mir-23b and mir-130b expression is downregulated in pituitary

7
1 3 miR-23b and miR-130b expression is downregulated in pituitary 4 adenomas 5 6 7 Vincenza Leone a Q1 , Concetta Langella a , Daniela D’Angelo a , Paula Mussnich a , Anne Wierinckx b,e , 8 Luigi Terracciano c , Gerald Raverot d , Joel Lachuer e , Sandra Rotondi f , Marie-Lise Jaffrain-Rea f,g , 9 Jacqueline Trouillas b,d , Alfredo Fusco a,10 a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università 11 degli Studi di Napoli ‘‘Federico II’’, Naples, Italy Q2 12 b INSERM U1052, Centre de Recherche en Cancerologie de Lyon, F-69000 Lyon, France and University Lyon, F-69000 Lyon, France 13 c Molecular Pathology Division Institute of Pathology, University Hospital Schönbeinstrasse, 40CH-4003 Basel, Switzerland 14 d INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-oncology and Neuroinflammation Team, F-69372 Lyon Cedex 08, France and University Lyon1, 15 F-69000 Lyon, France 16 e Profilexpert UNIV-US7 INSERM-UMS 3453 CNRS Lyon, France 17 f Department of Clinical Applied Sciences and Biotechnology, University of L’Aquila Via Vetoio, Coppito 2, 67100 L’Aquila, Italy 18 g Neuromed, IRCCS, Pozzilli, Italy 19 20 22 article info 23 Article history: 24 Received 21 October 2013 25 Received in revised form 5 March 2014 26 Accepted 5 March 2014 27 Available online xxxx 28 Keywords: 29 microRNA 30 Pituitary adenoma 31 HMGA2 32 CCNA2 33 34 abstract 35 MicroRNA (miRNA) deregulation plays a critical role in tumorigenesis. miR-23b and miR-130b are 36 induced by thyrotropin in thyroid cells in a cAMP-dependent manner. 37 The aim of our work has been to investigate the possible role of miR-23b and miR-130b in pituitary 38 tumorigenesis. We have analyzed their expression in a panel of pituitary adenomas (PAs) including GH 39 and NFPA adenomas. 40 We report that miR-23b and miR-130b are drastically reduced in GH, gonadotroph and NFPA adenomas 41 in comparison with normal pituitary gland. Interestingly, the overexpression of miR-23b and miR-130b 42 inhibits cell proliferation arresting the cells in the G1 and G2 phase of the cell cycle, respectively. More- 43 over, we demonstrate that miR-23b and miR-130b target HMGA2 and cyclin A2 (CCNA2) genes, respec- 44 tively. Finally, downregulation of miR-23b and miR-130b expression is associated with increased levels 45 of their respective targets in human PAs. 46 These findings suggest that miR-23b and miR-130b downregulation may contribute to pituitary 47 tumorigenesis. 48 Ó 2014 Elsevier Ireland Ltd. All rights reserved. 49 50 51 52 1. Introduction 53 Pituitary adenomas (PAs) are one of the most frequent 54 intracranial tumors with a prevalence of clinically apparent tumors 55 close to one in 1000 of the general population and are the third 56 most common intracranial tumor type after meningiomas and 57 gliomas (Scheithauer et al., 2006). GH- or ACTH-secreting PAs 58 represent 20–25% and 10% of pituitary tumors, respectively, pro- 59 lactinomas account to about 50% of PAs and TSH-secreting PAs 60 are rare (1%) (Arafah and Nasrallah, 2001). About one-third of 61 PAs are clinically non-functioning pituitary adenomas (NFPAs), 62 which do not exhibit signs of hormone hypersecretion. These are 63 mostly composed by the so called ‘‘null cell’’ PAs (devoided of 64 pituitary hormone immunoreactivity) and gonadotroph adenomas 65 (defined by FSH and/or LH immunoreactivity) (Trouillas et al., 66 1986). NFPA are usually large tumors diagnosed following local 67 compressive effects on brain structures and cranial nerves. 68 30–50% of the GH adenomas are histologically mixed GH/PRL cell 69 adenomas co-secreting PRL and GH (Lopes, 2010). Pituitary tumor- 70 igenesis is generally considered a model of the multi-step process 71 of carcinogenesis, in which molecular genetic alterations represent 72 the initializing event that transforms cells, and hormones and/or 73 growth factors promote cell proliferation (Melmed, 2003; Asa 74 and Ezzat, 2004). However, the molecular events leading to 75 pituitary tumor development are still unclear. Epigenetic events, 76 such as hypermethylation and/or microRNA (miRNA)-dependent http://dx.doi.org/10.1016/j.mce.2014.03.002 0303-7207/Ó 2014 Elsevier Ireland Ltd. All rights reserved. Corresponding author. Ad Q3 dress: Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, via Pansini 5, 80131 Naples, Italy. Tel.: +39 081 7463602; fax: +39 081 2296674. E-mail address: [email protected] (A. Fusco). Molecular and Cellular Endocrinology xxx (2014) xxx–xxx Contents lists available at ScienceDirect Molecular and Cellular Endocrinology journal homepage: www.elsevier.com/locate/mce MCE 8778 No. of Pages 7, Model 5G 3 April 2014 Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b expression is downregulated in pituitary adenomas. Molecular and Cellular Endocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

Upload: vominh

Post on 05-Jan-2017

233 views

Category:

Documents


1 download

TRANSCRIPT

1

3

4

5

6

7 Q1

8

9

1011 Q212131415161718

1920

2 2

2324252627

282930313233

3 4

51

52

53

54

55

56

57

58

59

60

61

Q3

Molecular and Cellular Endocrinology xxx (2014) xxx–xxx

MCE 8778 No. of Pages 7, Model 5G

3 April 2014

Contents lists available at ScienceDirect

Molecular and Cellular Endocrinology

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

miR-23b and miR-130b expression is downregulated in pituitaryadenomas

http://dx.doi.org/10.1016/j.mce.2014.03.0020303-7207/� 2014 Elsevier Ireland Ltd. All rights reserved.

⇑ Corresponding author. Address: Istituto di Endocrinologia ed OncologiaSperimentale del CNR, via Pansini 5, 80131 Naples, Italy. Tel.: +39 081 7463602;fax: +39 081 2296674.

E-mail address: [email protected] (A. Fusco).

Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b expression is downregulated in pituitary adenomas. Molecular and CEndocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

Vincenza Leone a, Concetta Langella a, Daniela D’Angelo a, Paula Mussnich a, Anne Wierinckx b,e,Luigi Terracciano c, Gerald Raverot d, Joel Lachuer e, Sandra Rotondi f, Marie-Lise Jaffrain-Rea f,g,Jacqueline Trouillas b,d, Alfredo Fusco a,⇑a Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Universitàdegli Studi di Napoli ‘‘Federico II’’, Naples, Italyb INSERM U1052, Centre de Recherche en Cancerologie de Lyon, F-69000 Lyon, France and University Lyon, F-69000 Lyon, Francec Molecular Pathology Division Institute of Pathology, University Hospital Schönbeinstrasse, 40CH-4003 Basel, Switzerlandd INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-oncology and Neuroinflammation Team, F-69372 Lyon Cedex 08, France and University Lyon1,F-69000 Lyon, Francee Profilexpert UNIV-US7 INSERM-UMS 3453 CNRS Lyon, Francef Department of Clinical Applied Sciences and Biotechnology, University of L’Aquila Via Vetoio, Coppito 2, 67100 L’Aquila, Italyg Neuromed, IRCCS, Pozzilli, Italy

3536373839404142434445

a r t i c l e i n f o

Article history:Received 21 October 2013Received in revised form 5 March 2014Accepted 5 March 2014Available online xxxx

Keywords:microRNAPituitary adenomaHMGA2CCNA2

46474849

a b s t r a c t

MicroRNA (miRNA) deregulation plays a critical role in tumorigenesis. miR-23b and miR-130b areinduced by thyrotropin in thyroid cells in a cAMP-dependent manner.

The aim of our work has been to investigate the possible role of miR-23b and miR-130b in pituitarytumorigenesis. We have analyzed their expression in a panel of pituitary adenomas (PAs) including GHand NFPA adenomas.

We report that miR-23b and miR-130b are drastically reduced in GH, gonadotroph and NFPA adenomasin comparison with normal pituitary gland. Interestingly, the overexpression of miR-23b and miR-130binhibits cell proliferation arresting the cells in the G1 and G2 phase of the cell cycle, respectively. More-over, we demonstrate that miR-23b and miR-130b target HMGA2 and cyclin A2 (CCNA2) genes, respec-tively. Finally, downregulation of miR-23b and miR-130b expression is associated with increased levelsof their respective targets in human PAs.

These findings suggest that miR-23b and miR-130b downregulation may contribute to pituitarytumorigenesis.

� 2014 Elsevier Ireland Ltd. All rights reserved.

50

62

63

64

65

66

67

68

69

70

71

72

1. Introduction

Pituitary adenomas (PAs) are one of the most frequentintracranial tumors with a prevalence of clinically apparent tumorsclose to one in 1000 of the general population and are the thirdmost common intracranial tumor type after meningiomas andgliomas (Scheithauer et al., 2006). GH- or ACTH-secreting PAsrepresent 20–25% and 10% of pituitary tumors, respectively, pro-lactinomas account to about 50% of PAs and TSH-secreting PAsare rare (1%) (Arafah and Nasrallah, 2001). About one-third ofPAs are clinically non-functioning pituitary adenomas (NFPAs),

73

74

75

76

which do not exhibit signs of hormone hypersecretion. These aremostly composed by the so called ‘‘null cell’’ PAs (devoided ofpituitary hormone immunoreactivity) and gonadotroph adenomas(defined by FSH and/or LH immunoreactivity) (Trouillas et al.,1986). NFPA are usually large tumors diagnosed following localcompressive effects on brain structures and cranial nerves.30–50% of the GH adenomas are histologically mixed GH/PRL celladenomas co-secreting PRL and GH (Lopes, 2010). Pituitary tumor-igenesis is generally considered a model of the multi-step processof carcinogenesis, in which molecular genetic alterations representthe initializing event that transforms cells, and hormones and/orgrowth factors promote cell proliferation (Melmed, 2003; Asaand Ezzat, 2004). However, the molecular events leading topituitary tumor development are still unclear. Epigenetic events,such as hypermethylation and/or microRNA (miRNA)-dependent

ellular

77

78

79

80

81

82

83

84

85

86

87

88

89

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

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

2 V. Leone et al. / Molecular and Cellular Endocrinology xxx (2014) xxx–xxx

MCE 8778 No. of Pages 7, Model 5G

3 April 2014

impairment of protein translation, are likely to be responsible forthe downregulation of gene and/or protein expression associatedwith pituitary tumorigenesis (Amaral et al., 2009; Tateno et al.,2010; Yacqub-Usman et al., 2012).

MiRNAs are a class of small non-coding RNAs which regulategene expression at post-transcriptional level. They bind to 30-untranslated (30-UTR) regions of target mRNAs, causing block oftranslation or mRNA degradation (Bartel, 2004). They play impor-tant roles in essential cellular processes such as differentiation, cellgrowth and cell death (Miska, 2005). Several studies have demon-strated altered expression of specific miRNAs in different types ofhuman neoplasias suggesting that they play a key role in tumori-genesis (Fabbri et al., 2008). Many studies have shown alterationsof miRNA expression in pituitary adenomas. Several of these dereg-ulated miRNAs may be involved in cell proliferation, apoptosis,cancer development and progression (Bottoni et al., 2007; Amaralet al., 2009; Butz et al., 2011; D’Angelo et al., 2012; Palmieriet al., 2012).

We have recently reported the identification of specific miRNAs,miR-23b, miR-130a and miR-130b, whose upregulation by TSH isrequired for thyroid cell growth and is protein kinase A-CREBdependent (Leone et al., 2012; Leone, unpublished results formiR-130b). We also identified SMAD3, a member of TGF-b path-way that has an inhibitor role in thyroid follicular cell proliferation,as target of miR-23b. Functional studies demonstrate that the over-expression of miR-23b promotes thyroid cell growth (Leone et al.,2012).

The aim of our work was to verify whether miR-23b and miR-miR-130b misexpression occurs in non-thyroid cell system, suchas pituitary adenomas, where alterations of the cAMP pathwayare frequent. Indeed, cAMP signaling is hyperactivated in GH-secreting adenomas, with an increased phosphorylation of thecAMP response element-binding protein (p-CREB) (Nishizawaet al., 2013; Gadelha et al., 2013). Moreover, it has been demon-strated that the arylhydrocarbon receptor–interacting protein(AIP), whose mutations have been linked o predisposition to pitu-itary adenomas, acts as a tumor suppressor by maintaining a lowcAMP signaling and concentration (Formosa et al., 2013). An in-creased TGF-b1 activity has also been reported in some PAs, whereTGF-b1 may represent a usefull serum marker for invasiveness(Elenkova et al., 2013; Katsuno et al., 2013). Futhermore, thesemiRNAs have been reported as downregulated in several humanneoplasias (Tong et al., 2009; Salvi et al., 2009; Kovaleva et al.,2012).

Here, we report that miR-23b and miR-130b are drastically re-duced in somatotroph, gonadotroph and null cell PAs in compari-son with normal pituitary gland. Moreover, we demonstrate thatmiR-23b and miR-130b target HMGA2 and cyclin A2 (CCNA2)genes, respectively, which are already known to play a critical rolein pituitary tumorigenesis.

179

180

181

182

183

184

185

186

187

188

189

190

191

2. Materials and methods

2.1. Tissue collection and RNA isolation

PAs were obtained from patients operated on for medical rea-sons. For each tumor, some fragments were either snap-frozen inliquid nitrogen or collected in RNA Later (Ambion) at surgery andstored at �80 �C until RNA extraction. Diagnostic immunohisto-chemistry for pituitary hormones was performed in all cases onparaffin-embedded sections and tumors were classified into soma-totroph (n = 15) and NFPA (n = 21) including gonadotroph (n = 16)and null cell (n = 5) PA (Lloyd et al., 2004). We declare that in-formed consent for the scientific use of biological material was ob-tained from all patients.

Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b exEndocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

Total RNA isolation from PAs was performed with TRIzol re-agent (Invitrogen) according to manufacturer’s instructions. RNAsfrom human normal pituitary glands were used as control. Normalpituitaries were obtained from autopsies of two females and threemales, aged between 50 and 60 years, devoid of endocrinediseases.

2.2. Bioinformatic prediction of miRNA target genes

Genes potentially targeted by the selected miRNAs were identi-fied by using different on-line available tools such as TargetScan(www.targetscan.org), miRanda (www.microrna.org) or miRwalk(www.umm.uni-heidelberg.de/apps/zmf/mirwalk). For more de-tails see Supplemental Information.

2.3. Cell lines and transfection

GH3 rat pituitary tumor cells secreting PRL and GH and HumanEmbryonic Kidney HEK-293 cells were cultured in DMEM contain-ing 10% FBS.

For transfection of miRNA oligonucleotides, cells were transfec-ted with 50 nmol/ml pre-miRNA precursors or control no-targetingscrambled oligonucleotides (Ambion) using siPORT neoFX Trans-fection Agent (Ambion).

2.4. Reverse Transcription and quantitative Real Time (qRT)-PCR

Reverse Transcription and qRT-PCR for mature miRNA wereperformed according to manufacturer’s instructions of miScriptSystem Kits (Qiagen) (for more details see Supplemental Informa-tion). qRT-PCR analyses for HMGA2 and CCNA2 expression wereperformed as previously described (De Martino et al., 2009). To cal-culate the relative expression levels we used the 2�DDCT method(Livak and Schmittgen, 2001). Primers for glucose-6-phosphatedehydrogenase (G6PD) were used for mRNA normalization andprimers for RNU6 (Qiagen) were used for miRNA normalization.The primers used to amplify the above mentioned genes are re-ported in Supplemental Information.

2.5. Western blotting and antibodies

Western blot analysis was performed as previously described(Lloyd et al., 2004) and the membranes were incubated with anti-bodies against CCNA2 (sc-751, Santa Cruz), a-tubulin (SC-8035,Santa Cruz), a-actin (sc-1616, Santa Cruz), Vinculin (sc-7649, SantaCruz), anti-HMGA2 antibody previously described (Finelli et al.,2002).

2.6. Luciferase target assays

Cells were co-transfected with the modified Firefly luciferasevectors described in Supplemental Information, along with theRenilla luciferase reporter plasmid and the miRNA oligonucleo-tides. Firefly and Renilla luciferase activities were measured 36 hafter transfection with the Dual-Luciferase Reporter Assay System(Promega). Firefly activity was normalized to Renilla activity ascontrol of transfection efficiency.

2.7. Growth curve assay

Exponentially growing GH3 and HEK-293 cells were plated in 6-well plates and transfected with 50 nmol/ml of pre-miR miRNAprecursor or scrambled oligonucleotide using siPORT neoFX. Cellswere counted in triplicate at 24, 48, 72 and 96 h after transfectionwith a Burker hemocytometer chamber.

pression is downregulated in pituitary adenomas. Molecular and Cellular

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

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

V. Leone et al. / Molecular and Cellular Endocrinology xxx (2014) xxx–xxx 3

MCE 8778 No. of Pages 7, Model 5G

3 April 2014

2.8. Flow cytometry

HEK-293 cells were plated and synchronized by serum depriva-tion for 48 h. Then, the cells were transfected with 50 nmol/ml ofpre-miR miRNA precursor or scrambled oligonucleotide using si-PORT neoFX and collected after 72 h.

After trypsinization, cells were washed in phosphate-bufferedsaline and fixed in 70% ethanol. Staining for DNA content was per-formed with 2 lg/ml propidium iodide and 20 lg/ml RNase A for30 min. For each measure 20,000 events have been analyzed. Weused a FACScan flow cytometer (Becton Dickinson) that was inter-faced with a Hewlett–Packard computer (Palo Alto, CA). Cell cycledata were analyzed with the CELL-FIT program (Becton Dickinson).

2.9. Statistical analysis

Student’s t-test was used to determine the differences betweentwo population of samples. Data are presented in the text asmean ± SEM and p < 0.05 was accepted as statistically significant.

To compare the differences between groups, one-way analysisof variance was used. The significance of differences was deter-mined by Analysis of Variances (ANOVA) followed by Dunnett’stest as the post hoc test using Graph Pad Prism 5.0. Data are pre-sented in the text as mean ± SEM and p < 0.05 was accepted as sta-tistically significant.

3. Results

3.1. miR-23b and miR-130b are downregulated in pituitary adenomas

To evaluate the possible role of miR-23b and miR-130b in pitu-itary tumorigenesis, we analyzed the expression of these miRNAs ina panel of 15 somatotroph PA and 21 NFPA in comparison with 5normal pituitary glands. As shown in Fig. 1, miR-23b was downreg-ulated in all tumors, except one GH (Panel A). MiR-130b was alsodownregulated in all tumors, apart from one NFPA (Panel B).

Fig. 1. Analysis of miR-23b and miR-130b expression in pituitary adenomas by qRT–PCRbetween somatotroph PA and NFPA (including gonadotroph (n = 16) and null cell (n = 5)samples was equal to 1. The range of variability of the expression of these miRNAs in northree independent experiments performed in triplicate. �p < 0.05 compared to normal p

Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b exEndocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

3.2. Identification of the target genes for the miR-23b and miR-130b

In order to understand the mechanism by which the downreg-ulation of miR-23b and miR-130b might be involved in pituitarytumorigenesis, we identified, using bioinformatic tools (miRanda,TargetScan and MiRwalk), several potential target genes. Interest-ingly, we found HMGA2 and CCNA2 genes as predicted targets formiR-23b and miR-130b, respectively. Therefore, we focused ourattention on these target genes since HMGA proteins are involvedin cell cycle dysregulation, which plays an important role in thedevelopment of PA (Fedele et al., 2006). Moreover, we have previ-ously demonstrated that HMGA2 plays a critical role in pituitarytumorigenesis. Indeed, the HMGA2 gene was found overexpressedin most human PAs (De Martino et al., 2009; Qian et al., 2009)and transgenic mice overexpressing HMGA2 developed mixedPRL/GH PAs (Fedele et al., 2002). A-type cyclins also have a criticalrole on the cell cycle regulation. In fact, by coupling successively toCDK2 and CDK1, they participate in the S/G2 transition and pro-gression through G2 (Yam et al., 2002).

In Fig. 2A we report the miRNA-targeting sites of miR-23b onHMGA2 30-UTR. To validate its influence on the expression of theHMGA2 gene, we have performed a time course, collecting HEK-293 cells transfected with miR-23b and relative scrambled, at 24,48, 72 and 96 h. As shown in Fig. 2B, transfection of miR-23b de-creased HMGA2 protein levels at 72 h as compared to the scram-bled oligonucleotide-transfected cells. Interestingly, as shown inFig. 2C, this was accompanied by a decrease in HMGA2 transcripts,even though there is not a perfect complementarity between themiR-23b and its target sequences. This result strongly suggeststhat miR-23b reduces HMGA2 protein level also by affectingHMGA2 mRNA stability. In Fig. 3A we report the miRNA-targetingsites of miR-130b on CCNA2 30-UTR. Similarly, we looked at timecourse of CCNA2 protein levels in cells transfected withmiR-130b. As shown in Fig. 3B, transfection of miR-130b decreasedCCNA2 protein levels at 48 and 72 h as compared to the scrambledoligonucleotide-transfected cells. In contrast, no significant

. The relative expression values indicate the relative change in the expression levelsPA) versus five normal pituitary glands, assuming that the mean value of the normalmal pituitary tissues is less than 10%. Each bar represents the mean value ± s.e. fromituitary.

pression is downregulated in pituitary adenomas. Molecular and Cellular

257

258

259

260

261

262

263

264

265

Fig. 2. miR-23b targets HMGA2. (A) Schematic representation of human HMGA230UTR and the relative position of the predicted miRNA-binding site. (B) Westernblot analysis of HMGA2 protein levels in HEK-293 cells transfected with indicatedmiRNA precursors or scrambled oligonucleotide and collected at 24, 48, 72 and96 h. a-tubulin was analyzed as loading control. A representative experiment isshown. The bars below represent the densitometric analysis performed usingImageJ software and normalized with tubulin. (C) qRT–PCR analysis of HMGA2mRNA in HEK-293 cells transfected and collected at 72 h. Relative expression valuesindicate the relative change in HMGA2 mRNA expression levels between miR-treated and scrambled oligonucleotide-treated cells, normalized with G6PD. Theerror bars represent the mean ± s.e of three independent experiments performed intriplicate. (D) Relative luciferase activity in HEK-293 cells transiently transfectedwith 30UTR-HMGA2 and 30UTR-HMGA2MUT along with the indicated miRNAoligonucleotides or with a no-targeting scrambled oligonucleotide. The relativeactivity of firefly luciferase expression was standardized to a transfection control,using Renilla luciferase. The scale bars represent the mean ± s.e. of three indepen-dent experiments performed in triplicate. p < 0.05 compared to scrambled oligo-nucleotide-transfected cells.

Fig. 3. miR-130b targets CCNA2. (A) Schematic representation of the 30-UTR sites ofthe CCNA2 gene targeted by miR-130b and the relative position of the predictedmiRNA-binding site. (B) Western blots of the CCNA2 protein expression in HEK-293cells transfected with miR-130b and a scrambled oligonucleotide and collected at24, 48, 72 and 96 h. a-actin was analyzed as loading control. A representativeexperiment is shown. The bars below represent the densitometric analysisperformed using ImageJ software and normalized with actin. (C) qRT–PCR analysisof CCNA2 mRNA in HEK-293 cells transfected and collected at 72 h. Relativeexpression values indicate the relative change in CCNA2 mRNA expression levelsbetween miRNA-treated and scrambled oligonucleotide-treated cells, normalizedwith G6PD. The error bars represent the mean ± s.e. p < 0.05 compared to scrambledoligonucleotide-transfected cells. (D) Relative luciferase activity in HEK-293 cellstransiently transfected with 30UTR-CCNA2- and 30UTR-CCNA2MUT along with miR-130b oligonucleotide. A no-targeting scrambled oligonucleotide was used as acontrol. The relative activity of firefly luciferase expression was standardized to atransfection control, using Renilla luciferase. The scale bars represent themean ± s.e. of three independent experiments performed in triplicate. �Significancevalues of p < 0.05 compared to scrambled oligonucleotide-transfected cells.

4 V. Leone et al. / Molecular and Cellular Endocrinology xxx (2014) xxx–xxx

MCE 8778 No. of Pages 7, Model 5G

3 April 2014

changes in the CCNA2 transcripts were observed (Fig. 3C). Thesedata are consistent with post-transcriptional regulation of theCCNA2 protein by miR-130b without effects on CCNA2 mRNAdegradation.

Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b exEndocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

In order to verify that a direct interaction between miRNAs andthe CCNA2 and HMGA2 mRNAs was responsible for protein leveldecrease, the 30-UTRs of CCNA2 and HMGA2 mRNA were inserteddownstream of the luciferase ORF, either in sense (30UTR-CCNA2and 30UTR-HMGA2) or in antisense (30UTR-CCNA2 MUT and

pression is downregulated in pituitary adenomas. Molecular and Cellular

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

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

V. Leone et al. / Molecular and Cellular Endocrinology xxx (2014) xxx–xxx 5

MCE 8778 No. of Pages 7, Model 5G

3 April 2014

30UTR-HMGA2 MUT) orientation. These reporter vectors weretransfected in HEK-293 cells together with miRNA oligonucleotideprecursors or a scrambled oligonucleotide. As shown in Fig. 2 and3D, the luciferase activity of the Luc-sense-30UTR constructs wasmarkedly decreased after transfection of both miRNAs, comparedwith the scrambled oligonucleotide, while the luciferase activityof Luc-antisense-30UTR constructs did not vary. This indicates thatthese miRNAs interfere with CCNA2 and HMGA2 translationthrough direct interaction with their 30-UTR.

3.3. The miR-23b and miR-130b inhibit cell proliferation

To understand the role of miRNA downregulation in pituitarytumorigenesis we analyzed their effects on cell proliferation. Theeffect of miR-23b expression on cell growth was studied on humanHEK-293 cells, we did not use the rat GH3 cells for this experimentsince HMGA2 is not expressed in these cells. As shown in Fig. 4A(left panel), a significant reduction of cell number was observed96 h after transient transfection with miR-23b compared withscrambled oligonucleotide-transfected cells. Even a more drasticreduction of the proliferation was obtained when the HMGA2 pro-tein expression was silenced by shRNA interference (data notshown). Conversely, the role of miR-130b was studied on GH3 cellstransiently transfected with miR-130b or the scrambled oligonu-cleotide. As shown in Fig. 4A (right panel), a significant reductionof cell number was also observed 96 h after transfection withmiR-130b compared with scrambled-oligonucleotide transfectedcells. We have also performed the same experiments on the HEK-293 cell line achieving almost identical results (SupplementaryFig. 1A).

Finally, to better characterize the effects of the analyzed miR-NAs on cell cycle progression, miRNA precursors or scrambled oli-gonucleotide were transfected in HEK-293 (Fig. 4B, left panel) andGH3 cells (Fig. 4B, right panel), and analyzed by flow cytometry.Interestingly, miR-23b-transfected cells displayed an increase in

Fig. 4. miR-23b and miR-130b inhibit cell proliferation. (A). Cell growth curves of HEK-2with miR-130b and counted each 24 h for 96 h after plating. Y-axis represents the mean o�Significance values of p < 0.05 compared to scrambled oligonucleotide-transfected cellspanel) cells transfected with the indicated miRNA precursors or scrambled oligonucleotidcytometry after propidium iodide staining. Each bar represents the mean ± s.e. from threcompared to scrambled oligonucleotide-transfected cells.

Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b exEndocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

the G1 phase population and a decrease in the S-phase, comparedwith scrambled-transfected cells. These results indicate that theoverexpression of this miRNA affects the G1-S transition of the cellcycle progression, as expected by its target (HMGA2). In contrast,miR-130b-transfected cells displayed a decreased number of cellsin the G1-phase, whereas the cell population in the G2-phasewas increased, compared with scrambled-oligonucleotide trans-fected cells. These findings are consistent with downregulation ofits target gene (CCNA2), which is involved in the regulation of S/G2 transition and progression through G2 phase of the cell cycle(Yam et al., 2002). We have also performed the same experimentson the HEK-293 cell line achieving almost identical results (Supple-mentary Fig. 1B).

3.4. miR-23b and miR-130b downregulation is associated with anincrease in HMGA2 and CCNA2, respectively, in human PAs

In order to further analyse the potential role of miRNA down-regulation in human pituitary tumorigenesis, the expression ofCCNA2 was studied by western blot in a subset of GH-PAs showinga marked downregulation of miR-130b. As shown in Fig. 5A, an in-creased CCNA2 protein expression was observed in all cases, incomparison with normal pituitary. Similarly, we previously re-ported increased HMGA2 protein levels in GH-PAs, in comparisonwith normal pituitary (D’Angelo et al., 2012). In this study, wefound that miR-23b downregulation was associated with overex-pression of HMGA2 mRNA evaluated by qRT-PCR in GH, gonadotro-phin and null cell PAs (Fig. 5 B).

4. Discussion

In this study, we have analyzed the expression of miR-23b andmiR-130b in pituitary adenomas. Both were found to be downreg-ulated in GH, gonadotroph and null cell PA in comparison with

93 (left panel) cells transfected with miR-23b or GH3 (right panel) cells transfectedf viable cell count ± s.e. from three independent experiments performed in triplicate.. (B) Flow cytometric analysis of synchronized HEK-293 (left panel) and GH3 (righte. After transfection, the DNA of the transfected cells was analyzed 72 h later by flowe independent experiments performed in triplicate. �Significance values of p < 0.05

pression is downregulated in pituitary adenomas. Molecular and Cellular

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

367

368

369

370

371

372

373

374

375

376

377

378

379Q4

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

Fig. 5. miR-23b and miR-130b downregulation is associated with an increase in HMGA2 and CCNA2 in human PAs compared with normal pituitary (NP). (A) Western blotanalysis of CCNA2 protein expression in a subset of somatotroph PA. a-tubulin has been used as loading control. (B) qRT–PCR analysis of HMGA2 mRNA levels in somatotrophPA and NFPA (including gonadotroph (n = 16) and null cell (n = 5) PA) and in normal pituitary tissue (5 samples). The relative expression values indicate the relative change inthe expression levels between adenomas and normal pituitary assuming that the value of normal pituitary tissue is equal to 1.

6 V. Leone et al. / Molecular and Cellular Endocrinology xxx (2014) xxx–xxx

MCE 8778 No. of Pages 7, Model 5G

3 April 2014

normal pituitary gland, in contrast with the results obtained inthyroid cells stimulated by TSH (Leone et al., 2012) and thyroidadenomas (Leone, manuscript in preparation) showing upregula-tion of these miRNAs. We also identified the HMGA2 gene as targetof miR-23b. Overexpression of HMGA2 is a frequent event in PAsand high levels of HMGA2 protein have been correlated with tumorsize, invasiveness and higher Ki-67 index in PRL, silent ACTH- andgonadotroph adenomas (Qian et al., 2009). We have previouslyshown that HMGA2 overexpression in prolactinomas was fre-quently associated with alterations of the HMGA2 gene, locatedin chromosome 12, including amplification, rearrangements andtrisomy of chromosome 12 (Finelli et al., 2002). In this study, wedemonstrate that miR-23b interaction with HMGA2 mRNA 30 UTRin vitro is directly responsible for HMGA2 protein decrease. In addi-tion, miR-23b was found to affect HMGA2 mRNA stability in vitroand an opposite behavior between HMGA2 transcripts and theexpression of miR-23b was observed in human PAs. Therefore,miR-23b downregulation can, at least partially, account for HMGA2overexpression in NFPAs where genomic HMGA2 alterations havebeen rarely observed (Pierantoni et al., 2005). Then, downregula-tion of miRNAs targeting HMGA2 represents an alternative mecha-nism of HMGA2 overexpression in PA and may potentiallysynergize with HMGA2 gene alterations to induce high HMGA2protein levels. Moreover, we identified the CCNA2 gene as targetof miR-130b. A-type cyclins exert a critical role on the cell cycleregulation. In fact, Cyclin A is the only cyclin able to regulate multi-ple steps of the cell cycle, since it participates in the G1/S transitionand progression through G2 phase of the cell cycle (Yam et al.,2002).

In this study, we demonstrate that miR-130b interaction withCCNA2 mRNA 30 UTR in vitro is directly responsible for CCNA2 pro-tein decrease, with no effect on CCNA2 mRNA stability. Accordingly,CCNA2 was found to be overexpressed in a subset of GH-secretingPAs showing a marked downregulation of miR-130b. Functionalstudies suggest a role for the downregulation of these miRNAs in

Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b exEndocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

tumorigenesis. Indeed, overexpression of both miR-23b and miR-130b was found to inhibit cell growth. Moreover, FACS analysisdemonstrated that, consistently with their identified target genes,miR-23b retains the cells in the G1 phase of the cell cycle, whereasmiR-130b retains the cells in the G2 phase. These findings furthersupport the hypothesis that downregulation of miR-23b andmiR-130b may contribute to development of human PAs throughan overexpression of HMGA2 and CCNA2, respectively. However,we have to consider that downregulation of these miRNAs maycontribute to pituitary tumorigenesis also by targeting other genescoding for proteins involved in cell proliferation as SMAD3, a mem-ber of TGF-b pathway that we identified as target of miR-23b.Indeed, it is well known that each miRNA regulates the expressionof several genes.

Interestingly, recent studies have demonstrated that miR-23band miR-130b are also downregulated in prolactinomas (Chenet al., 2012a,b), suggesting this might represent a general eventin pituitary tumorigenesis. It is noteworthy, to observe that bothmiRNAs were not reported in our previous study where the miRNAexpression profile of 12 GH-adenomas was analyzed. Unfortu-nately, these techniques are not completely precise yet, and thisalso accounts for different results obtained by different groupsanalyzing the same tumor histotypes. Moreover, we did not ana-lyze miR-23b in the study by Palmieri et al., where we focused juston a limited number of miRNAs able to target the genes coding forthe HMGA proteins.

Notably, both miR-23b and miR-130b have been found deregu-lated in several human neoplasias. Indeed, miR-23b is overexpres-sed in bladder (Gottardo et al., 2007) and oral squamous cellcarcinoma (Scapoli et al., 2010) in breast cancer (Paris et al.,2012) where it acts as a potential oncomir. Conversely, it is down-regulated in prostate cancer (Tong et al., 2009), hepatocellular car-cinoma (Salvi et al., 2009) where it acts as a tumor suppressor bytargeting the urokinase-type plasminogen activator (uPA) and c-met (Salvi et al., 2009). Also miR-130 is upregulated in several

pression is downregulated in pituitary adenomas. Molecular and Cellular

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

424425426427428429430431432433434435436437438439440441442443444445446447448449450451452453454455456457458459460461462463464465466467468

469470471472473474475476477478479480481482483484485486487488489490491492493494495496497498499

V. Leone et al. / Molecular and Cellular Endocrinology xxx (2014) xxx–xxx 7

MCE 8778 No. of Pages 7, Model 5G

3 April 2014

types of cancer, such as nonsmall cell lung cancer (Yang et al.,2010), pancreatic (Chen et al., 2012a,b) and breast cancer (Shiet al., 2011), whereas is down-regulated in chronic lymphocyticleukemia (Kovaleva et al., 2012).

In conclusion, we report the identification of miRNAs drasticallyand constantly downregulated in PAs. Since these miRNAs targetgenes, such as HMGA2 and CCNA2, whose overexpression plays acritical role in pituitary tumorigenesis, it is reasonable to retainthat their downregulation might contribute to this process sug-gesting an approach to the therapy of PAs based on the restorationof the downregulated miRNAs.

Acknowledgments

This work was supported by grants from Associazione Italianaper la Ricerca sul Cancro (AIRC) (IG 5346), the Project ‘‘Invec-chiamemto’’ of the National Research Council (CNR) and from theItalian Ministry of Economy and Finance to the CNR for the ProjectFaReBio di Qualità. D. D’Angelo is a recipient of a fellowship fromItalian Foundation for Cancer research (FIRC).

We are grateful to E. Jouhanneau, V. Esposito e F. Giangasperofor adenomas collection and analysis and Mario Berardone forartwork.

500501502503504505506507508509510511512513514515516517518519520521522523524525526527528529530531532533534535536537538539540541542543544545546547548549550551552553554

Appendix A. Supplementary material

Supplementary data associated with this article can be found, inthe online version, at http://dx.doi.org/10.1016/j.mce.2014.03.002.

References

Amaral, F.C., Torres, N., Saggioro, F., Neder, L., Machado, H.R., Silva Jr., W.A., Moreira,A.C., Castro, M., 2009. MicroRNAs differentially expressed in ACTH-secretingpituitary tumors. J. Clin. Endocrinol. Metab. 94, 320–323.

Arafah, B.M., Nasrallah, M.P., 2001. Pituitary tumors: pathophysiology, clinicalmanifestations and management. Endocr. Relat. Cancer 8, 287–305.

Asa, S.L., Ezzat, S., 2004. Molecular basis of pituitary development and cytogenesis.Front Horm. Res. 32, 1–19.

Bartel, D.P., 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell116, 281–297.

Bottoni, A., Zatelli, M.C., Ferracin, M., Tagliati, F., Piccin, D., Vignali, C., Calin, G.A.,Negrini, M., Croce, C.M., Degli Uberti, E.C., 2007. Identification of differentiallyexpressed microRNAs by microarray: a possible role for microRNA genes inpituitary adenomas. J. Cell. Physiol. 210, 370–377.

Butz, H., Likó, I., Czirják, S., Igaz, P., Korbonits, M., Rácz, K., Patócs, A., 2011.MicroRNA profile indicates downregulation of the TGFb pathway in sporadicnon-functioning pituitary adenomas. Pituitary 14, 112–124.

Chen, Y.X., Li, Q., Wang, C.D., Su, Z.P., Li, W.Q., Chen, X.B., Wu, Z.B., 2012a.Differential expression analysis of prolactinoma-related microRNAs. ZhonghuaYi Xue Za Zhi 92, 320–323.

Chen, Z., Chen, L.Y., Dai, H.Y., Wang, P., Gao, S., Wang, K., 2012b. MiR-301a promotespancreatic cancer cell proliferation by directly inhibiting bim expression. J. Cell.Biochem. 113, 3229–3235.

D’Angelo, D., Palmieri, D., Mussnich, P., Roche, M., Wierinckx, A., Raverot, G., Fedele,M., Croce, C.M., Trouillas, J., Fusco, A., 2012. Altered microRNA expression profilein human pituitary GH adenomas: down-regulation of miRNA targetingHMGA1, HMGA2, and E2F1. J. Clin. Endocrinol. Metab. 97, E1128–1138.

De Martino, I., Visone, R., Wierinckx, A., Palmieri, D., Ferraro, A., Cappabianca, P.,Chiappetta, G., Forzati, F., Lombardi, G., Colao, A., Trouillas, J., Fedele, M., Fusco,A., 2009. HMGA proteins up-regulate CCNB2 gene in mouse and humanpituitary adenomas. Cancer Res. 69, 1844–1850.

Elenkova, A.P., Atanassova, I., Kirilov, G., Vasilev, V., Kalinov, K., Zacharieva, S., 2013.Transforming growth factor beta 1 (TGF-b1) is not a reliable biomarker forvalvular fibrosis but could be a potential serum marker for invasiveness ofprolactinomas (pilot study). Eur. J. Endocrinol. 169, 299–306.

Fabbri, M., Croce, C.M., Calin, G.A., 2008. MicroRNAs. Cancer J. 14, 1–6.Fedele, M., Battista, S., Kenyon, L., Baldassarre, G., Fidanza, V., Klein-Szanto, A.J.P.,

Parlow, A.F., Visone, R., Pierantoni, G.M., Outwater, E., Santoro, M., Croce, C.M.,Fusco, A., 2002. Overexpression of the HMGA2 gene in transgenic mice leads tothe onset of pituitary adenomas. Oncogene 21, 3190–3198.

Fedele, M., Visone, R., De Martino, I., Troncone, G., Palmieri, D., Battista, S.,Ciarmiello, A., Pallante, P., Arra, C., Melillo, R.M., Helin, K., Croce, C.M., Fusco, A.,2006. HMGA2 induces pituitary tumorigenesis by enhancing E2F1 activity.Cancer Cell 9, 459–471.

Finelli, P., Pierantoni, G.M., Giardino, D., Losa, M., Rodeschini, O., Fedele, M., Valtorta,E., Mortini, P., Croce, C.M., Larizza, L., Fusco, A., 2002. The High Mobility Group

555

Please cite this article in press as: Leone, V., et al. miR-23b and miR-130b exEndocrinology (2014), http://dx.doi.org/10.1016/j.mce.2014.03.002

A2 gene is amplified and overexpressed in human prolactinomas. Cancer Res.62, 2398–2405.

Formosa, R., Xuereb-Anastasi, A., Vassallo, J., 2013. Aip regulates cAMP signallingand GH secretion in GH3 cells. Endocr. Relat. Cancer 20, 495–505.

Gadelha, M.R., Trivellin, G., Hernández Ramírez, L.C., Korbonits, M., 2013. Geneticsof pituitary adenomas. Front Horm. Res. 41, 111–140.

Gottardo, F., Liu, C.G., Ferracin, M., Calin, G.A., Fassan, M., Bassi, P., Sevignani, C.,Byrne, D., Negrini, M., Pagano, F., Gomella, L.G., Croce, C.M., Baffa, R., 2007.Micro-RNA profiling in kidney and bladder cancers. Urol. Oncol. 25, 387–392.

Katsuno, Y., Lamouille, S., Derynck, R., 2013. TGF-b signaling and epithelial–mesenchymal transition in cancer progression. Curr. Opin. Oncol. 25, 76–84.

Kovaleva, V., Mora, R., Park, Y.J., Plass, C., Chiramel, A.I., Bartenschlager, R., Döhner,H., Stilgenbauer, S., Pscherer, A., Lichter, P., Seiffert, M., 2012. MiRNA-130atargets ATG2B and DICER1 to inhibit autophagy and trigger killing of chroniclymphocytic leukemia cells. Cancer Res. 72, 1763–1772.

Leone, V., D’Angelo, D., Pallante, P., Croce, C.M., Fusco, A., 2012. Thyrotropinregulates thyroid cell proliferation by up-regulating miR-23b and miR-29b thattarget SMAD3. J. Clin. Endocrinol. Metab. 97, 3292–3301.

Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data usingreal-time quantitative PCR and the 2(�Delta Delta C(T)) method. Methods 25,402–408.

Lloyd, R.V., Kovacs, K., Young, Jr W.F., Farrel, W.E., Asa, S.L., Trouillas, J.,Kontogeorgos, G., Sano, T., Scheithauer, B., Horvath, E., 2004. Pituitarytumours: introduction. In chapter 1: tumours of the pituitary. Pathology andgenetics of tumours of endocrine organs. World Hearth OrganizationClassification of Tumours. Delellis RA, Lloyd RV, Heitz PU, Eng C (Eds.), IARCPress, Lyon, p. 10–13.

Lopes, M.B., 2010. Growth hormone-secreting adenomas: pathology and cellbiology. Neurosurg. Focus 29, E2.

Melmed, S., 2003. Mechanisms for pituitary tumorigenesis: the plastic pituitary. J.Clin. Invest. 112, 1603–1618.

Miska, E.A., 2005. How microRNAs control cell division, differentiation and death.Curr. Opin. Genet. Dev. 15, 563–568.

Nishizawa, H., Fukuoka, H., Iguchi, G., Inoshita, N., Yamada, S., Takahashi, Y., 2013.AIP mutation identified in a patient with acromegaly caused by pituitarysomatotroph adenoma with neuronal choristoma. Exp. Clin. Endocrinol.Diabetes 121, 295–299.

Palmieri, D., D’Angelo, D., Valentino, T., De Martino, I., Ferraro, A., Wierinckx, A.,Fedele, M., Trouillas, J., Fusco, A., 2012. Downregulation of HMGA-targetingmicroRNAs has a critical role in human pituitary tumorigenesis. Oncogene 23(31), 3857–3865.

Paris, O., Ferraro, L., Grober, O.M., Ravo, M., De Filippo, M.R., Giurato, G., Nassa, G.,Tarallo, R., Cantarella, C., Rizzo, F., Di Benedetto, A., Mottolese, M., Benes, V.,Ambrosino, C., Nola, E., Weisz, A., 2012. Direct regulation of microRNAbiogenesis and expression by estrogen receptor beta in hormone-responsivebreast cancer. Oncogene 31, 4196–4206.

Pierantoni, G.M., Finelli, P., Valtorta, E., Giardino, D., Rodeschini, O., Esposito, F.,Losa, M., Fusco, A., Larizza, L., 2005. High-mobility group A2 gene expression isfrequently induced in non-functioning pituitary adenomas (NFPAs), even in theabsence of chromosome 12 polysomy. Endocr. Relat. Cancer 12, 867–874.

Qian, Z.R., Asa, S.L., Siomi, H., Siomi, M.C., Yoshimoto, K., Yamada, S., Wang, E.L.,Rahman, M.M., Inoue, H., Itakura, M., Kudo, E., Sano, T., 2009. Overexpression ofHMGA2 relates to reduction of the let-7 and its relationship toclinicopathological features in pituitary adenomas. Mod. Pathol. 22, 431–441.

Salvi, A., Sabelli, C., Moncini, S., Venturin, M., Arici, B., Riva, P., Portolani, N., Giulini,S.M., De Petro, G., Barlati, S., 2009. MicroRNA-23b mediates urokinase and c-met downmodulation and a decreased migration of human hepatocellularcarcinoma cells. FEBS J. 276, 2966–2982.

Scapoli, L., Palmieri, A., Lo Muzio, L., Pezzetti, F., Rubini, C., Girardi, A., Farinella, F.,Mazzotta, M., Carinci, F., 2010. MicroRNA expression profiling of oral carcinomaidentifies new markers of tumor progression. Int. J. Immunopathol. Pharmacol.23, 1229–1234.

Scheithauer, B.W., Gaffey, T.A., Lloyd, R.V., Sebo, T.J., Kovacs, K.T., Horvath, E.,Yapicier, O., Young Jr., W.F., Meyer, F.B., Kuroki, T., Riehle, D.L., Laws Jr., E.R.,2006. Pathobiology of pituitary adenomas and carcinomas. Neurosurgery 59,341–353.

Shi, W., Gerster, K., Alajez, N.M., Tsang, J., Waldron, L., Pintilie, M., Hui, A.B., Sykes, J.,P’ng, C., Miller, N., McCready, D., Fyles, A., Liu, F.F., 2011. MicroRNA-301mediates proliferation and invasion in human breast cancer. Cancer Res. 71,2926–2937.

Tateno, T., Zhu, X., Asa, S.L., Ezzat, S., 2010. Chromatin remodeling and histonemodifications in pituitary tumors. Mol. Cell. Endocrinol. 15 (326), 66–70.

Tong, A.W., Fulgham, P., Jay, C., Chen, P., Khalil, I., Liu, S., Senzer, N., Eklund, A.C.,Han, J., Nemunaitis, J., 2009. MicroRNA profile analysis of human prostatecancers. Cancer Gene Ther. 16, 206–216.

Trouillas, J., Girod, C., Sassolas, G., Claustrat, B., 1986. The human gonadotropicadenoma: pathologic diagnosis and hormonal correlations in 26 tumors. Semin.Diagn. Pathol. 3, 42–57.

Yacqub-Usman, K., Richardson, A., Duong, C.V., Clayton, R.N., Farrell, W.E., 2012. Thepituitary tumour epigenome: aberrations and prospects for targeted therapy.Nat. Rev. Endocrinol. 24 (8), 486–494.

Yam, C.H., Fung, T.K., Poon, R.Y., 2002. Cyclin A in cell cycle control and cancer. Cell.Mol. Life Sci. 59, 1317–1326.

Yang, X.C., Tian, L.L., Wu, H.L., Jiang, X.Y., Du, L.Q., Zhang, H., Wang, Y.Y., Wu, H.Y., Li,D.G., She, Y., Liu, Q.F., Fan, F.Y., Meng, A.M., 2010. Expression of miRNA-130a innonsmall cell lung cancer. Am. J. Med. Sci. 340, 385–388.

pression is downregulated in pituitary adenomas. Molecular and Cellular