miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in...

7
miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Wei a,1 , Long Cui b,1 , Zubing Mei b , Min Liu a , Daoxiang Zhang a,b,c,a Department of Pathology of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, China b Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, China c Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China article info Article history: Received 18 January 2014 Revised 25 February 2014 Accepted 4 March 2014 Available online 28 March 2014 Edited by Tamas Dalmay Keywords: miR-181a PTEN Glycolysis abstract Cancer cell metabolism is often characterized by a shift from an oxidative to a glycolytic bioenerget- ics pathway, a phenomenon known as the warburg effect. Whether the deregulation of miRNAs con- tributes to the warburg effect remains largely unknown. Here we show that miR-181a expression is increased and thus induces a metabolic shift in colon cancer cells. miR-181a performs this function by inhibiting the expression of PTEN, leading to an increase of phosphorylated AKT which triggers metabolic shift. The increase of lactate production induced by miR-181a results in the rapid growth of cancer cells. These results identify miR-181a as a molecular switch involved in the orchestration of the warburg effect in colon cancer cells via the PTEN/AKT pathway. Ó 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. 1. Introduction The metabolism of cancer cells is significantly different from that of normally differentiated cells [1]. Normally differentiated cells rely primarily on the oxidation of pyruvate in the mitochon- dria to generate energy for cellular physiology; however, even with sufficient oxygen, rapidly growing cancer cells rely on aerobic gly- colysis to generate energy. This phenomenon is termed ‘‘the War- burg effect’’ [2]. This metabolic shift towards enhanced glycolysis is believed to be the result of adaptations to support the continu- ous proliferation of cancer cells. Flux along other key metabolic pathways, such as the glutaminolysis pathway, is also elevated [3]. There is a growing body of evidence indicating that repro- grammed cancer metabolism is a potential therapeutic target. microRNAs (miRNAs) are an evolutionarily conserved group of small non-coding RNAs (18–24 nucleotides) that suppress gene expression [4]. miRNA genes are usually transcribed by RNA poly- merase II as longer miRNA precursors called pri-miRNAs. Once transcribed, pri-miRNAs are recognized by Drosha and processed into pre-miRNAs, which are exported from the nucleus to the cytoplasm through the protein Exportin-5. In the cytoplasm, pre- miRNAs are cleaved into mature miRNAs by Dicer endonucleases. Mature miRNAs directly bind with their target genes via se- quence-specific interactions with 3 0 untranslated regions (UTR) of cognate mRNA targets, resulting in suppression of mRNA transla- tion or mRNA decay [5]. miRNAs are now known as essential regulators of development and physiological processes. By regulating oncogenic and tumor suppressor signal pathways, miRNAs might also play significant roles in cancer development. Recently, a large number of miRNAs were identified as important natural regulators of metabolism [6]. For example, miR-33a and miR-33b play a key role in regulat- ing fatty acid degradation and cholesterol homeostasis by acting in concert with their host genes, the sterol-regulatory element- binding protein (SREBP) transcription factors [7]; numerous miR- NAs, (e.g., miR-21, miR-143, miR-130, miR-27a) are reported to be involved in white-adipocyte differentiation [8]; and inhibition of miR-122 is thought to decrease plasma triglyceride and choles- terol concentrations [9]. Aberrant expression of miR-181a has been reported in several types of cancer and appears to be significantly associated with the clinical outcome of human cancer patients. For example, miR-181a is significantly downregulated in primary glioblastomas and in non-small cell lung cancers [10,11], but it is significantly overexpressed in breast cancer cells, hepatocellular carcinoma cells, and human gastric cancer tissues [12–14]. Overexpression of miR-181a is correlated with lymph node metastasis, vascular invasion, and poor survival [15]. The direct role of miR-181a in metabolism has not yet been reported. In this study, we found that miR-181a is overexpressed http://dx.doi.org/10.1016/j.febslet.2014.03.037 0014-5793/Ó 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. Corresponding author at: Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China. E-mail address: [email protected] (D. Zhang). 1 These authors contributed equally to this work. FEBS Letters 588 (2014) 1773–1779 journal homepage: www.FEBSLetters.org

Transcript of miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in...

Page 1: miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Weia,1, Long Cuib,1, Zubing

FEBS Letters 588 (2014) 1773–1779

journal homepage: www.FEBSLetters .org

miR-181a mediates metabolic shift in colon cancer cellsvia the PTEN/AKT pathway

http://dx.doi.org/10.1016/j.febslet.2014.03.0370014-5793/� 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.

⇑ Corresponding author at: Department of Biochemistry & Molecular Cell Biology,Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road,Shanghai 200025, China.

E-mail address: [email protected] (D. Zhang).1 These authors contributed equally to this work.

Zhonghua Wei a,1, Long Cui b,1, Zubing Mei b, Min Liu a, Daoxiang Zhang a,b,c,⇑a Department of Pathology of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Chinab Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Chinac Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China

a r t i c l e i n f o

Article history:Received 18 January 2014Revised 25 February 2014Accepted 4 March 2014Available online 28 March 2014

Edited by Tamas Dalmay

Keywords:miR-181aPTENGlycolysis

a b s t r a c t

Cancer cell metabolism is often characterized by a shift from an oxidative to a glycolytic bioenerget-ics pathway, a phenomenon known as the warburg effect. Whether the deregulation of miRNAs con-tributes to the warburg effect remains largely unknown. Here we show that miR-181a expression isincreased and thus induces a metabolic shift in colon cancer cells. miR-181a performs this functionby inhibiting the expression of PTEN, leading to an increase of phosphorylated AKT which triggersmetabolic shift. The increase of lactate production induced by miR-181a results in the rapid growthof cancer cells. These results identify miR-181a as a molecular switch involved in the orchestrationof the warburg effect in colon cancer cells via the PTEN/AKT pathway.� 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.

1. Introduction Mature miRNAs directly bind with their target genes via se-

The metabolism of cancer cells is significantly different fromthat of normally differentiated cells [1]. Normally differentiatedcells rely primarily on the oxidation of pyruvate in the mitochon-dria to generate energy for cellular physiology; however, even withsufficient oxygen, rapidly growing cancer cells rely on aerobic gly-colysis to generate energy. This phenomenon is termed ‘‘the War-burg effect’’ [2]. This metabolic shift towards enhanced glycolysisis believed to be the result of adaptations to support the continu-ous proliferation of cancer cells. Flux along other key metabolicpathways, such as the glutaminolysis pathway, is also elevated[3]. There is a growing body of evidence indicating that repro-grammed cancer metabolism is a potential therapeutic target.

microRNAs (miRNAs) are an evolutionarily conserved group ofsmall non-coding RNAs (18–24 nucleotides) that suppress geneexpression [4]. miRNA genes are usually transcribed by RNA poly-merase II as longer miRNA precursors called pri-miRNAs. Oncetranscribed, pri-miRNAs are recognized by Drosha and processedinto pre-miRNAs, which are exported from the nucleus to thecytoplasm through the protein Exportin-5. In the cytoplasm, pre-miRNAs are cleaved into mature miRNAs by Dicer endonucleases.

quence-specific interactions with 30 untranslated regions (UTR) ofcognate mRNA targets, resulting in suppression of mRNA transla-tion or mRNA decay [5].

miRNAs are now known as essential regulators of developmentand physiological processes. By regulating oncogenic and tumorsuppressor signal pathways, miRNAs might also play significantroles in cancer development. Recently, a large number of miRNAswere identified as important natural regulators of metabolism[6]. For example, miR-33a and miR-33b play a key role in regulat-ing fatty acid degradation and cholesterol homeostasis by acting inconcert with their host genes, the sterol-regulatory element-binding protein (SREBP) transcription factors [7]; numerous miR-NAs, (e.g., miR-21, miR-143, miR-130, miR-27a) are reported tobe involved in white-adipocyte differentiation [8]; and inhibitionof miR-122 is thought to decrease plasma triglyceride and choles-terol concentrations [9].

Aberrant expression of miR-181a has been reported in severaltypes of cancer and appears to be significantly associated withthe clinical outcome of human cancer patients. For example,miR-181a is significantly downregulated in primary glioblastomasand in non-small cell lung cancers [10,11], but it is significantlyoverexpressed in breast cancer cells, hepatocellular carcinomacells, and human gastric cancer tissues [12–14]. Overexpressionof miR-181a is correlated with lymph node metastasis, vascularinvasion, and poor survival [15].

The direct role of miR-181a in metabolism has not yet beenreported. In this study, we found that miR-181a is overexpressed

Page 2: miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Weia,1, Long Cuib,1, Zubing

Fig. 1. miR-181a is overexpressed in colon cancer. (A) Heatmap of selected miRNAexpression in colon cancer cells compared with selected miRNA expression innormal tissue. T: tumor; N: normal tissue. (B) Real-time PCR analysis of miR-181acontent in colon cancer tissue compared with miR-181a content in normal colontissue.

1774 Z. Wei et al. / FEBS Letters 588 (2014) 1773–1779

in human colon cancer tissue. Ectopic overexpression of miR-181aenhanced cell proliferation through increased glycolysis. A lucifer-ase reporter assay showed that miR-181a suppressed PTEN expres-sion by directly targeting its 30-UTR, thus resulting in increasedAKT phosphorylation. These data suggest that miR-181a causesan increase in lactate production and promotes cell proliferationthrough the PTEN/AKT pathway.

2. Materials and methods

2.1. Cell culture and tissue samples

HCT15 and HCT116 colon cancer cells were purchased from theAmerican Type Culture Collection (ATCC). HCT15 and HCT116 cellswere cultured in Dulbecco’s Modified Eagle’s Medium (DMEM;Invitrogen, California) supplemented with 10% fetal bovine serum(FBS; Invitrogen, California), 4.5 g/L glucose, and 100 units/mlpenicillin/streptomycin in a tissue incubator maintained at 37 �C,5% CO2, and 90% humidity. LY294002 and 2-DG were obtainedfrom Sigma–Aldrich and dissolved in dimethyl sulfoxide (DMSO).

Colon cancer and adjacent non-tumor tissue samples were col-lected from patients undergoing surgical resection at the ShanghaiJiao Tong University Affiliated Sixth People’s Hospital. The specimenswere immediately frozen and stored at �80 �C in an ultra-low-temperature freezer. Pathological examination and quantitativereal-time PCR were used to confirm cancer diagnosis.

2.2. Transfections

All transfections were performed using Lipofectamine 2000(Invitrogen, Carlsbad, CA, USA) according to the manufacturer’sinstructions. The miR-181a inhibitor, the negative controls andthe miR-181a mimics were purchased from GenePharma (China,Shanghai). The miR-181a inhibitor and the miR-181a mimics wereused at a final concentration of 50 nM.

2.3. Luciferase reporter assay

For the luciferase reporter assay, 293T cells were seeded in a 24-well plate and grown to 80–90% confluence. Lipofectamine 2000was then used to cotransfect cells with either 50 nM aliquots ofthe miR-181a mimics or 50 nM aliquots of a scramble and either40 ng of pmirGLO-PTEN-30UTR-WT or 40 ng of pmirGLO-PTEN-30UTR-MUT, according to the manufacturer’s instructions. After48 h, the cells were collected and lysed with a passive lysis bufferand then analyzed using a Dual-Luciferase Reporter Assay System(Promega). Luciferase activity was detected using a M200 micro-plate fluorescence reader. The constitutive expression of Renillaluciferase was used as an internal control to correct for differencesin harvest efficiencies and transfection. Transfections were per-formed in duplicate and repeated in at least three independentexperiments.

2.4. RNA extraction and real-time PCR

The culture medium was removed, and the cells were immedi-ately washed with ice-cold PBS. Subsequently, 1 ml of TRIzol re-agent was added. and the cellular RNA was extracted accordingto the acid guanidinium thiocyanate-phenol–chloroform method.MMLV-RT reactions were used to synthesize cDNA from 1 lg of to-tal RNA according to the manufacturer’s instructions. Real-timequantitative reactions were performed in triplicate in a 96-wellplate containing 1 ll of synthesized cDNA, SYBR Green PCR mix,and gene-specific forward and reverse PCR primers. The resultswere normalized using b-actin. The melting curves were analyzed

to verify the specificity of the RT-PCR reaction and the absence ofprimer dimer formation. The primers used for detection are as fol-lows: mTOR-F: 50-TCGCTGAAGTCACACAGACC-30; mTOR-R: 50-CTTTGGCATATGCTCGGCAC-30; rictor-F: 50-ACAACTGGGATGCTGTGAGG-30; rictor-R: 50-CCAAACCGGTCATCCTCCAA-30; mSin-F: 50-TTGGCCCACAGAAAGGATGG-30; mSin-R: 50-TGAGCTGTGTTTGAGCGTCT-30; mLST8-F: 50-GGTGAATGCCTTGGAGGTCA-30; mLST8-R: 50-TGCGTTCACCTGGAAGATCC-30; PRR5-F: 50-AGTTCATGAGTTCGCCCAGC-30; PRR5-R: 50-GTGAAGAAGGACCCCAGCTC; PTEN-F: 50-GTGGTCTGCCAGCTAAAGGT-30; PTEN-R: 50-TCACCACACACAGGTAACGG-30.

For a quantification of miRNAs, a TaqMan MicroRNA reverse tran-scription kit was used according to the manufacturer’s instructions.The results were normalized to RNU48. The stem-loop primersused for miR-181a and RNU48 are as follows: miR-181a-stem-loopprimer: 50-GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACACTCAC-30; miR-181a-F: 50-GCGGCGAACATTCAACGATG-30; miR-181a-R: 50-GTGCAGGGTCCGAGG-30; RNU48-F: 50-TGATGATGACCCCAGGTAACTC-30; RNU48-R: 50-GAGCGCTGCGGTGATG-30.

2.5. Western blots

Protein was harvested with RIPA buffer, quantified, and then re-solved on an SDS–PAGE gel. The protein was then transferred ontoa membrane, which was blocked and incubated with an appropri-ate primary antibody. After being washed, the membranes wereincubated with a secondary antibody. After three more washes,the proteins were detected by chemiluminescence.

Page 3: miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Weia,1, Long Cuib,1, Zubing

Fig. 2. miR-181a promotes the proliferation of colon cancer cells. (A) Real-time PCR analysis of miR-181a expression in HCT15 and HCT116 cancer cells transfected with miR-181a mimics or a miR-181 inhibitor. (B) CCK8 assay results show that the growth rate of miR-181a-overexpressing colon cancer cells is faster that the growth rate of controlcells. (C) CCK8 assay results show that the growth rate of miR-181a knockdown colon cancer cells is slower that the growth rate of control cells. (D) Western blot assays showthat cyclin D1 expression is induced by miR-181a in colon cancer cells.

Z. Wei et al. / FEBS Letters 588 (2014) 1773–1779 1775

2.6. Cell viability assays

Five-thousand cells/well were seeded into a 96-well plate andtreated with miR-181a mimics or a miR-181a inhibitor. After incu-bation for 24 h, 48 h, 72 h, or 96 h, the cells were labeled with CellCounting Kit-8 (CCK8) (DOJINDO. Tabaru, JAPAN) for 2 h. Cell via-bility was measured at an absorbance of 450 nm.

2.7. Measurement of extracellular lactate

Fifty-thousand cells that had been transfected with miR-181amimics or a miR-181a inhibitor were seeded in 60-mm dishesand incubated in DMEM with 10% FBS overnight. Then, the media

was removed, and the cells were incubated in DMEM without FBS.After incubation for 1 h, the supernatant was collected. The con-centration of lactate in the supernatant was quantified using a col-orimetric assay according to the manufacturer’s instructions(Lactate Assay Kit, Biovision).

2.8. Measurement of glucose uptake

After 12 h of serum starvation, cells were cultured in DMEMcontaining 25 mM glucose. To measure the rate of glucose uptake,the cells were washed three times with PBS and then incubated for3 h in DMEM containing 1 mCi/ml 2-deoxy-2[1,2-3H] glucose(PerkinElmer Life Sciences). The cells were washed three times

Page 4: miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Weia,1, Long Cuib,1, Zubing

1776 Z. Wei et al. / FEBS Letters 588 (2014) 1773–1779

with ice-cold PBS and solubilized in 1% SDS. A scintillation counterwas used to determine the radioactivity of each aliquot. Each assaywas performed in triplicate.

2.9. Statistical analyses

Statistical analysis was performed with GraphPad Prism 5software. Student’s t-tests were used to determine statisticalsignificance. The significance level was set at P < 0.05 for allanalyses.

3. Results

3.1. miR-181a is overexpressed in colon cancer

First, we analyzed the miRNA expression profile from a previ-ously published dataset (Gene expression omnibus accessionGSE7828) and found that miR-181a was overexpressed in coloncancers (Fig. 1A). Next, we confirmed the expression of miR-181ain colon cancer tissues. qRT-PCR analysis showed that among eightmatched sample pairs, miR-181a expression was significantlyhigher in all clinical colon cancer tissues compared with adjacentnon-tumor tissue acquired from a site 4 cm away from the tumor’smargin (Fig. 1B).

3.2. miR-181a promotes the proliferation of colon cancer cells

To assess the role of miR-181a in the growth of colon cancercells, we transfected HCT15 and HCT116 colon cancer cells with50 nM aliquots of miR-181a mimics or 50 nM aliquots of a

Fig. 3. miR-181a induces a metabolic shift in colon cancer cells. (A) Lactate productionshow that miR-181a increases the rate of glucose uptake. (C) Western blot assays show th(D) CCK8 assays show that inhibition of glycolysis with 2-DG blocks miR-181a-induced

miR-181a inhibitor. The qRT-PCR analysis revealed that miR-181aexpression was dramatically and specifically increased in the mi-mic-transfected cells and decreased in the inhibitor-transfectedcells (Fig. 2A). A CCK8 assay showed that cell proliferation wasgreater in miR-181a-overexpressing colon cancer cells than inthe control cells (Fig. 2B). We then asked whether reducing miR-181a in colon cancer cells inhibited cell proliferation. To addressthis question, we treated HCT116 and HCT15 cancer cells with amiR-181a inhibitor. As expected, loss of miR-181a function atten-uated cell proliferation (Fig. 2C).

To further explore the effect of miR-181a on cell cycle progres-sion, we used Western blot analysis to demonstrate that overex-pression of miR-181a enhanced the level of cyclin D1, a criticalcell cycle component (Fig. 2D). Taken together, these results sug-gest that miR-181a enhanced cell proliferation.

3.3. miR-181a induces a metabolic shift in colon cancer cells

We next sought to determine whether miR-181a was capable ofinducing a metabolic shift in colon cancer cells. Induction of miR-181a in colon cancer cells increased glucose uptake and lactatesecretion (Fig. 3A and B). Conversely, the use of an inhibitor toknockdown miR-181a expression reduced lactate secretion andglucose uptake (Fig. 3A and B). Glut1 and HK2 are key cell glycol-ysis genes. Western blot analysis showed that miR-181a increasedthe expression of Glut1 and HK2 (Fig. 3C).

To determine whether a miR-181a-induced metabolic switch isrequired for miR-181a-dependent cell proliferation, we treated miR-181a mimic-transfected HCT116 cancer cells with 2-deoxyglucose(2-DG), a glucose analog that inhibits glycolysis via its action on

assays show that miR-181a stimulates lactate secretion. (B) Glucose uptake assaysat the expression of HK2 and Glut1 is upregulated in miR-181a overexpressing cells.increases in colon cancer cell proliferation.

Page 5: miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Weia,1, Long Cuib,1, Zubing

Z. Wei et al. / FEBS Letters 588 (2014) 1773–1779 1777

hexokinase, and analyzed cell numbers with a CCK8 assay. The re-sults demonstrated that inhibition of miR-181a-induced glycolysisis sufficient to block miR-181a-induced cell proliferation (Fig. 3D).Taken together, these results suggest that miR-181a-induced glycol-ysis is responsible for the observed increase in colon cancer cellproliferation.

3.4. Phosphorylated AKT is required for the metabolic shift induced bymiR-181a

Previous reports have demonstrated that hypoxia inducible fac-tors (HIFs) and the proto-oncogene c-Myc play a central role in reg-ulating cell glycolysis. We detected HIF1-a and c-Myc proteinlevels in two miR-181a-overexpressing cell lines: HCT15 andHCT116. Western blot analysis revealed no differences in the pro-tein expression of HIF1-a and c-Myc between miR-181a-over-expressing colon cancer cells and control cells (Fig. 4A). Recentstudies have indicated that miR-181a expression increases Erk1/2, AKT, and Src signaling in breast cancer cells. In addition, AKTactivity renders cancer cells dependent on aerobic glycolysis forcontinued growth and survival [15,16]. Therefore, we hypothesizedthat miR-181a would stimulate cancer cell glycolysis by increasingAKT activity. Consistent with this prediction, overexpression ofmiR-181a in colon cancer cells was sufficient to induce AKT S473phosphorylation without affecting the total AKT level (Fig. 4A).These data suggest that increased miR-181a expression is associ-ated with an increase in AKT S473 phosphorylation. To determinewhether AKT activity might account for the metabolic shift in-duced by miR-181a, we treated miR-181a-overexpressing cellswith LY294002, a cell-permeable specific phosphatidylinositol3-kinase inhibitor. Western blot analysis showed that LY294002significantly inhibits the phosphorylation of AKT and decreases

Fig. 4. Phosphorylated AKT is required for the miR-181a-induced metabolic shift. (A) Waffect total AKT, c-Myc, or HIF1-a expression. (B) Western blot assays show that inhibitioHK2 and Glut1. (C) Inhibition of AKT phosphorylation with LY294002 suppresses miR-181LY294002 suppresses miR-181a-induced increases in the rate of glucose uptake.

miR-181a-induced expression of Glut1 and HK2 (Fig. 4B). Thesedata indicate that the inhibition of AKT activity in miR-181a-over-expressing cells is sufficient to decrease colon cancer-inducedalterations in the rate of glycolysis via a reduction of Glut1 andHK2 expression. Furthermore, LY294002 inhibited the increasedrate of glucose and lactate production observed in miR-181a-overexpressing cells (Fig. 4C and D). These data suggest thatAKT activity is required for miR-181a-induced changes inglycolysis.

3.5. miR-181a suppresses expression of PTEN

Three different mRNA target-predicting algorithms (TargetScan,miRanda, and targetRank) were used to identify potential directtargets of miR-181a based on binding sites in the 30UTR. We se-lected candidate genes that were associated with AKT phosphory-lation. mRNA target-predicting algorithms identified an areawithin the 30UTR of PTEN as a potential target of miR-181a.qRT-PCR analysis was used to verify PTEN as a target of miR-181a. mRNA expression of PTEN was decreased in miR-181a-over-expressing cells; however, there were no significant changes in themRNA expression of the essential components of mTORC2 (i.e.,mTOR, rictor, mSin1, mLST8, PRR5), which phosphorylates mem-bers of the AGC kinase family, including AKT (Fig. 5A). Western blotanalysis also confirmed that expression of miR-181a decreased theprotein level of PTEN (Fig. 5B).

To further determine whether PTEN is a direct target of miR-181a, we constructed a wild-type dual-luciferase UTR vector thatcontained the 30UTR of PTEN and a mutant dual-luciferase UTR vec-tor that harbored mutations in the miR-181a binding sites of the30UTR of PTEN (PTEN-mut) (Fig. 5C). As expected, miR-181a effec-tively suppressed the luciferase reporter activity of the wild-type

estern blot assays show that miR-181a increases AKT phosphorylation but does notn of AKT phosphorylation with LY294002 blocks miR-181a-induced upregulation ofa-induced increases in lactate secretion. (D) Inhibition of AKT phosphorylation with

Page 6: miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Weia,1, Long Cuib,1, Zubing

Fig. 5. miR-181a suppresses expression of PTEN. (A) Real-time PCR shows that overexpression of miR-181a significantly downregulates mRNA expression of PTEN but doesnot affect mRNA expression of mTORC2 components. (B) Western blot assays suggest that overexpression of miR-181a suppresses the PTEN expression. (C) This sequence inthe 30UTR of PTEN was predicted to bind to miR-181a. The nucleotides in res were mutated into their complementary nucleotides in Mut 30UTR. (D) Dual-luciferase assaysshow that miR-181a suppresses luciferase activity in cells containing the 30UTR wild type vector, but miR-181a does not affect luciferase activity in cells containing the 30UTRmutant vector.

1778 Z. Wei et al. / FEBS Letters 588 (2014) 1773–1779

vector (Fig. 5D), but this decreased luciferase activity was reversedwhen cells were transfected with the mutant vector (Fig. 5D).Taken together, these results demonstrate that miR-181a directlyregulates PTEN expression through binding sites in the 30UTR.

4. Discussion

miRNAs have attracted increased attention due to their keyrole in many biological events, including cell proliferation, cellmotility, differentiation, and apoptosis [17]. In recent years, thedysregulation of miR-181a has been associated with a variety ofhuman cancers. Significant miR-181a downregulation has been re-ported in primary glioblastomas, squamous cell cancers, and non-small cell lung carcinomas. However, significant miR-181a overex-pression has been reported in MCF-7 breast cancer cells, humangastric cancer tissues, and HCC cells. Therefore, the function ofmiR-181a in tumorigenesis remains unclear.

In this study, we found that miR-181a expression was higher incolon cancer tissues compared with adjacent normal tissues(Fig. 1B). This elevation was consistent with the miRNA profile dif-ferences between colon adenocarcinomas and paired non-tumor-ous colon cells (GSE7828). Previous reports have demonstratedthat miR-181a promotes gastric cancer cell proliferation and col-ony formation in vitro and HCC xenograft growth in nude mice[14]. Therefore, we hypothesized that miR-181a may act as anoncomir to enhance cell proliferation. The results of the CCK8 assayindicated that miR-181a promoted cell proliferation (Fig. 2B andC). Cyclin D1, a cell cycle checkpoint protein [18], was also upreg-ulated in miR-181a-overexpressing cells, further confirming thatmiR-181a regulated cell growth. However, further study is re-quired to determine how miR-181a influences the expression ofCyclin D1.

The molecular mechanisms responsible for shifting energymetabolism to the advantage of cancer cells are complex and onlypartially understood. In this study, we demonstrated that miR-181a plays an important role in reprogramming the metabolismof colon cells. Overexpression of miR-181a increases glucose

uptake and lactate secretion, while knockdown of miR-181adecreases glucose uptake and lactate secretion (Fig. 3A and B).The altered metabolism induced by miR-181a is required forcancer cell growth (Fig. 3D).

Moreover, we found that PTEN is a direct target of miR-181a.The suppression of PTEN increases AKT S473 phosphorylation,which is involved in glycolysis [19]. Inhibition of AKT S473 phos-phorylation with LY294002 blocks miR-181a-induced glycolysis(Fig. 4C and D). Previous reports have demonstrated that mTORC2regulates cell glycolysis by phosphorylating AKT [19], but in thisstudy, we found no significant changes in the critical componentsof mTORC2, including mTOR, rictor, mSin1, mLST8, and PRR5. Like-wise, we did not detect significant changes in the expression ofc-Myc and HIF1-a, both of which are well-established transcrip-tional factors. Dysregulation of miR-181a in colon cancers has beenextensively investigated for its role in aerobic glycolysis.

In summary, our study provides direct evidence that miR-181apromotes colon cancer cell glycolysis through the miR-181a-PTEN-AKT cascade, thus promoting cell proliferation. These results iden-tify miR-181a as a molecular switch involved in the orchestrationof the Warburg effect in colon cancer cells and as a potential ther-apeutic target for colon cancer.

References

[1] Kroemer, G. and Pouyssegur, J. (2008) Tumor cell metabolism: cancer’sAchilles’ heel. Cancer Cell 13, 472–482.

[2] Vander Heiden, M.G., Cantley, L.C. and Thompson, C.B. (2009) Understandingthe Warburg effect: the metabolic requirements of cell proliferation. Science324, 1029–1033.

[3] Munoz-Pinedo, C., El Mjiyad, N. and Ricci, J.E. (2012) Cancer metabolism:current perspectives and future directions. Cell Death Dis. 3, e248.

[4] Ameres, S.L. and Zamore, P.D. (2013) Diversifying microRNA sequence andfunction. Nat. Rev. Mol. Cell Biol. 14, 475–488.

[5] Bartel, D.P. (2009) MicroRNAs: target recognition and regulatory functions.Cell 136, 215–233.

[6] Rottiers, V. and Naar, A.M. (2012) MicroRNAs in metabolism and metabolicdisorders. Nat. Rev. Mol. Cell Biol. 13, 239–250.

[7] Rayner, K.J., Suarez, Y., Davalos, A., Parathath, S., Fitzgerald, M.L., Tamehiro, N.,Fisher, E.A., Moore, K.J. and Fernandez-Hernando, C. (2010) MiR-33 contributesto the regulation of cholesterol homeostasis. Science 328, 1570–1573.

Page 7: miR-181a mediates metabolic shift in colon cancer cells ... · miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway Zhonghua Weia,1, Long Cuib,1, Zubing

Z. Wei et al. / FEBS Letters 588 (2014) 1773–1779 1779

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

[9] Esau, C., Davis, S., Murray, S.F., Yu, X.X., Pandey, S.K., Pear, M., Watts, L., Booten,S.L., Graham, M., McKay, R., Subramaniam, A., Propp, S., Lollo, B.A., Freier, S.,Bennett, C.F., Bhanot, S. and Monia, B.P. (2006) MiR-122 regulation of lipidmetabolism revealed by in vivo antisense targeting. Cell Metab. 3, 87–98.

[10] Ciafre, S.A., Galardi, S., Mangiola, A., Ferracin, M., Liu, C.G., Sabatino, G., Negrini,M., Maira, G., Croce, C.M. and Farace, M.G. (2005) Extensive modulation of aset of microRNAs in primary glioblastoma. Biochem. Biophys. Res. Commun.334, 1351–1358.

[11] Gao, W., Yu, Y., Cao, H., Shen, H., Li, X., Pan, S. and Shu, Y. (2010) Deregulatedexpression of miR-21, miR-143 and miR-181a in non small cell lung cancer isrelated to clinicopathologic characteristics or patient prognosis. Biomed.Pharmacother. 64, 399–408.

[12] Jiao, X., Zhao, L., Ma, M., Bai, X., He, M., Yan, Y., Wang, Y., Chen, Q., Zhao, X., Zhou,M., Cui, Z., Zheng, Z., Wang, E. and Wei, M. (2013) MiR-181a enhances drugsensitivity in mitoxantone-resistant breast cancer cells by targeting breastcancer resistance protein (BCRP/ABCG2). Breast Cancer Res. Treat. 139, 717–730.

[13] Meng, F., Glaser, S.S., Francis, H., DeMorrow, S., Han, Y., Passarini, J.D., Stokes,A., Cleary, J.P., Liu, X., Venter, J., Kumar, P., Priester, S., Hubble, L., Staloch, D.,

Sharma, G., Liu, C.G. and Alpini, G. (2012) Functional analysis of microRNAs inhuman hepatocellular cancer stem cells. J. Cell. Mol. Med. 16, 160–173.

[14] Zhang, X., Nie, Y., Du, Y., Cao, J., Shen, B. and Li, Y. (2012) MicroRNA-181apromotes gastric cancer by negatively regulating tumor suppressor KLF6.Tumour Biol. 33, 1589–1597.

[15] Taylor, M.A., Sossey-Alaoui, K., Thompson, C.L., Danielpour, D. and Schiemann,W.P. (2013) TGF-beta upregulates miR-181a expression to promote breastcancer metastasis. J. Clin. Invest. 123, 150–163.

[16] Elstrom, R.L., Bauer, D.E., Buzzai, M., Karnauskas, R., Harris, M.H., Plas, D.R.,Zhuang, H., Cinalli, R.M., Alavi, A., Rudin, C.M. and Thompson, C.B. (2004) Aktstimulates aerobic glycolysis in cancer cells. Cancer Res. 64, 3892–3899.

[17] Jansson, M.D. and Lund, A.H. (2012) MicroRNA and cancer. Mol. Oncol. 6, 590–610.

[18] Baldin, V., Lukas, J., Marcote, M.J., Pagano, M. and Draetta, G. (1993) Cyclin D1is a nuclear protein required for cell cycle progression in G1. Genes Dev. 7,812–821.

[19] Hagiwara, A., Cornu, M., Cybulski, N., Polak, P., Betz, C., Trapani, F., Terracciano,L., Heim, M.H., Ruegg, M.A. and Hall, M.N. (2012) Hepatic mTORC2 activatesglycolysis and lipogenesis through Akt, glucokinase, and SREBP1c. Cell Metab.15, 725–738.