498499500501502503504505506507508509510511512513514515516517518519520521522523524525526527528529530531532533534535536537538539540541542543544545546547548549550551552553554555556557558559560561562563564565566567568569

6 B. Chan et al. / Cancer Letters xxx (2014) xxx–xxx

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

[13] R. Martin, P. Smibert, A. Yalcin, D.M. Tyler, U. Schäfer, T. Tuschl, E.C. Lai,Drosophila pasha mutant distinguishes the canonical microRNA and mirtronpathways, Mol. Cell. Biol. 29 (3) (2009) 861–870.

[14] R. Yi, Y. Qin, I.G. Macara, B.R. Cullen, Exportin-5 mediates the nuclear exportof pre-microRNAs and short hairpin RNAs, Genes Dev. 17 (24) (2003) 3011–3016.

[15] M.T. Bohnsack, K. Czaplinski, D. Gorlich, Exportin 5 is a RanGTP-dependentdsRNA-binding protein that mediates nuclear export of pre-miRNAs, RNA 10(2) (2004) 185–191.

[16] G. Hutvágner, J. McLachlan, A.E. Pasquinelli, E. Bálint, T. Tuschl, P.D. Zamore, Acellular function for the RNA-interference enzyme Dicer in the maturation ofthe let-7 small temporal RNA, Science 293 (5531) (2001) 834–838.

[17] T.P. Chendrimada, R.I. Gregory, E. Kumaraswamy, J. Norman, N. Cooch, K.Nishikura, R. Shiekhattar, TRBP recruits the Dicer complex to Ago2 formicroRNA processing and gene silencing, Nature 436 (7051) (2005) 740–744.

[18] K. Saito, A. Ishizuka, H. Siomi, M.C. Siomi, Processing of pre-microRNAs by theDicer-1-Loquacious complex in Drosophila cells, PLoS Biol. 3 (7) (2005) e235.

[19] K. Okamura, A. Ishizuka, H. Siomi, M.C. Siomi, Distinct roles for Argonauteproteins in small RNA-directed RNA cleavage pathways, Genes Dev. 18 (14)(2004) 1655–1666.

[20] A. Lujambio, S.W. Lowe, The microcosmos of cancer, Nature 482 (7385)(2012) 347–355.

[21] H. Hermeking, MicroRNAs in the p53 network: micromanagement of tumoursuppression, Nat. Rev. Cancer 12 (9) (2012) 613–626.

[22] H.S. Seol, Y. Akiyama, S. Shimada, H.J. Lee, T.I. Kim, S.M. Chun, S.R. Singh, S.J.Jang, Epigenetic silencing of microRNA-373 to epithelial-mesenchymaltransition in non-small cell lung cancer through IRAK2 and LAMP1 axes,Cancer Lett. 353 (2) (2014) 232–241.

[23] X. Gao, W. Jin, The emerging role of tumor-suppressive microRNA-218 intargeting glioblastoma stemness, Cancer Lett. 353 (1) (2014) 25–31.

[24] Y. Su, X. Li, W. Ji, B. Sun, C. Xu, Z. Li, G. Qian, C. Su, Small molecule with bigrole: MicroRNAs in cancer metastatic microenvironments, Cancer Lett. 344(2) (2014) 147–156.

[25] R.M. O’Connell, D.S. Rao, A.A. Chaudhuri, D. Baltimore, Physiological andpathological roles for microRNAs in the immune system, Nat. Rev. Immunol.10 (2) (2010) 111–122.

[26] J. Shen, S.A. Stass, F. Jiang, MicroRNAs as potential biomarkers in human solidtumors, Cancer Lett. 329 (2) (2013) 125–136.

[27] O. Dumortier, C. Hinault, E. Van Obberghen, MicroRNAs and metabolismcrosstalk in energy homeostasis, Cell Metab. 18 (3) (2013) 312–324.

[28] S.A. Ross, C.D. Davis, The emerging role of microRNAs and nutrition inmodulating health and disease, Annu. Rev. Nutr. 34 (2014) 305–336.

[29] S. Chen, Y. Xue, X. Wu, C. Le, A. Bhutkar, E.L. Bell, F. Zhang, R. Langer, P.A.Sharp, Global microRNA depletion suppresses tumor angiogenesis, GenesDev. 28 (10) (2014) 1054–1067.

[30] S.R. Singh, P. Rameshwar, MicroRNA in Development and in the Progressionof Cancer, Springer, New York, 2014.

[31] P.K. Singh, R.E. Brand, K. Mehla, MicroRNAs in pancreatic cancer metabolism,Nat. Rev. Gastroenterol. Hepatol. 9 (6) (2012) 334–344.

[32] O. Warburg, On the origin of cancer cells, Science 123 (3191) (1956) 309–314.[33] J. Singleterry, A. Sreedhar, Y. Zhao, Components of cancer metabolism and

therapeutic interventions, Mitochondrion 17 (2014) 50–55.[34] P. Swietach, R.D. Vaughan-Jones, A.L. Harris, Regulation of tumor pH and the

role of carbonic anhydrase 9, Cancer Metastasis Rev. 26 (2) (2007) 299–310.[35] K.A. Nath, E.O. Ngo, R.P. Hebbel, A.J. Croatt, B. Zhou, L.M. Nutter, Alpha-

Ketoacids scavenge H2O2 in vitro and in vivo and reduce menadione-inducedDNA injury and cytotoxicity, Am. J. Physiol. 268 (1995). C227-36.

[36] G.A. Calin, C.M. Croce, MicroRNA signatures in human cancers, Nat. Rev.Cancer 6 (11) (2006) 857–866.

[37] M.S. Kumar, J. Lu, K.L. Mercer, et al., Impaired microRNA processing enhancescellular transformation and tumorigenesis, Nat. Genet. 39 (2007) 673–677.

[38] R.A. Cairns, I.S. Harris, T.W. Mak, Regulation of cancer cell metabolism, Nat.Rev. Cancer 11 (2) (2011) 85–95.

[39] B. Chen, H. Li, X. Zeng, P. Yang, X. Liu, X. Zhao, S. Liang, Rolesof microRNA on cancer cell metabolism, J. Transl. Med. 20 (10) (2012) 228.

[40] J. Bienertova-Vasku, J. Sana, O. Slaby, The role of microRNAs in mitochondriain cancer, Cancer Lett. 336 (1) (2013) 1–7.

[41] M. Hatziapostolou, C. Polytarchou, D. Iliopoulos, MiRNAs link metabolicreprogramming to oncogenesis, Trends Endocrinol. Metab. 24 (7) (2013) 361–373.

[42] R.K. Reyes, T. Motiwala, S.T. Jacob, Regulation of glucose metabolism inhepatocarcinogenesis by microRNAs, Gene Expr. 16 (2) (2014) 85–92.

[43] M. Tomasetti, L. Nocchi, S. Staffolani, N. Manzella, M. Amati, J. Goodwin, K.Kluckova, M. Nguyen, E. Strafella, M. Bajzikova, M. Peterka, S. Lettlova, J.Truksa, W. Lee, L.F. Dong, L. Santarelli, J. Neuzil, MicroRNA-126 suppressesmesothelioma malignancy by targeting IRS1 and interfering with themitochondrial function, Antioxid. Redox Signal. (Apr 23) (2014). Epubahead of print.

[44] M. Wu, C. Chan, Prediction of therapeutic microRNA based on the humanmetabolic network, Bioinformatics (Jan 22) (2014). Epub ahead of print.

[45] H.C. Bi, Y.Z. Pan, J.X. Qiu, K.W. Krausz, F. Li, C.H. Johnson, C.T. Jiang, F.J.Gonzalez, A.M. Yu, N-methylnicotinamide and nicotinamide N-methyltransferase are associated with microRNA-1291-altered pancreaticcarcinoma cell metabolome and suppressed tumorigenesis, Carcinogenesis35 (10) (2014) 2264–2272.

Please cite this article in press as: B. Chan et al., Roles of microRNAj.canlet.2014.10.011

[46] L.H. Jin, C. Wei, Role of MicroRNAs in the Warburg Effect and MitochondrialMetabolism in Cancer, Asian Pac. J. Cancer Prev. 15 (17) (2014) 7015–7019.

[47] H. Tang, M. Lee, O. Sharpe, L. Salamone, E.J. Noonan, C.D. Hoang, S. Levine,W.H. Robinson, J.B. Shrager, Oxidative stress-responsive microRNA-320regulates glycolysis in diverse biological systems, FASEB J. 26 (11) (2012)4710–4721.

[48] A. Singh, C. Happel, S.K. Manna, G. Acquaah-Mensah, J. Carrerero, S. Kumar, P.Nasipuri, K.W. Krausz, N. Wakabayashi, R. Dewi, L.G. Boros, F.J. Gonzalez, E.Gabrielson, K.K. Wong, G. Girnun, S. Biswal, Transcription factor NRF2regulates miR-1 and miR-206 to drive tumorigenesis, J Clin Invest. 123 (7)(2013) 2921–2934.

[49] Z. Wei, L. Cui, Z. Mei, M. Liu, D. Zhang, MiR-181a mediates metabolic shift incolon cancer cells via the PTEN/AKT pathway, FEBS Lett. 588 (9) (2014) 1773–1779.

[50] Y.Y. Park, S.B. Kim, H.D. Han, B.H. Sohn, J.H. Kim, J. Liang, Y. Lu, C. Rodriguez-Aguayo, G. Lopez-Berestein, G.B. Mills, A.K. Sood, J.S. Lee, Tat-activatingregulatory DNA-binding protein regulates glycolysis in hepatocellularcarcinoma by regulating the platelet isoform of phosphofructokinasethrough microRNA 520, Hepatology 58 (1) (2013) 182–191.

[51] D.W. Dai, Q. Lu, L.X. Wang, W.Y. Zhao, Y.Q. Cao, Y.N. Li, G.S. Han, J.M. Liu, Z.J.Yue, Decreased miR-106a inhibits glioma cell glucose uptake andproliferation by targeting SLC2A3 in GBM, BMC Cancer 14 (13) (2013) 478.

[52] E. Velez, X. Yong, MiR-17-3p regulates metabolism of prostate cancer cell,FASEB J. 25 (903) (2011) 7.

[53] A. Ahmad, A. Aboukameel, D. Kong, Z. Wang, S. Sethi, W. Chen, F.H. Sarkar, A.Raz, Phosphoglucose isomerase/autocrine motility factor mediates epithelial-mesenchymal transition regulated by miR-200 in breast cancer cells, CancerRes. 71 (9) (2011) 3400–3409.

[54] M.L. Macheda, S. Rogers, Best JD Molecular and cellular regulation of glucosetransporter (GLUT) proteins in cancer, J. Cell. Physiol. 202 (3) (2005) 654–662.

[55] X. Fei, M. Qi, B. Wu, Y. Song, Y. Wang, T. Li, MicroRNA-195-5p suppressesglucose uptake and proliferation of human bladder cancer T24 cells byregulating GLUT3 expression, FEBS Lett. 586 (4) (2012) 392–397.

[56] X. Yang, Y. Cheng, P. Li, J. Tao, X. Deng, X. Zhang, M. Gu, Q. Lu, C. Yin, Alentiviral sponge for miRNA-21 diminishes aerobic glycolysis in bladdercancer T24 cells via the PTEN/PI3K/AKT/mTOR axis, Tumour Biol. (Sep 30)(2014). Epub ahead of print.

[57] T. Yamasaki, N. Seki, H. Yoshino, T. Itesako, Y. Yamada, S. Tatarano, H. Hidaka,T. Yonezawa, M. Nakagawa, H. Enokida, Tumor-suppressive microRNA-1291directly regulates glucose transporter 1 in renal cell carcinoma, Cancer Sci.104 (11) (2013 Nov) 1411–1419.

[58] S.P. Mathupala, Y.H. Ko, P.L. Pedersen, Hexokinase-2 bound to mitochondria:cancer’s stygian link to the ‘‘Warburg Effect’’ and a pivotal target for effectivetherapy, Semin. Cancer Biol. 19 (1) (2009) 17–24.

[59] A. Peschiaroli, A. Giacobbe, A. Formosa, E.K. Markert, L. Bongiorno-Borbone,A.J. Levine, E. Candi, A. D’Alessandro, L. Zolla, Finazzi Agrò A, Melino G. miR-143 regulates hexokinase 2 expression in cancer cells, Oncogene 32 (6)(2013) 797–802.

[60] S. Jiang, L.F. Zhang, H.W. Zhang, S. Hu, M.H. Lu, S. Liang, B. Li, Y. Li, D. Li, E.D.Wang, Liu MF.A novel miR-155/miR-143 cascade controls glycolysis byregulating hexokinase 2 in breast cancer cells, EMBO J. 31 (8) (2012) 1985–1998.

[61] R. Fang, T. Xiao, Z. Fang, Y. Sun, F. Li, Y. Gao, Y. Feng, L. Li, Y. Wang, X. Liu, H.Chen, X.Y. Liu, H. Ji, MicroRNA-143 (miR-143) regulates cancer glycolysis viatargeting hexokinase 2 gene, J. Biol. Chem. 287 (27) (2012) 23227–23235.

[62] L.H. Gregersen, A. Jacobsen, L.B. Frankel, J. Wen, A. Krogh, A.H. Lund,MicroRNA-143 down-regulates Hexokinase 2 in colon cancer cells, BMCCancer 12 (2012) 232.

[63] S. Zhao, H. Liu, Y. Liu, J. Wu, C. Wang, X. Hou, X. Chen, G. Yang, L. Zhao, H. Che,Y. Bi, H. Wang, F. Peng, J. Ai, MiR-143 inhibits glycolysis and depletesstemness of glioblastoma stem-like cells, Cancer Lett. 333 (2) (2013) 253–260.

[64] J.X. Jiang, S. Gao, Y.Z. Pan, C. Yu, C.Y. Sun, Overexpression of microRNA-125bsensitizes human hepatocellular carcinoma cells to 5-fluorouracil throughinhibition of glycolysis by targeting hexokinase II, Mol. Med. Rep. 10 (2)(2014) 995–1002.

[65] E. Tili, J.J. Michaille, Z. Luo, S. Volinia, L.Z. Rassenti, T.J. Kipps, C.M. Croce, Thedown-regulation of miR-125b in chronic lymphocytic leukemias leads tometabolic adaptation of cells to a transformed state, Blood 120 (13) (2012)2631–2638.

[66] N.H. Kim, Y.H. Cha, S.E. Kang, Y. Lee, I. Lee, S.Y. Cha, J.K. Ryu, J.M. Na, C. Park,H.G. Yoon, G.J. Park, J.I. Yook, H.S. Kim, P53 regulates nuclear GSK-3 levelsthrough miR-34-mediated Axin2 suppression in colorectal cancer cells, CellCycle 12 (10) (2013) 1578–1587.

[67] C. Coulouarn, V.M. Factor, J.B. Andersen, M.E. Durkin, S.S. Thorgeirsson, Loss ofmiR-122 expression in liver cancer correlates with suppression of the hepaticphenotype and gain of metastatic properties, Oncogene 28 (40) (2009) 3526–3536.

[68] G.A. Calin, A. Cimmino, M. Fabbri, M. Ferracin, S.E. Wojcik, M. Shimizu, C.Taccioli, N. Zanesi, R. Garzon, R.I. Aqeilan, H. Alder, S. Volinia, L. Rassenti, X.Liu, C.G. Liu, T.J. Kipps, M. Negrini, C.M. Croce, MiR-15a and miR-16-1 clusterfunctions in human leukemia, Proc. Natl. Acad. Sci. USA 105 (13) (2008)5166–5171.

[69] T. Ichimi, H. Enokida, Y. Okuno, R. Kunimoto, T. Chiyomaru, K. Kawamoto, K.Kawahara, K. Toki, K. Kawakami, K. Nishiyama, G. Tsujimoto, M. Nakagawa, N.

in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/

570571572573574575576577578579580581582583584585586587588589590591592593594595596597598599600601602603604605606607608609610611612613614615616617618619620621622623624625626627628629630631632633634635636637638639640641642643644645646647648649650651652653654

655656657658659660661662663664665666667668669670671672673674675676677678679680681682683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738739

B. Chan et al. / Cancer Letters xxx (2014) xxx–xxx 7

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

Seki, Identification of novel microRNA targets based on microRNA signaturesin bladder cancer, Int. J. Cancer 125 (2) (2009) 345–352.

[70] M.K. Muniyappa, P. Dowling, M. Henry, P. Meleady, P. Doolan, P. Gammell, M.Clynes, N. Barron, MiRNA-29a regulates the expression of numerous proteinsand reduces the invasiveness and proliferation of human carcinoma cell lines,Eur. J. Cancer 45 (17) (2009) 3104–3118.

[71] H.R. Christofk, M.G. Vander Heiden, M.H. Harris, A. Ramanathan, R.E.Gerszten, R. Wei, M.D. Fleming, S.L. Schreiber, L.C. Cantley, The M2 spliceisoform of pyruvate kinase is important for cancer metabolism and tumourgrowth, Nature 452 (7184) (2008) 230–233.

[72] Y. Sun, X. Zhao, Y. Zhou, Y. Hu, MiR-124, miR-137 and miR-340 regulatecolorectal cancer growth via inhibition of the Warburg effect, Oncol. Rep. 28(4) (2012) 1346–1352.

[73] A.M. Liu, Z. Xu, F.H. Shek, K.F. Wong, N.P. Lee, R.T. Poon, J. Chen, J.M. Luk, MiR-122 targets pyruvate kinase M2 and affects metabolism of hepatocellularcarcinoma, PLoS ONE 9 (1) (2014) e86872.

[74] T.S. Wong, X.B. Liu, A. Chung-Wai Ho, A. Po-Wing Yuen, R. Wai-Man Ng, W.Ignace Wei, Identification of pyruvate kinase type M2 as potentialoncoprotein in squamous cell carcinoma of tongue through microRNAprofiling, Int. J. Cancer 123 (2) (2008) 251–257.

[75] B. Kefas, L. Comeau, N. Erdle, E. Montgomery, S. Amos, B. Purow, Pyruvatekinase M2 is a target of the tumor-suppressive microRNA-326 and regulatesthe survival of glioma cells, Neuro Oncol. 12 (11) (2010) 1102–1112.

[76] W. Li, J. Wang, Q.D. Chen, X. Qian, Q. Li, Y. Yin, Z.M. Shi, L. Wang, J. Lin, L.Z. Liu,B.H. Jiang, Insulin promotes glucose consumption via regulation of miR-99a/mTOR/PKM2 pathway, PLoS ONE 8 (6) (2013) e64924.

[77] K.K. Li, J.C. Pang, A.K. Ching, C.K. Wong, X. Kong, Y. Wang, L. Zhou, Z. Chen, H.K.Ng, MiR-124 is frequently down-regulated in medulloblastoma and is anegative regulator of SLC16A1, Hum. Pathol. 40 (9) (2009) 1234–1243.

[78] T. Kinoshita, N. Nohata, H. Yoshino, T. Hanazawa, N. Kikkawa, L. Fujimura, T.Chiyomaru, K. Kawakami, H. Enokida, M. Nakagawa, Y. Okamoto, N. Seki,Tumor suppressive microRNA-375 regulates lactate dehydrogenase B inmaxillary sinus squamous cell carcinoma, Int. J. Oncol. 40 (1) (2012) 185–193.

[79] B.R. Wilfred, W.X. Wang, P.T. Nelson, Energizing miRNA research: a review ofthe role of miRNAs in lipid metabolism, with a prediction that miR-103/107regulates human metabolic pathways, Mol. Genet. Metab. 91 (3) (2007) 209–217.

[80] H. Tanaka, T. Sasayama, K. Tanaka, S. Nakamizo, M. Nishihara, K. Mizukawa,M. Kohta, J. Koyama, S. Miyake, M. Taniguchi, K. Hosoda, E. Kohmura,MicroRNA-183 upregulates HIF-1a by targeting isocitrate dehydrogenase 2(IDH2) in glioma cells, J. Neurooncol. 111 (3) (2013) 273–283.

[81] E. Favaro, A. Ramachandran, R. McCormick, H. Gee, C. Blancher, M. Crosby, C.Devlin, C. Blick, F. Buffa, J.L. Li, B. Vojnovic, R. Pires das Neves, P. Glazer, F.Iborra, M. Ivan, J. Ragoussis, A.L. Harris, MicroRNA-210 regulatesmitochondrial free radical response to hypoxia and krebs cycle in cancercells by targeting iron sulfur cluster protein ISCU, PLoS One 5 (4) (2010)e10345.

[82] Z. Chen, Y. Li, H. Zhang, P. Huang, R. Luthra, Hypoxia-regulated microRNA-210modulates mitochondrial function and decreases ISCU and COX10 expression,Oncogene 29 (30) (2010) 4362–4368.

[83] M.P. Puisségur, N.M. Mazure, T. Bertero, L. Pradelli, S. Grosso, K. Robbe-Sermesant, T. Maurin, K. Lebrigand, B. Cardinaud, V. Hofman, S. Fourre, V.Magnone, J.E. Ricci, J. Pouysségur, P. Gounon, P. Hofman, P. Barbry, B. Mari,MiR-210 is overexpressed in late stages of lung cancer and mediatesmitochondrial alterations associated with modulation of HIF-1 activity, CellDeath Differ. 18 (3) (2011) 465–478.

[84] S.Y. Chan, Y.Y. Zhang, C. Hemann, C.E. Mahoney, J.L. Zweier, J. Loscalzo,MicroRNA-210 controls mitochondrial metabolism during hypoxia byrepressing the iron-sulfur cluster assembly proteins ISCU1/2, Cell Metab. 10(4) (2009) 273–284.

[85] L.J. Eichner, M.C. Perry, C.R. Dufour, N. Bertos, M. Park, J. St-Pierre, V. Giguere,MiR-378(⁄) mediates metabolic shift in breast cancer cells via the PGC-1b/ERRc transcriptional pathway, Cell Metab. 12 (4) (2010) 353–361.

[86] M. Donadelli, I. Dando, C. Fiorini, M. Palmieri, Regulation of miR-23bexpression and its dual role on ROS production and tumour development,Cancer Lett. 349 (2) (2014) 107–113.

[87] Y. Xu, F. Fang, J. Zhang, S. Josson, St Clair WH, St Clair DK. miR-17⁄ suppressestumorigenicity of prostate cancer by inhibiting mitochondrial antioxidantenzymes, PLoS ONE 5 (12) (2010) e14356.

[88] H. Ebi, T. Sato, N. Sugito, Y. Hosono, Y. Yatabe, Y. Matsuyama, T. Yamaguchi, H.Osada, M. Suzuki, T. Takahashi, Counterbalance between RB inactivation andmiR-17-92 overexpression in reactive oxygen species and DNA damageinduction in lung cancers, Oncogene 28 (38) (2009) 3371–3379.

[89] J. Burchard, C. Zhang, A.M. Liu, R.T. Poon, N.P. Lee, K.F. Wong, P.C. Sham, B.Y.Lam, M.D. Ferguson, G. Tokiwa, R. Smith, B. Leeson, R. Beard, J.R. Lamb, L. Lim,M. Mao, H. Dai, J.M. Luk, MicroRNA-122 as a regulator of mitochondrialmetabolic gene network in hepatocellular carcinoma, Mol. Syst. Biol. 24 (6)(2010) 402.

[90] S. Venkataraman, I. Alimova, R. Fan, P. Harris, N. Foreman, R. Vibhakar,MicroRNA 128a increases intracellular ROS level by targeting Bmi-1 andinhibits medulloblastoma cancer cell growth by promoting senescence, PLoSONE 5 (6) (2010) e10748.

[91] X. Zhang, W.L. Ng, P. Wang, L. Tian, E. Werner, H. Wang, P. Doetsch, Y. Wang,MicroRNA-21 modulates the levels of reactive oxygen species by targetingSOD3 and TNFa, Cancer Res. 72 (18) (2012) 4707–4713.

Please cite this article in press as: B. Chan et al., Roles of microRNAj.canlet.2014.10.011

[92] B. Mateescu, L. Batista, M. Cardon, T. Gruosso, Y. de Feraudy, O. Mariani, A.Nicolas, J.P. Meyniel, P. Cottu, X. Sastre-Garau, F. Mechta-Grigoriou, MiR-141and miR-200a act on ovarian tumorigenesis by controlling oxidative stressresponse, Nat. Med. 17 (12) (2011) 1627–1635.

[93] S.Z. Li, Y.Y. Hu, J. Zhao, Y.B. Zhao, J.D. Sun, Y.F. Yang, C.C. Ji, Z.B. Liu, W.D. Cao,Y. Qu, W.P. Liu, G. Cheng, Z. Fei, MicroRNA-34a induces apoptosis in thehuman glioma cell line, A172, through enhanced ROS productionand NOX2 expression, Biochem. Biophys. Res. Commun. 444 (1) (2014) 6–12.

[94] P. Gao, I. Tchernyshyov, T.C. Chang, Y.S. Lee, K. Kita, T. Ochi, K.I. Zeller, A.M. DeMarzo, J.E. Van Eyk, J.T. Mendell, C.V. Dang, C-Myc suppression of miR-23a/benhances mitochondrial glutaminase expression and glutaminemetabolism,Nature 458 (7239) (2009) 762–765.

[95] B. Wang, S.H. Hsu, W. Frankel, K. Ghoshal, S.T. Jacob, Stat3-mediatedactivation of microRNA-23a suppresses gluconeogenesis in hepatocellularcarcinoma by down-regulating glucose-6-phosphatase and peroxisomeproliferator-activated receptor gamma, coactivator 1 alpha, Hepatology 56(1) (2012) 186–197.

[96] W. Liu, A. Le, C. Hancock, A.N. Lane, C.V. Dang, T.W. Fan, J.M. Phang,Reprogramming of proline and glutamine metabolism contributes to theproliferative and metabolic responses regulated by oncogenic transcriptionfactor c-MYC, Proc. Natl. Acad. Sci. USA 109 (23) (2012) 8983–8988.

[97] W. Hu, C. Zhang, R. Wu, Y. Sun, A. Levine, Z. Feng, Glutaminase 2, a novel p53target gene regulating energy metabolism and antioxidant function, Proc.Natl. Acad. Sci. USA 107 (16) (2010) 7455–7460.

[98] P. Wang, L. Zhang, Z. Chen, Z. Meng, MicroRNA targets autophagy inpancreatic cancer cells during cancer therapy, Autophagy 9 (12) (2013)2171–2172.

[99] N. Rotllan, C. Fernández-Hernando, MicroRNA regulation of cholesterolmetabolism, Cholesterol 2012 (2012) 847849.

[100] E. Tili, J.J. Michaille, Z. Luo, S. Volinia, L.Z. Rassenti, T.J. Kipps, C.M. Croce, Thedown-regulation of miR-125b in chronic lymphocytic leukemias leads tometabolic adaptation of cells to a transformed state, Blood 120 (13) (2012)2631–2638.

[101] X. Li, Y.T. Chen, S. Josson, N.K. Mukhopadhyay, J. Kim, M.R. Freeman, W.C.Huang, MicroRNA-185 and 342 inhibit tumorigenicity and induce apoptosisthrough blockade of the SREBP metabolic pathway in prostate cancer cells,PLoS ONE 8 (8) (2013) e70987.

[102] M. Cui, Y. Wang, B. Sun, Z. Xiao, L. Ye, X. Zhang, MiR-205 modulates abnormallipid metabolism of hepatoma cells via targeting acyl-CoA synthetase long-chain family member 1 (ACSL1) mRNA, Biochem. Biophys. Res. Commun. 444(2) (2014) 270–275.

[103] S.H. Hsu, B. Wang, J. Kota, J. Yu, S. Costinean, H. Kutay, L. Yu, S. Bai, K. La Perle,R.R. Chivukula, H. Mao, M. Wei, K.R. Clark, J.R. Mendell, M.A. Caligiuri, S.T.Jacob, J.T. Mendell, K. Ghoshal, Essential metabolic, anti-inflammatory, andanti-tumorigenic functions of miR-122 in liver, J. Clin. Invest. 122 (8) (2012)2871–2883.

[104] W.C. Tsai, S.D. Hsu, C.S. Hsu, T.C. Lai, S.J. Chen, R. Shen, Y. Huang, H.C. Chen,C.H. Lee, T.F. Tsai, M.T. Hsu, J.C. Wu, H.D. Huang, M.S. Shiao, M. Hsiao, A.P.Tsou, MicroRNA-122 plays a critical role in liver homeostasis andhepatocarcinogenesis, J. Clin. Invest. 122 (8) (2012) 2884–2897.

[105] A. Bourdon, L. Minai, V. Serre, J.P. Jais, E. Sarzi, S. Aubert, D. Chrétien, P. deLonlay, V. Paquis-Flucklinger, H. Arakawa, Y. Nakamura, A. Munnich, A. Rötig,Mutation of RRM2B, encoding p53-controlled ribonucleotide reductase(p53R2), causes severe mitochondrial DNA depletion, Nat. Genet. 39 (6)(2007) 776–780.

[106] D.G. Zhang, J.N. Zheng, D.S. Pei, P53/microRNA-34-induced metabolicregulation: new opportunities in anticancer therapy, Mol. Cancer 13 (2014)115.

[107] M. Sachdeva, S. Zhu, F. Wu, H. Wu, V. Walia, S. Kumar, R. Elble, K. Watabe, Y.Y.Mo, P53 represses c-Myc through induction of the tumor suppressor miR-145, Proc. Natl. Acad. Sci. USA 106 (9) (2009) 3207–3212.

[108] H. Shim, C. Dolde, B.C. Lewis, C.S. Wu, G. Dang, R.A. Jungmann, R. Dalla-Favera, C.V. Dang, C-Myc transactivation of LDH-A: implications for tumormetabolism and growth, Proc. Natl. Acad. Sci. USA 94 (13) (1997) 6658–6663.

[109] K.A. O’Donnell, E.A. Wentzel, K.I. Zeller, C.V. Dang, J.T. Mendell, C-Myc-regulated microRNAs modulate E2F1 expression, Nature 435 (7043) (2005)839–843.

[110] J. Godlewski, M.O. Nowicki, A. Bronisz, G. Nuovo, J. Palatini, M. De Lay, J. VanBrocklyn, M.C. Ostrowski, E.A. Chiocca, S.E. Lawler, MicroRNA-451 regulatesLKB1/AMPK signaling and allows adaptation to metabolic stress in gliomacells, Mol. Cell 37 (5) (2010) 620–632.

[111] D. Iliopoulos, C. Polytarchou, M. Hatziapostolou, F. Kottakis, I.G. Maroulakou,K. Struhl, P.N. Tsichlis, MicroRNAs differentially regulated by Akt isoformscontrol EMT and stem cell renewal in cancer cells, Sci. Signal 2 (92) (2009)ra62.

[112] A.K. Nagaraja, C.J. Creighton, Z. Yu, H. Zhu, P.H. Gunaratne, J.G. Reid, E. Olokpa,H. Itamochi, N.T. Ueno, S.M. Hawkins, M.L. Anderson, M.M. Matzuk, A linkbetween mir-100 and FRAP1/mTOR in clear cell ovarian cancer, Mol.Endocrinol. 24 (2) (2010) 447–463.

[113] F. Fornari, M. Milazzo, P. Chieco, M. Negrini, G.A. Calin, G.L. Grazi, D. Pollutri,C.M. Croce, L. Bolondi, L. Gramantieri, MiR-199a-3p regulates mTOR and c-Met to influence the doxorubicin sensitivity of human hepatocarcinoma cells,Cancer Res. 70 (12) (2010) 5184–5193.

[114] L. Bao, Y. Yan, C. Xu, W. Ji, S. Shen, G. Xu, Y. Zeng, B. Sun, H. Qian, L. Chen, M.Wu, C. Su, J. Chen, MicroRNA-21 suppresses PTEN and hSulf-1 expression and

in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/

740741742743744745746747748749750751752753754755756757758759760761762763764765766767768769770771772773774775776

777778779780781782783784785786787788789790791792793794795796797798799800801802803804805806807808809810811812

813

8 B. Chan et al. / Cancer Letters xxx (2014) xxx–xxx

CAN 12080 No. of Pages 8, Model 5G

23 October 2014

promotes hepatocellular carcinoma progression through AKT/ERK pathways,Cancer Lett. 337 (2) (2013) 226–236.

[115] W. Zeng, P. Liu, W. Pan, S.R. Singh, Y. Wei, Hypoxia and hypoxia induciblefactors in tumor metabolism, Cancer Lett. (Feb 4) (2014). pii:S0304-3835(14)00089-5.

[116] D. Gaglio, C.M. Metallo, P.A. Gameiro, K. Hiller, L.S. Danna, C. Balestrieri, L.Alberghina, G. Stephanopoulos, F. Chiaradonna, Oncogenic K-Ras decouplesglucose and glutamine metabolism to support cancer cell growth, Mol. Syst.Biol. 7 (2011) 523.

[117] H. Ying, A.C. Kimmelman, C.A. Lyssiotis, S. Hua, G.C. Chu, E. Fletcher-Sananikone, J.W. Locasale, J. Son, H. Zhang, J.L. Coloff, H. Yan, W. Wang, S.Chen, A. Viale, H. Zheng, J.H. Paik, C. Lim, A.R. Guimaraes, E.S. Martin, J. Chang,A.F. Hezel, S.R. Perry, J. Hu, B. Gan, Y. Xiao, J.M. Asara, R. Weissleder, Y.A.Wang, L. Chin, L.C. Cantley, R.A. DePinho, Oncogenic Kras maintainspancreatic tumors through regulation of anabolic glucose metabolism, Cell149 (3) (2012) 656–670.

[118] S. Yu, Z. Lu, C. Liu, Y. Meng, Y. Ma, W. Zhao, J. Liu, J. Yu, J. Chen, MiRNA-96suppresses KRAS and functions as a tumor suppressor gene in pancreaticcancer, Cancer Res. 70 (14) (2010) 6015–6025.

[119] W.T. Liao, Y.P. Ye, N.J. Zhang, T.T. Li, S.Y. Wang, Y.M. Cui, L. Qi, P. Wu, H.L. Jiao,Y.J. Xie, C. Zhang, J.X. Wang, Y.Q. Ding, MicroRNA-30b functions as a tumoursuppressor in human colorectal cancer by targeting KRAS, PIK3CD and BCL2,J. Pathol. 232 (4) (2014) 415–427.

[120] P. Soon, H. Kiaris, MicroRNAs in the tumour microenvironment: big role forsmall players, Endocr. Relat. Cancer 20 (5) (2013). R257-67.

[121] J. Chou, P. Shahi, Z. Werb, MicroRNA-mediated regulation of thetumor microenvironment, Cell Cycle 12 (20) (2013) 3262–3271.

[122] T. Fiaschi, P. Chiarugi, Oxidative stress, tumor microenvironment, andmetabolic reprogramming: a diabolic liaison, Int. J. Cell Biol. 2012 (2012)762825.

[123] S.J. Lunt, N. Chaudary, R.P. Hill, The tumor microenvironment and metastaticdisease, Clin. Exp. Metastasis 26 (1) (2009) 19–34.

[124] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell144 (5) (2011) 646–674.

[125] P. Sun, J.W. Hu, W.J. Xiong, J. Mi, MiR-186 regulates glycolysis through Glut1during the formation of cancer-associated fibroblasts, Asian Pac. J. CancerPrev. 15 (10) (2014) 4245–4250.

Please cite this article in press as: B. Chan et al., Roles of microRNAj.canlet.2014.10.011

[126] F. Sotgia, D. Whitaker-Menezes, U.E. Martinez-Outschoorn, N. Flomenberg,R.C. Birbe, A.K. Witkiewicz, A. Howell, N.J. Philp, R.G. Pestell, M.P. Lisanti,Mitochondrial metabolism in cancer metastasis: visualizing tumor cellmitochondria and the ‘‘reverse Warburg effect’’ in positive lymph nodetissue, Cell Cycle 11 (7) (2012) 1445–1454.

[127] M. Tomasetti, J. Neuzil, L. Dong, MicroRNAs as regulators of mitochondrialfunction: role in cancer suppression, Biochim. Biophys. Acta 1840 (4) (2014)1441–1453.

[128] L.M. Phan, S.C. Yeung, M.H. Lee, Cancer metabolic reprogramming:importance, main features, and potentials for precise targeted anti-cancertherapies, Cancer Biol. Med. 11 (1) (2014) 1–19.

[129] A. Drakaki, M. Hatziapostolou, D. Iliopoulos, Therapeutically targetingmicroRNAs in liver cancer, Curr. Pharm. Des. 19 (7) (2013) 1180–1191.

[130] C. Pulito, S. Donzelli, P. Muti, L. Puzzo, S. Strano, G. Blandino, MicroRNAs andcancer metabolism reprogramming: the paradigm of metformin, Ann. Transl.Med. 2 (6) (2014) 58.

[131] M.V. Iorio, C.M. Croce, MicroRNA dysregulation in cancer: diagnostics,monitoring and therapeutics: a comprehensive review, EMBO Mol. Med. 4(3) (2012) 143–159.

[132] J.K. Park, T. Kogure, G.J. Nuovo, J. Jiang, L. He, J.H. Kim, M.A. Phelps, T.L.Papenfuss, C.M. Croce, T. Patel, T.D. Schmittgen, MiR-221 silencing blockshepatocellular carcinoma and promotes survival, Cancer Res. 71 (24) (2011)7608–7616.

[133] J. Kota, R.R. Chivukula, K.A. O’Donnell, E.A. Wentzel, C.L. Montgomery, H.W.Hwang, T.C. Chang, P. Vivekanandan, M. Torbenson, K.R. Clark, J.R. Mendell,J.T. Mendell, Therapeutic microRNA delivery suppresses tumorigenesis in amurine liver cancer model, Cell 137 (6) (2009) 1005–1017.

[134] P. Trang, J.F. Wiggins, C.L. Daige, C. Cho, M. Omotola, D. Brown, J.B. Weidhaas,A.G. Bader, F.J. Slack, Systemic delivery of tumor suppressor microRNAmimics using a neutral lipid emulsion inhibits lung tumors in mice, Mol.Ther. 19 (6) (2011) 1116–1122.

[135] F. Takeshita, L. Patrawala, M. Osaki, R.U. Takahashi, Y. Yamamoto, N. Kosaka,M. Kawamata, K. Kelnar, A.G. Bader, D. Brown, T. Ochiya, Systemic delivery ofsynthetic microRNA-16 inhibits the growth of metastatic prostate tumors viadownregulation of multiple cell-cycle genes, Mol. Ther. 18 (1) (2010) 181–187.

in cancer metabolism, Cancer Lett. (2014), http://dx.doi.org/10.1016/