14
Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai 1 , Iris Ming-Jing Xu 1 , David Kung-Chun Chiu 1 , Aki Pui-Wah Tse 1 , Larry Lai Wei 1 , Cheuk-Ting Law 1 , Derek Lee 1 , Chun-Ming Wong 1,2 , Maria Pik Wong 1 , Irene Oi-Lin Ng 1,2 , and Carmen Chak-Lui Wong 1,2 Abstract Purpose: Hepatocellular carcinoma (HCC) lacks effective cura- tive therapy. Hypoxia is commonly found in HCC. Hypoxia elicits a series of protumorigenic responses through hypoxia-inducible factor-1 (HIF1). Better understanding of the metabolic adapta- tions of HCC cells during hypoxia is essential to the design of new therapeutic regimen. Experimental Design: Expressions of genes involved in the electron transport chain (ETC) in HCC cell lines (20% and 1% O 2 ) and human HCC samples were analyzed by transcriptome sequencing. Expression of NDUFA4L2, a less active subunit in complex I of the ETC, in 100 pairs of HCC and nontumorous liver tissues were analyzed by qRT-PCR. Student t test and KaplanMeier analyses were used for clinicopathologic correlation and survival studies. Orthotopic HCC implantation model was used to evaluate the efciency of HIF inhibitor. Results: NDUFA4L2 was drastically overexpressed in human HCC and induced by hypoxia. NDUFA4L2 overexpression was closely associated with tumor microsatellite formation, absence of tumor encapsulation, and poor overall survival in HCC patients. We conrmed that NDUFA4L2 was HIF1-regulated in HCC cells. Inactivation of HIF1/NDUFA4L2 increased mitochon- drial activity and oxygen consumption, resulting in ROS accu- mulation and apoptosis. Knockdown of NDUFA4L2 markedly suppressed HCC growth and metastasis in vivo. HIF inhibitor, digoxin, signicantly suppressed growth of tumors that expressed high level of NDUFA4L2. Conclusions: Our study has provided the rst clinical relevance of NDUFA4L2 in human cancer and suggested that HCC patients with NDUFA4L2 overexpression may be suitable candidates for HIF inhibitor treatment. Clin Cancer Res; 22(12); 310517. Ó2016 AACR. Introduction Hepatocellular carcinoma (HCC), a malignancy derived from hepatocytes, accounts for 90% of primary liver cancer. It is the fth most common cancer and the third leading cause of cancer deaths in the world (1). Majority of deaths in HCC are attributed to its asymptomatic nature that delays diagnosis and treatment. Most HCC patients are not suitable for the only promising curative therapies, surgical resection, and liver trans- plantation. Sorafenib, an oral multikinase inhibitor and the only FDA-approved drug for advanced HCC patients, could modestly prolong the survival of patients for 3 months (2, 3). Liver is an organ responsible for many important metabolic functions in the body, such as the Cori-cycle, glycogen metab- olism, and blood glucose homeostasis. Hepatocarcinogenesis is accompanied by the loss of normal metabolic functions in the liver and the acquisition of new metabolic functions which favor cancer growth. Exploration of the molecular contexts associated with these metabolic changes will help to identify novel targets for HCC treatment. Hypoxia, or oxygen (O 2 ) deprivation, is frequently found in regions of HCC that are distant from functional vasculature. Palliative therapies, such as transcatheter arterial (chemo) embolization (TAE/TACE) and hepatic artery ligation, that involve restriction of blood supply to the tumors adversely induce hypoxia (4). To overcome the shortage of O 2 , cells adapt to hypoxia through HIFs which are composed of the constitu- tively expressed HIF1b subunit and the oxygen labile subunit HIF1/2a (5). In the presence of O 2 , HIF1/2a is hydroxylated by prolyl hydroxylases (6), facilitating binding of von HippelLindau (VHL), which polyubiquitinates HIF1/2a for proteaso- mal degradation (7). In the absence of O 2 , stabilized HIF1/2a dimerizes with HIF1b to initiate transcription of genes related to hypoxia adaptive responses that advantage cancer develop- ment (8). Although it is known that upregulation of HIF1 is closely associated with poor clinical outcome in HCC patients (4), the detailed molecular mechanisms by which HIF1 pro- motes HCC progression remain poorly understood. HIF1a transcriptionally activates many metabolic genes, allowing the cells to adapt to hypoxia, by shunting the glucose intermediates into glycolysis instead of the tricarboxylic acid (TCA) cycle (9). These metabolic genes include glucose trans- porter (GLUT1), hexokinase 2 (HK2), lactate dehydrogenase A 1 Department of Pathology,The University of Hong Kong, Hong Kong. 2 State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Authors: Carmen Chak-Lui Wong, The University of Hong Kong, L-704 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, Hong Kong. Phone: 852-39179014; Fax: 852-28195375l; E-mail: [email protected]; Irene Oi-Lin Ng, [email protected] doi: 10.1158/1078-0432.CCR-15-1987 Ó2016 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 3105 Research. on September 27, 2020. © 2016 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

Biology of Human Tumors

NDUFA4L2 Fine-tunes Oxidative Stress inHepatocellular CarcinomaRobin Kit-Ho Lai1, Iris Ming-Jing Xu1, David Kung-Chun Chiu1, Aki Pui-Wah Tse1,Larry Lai Wei1, Cheuk-Ting Law1, Derek Lee1, Chun-Ming Wong1,2,Maria Pik Wong1, Irene Oi-Lin Ng1,2, and Carmen Chak-Lui Wong1,2

Abstract

Purpose:Hepatocellular carcinoma (HCC) lacks effective cura-tive therapy.Hypoxia is commonly found inHCC.Hypoxia elicitsa series of protumorigenic responses through hypoxia-induciblefactor-1 (HIF1). Better understanding of the metabolic adapta-tions of HCC cells during hypoxia is essential to the design of newtherapeutic regimen.

Experimental Design: Expressions of genes involved in theelectron transport chain (ETC) inHCCcell lines (20%and1%O2)and human HCC samples were analyzed by transcriptomesequencing. Expression of NDUFA4L2, a less active subunit incomplex I of the ETC, in 100 pairs of HCC and nontumorous livertissues were analyzed by qRT-PCR. Student t test and Kaplan–Meier analyses were used for clinicopathologic correlation andsurvival studies. Orthotopic HCC implantation model was usedto evaluate the efficiency of HIF inhibitor.

Results: NDUFA4L2 was drastically overexpressed in humanHCC and induced by hypoxia. NDUFA4L2 overexpression wasclosely associated with tumor microsatellite formation, absenceof tumor encapsulation, and poor overall survival in HCCpatients. We confirmed that NDUFA4L2 was HIF1-regulated inHCC cells. Inactivation of HIF1/NDUFA4L2 increasedmitochon-drial activity and oxygen consumption, resulting in ROS accu-mulation and apoptosis. Knockdown of NDUFA4L2 markedlysuppressed HCC growth and metastasis in vivo. HIF inhibitor,digoxin, significantly suppressed growth of tumors that expressedhigh level of NDUFA4L2.

Conclusions:Our study has provided the first clinical relevanceof NDUFA4L2 in human cancer and suggested that HCC patientswith NDUFA4L2 overexpression may be suitable candidates forHIF inhibitor treatment. ClinCancer Res; 22(12); 3105–17.�2016AACR.

IntroductionHepatocellular carcinoma (HCC), a malignancy derived

from hepatocytes, accounts for 90% of primary liver cancer.It is the fifth most common cancer and the third leading causeof cancer deaths in the world (1). Majority of deaths in HCC areattributed to its asymptomatic nature that delays diagnosis andtreatment. Most HCC patients are not suitable for the onlypromising curative therapies, surgical resection, and liver trans-plantation. Sorafenib, an oral multikinase inhibitor and theonly FDA-approved drug for advanced HCC patients, couldmodestly prolong the survival of patients for 3 months (2, 3).Liver is an organ responsible for many important metabolicfunctions in the body, such as the Cori-cycle, glycogen metab-olism, and blood glucose homeostasis. Hepatocarcinogenesis is

accompanied by the loss of normal metabolic functions in theliver and the acquisition of new metabolic functions whichfavor cancer growth. Exploration of the molecular contextsassociated with these metabolic changes will help to identifynovel targets for HCC treatment.

Hypoxia, or oxygen (O2) deprivation, is frequently found inregions of HCC that are distant from functional vasculature.Palliative therapies, such as transcatheter arterial (chemo)embolization (TAE/TACE) and hepatic artery ligation, thatinvolve restriction of blood supply to the tumors adverselyinduce hypoxia (4). To overcome the shortage of O2, cells adaptto hypoxia through HIFs which are composed of the constitu-tively expressed HIF1b subunit and the oxygen labile subunitHIF1/2a (5). In the presence of O2, HIF1/2a is hydroxylated byprolyl hydroxylases (6), facilitating binding of von Hippel–Lindau (VHL), which polyubiquitinates HIF1/2a for proteaso-mal degradation (7). In the absence of O2, stabilized HIF1/2adimerizes with HIF1b to initiate transcription of genes relatedto hypoxia adaptive responses that advantage cancer develop-ment (8). Although it is known that upregulation of HIF1 isclosely associated with poor clinical outcome in HCC patients(4), the detailed molecular mechanisms by which HIF1 pro-motes HCC progression remain poorly understood.

HIF1a transcriptionally activates many metabolic genes,allowing the cells to adapt to hypoxia, by shunting the glucoseintermediates into glycolysis instead of the tricarboxylic acid(TCA) cycle (9). These metabolic genes include glucose trans-porter (GLUT1), hexokinase 2 (HK2), lactate dehydrogenase A

1Department of Pathology, The University of Hong Kong, Hong Kong.2State Key Laboratory for Liver Research, The University of HongKong, Hong Kong.

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Authors: Carmen Chak-LuiWong, The University of Hong Kong,L-704 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam,Hong Kong, Hong Kong. Phone: 852-39179014; Fax: 852-28195375l; E-mail:[email protected]; Irene Oi-Lin Ng, [email protected]

doi: 10.1158/1078-0432.CCR-15-1987

�2016 American Association for Cancer Research.

ClinicalCancerResearch

www.aacrjournals.org 3105

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 2: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

(LDHA), and pyruvate dehydrogenase kinase 1 (PDK1). GLUT1facilitates glucose uptake (10). HK2 catalyzes the phosphory-lation of glucose to glucose-6-phosphate, the first step of glycol-ysis (11, 12). LDHA converts pyruvate to lactate (13, 14). PDK1inactivates pyruvate dehydrogenase to prevent pyruvate conver-sion to acetyl CoA. These processes restrict the entrance of glucoseintermediates into the TCA cycle, thereby reducing the activity ofoxidative phosphorylation (OXPHOS) in the mitochondria (9).Interestingly, all these metabolic genes are involved in cancerprogression.

The OXPHOS system comprises four electron transport chain(ETC) complexes, including complex I (NADH–ubiquinone oxi-doreductase), complex II (succinate:ubiquinone oxidoreductase),complex III (ubiquinol–cytochrome c reductase), and complex IV(cytochrome c oxidase). Most ATP in the cells is produced whenelectrons transfer through these complexes to the ultimate elec-tron acceptor, O2. Complex I, a 1 MDa complex of 45 subunits, isthe first step where OXPHOS takes place. Complex I catalyzes thetransfer of electrons from NADH to flavoprotein through eightiron–sulfur clusters, and finally to ubiquinone. Complex I is amajor ROS-producing site. Low level of ROS has been shown toactivate signaling pathways, such as MAPK-ERK, JNK, p38MAPK,and FAK, to promote cancer cell survival and metastasis (15),whereas excessive ROS accumulation suppresses cancer cellgrowth through induction of G2–Mcell-cycle arrest and apoptosis(16–18). Most of the chemotherapeutic and radiotherapeuticstrategies against cancer cells are mediated through ROS induc-tion (19, 20).

HIF1a regulates several ETC components to minimize ROSproduction and optimize mitochondrial respiration. HIF1ainduces the switching of COX subunit 4 isoforms from COX4-1 toCOX4-2 in complex IV tomaximize the efficiencyofOXPHOSunder hypoxia (21). HIF1a induces expression of a miRNA, miR-210, which suppresses iron–sulfur cluster assembly proteins(ISCU1/2) in complexes I and III to reduce oxygen consumption,ROS production, and apoptosis during hypoxic stress (22). Main-ly demonstrated in mouse embryonic fibroblasts, HIF1a inducedETC complex I subunit, NADH dehydrogenase (ubiquinone) 1

alpha subcomplex 4–like 2 (NDUFA4L2), to reduce complex Iactivity and ROS production (23).

Currently, knowledge about NDUFA4L2, particularly on itsroles in cancer development, is scarce. NDUFA4L2 was found tobe one of the seven biomarkers that distinguish medullarythyroid carcinoma from head and neck paraganglioma (PGL;ref. 24). Earlier study showed that NDUFA4L2 is overexpressedin PGL tumors that lack VHL (25). Nonetheless, its clinicalimplications and in vivo roles in cancers, particularly in HCC,have never been thoroughly studied. This study uncovers theclinical relevance and roles of NDUFA4L2 in REDOX homeo-stasis in HCC. Furthermore, we have successfully demonstratedthat targeting HIF1/NDUFA4L2 pathway by HIF inhibitorrepresents a novel therapeutic strategy for HCC.

Materials and MethodsPatient samples

HumanHCC and the corresponding paired nontumorous livertissues were collected at Queen Mary Hospital, the University ofHong Kong during surgical resection. Human lung squamous cellcarcinoma (SCC) and the corresponding paired nontumorouslung tissues samples were kindly provided by Dr. Maria P. Wong(the University of Hong Kong). Use of human samples wasapproved by the Institutional Review Board of the University ofHong Kong/Hospital Authority Hong Kong West Cluster.

Cell linesHuman HCC cell line, PLC/PRF/5, and human cervical cancer

cell line, HeLa, were obtained from the American Type CultureCollect (ATCC) and cultured according to ATCC recommenda-tions.Metastatic humanHCC cell line,MHCC97L, was a gift fromDr. Z.Y. Tang (Fudan University of Shanghai). All the cell linesused were authenticated by the AuthentiFiler PCR AmplificationKit (Applied Biosystems) on September 1st, 2014. All cell linesused in this article were thawed from the authenticated cell stockand used within four passages.

Clinicopathologic correlation and patients' survival analysisThe clinicopathologic features of HCC patients were ana-

lyzed by pathologist as we previously described (26). Theparameters included age, gender, tumor size, cellular differen-tiation by Edmondson grading, direct liver invasion, absence oftumor encapsulation, presence of tumor microsatellite forma-tion, venous invasion, and background liver disease. Overallsurvival was calculated from the date of surgical resection to thedate of death or last follow-up. The prognostic significance ofNDUFA4L2 overexpression was determined by the Kaplan–Meier method followed by the log-rank test, as we previouslydescribed (27, 28). All statistical tests were performed bySPSS20.0. The demographic data of HCC patients were sum-marized in Supplementary Table S1.

Fluorescence imaging and flow cytometry analysisCells were grown on glass coverslips in 6-well culture plate

and cultured in 20% and 1%O2 for 24 hours. Cells were stainedwith 2 mmol/L 5,5',6,6'-tetrachloro-1,1',3,3' -tetraethyl-benzi-midazolylcarbocyanine chloride (JC-1; Invitrogen). Imageswere captured and scanned under 20�magnification with LSM510 Meta laser scanning microscope (Carl Zeiss) connected to

Translational Relevance

Hepatocellular carcinoma (HCC) frequently experienceshypoxia. Hypoxia results in an inefficient transfer of electronsduring oxidative phosphorylation leading to increased oxida-tive stress. In this study, we demonstrated that a less activecomplex I subunit in the electron transport chain, NDUFA4L2,was significantly overexpressed in HCC and other humancancer types. Overexpression of NDUFA4L2 was associatedwith aggressive clinical features in human HCC and shorteroverall survival in HCC patients. A series of in vitro and in vivoassays converged to show that NDUFA4L2 reduced ROS-mediated apoptosis to confer HCC cells growth advantages.As NDUFA4L2 is a direct transcriptional target of HIF, wefound that HIF inhibitor, digoxin, profoundly inhibitedgrowth of tumors that expressed high level of NDUFA4L2 inorthotopic model. Our findings suggested that cancer patientswith NDUFA4L2 overexpression may be suitable candidatesfor HIF inhibitor treatment.

Lai et al.

Clin Cancer Res; 22(12) June 15, 2016 Clinical Cancer Research3106

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 3: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

Axiocam microscope camera (Carl Zeiss). For flow cytometryanalysis, cells were stained with 2 mmol/L JC-1.

Oxygen consumption assayCells were grown on 96-well culture plate and cultured in

20% and 1% O2 for 24 hours. Cells were stained with oxygen-sensitive probe MitoXpress-Intra (NanO2) and washed accord-ing to the manufacturer's instruction (Luxcel Bioscience). Phos-phorescence was measured by a multilabel reader Victor2

(Perkin-Elmer Life Sciences) with 340 nm excitation and 642nm emission filters.

ROS measurementTrypsinized cells were washed with PBS and stained with 2

mmol/L general ROS indicator chloromethyl-20,70-dichlorodihy-drofluorescein diacetate (CM-H2DCFDA; Life Technologies) or 5mmol/L mitochondrial ROS indicator MitoSOX (MolecularProbes) for 5 and 10 minutes, respectively, and analyzed by flowcytometer FACSCanto II Analyzer (BD Biosciences). For MitoSOXstaining, cells were washed with staining buffer 3 times beforeflow cytometry analysis. Data from flow cytometry studies wereanalyzed by FlowJo software. For antioxidant treatment, 0.5, 2, or5 mmol/L of N-acetyl-L-cysteine (NAC) or L-ascorbic acid (SigmaAldrich) were incubated at 37�C for 24 hours prior to ROSmeasurement. For in vivo ROS measurement, tumors harvestedfrom mice were dissociated with gentleMACS dissociator, andsingle-cell suspensions were stained with 2 mmol/L CM-H2DCFDA for flow cytometry analysis.

Apoptosis and cell proliferation assaysFor apoptosis measurement, 2.5 � 105 cells were cultured in

20% and 1%O2 for 24 hours. Apoptosis was determined by flowcytometry after staining with propidium iodide (Calbiochem)and Annexin V (MBL International Corporation). For antioxidanttreatment, 0.1 mmol/L glutathione ethyl ester (GSH-EE; CaymanChemical) was added toMHCC97L-shL2 cells and cultured in 1%O2 for 40hours. For cell proliferationmeasuredby cell counting, 2� 104 MHCC97L-NTC and -shL2 cells were grown on 12-wellculture plate and cultured in 20%and1%O2 for 4days. Cellsweretrypsinized and counted with Z1 coulter particle counter (Beck-manCoulter) every 24hours. For cell proliferationmeasuredby5-bromo-20-deoxyuridine (BrdUrd) assay, 2.5 � 104 MHCC97L-NTC and -shL2 cells were grown on 96-well culture plate andcultured in 20% and 1% O2 for 24 hours. BrdUrd labeling andcolorimetric measurement were performed according to theman-ufacturer's instructions (Roche Life Science).

Animal studiesAll animal studies were approved by the Committee on the

Use of Live Animals in Teaching and Research, the University ofHong Kong, and followed under the Animals (Control ofExperiments) Ordinance of Hong Kong. For orthotopic livertumor injection in nude mice, 1 � 106 luciferase-labeledMHCC97L cells were injected into the left lobes of the liversof 6- to 8-week-old BALB/c nude mice. Six weeks after injection,mice were administered with 100 mg/kg D-luciferin by peri-toneal injection 5 minutes before bioluminescent imaging(IVIS 100 Imaging System; Xenogen). Livers and lungs wereharvested for ex-vivo imaging and histologic analysis. Liverswere harvested for ex-vivo imaging and histologic analysis as

described above. For subcutaneous tumor model, 1 � 106

MHCC97L cells were injected subcutaneously to the flanks ofnude mice. Tumor volume was calculated with the formula:length (mm) � width (mm) � depth (mm) � 0.52 (mm3). Forpharmacologic studies, drug administration began a week afterorthotopic or subcutaneous injection. Mice were administeredwith 1.2 mg/kg/day digoxin or vehicles (saline) by intraperi-toneal injection for 21 consecutive days.

Transcriptome sequencingTranscriptome sequencing was performed in 16 pairs of

HCC tissues and corresponding NT liver tissues or MHCC97Lcells exposed to 20% and 1% O2 for 24 hours. TruSeq standardmRNA sample Prep kit (Illumina) was used for polyA þmRNAlibrary preparation. Illumina HiSeq2000 was employed for100 bp paired-end sequencing (Axeq Technologies), and datawere analyzed by TopHat-Cufflinks pipeline (29). Values wereindicated by FPKM (fragments per kilobase transcript sequenceper million mapped reads). Pathway analysis was performed bythe Database for Annotation, Visualization and IntegratedDiscovery (DAVID).

Statistical analysisStatistical analysis was performed by two-tailed Student t test

or Wilcoxon's signed rank test using GraphPad Prism 5.0(GraphPad Software Inc.). All functional assays are represen-tative of �3 independent experiments and expressed as mean �SEM. P value less than 0.05 was considered to be statisticallysignificant.

Quantitative real-time PCR, establishment of knockdownand knockout HCC cells, in vitro assays, and TCGA/Oncomine data

The following are described in Supplementary Materialsand Methods: quantitative real-time PCR (qRT-PCR), chromatinimmunoprecipitation (ChIP) assay, establishment of HIF orNDUFA4L2 knockdown andHIF knockout HCC cells, mitochon-drial membrane potential, mitochondrial mass, migration assay,histology, and The Cancer Genome Atlas (TCGA)/Oncominedata.

ResultsMitochondrial NDUFA4L2 is frequently overexpressed inhuman HCC and other human solid cancers

To better understand the hypoxia-adaptation system inHCC, we performed transcriptome sequencing to compare thegene expression profiles of a HCC cell line, MHCC97L, thatwere exposed to normoxia (20% O2) and hypoxia (0.1% and1% O2). Among the upregulated genes, mitochondrial NDU-FA4L2 was one of the five most abundant genes in hypoxicconditions, indicating the biologic relevance of this gene inhuman HCC. Other well-characterized HIF target genes,including ANPTGL4 (30), PLOD2 (31), VEGF (32), CA9(33), and P4HA2 (34), were on the top 20 of the list. Intrigu-ingly, when we interrogated the expression of genes that areinvolved in the mitochondrial complex I, only the subunitNDUFA4L2 was distinctly and dramatically upregulated inhypoxia (both 0.1% and 1% O2), whereas all the other sub-units remained unaffected or downregulated (Fig. 1A). Wefurther examined the transcriptome sequencing data on 16

NDUFA4L2 in HCC Development

www.aacrjournals.org Clin Cancer Res; 22(12) June 15, 2016 3107

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 4: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

Lai et al.

Clin Cancer Res; 22(12) June 15, 2016 Clinical Cancer Research3108

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 5: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

Figure 1.NDUFA4L2 is overexpressed in human HCC and other human solid cancers. A, transcriptome sequencing data comparing FPKM values of genes encodingmitochondrial complex I subunits in MHCC97L cells cultured in (left) 0.1% O2 or (middle) 1% O2 relative to 20% O2 and (right) 16 pairs of human HCCand the corresponding NT tissues. B, mRNA expression levels of NDUFA4L2 normalized with hypoxanthine-guanine phosphoribosyltransferase in anexpanded cohort of 100 cases of paired human HCC, and corresponding NT tissues were determined by qRT-PCR. Waterfall plot shows that NDUFA4L2is overexpressed in 71% of human HCC samples by at least 2-fold. C, clinicopathologic analysis shows that NDUFA4L2 overexpression is closelyassociated with more aggressive features in HCC, including the presence of tumor microsatellite formation and the absence of tumor encapsulation.HCC patients with high NDUFA4L2 expression (= median) tend to have lower 5-year overall survival rate (Student t test; P ¼ 0.086). D, mRNA expressionlevels of NDUFA4L2 obtained from TCGA database of 49 paired human HCC and corresponding NT tissues (RSEM, RNA-Seq by Expectation Maximization).E, mRNA expression levels of NDUFA4L2 normalized with b-actin in 31 cases of paired human lung SCC, and corresponding NT tissues weredetermined by qRT-PCR. Waterfall plot shows that NDUFA4L2 is overexpressed in 65% of human lung SCC samples by at least 2-fold. (Wilcoxon's signedrank test; ��� , P < 0.001 for B, D, and E. Student t test; � , P < 0.05; �� , P < 0.01 for C).

Figure 2.NDUFA4L2 is regulated by HIF1a but not HIF2a in HCC. A, mRNA and protein expression of NDUFA4L2 in MHCC97L-EV, -shHIF1a, and -shHIF2a cellscultured in 20% and 1% O2 for 24 and 48 hours, respectively. B, NDUFA4L2 protein expression in MHCC97L-WT (parental) and -HIF1a�/� (KO) cellscultured in 20% and 1% O2 for 48 hours. C, NDUFA4L2 mRNA and protein expression in MHCC97L cells treated with HIF inhibitor digoxin at the indicateddoses. D, putative HREs are mapped in the promoter of NDUFA4L2. Core HIF binding sequences (50-RCGTG-30) are highlighted as bold with underlines,while ancillary sequence is highlighted as bold. E, ChIP assay was performed with IgG and HIF1a antibodies in MHCC97L cells cultured in 20% and 1%O2. qRT-PCR was performed after immunoprecipitation with anti-IgG or anti-HIF1a antibodies, represented as fold enrichment normalized to 20% O2

(results represent mean � SEM. Student t test; N ¼ 3 independent experiments: �, P < 0.05; �� , P < 0.01; ��� , P < 0.001 as indicated).

NDUFA4L2 in HCC Development

www.aacrjournals.org Clin Cancer Res; 22(12) June 15, 2016 3109

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 6: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

Lai et al.

Clin Cancer Res; 22(12) June 15, 2016 Clinical Cancer Research3110

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 7: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

primary HCCs and their corresponding nontumorous (NT)livers and found that NDUFA4L2 was substantially overex-pressed in HCC tissues (Fig. 1A). Moreover, overexpression ofNDUFA4L2 mRNA was validated by qRT-PCR in a separate,larger cohort of 100 pairs of HCC samples (Fig. 1B). Further-more, overexpression of NDUFA4L2 in HCC tumors was sig-nificantly associated with aggressive pathologic features,including presence of tumor microsatellite formation (Studentt test; P ¼ 0.002) and absence of tumor encapsulation (Studentt test; P ¼ 0.039; Fig. 1C and Supplementary Table S4). Moreimportantly, overexpression of NDUFA4L2 tended to be asso-ciated with poorer overall survival of HCC patients (Student ttest; P ¼ 0.086; Fig. 1C). TCGA database, which includestranscriptome sequencing data of 49 pairs of HCC and NTliver tissues from an independent cohort, echoed with our in-house data (Fig. 1D). Intriguingly, when we studied the micro-array data available in the Oncomine database, we noticed thatNDUFA4L2 was also overexpressed in other human solidcancers, including renal cell carcinoma (RCC), lung SCC, aswell as colorectal carcinoma (Supplementary Fig. S1). Theoverexpression of NDUFA4L2 mRNA in lung SCC was furtherconfirmed in our 31 pairs of lung SCC tissues and their normalcounterparts by qRT-PCR (Fig. 1E). Collectively, these resultsdemonstrated the clinical relevance of NDUFA4L2 in humanHCC and other human solid cancers.

HIF1a, but not HIF2a, positively regulates NDUFA4L2 inhypoxia

To investigate whether NDUFA4L2 expression was mediatedby HIFs under hypoxia, we generated HCC cells, MHCC97L,that stably expressed shRNA against HIF1a (-shHIF1a) orHIF2a (-shHIF2a; Supplementary Fig. S2A). The hypoxia-induced NDUFA4L2 expression was markedly abolished whenHIF1a, but not HIF2a, was knocked down (Fig. 2A). We furtherestablished HIF1a knockout cells by Transcription Activator-likeEffector Nuclease approach and found that hypoxia-inducedNDUFA4L2 expression was drastically abrogated (Fig. 2B andSupplementary Fig. S2B). Digoxin inhibited HIF1 protein syn-thesis and profoundly inhibited hypoxia-induced NDUFA4L2expression in a dose-dependent manner (Fig. 2C). In addition,by in silico analysis, we identified two potential hypoxia re-sponse elements (HRE) containing the core HIF-bindingsequence motif 50-RCGTG-30 at the promoter of NDUFA4L2(Fig. 2D). ChIP assay in cell lines of different origins, MHCC97Land HeLa cells, indicated that DNA was enriched when HIF1a

antibody was used, as compared with the IgG control (Fig. 2Eand Supplementary Fig. S3). Intriguingly, we consistentlydetected a band beneath the expected protein size of NDU-FA4L2. This protein was only expressed in normoxia but not inhypoxia (Figs. 2A, B, and C, and 3A and B). NDUFA4L2 shared67% amino acid sequence with its paralogue NDUFA4, whichis 6 amino acids shorter than NDUFA4L2. To confirm theidentity of the unknown protein beneath NDUFA4L2, weknocked down NDUFA4 (-shA4-30 and -shA4-88) in MHCC97Lcells and confirmed that the lower band was NDUFA4 (Sup-plementary Fig. S4A and S4B). These results demonstrated thatNDUFA4L2 was preferentially expressed under hypoxia solelythrough HIF1a, whereas NDUFA4 protein was preferentiallyexpressed under normoxia.

NDUFA4L2 reduces mitochondrial activity by decreasingoxygen consumption and prevents excessive ROS productionunder hypoxia

To investigate the functions of hypoxia-induced NDUFA4L2in mitochondria in vitro, we generated NDUFA4L2 loss-of-function and gain-of-function HCC cell models. For NDU-FA4L2 loss-of-function HCC cell model, shRNA against NDU-FA4L2 (shL2) or nontarget control (NTC) was stably expressedin MHCC97L cells by lentiviral transfection approach (Fig. 3A).For NDUFA4L2 gain-of-function HCC cell model, NDUFA4L2and empty vector (EV) were stably expressed in anotherHCC cell line, which expressed a lower level of NDUFA4L2,PLC/PRF/5 (PLC; Fig. 3B). As NDUFA4L2 is located in thefirst complex of the ETC, we asked if NDUFA4L2 would affectthe mitochondrial activity by staining the cells with a probe,JC-1, which accumulates in active mitochondria to indicatemitochondrial potential. By fluorescence imaging, we showedthat the mitochondrial membrane potential was decreasedin hypoxia, but was elevated when NDUFA4L2 was knockeddown (Fig. 3C). Flow cytometry analysis further confirmed theresult (Fig. 3C). Similar observation was obtained with tetra-methylrhodamine ethyl ester, another fluorescent probe thatmeasures the mitochondrial membrane potential (Supple-mentary Fig. S5A). Consistently, mitochondrial mass wasincreased in NDUFA4L2 knockdown cells as indicated by thenonyl acridine orange (NAO) staining (Fig. 3D). Consistently,we found that oxygen consumption in the mitochondria wassignificantly elevated when NDUFA4L2 was knocked downunder hypoxia (Fig. 3E). HIF1a knockout mirrored the effectof NDUFA4L2 knockdown in oxygen consumption (Fig. 3E),

Figure 3.NDUFA4L2 reduces mitochondrial activity, oxygen consumption, and ROS production in hypoxia. A, mRNA and protein expression of NDUFA4L2 in MHCC97L-NTC and -shL2 cells exposed to 20% and 1% O2 for 24 and 48 hours, respectively (results represent mean � SEM. Student t test; N ¼ 3 independentexperiments: ��� , P < 0.001 as indicated). B, mRNA and protein expression of NDUFA4L2 in PLC-EV and -NDUFA4L2 cells cultured in 20% O2 for 24and 48 hours, respectively (results represent mean � SEM. Student t test; N ¼ 3 independent experiments: ��� , P < 0.001 as indicated). C, Left,representative fluorescent images of JC-1 stained MHCC97L-NTC and -shL2 cells that were exposed to 20% and 1% O2. Right, mitochondrial membranepotential was further determined with JC-1 staining by flow cytometry analysis (results represent mean � SEM. Student t test; N ¼ 5 independentexperiments: ��� , P < 0.001 as indicated). Scale, 50 mm. D, mitochondrial mass was measured with NAO staining by flow cytometry analysis (resultsrepresent mean � SEM. Student t test; N ¼ 5 independent experiments: �� , P < 0.01 as indicated). E, oxygen consumption was measured with

MitoXpress�-Intra (NanO2) in MHCC97L-NTC and -shL2 cells or MHCC97L-WT and -HIF1a�/� (KO) cells exposed to 20% and 1% O2 (results representmean � SEM. Student t test; N ¼ 3 independent experiments: � , P < 0.05; �� , P < 0.01 as indicated). F, ROS level was measured with CM-H2DCFDA orMitoSox staining by flow cytometry analysis in (i and ii) MHCC97L-NTC and -shL2 cells cultured in 20% and 1% O2, (iii) PLC-EV and -NDUFA4L2 cellscultured in 20% O2, and (iv) PLC-EV and -CA5 cells cultured in 20% O2 (results represent mean � SEM. Student t test; N ¼ 3 independent experiments:�� , P < 0.01 as indicated). Cells for all metabolic assays were cultured for 24 hours. Values obtained in mitochondrial membrane potential, mass, andROS were normalized to 20% O2 MHCC97L-NTC or PLC-EV.

NDUFA4L2 in HCC Development

www.aacrjournals.org Clin Cancer Res; 22(12) June 15, 2016 3111

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 8: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

Lai et al.

Clin Cancer Res; 22(12) June 15, 2016 Clinical Cancer Research3112

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 9: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

further supporting the regulatory role of HIF1a on NDUFA4L2.Hypoxia triggers an imbalanced electron flow through theETC leading to generation of ROS in the mitochondria. Hence,we evaluated the intracellular ROS level by the general ROSindicator, chloromethyl-2',7'-dichlorodihydrofluorescein dia-cetate (CM-H2DCFDA), and mitochondrial ROS indicator,MitoSOX. ROS level was elevated when cells were exposed tohypoxia and was further induced when NDUFA4L2 wasknocked down especially under hypoxic condition (Fig. 3F).Reversely, a significant reduction of ROS was found in NDU-FA4L2-overexpressing HCC cells (Fig. 3F). Similar trend wasobserved in HCC cells that expressed the constitutively activeform of HIF1a (CA5; Fig. 3F). To eliminate off-target effect,siRNAs targeting different sequences of NDUFA4L2 were trans-fected into MHCC97L cells (Supplementary Fig. S5B), andsimilar metabolic consequences were found as compared withstable knockdown of NDUFA4L2 (Supplementary Fig. S5Cand S5D).

NDUFA4L2 maintains redox homeostasis and promotes cellproliferation and survival

Knockdown of NDUFA4L2 resulted in a dramatic increaseof ROS, which we speculated would be harmful to HCC cells.To demonstrate the roles of ROS in HCC cells, we employedtwo antioxidants, NAC and L-ascorbic acid. ROS induced byNDUFA4L2 knockdown was rescued dose dependently byboth antioxidants (Fig. 4A). To further demonstrate the reg-ulation of HIF1a on NDUFA4L2 in ROS scavenging, we re-expressed NDUFA4L2 in HIF1a knockdown PLC cells. ROSwas dramatically induced in HIF1a knockdown cells underhypoxia and was significantly rescued upon re-expression ofNDUFA4L2 (Fig. 4B). Excessive accumulation of ROS cancause cell senescence and apoptosis, eventually leading to celldeath (16–18). We therefore evaluated apoptosis by AnnexinV–propidium iodide (PI) staining in MHCC97L-NTC and-shL2 cells. MHCC97L-NTC cells showed higher Annexin Vand PI staining under hypoxic than normoxic condition,while knockdown of NDUFA4L2 further increased apoptosis(Fig. 4C), which could be rescued by extracellular GSH-EE, amodified and more permeable form of glutathione, an anti-oxidant that counteracts mitochondrial ROS (SupplementaryFig. S6A and S6B). To further confirm that increased apoptosiswas caused by ROS, we exogenously treated the cells with H2

O2, a common source of ROS (35). Apoptosis was elicited byH2O2 drastically in MHCC97L-NTC cells and was furtherelevated in -shL2 cells (Supplementary Fig. S6C). Consistently,both the cell counting and BrdUrd assays showed that cellproliferation was substantially reduced when NDUFA4L2was knocked down especially under hypoxic condition (Fig.

4D and Supplementary Fig. S6D). More interestingly, cellproliferation was restored by NAC in the NDUFA4L2 knock-down HCC cells (Fig. 4E), suggesting that impairment of cellproliferation was associated with ROS. All these data con-verged to show that NDUFA4L2 was responsible to reduceROS under hypoxia, thereby maintaining cell survival andpromoting cell proliferation.

NDUFA4L2 promotes tumor growth, reduces oxidative stress,and enhances lung metastasis

To demonstrate the functions of NDUFA4L2 in tumorgrowth in vivo, we orthotopically injected the luciferase-labeledMHCC97L-NTC and -shL2 cells into nude mice. MHCC97L-NTC tumors grew substantially larger than the MHCC97L-shL2tumors (Fig. 5A and 5B). To evaluate the proliferative output ofMHCC97L-NTC and -shL2 cells in vivo, we stained the tumorswith proliferative marker (Ki67). We found that knockdown ofNDUFA4L2 markedly reduced Ki67 staining (Fig. 5C). Wefurther dissociated the tumors and measured the ROS level ofthe tumors derived from MHCC97L-NTC and -shL2 cell lines.Consistent with our in vitro data, knockdown of NDUFA4L2elevated ROS in vivo (Fig. 5D). Moreover, knockdown of NDU-FA4L2 markedly repressed growth of metastatic lesions in thelungs of the tumor-bearing mice as indicated by Xenogenimaging (4 of 6 in MHCC97L-NTC and 1 of 6 in -shL2group; Fig. 5E). We further found that knockdown of NDU-FA4L2 significantly reduced cell-migratory ability of HCC cellsin Transwell assay. Interestingly, the migratory ability of HCCcells was inversely correlated with the amount of intracellularROS (Supplementary Fig. S7A and Fig. 3E), suggesting that ROSmay also affect cell motility.

Pharmacologic inhibition of HIF suppresses progression oftumors that express high level of NDUFA4L2

So far, we found that hypoxia-induced NDUFA4L2 tightlyregulated REDOX homeostasis and survival in HCC cells, andHIF1a is the central regulator of NDUFA4L2. We thereforereasoned that HIF inhibitor could repress the tumors thatexpress high levels of NDUFA4L2 by damaging HCC cellsthrough elevating ROS. We evaluated the ROS levels ofMHCC97L-NTC and -shL2 cells treated with an HIF inhibitor,digoxin, under hypoxic condition. We observed that digoxinwas more efficient in elevating the ROS level in MHCC97L-NTCcells than -shL2 cells (Fig. 6A). Next, we evaluated the effect ofdigoxin in vivo. We performed orthotopic implantation of theluciferase-labeled MHCC97L-NTC and -shL2 cells in nude mice.One week after implantation, mice were administered with1.2 mg/kg/day digoxin or vehicle control (saline) through i.p.injection for 21 days. Digoxin markedly repressed growth of

Figure 4.NDUFA4L2 reduces intracellular ROS to enhance HCC cell survival. A, CM-H2DCFDA staining showing the ROS levels of MHCC97L-NTC and -shL2 cellsexposed to 1% O2 for 24 hours in the presence of indicated concentrations of NAC and ascorbic acid. Top, representative flow cytometry analysisshowing the ROS levels in the indicated subclones. Bottom, histograms summarizing the ROS levels in the indicated subclones. The ROS level was relativeto 1% O2 MHCC97L-NTC. B, CM-H2DCFDA staining showing the ROS levels of PLC-EV, -shHIF1a, and HIF1a knockdown with NDUFA4L2 overexpressing(-shHIF1a þ NDUFA4L2) cells exposed to 1% O2 for 24 hours. The ROS level was relative to 20% O2 PLC-EV. Top, representative flow cytometryanalysis showing the ROS levels in the indicated subclones. Bottom, histograms summarizing the ROS levels in the indicated subclones. C, Annexin Vand PI staining showing the percentage of apoptotic cells in MHCC97L-NTC and -shL2 cells exposed to 20% and 1% O2 for 24 hours. D, cell proliferationof MHCC97L-NTC and -shL2 cells exposed to 20% and 1% O2 for 96 hours. E, BrdUrd assay of MHCC97L-NTC and -shL2 cells treated with indicatedconcentrations of NAC was exposed to 1% O2 for 24 hours (results represent mean � SEM. Student t test; N ¼ 3 independent experiments: �, P < 0.05;�� , P < 0.01; ��� , P < 0.001 as indicated).

NDUFA4L2 in HCC Development

www.aacrjournals.org Clin Cancer Res; 22(12) June 15, 2016 3113

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 10: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

tumors with a more prominent effect on the MHCC97L-NTCgroup as compared with MHCC97L-shL2 group (Fig. 6B).Similarly, digoxin decreased the growth of subcutaneous tumors

derived from MHCC97L-EV cells more significantly as com-pared with MHCC97L-shHIF1a cells (Supplementary Fig. S7B).Meanwhile, body weights of the animals were not affected by

Figure 5.Knockdown of NDUFA4L2 suppresses tumor growth and induces oxidative stress in vivo. A, bioluminescence and (B) tumor volume of the orthotopictumors derived from MHCC97L-NTC and -shL2 cells. Scale, 1 cm. C, Left, representative IHC pictures of Ki67 positivity in tumors. Cells were scoredfrom 0 to 3 based on the staining intensity as represented by the arrows. Scale, 100 mm. Right, more than 1,000 cells were scored in each animal, andaverage percentage was calculated for each score. D, workflow of the measurement of intracellular ROS level in mouse model. Nude mice wereorthotopically injected with MHCC97L-NTC and -shL2 cells. CM-H2DCFDA staining showing the ROS levels of the dissociated orthotopic tumorsderived from MHCC97L-NTC and -shL2. E, bioluminescence of the lungs harvested from orthotopic tumor-bearing mice implanted with MHCC97L-NTCand -shL2 cells (Student t test; N ¼ 6 per group: � , P < 0.05; �� , P < 0.01).

Lai et al.

Clin Cancer Res; 22(12) June 15, 2016 Clinical Cancer Research3114

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 11: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

digoxin (Supplementary Fig. S7C). These results suggested thatHIF inhibitor blocks HCC progression at least partially throughHIF1a/NDUFA4L2.

Potential gene targets of NDUFA4L2To explore novel functions of NDUFA4L2, we performed

transcriptome sequencing to compare the global gene expressionprofiles ofMHCC97L-NTC andMHCC97L-shL2 cells. The expres-sions of 629 genes were found to be decreased inMHCC97L-shL2cells for more than 0.5-fold as compared with MHCC97L-NTC(Supplementary Table S5). The expressions of 683 genes werefound to be increased for more than 2-fold in MHCC97L-shL2cells relative to MHCC97L-NTC (Supplementary Table S6).Intriguingly, pathway analysis by DAVID suggested that NDU-

FA4L2 may regulate multiple biologic processes, including mem-brane potential and membrane depolarization (SupplementaryTable S7). Of note, NRF2, a transcription factor that activatesantioxidant genes, was found to be slightly induced (FPKM29.246 in MHCC97L-NTC vs. FPKM 34.151 in -shL2), suggestingthat a compensatory anti–oxidant-producing mechanism mightbe involved in response to the high ROS level upon NDUFA4L2knockdown.

DiscussionROS are fundamentally produced from O2 when it is con-

sumed in different metabolic reactions. These reactions takeplace in the mitochondria at the ETC for ATP generation,

Figure 6.HIF inhibitors suppress growth of tumors which express high level of NDUFA4L2. A, CM-H2DCFDA staining showing the ROS levels of MHCC97L-NTC and-shL2 cells exposed to 1% O2 for 24 hours in the presence of indicated concentrations of HIF inhibitor digoxin (results represent mean � SEM. Studentt test; N ¼ 3 independent experiments: �� , P < 0.01; ��� , P < 0.001 as indicated). B, tumor volumes of orthotopic tumors derived from MHCC97L-NTCand -shL2 cells in mice that were administered with digoxin (1.2 mg/kg/day, i.p.) or vehicle (saline) for 21 consecutive days (Student t test; N ¼ 6 per group:� , P < 0.05; ��� , P < 0.001 as indicated). Scale, 1 cm. C, regulation and role of NDUFA4L2 in human HCC. i, hypoxia elicits an imbalanced electronflow through the ETC, leading to excessive ROS accumulation which is detrimental to HCC cells. ii, HIF1a overcomes by transcriptionally activatingNDUFA4L2 in the mitochondrial complex I selectively to reduce the mitochondrial activity and the electron flux through ETC, thereby reducing ROSand ROS-induced apoptosis. This metabolic adaptation of oxidative stress mediated by HIF1a under hypoxia conferred survival advantage to the rapidlygrowing HCC cells which frequently experience hypoxia.

NDUFA4L2 in HCC Development

www.aacrjournals.org Clin Cancer Res; 22(12) June 15, 2016 3115

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 12: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

endoplasmic reticulum during disulfide bond formation forprotein folding, and peroxisomes during b-oxidation of fattyacids. Under hypoxia, these metabolic events cannot be carriedout properly, leading to the generation of O2-containing freeradicals. In the mitochondria that experiences shortage of theultimate electron acceptor, O2, electrons cannot be efficientlypass through the ETC, leading to rapid accumulation of ROS incomplexes I and III. Hypoxia is a common finding in HCC, andour study revealed the molecular mechanisms by which hyp-oxic HCC cells modulate ROS level in the mitochondria. Ourstudy showed that HCC cells, by utilizing NDUFA4L2 in thecomplex I of the ETC, reduced the activity of the mitochondriato prevent ROS accumulation and apoptosis (Fig. 6C). Notonly have we confirmed that this was dependent on HIF1a inHCC cells, more importantly, our study unprecedentedlyrevealed the clinical relevance of NDUFA4L2 in human HCC.Apart from HCC, we also showed that NDUFA4L2 was over-expressed in multiple solid cancers, suggesting that our find-ings do not limit to HCC but applies to other solid cancermodels that frequently experience hypoxia. Notably, we andTello and colleagues. consistently found that NDUFA4 andNDUFA4L2 have opposite expression patterns at normoxiaand hypoxia in different cell models (23). Studies have docu-mented that HIF1a could induce expression of siah E3 ubi-quitin protein ligase 2 (SIAH2) and mitochondrial LON pep-tidase to degrade mitochondrial proteins such as a-ketogluta-rate dehydrogenase and COX4-1 to reduce mitochondrial fluxunder hypoxia, respectively (21, 36). Whether NDUFA4 isdegraded in a similar fashion remains an interesting topic tobe addressed.

Prenatal lethality in NDUFA4L2 knockout mice has greatlylimited the expansion of knowledge on the in vivo functions ofthis complex I subunit (23). Our study, using orthotopic livercancer model, has provided the first in vivo evidence to confirmthe protumorigenic functions of NDUFA4L2. Our study alsodemonstrated that digoxin profoundly blocked growth oforthotopic tumors that expressed high level of HIF1a/NDU-FA4L2. Digoxin has been shown to inhibit HIF1a synthesis andis well-tolerated in patients (30). Digoxin has been used for thetreatment of congestive heart failure and is currently underclinical trial for the treatment of breast cancer (ClinicalTrials.gov identifier: NCT01763931). Our study suggested that digox-in may be the most suitable candidate for patients whoseprimary HCCs express high levels of HIF1a or NDUFA4L2.

Worth mentioning, complex I inhibitors including metforminhave promising antitumor effects in the clinical setting. Metfor-min has been the most frequently prescribed antidiabetic med-ication in the world. By reducing the ATP production in the ETC,metformin activates tumor suppressor 50-AMP-activated proteinkinase (AMPK; ref. 37). Reports have indicated that metforminreduced cancer incidence (38, 39). Metformin also reduced pro-liferative output of breast cancer cells in nondiabetic cancerpatients (40). These interesting findings highlight an interestingdilemma—is energy or REDOX homeostasis more important forcancer survival? Accumulating studies suggested that ATP is notthe limiting factor while excessive ROS is cell destructive.Unquestionably, cancer cells could not survive without energy;therefore, cancer cells need to acquire a tightly controlled homeo-static balance for the ETC, a place that generates both ATP andROS, to sustain tumor growth. Drugs that could tip this balancemerits exploration for their efficiency in cancer treatment.

REDOX homeostasis can be achieved by twomechanisms: (i)reduction of ROS production and (ii) elevation of antioxidantproduction. Our study showed that NDUFA4L2 directlydecreased the generation of ROS in the mitochondria of HCCcells. Meanwhile, genes that are associated with the productionand utilization of antioxidants, including NADPH, glutathione,and thioredoxin, have been implicated in HCC. Glucose 6phosphate dehydrogenase, a critical enzyme in the pentosephosphate pathway generating NADPH, has been shown tobe overexpressed in HCC in a PTEN-dependent manner (41).Overexpression of thioredoxin and thioredoxin-related REDOXmolecules, glutaredoxin and peroxiredoxin, was documentedin HCC and was associated with increased proliferative output,aggressive clinicopathologic parameters, presence of metastasis,and poor prognosis in HCC patients (42, 43). Elevation ofcysteine/glutamate transporter, xCT, which is important forglutathione production, was found to be associated with poorclinical outcome in HCC, and disruption of xCT in HCC cellsincreased ROS-mediated autophagic cell death (44). A recentstudy has elegantly demonstrated that cotreatment of inhibitorsblocking different pathways that synthesize antioxidants couldsynergistically prevent tumor growth (45). Along similar lines,our current study suggested that raising ROS level through HIFinhibitor, which suppressed HIF1a/NDUFA4L2 pathway,represents an attractive therapeutic strategy for HCC treatment.In the coming decade, combination therapies targeting multi-ple machineries that buffer ROS represent an exciting transla-tion research direction.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: R.K.-H. Lai, I.O.-L. Ng, C.C.-L. WongDevelopment of methodology: R.K.-H. Lai, C.C.-L. WongAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): R.K.-H. Lai, I. M.-J. Xu, D.K.-C. Chiu, A.P.-W. Tse,M.P. Wong, C.C.-L. WongAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): R.K.-H. Lai, D.K.-C. Chiu, A.P.-W. Tse, C.-T. Law,C.-M. Wong, C.C.-L. WongWriting, review, and/or revision of the manuscript: R.K.-H. Lai, D. Lee,M.P. Wong, I.O.-L. Ng, C.C.-L. WongAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): L.L. Wei, C.-T. Law, I.O.-L. Ng, C.C.-L. WongStudy supervision: I.O.-L. Ng, C.C.-L. Wong

AcknowledgmentspQCXIN HIF1a (CA5) construct is a generous gift from Professor Gregg L.

Semenza. The authors thank the Core Facility of LKS Faculty of Medicine fortheir technical support. They also thank Dr. Wilson Yick-Pang Ching and Dr.Yuan Zhou for the valuable advice and technical help.

Grant SupportThis work was supported by the Small Project Funding of the University of

HongKong 2012 and theHongKongResearchGrants Council General ResearchFund (HKU 781213M).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received August 19, 2015; revised December 15, 2015; accepted January 4,2016; published OnlineFirst January 27, 2016.

Lai et al.

Clin Cancer Res; 22(12) June 15, 2016 Clinical Cancer Research3116

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 13: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

References1. Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet 2012;379:

1245–55.2. Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safety

of sorafenib in patients in the Asia-Pacific region with advanced hepato-cellular carcinoma: a phase III randomised, double-blind, placebo-con-trolled trial. Lancet Oncol 2009;10:25–34.

3. Llovet JM, Ricci S,Mazzaferro V,Hilgard P, Gane E, Blanc JF, et al. Sorafenibin advanced hepatocellular carcinoma. N Engl J Med 2008;359:378–90.

4. Wong CC, Kai AK, Ng IO. The impact of hypoxia in hepatocellularcarcinoma metastasis. Front Med. 2014;8:33–41.

5. Semenza GL. Hypoxia-inducible factors in physiology and medicine. Cell2012;148:399–408.

6. Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O'Rourke J, Mole DR,et al. C. elegans EGL-9 and mammalian homologs define a family ofdioxygenases that regulate HIF by prolyl hydroxylation. Cell 2001;107:43–54.

7. Kaelin WGJr, Ratcliffe PJ. Oxygen sensing by metazoans: the centralrole of the HIF hydroxylase pathway. Mol Cell 2008;30:393–402.

8. Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is abasic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension.Proc Natl Acad Sci U S A 1995;92:5510–4.

9. Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expres-sion of pyruvate dehydrogenase kinase: a metabolic switch required forcellular adaptation to hypoxia. Cell Metab 2006;3:177–85.

10. Iyer NV, Kotch LE, Agani F, Leung SW, Laughner E, Wenger RH, et al.Cellular and developmental control of O2 homeostasis by hypoxia-induc-ible factor 1 alpha. Genes Dev 1998;12:149–62.

11. Kim JW, Gao P, Liu YC, Semenza GL, Dang CV. Hypoxia-inducible factor 1and dysregulated c-Myc cooperatively induce vascular endothelial growthfactor and metabolic switches hexokinase 2 and pyruvate dehydrogenasekinase 1. Mol Cell Biol 2007;27:7381–93.

12. Semenza GL, Roth PH, Fang HM, Wang GL. Transcriptional regulation ofgenes encoding glycolytic enzymes by hypoxia-inducible factor 1. J BiolChem 1994;269:23757–63.

13. Fantin VR, St-Pierre J, Leder P. Attenuation of LDH-A expression uncovers alink between glycolysis, mitochondrial physiology, and tumor mainte-nance. Cancer Cell 2006;9:425–34.

14. Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, Maire P,et al. Hypoxia response elements in the aldolase A, enolase 1, and lactatedehydrogenase A gene promoters contain essential binding sites for hyp-oxia-inducible factor 1. J Biol Chem 1996;271:32529–37.

15. Tochhawng L, Deng S, Pervaiz S, Yap CT. Redox regulation of cancer cellmigration and invasion. Mitochondrion 2013;13:246–53.

16. Ames BN. Dietary carcinogens and anticarcinogens. Oxygen radicals anddegenerative diseases. Science 1983;221:1256–64.

17. HamptonMB, Orrenius S. Dual regulation of caspase activity by hydrogenperoxide: implications for apoptosis. FEBS Lett 1997;414:552–6.

18. Sena LA, Chandel NS. Physiological roles ofmitochondrial reactive oxygenspecies. Mol Cell 2012;48:158–67.

19. Renschler MF. The emerging role of reactive oxygen species in cancertherapy. Eur J Cancer 2004;40:1934–40.

20. Toler SM, Noe D, Sharma A. Selective enhancement of cellular oxidativestress by chloroquine: implications for the treatment of glioblastomamultiforme. Neurosurg Focus 2006;21:E10.

21. Fukuda R, Zhang H, Kim JW, Shimoda L, Dang CV, Semenza GL. HIF-1regulates cytochromeoxidase subunits to optimize efficiency of respirationin hypoxic cells. Cell 2007;129:111–22.

22. Chan SY, Zhang YY, Hemann C, Mahoney CE, Zweier JL, Loscalzo J.MicroRNA-210 controls mitochondrial metabolism during hypoxia byrepressing the iron-sulfur cluster assembly proteins ISCU1/2. Cell Metab2009;10:273–84.

23. Tello D, Balsa E, Acosta-Iborra B, Fuertes-Yebra E, Elorza A, Ordonez A,et al. Induction of the mitochondrial NDUFA4L2 protein by HIF-1alphadecreases oxygen consumption by inhibiting Complex I activity. CellMetab 2011;14:768–79.

24. Castelblanco E,Gallel P, Ros S, Gatius S, Valls J, De-Cubas AA, et al. Thyroidparaganglioma. Report of 3 cases and description of an immunohisto-chemical profile useful in the differential diagnosis withmedullary thyroidcarcinoma, based on complementary DNA array results. Hum Pathol2012;43:1103–12.

25. Favier J, Briere JJ, Burnichon N, Riviere J, Vescovo L, Benit P, et al. TheWarburg effect is genetically determined in inherited pheochromocyto-mas. PloS one 2009;4:e7094.

26. Ng IO, Lai EC, Fan ST,NgMM,SoMK.Prognostic significance of pathologicfeatures of hepatocellular carcinoma. A multivariate analysis of 278patients. Cancer 1995;76:2443–8.

27. Wong CC, Au SL, Tse AP, Xu IM, Lai RK, Chiu DK, et al. Switching ofpyruvate kinase isoform L to m2 promotes metabolic reprogramming inhepatocarcinogenesis. PloS one 2014;9:e115036.

28. Wong CC, Wong CM, Tung EK, Au SL, Lee JM, Poon RT, et al. ThemicroRNA miR-139 suppresses metastasis and progression of hepato-cellular carcinoma by down-regulating Rho-kinase 2. Gastroenterology2011;140:322–31.

29. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ,et al. Transcript assembly and quantification by RNA-Seq reveals unan-notated transcripts and isoform switching during cell differentiation. NatBiotechnol 2010;28:511–5.

30. Zhang H, Wong CC, Wei H, Gilkes DM, Korangath P, Chaturvedi P, et al.HIF-1-dependent expression of angiopoietin-like 4 and L1CAM mediatesvascular metastasis of hypoxic breast cancer cells to the lungs. Oncogene2012;31:1757–70.

31. Gilkes DM, Bajpai S, Wong CC, Chaturvedi P, Hubbi ME, Wirtz D, et al.Procollagen lysyl hydroxylase 2 is essential for hypoxia-induced breastcancer metastasis. Mol Cancer Res 2013;11:456–66.

32. Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, et al.Activation of vascular endothelial growth factor gene transcription byhypoxia-inducible factor 1. Mol Cell Biol 1996;16:4604–13.

33. Giatromanolaki A, Koukourakis MI, Sivridis E, Pastorek J, Wykoff CC,Gatter KC, et al. Expression of hypoxia-inducible carbonic anhydrase-9relates to angiogenic pathways and independently to poor outcome innon-small cell lung cancer. Cancer Res 2001;61:7992–8.

34. Chaturvedi P, Gilkes DM, Wong CC, Luo W, Zhang H, Wei H, et al.Hypoxia-inducible factor-dependent breast cancer-mesenchymal stemcell bidirectional signaling promotes metastasis. J Clin Invest 2013;123:189–205.

35. MurphyMP.Howmitochondria produce reactive oxygen species. BiochemJ 2009;417:1–13.

36. Ma B, Chen Y, Chen L, Cheng H, Mu C, Li J, et al. Hypoxia regulatesHippo signalling through the SIAH2 ubiquitin E3 ligase. Nat Cell Bio2015;17:95–103.

37. Pernicova I, Korbonits M.Metformin–mode of action and clinical implica-tions for diabetes and cancer. Nat Rev Endocrinol 2014;10:143–56.

38. Lee JH, Kim TI, Jeon SM, Hong SP, Cheon JH, Kim WH. The effects ofmetformin on the survival of colorectal cancer patients with diabetesmellitus. Int J Cancer 2012;131:752–9.

39. HeX, Esteva FJ, Ensor J,Hortobagyi GN, LeeMH, Yeung SC.Metformin andthiazolidinediones are associated with improved breast cancer-specificsurvival of diabetic women with HER2þ breast cancer. Ann Oncol 2012;23:1771–80.

40. Hadad S, Iwamoto T, Jordan L, Purdie C, Bray S, Baker L, et al. Evidence forbiological effects of metformin in operable breast cancer: a pre-operative,window-of-opportunity, randomized trial. Breast Cancer Res Treat 2011;128:783–94.

41. Hong X, Song R, Song H, Zheng T,Wang J, Liang Y, et al. PTEN antagonisesTcl1/hnRNPK-mediated G6PD pre-mRNA splicing which contributes tohepatocarcinogenesis. Gut 2014;63:1635–47.

42. Mollbrink A, Jawad R, Vlamis-Gardikas A, Edenvik P, Isaksson B,Danielsson O, et al. Expression of thioredoxins and glutaredoxins inhuman hepatocellular carcinoma: correlation to cell proliferation,tumor size and metabolic syndrome. Int J Immunopathol Pharmacol2014;27:169–83.

43. Sun QK, Zhu JY, Wang W, Lv Y, Zhou HC, Yu JH, et al. Diagnostic andprognostic significance of peroxiredoxin 1 expression in human hepato-cellular carcinoma. Med Oncol 2014;31:786.

44. Guo W, Zhao Y, Zhang Z, Tan N, Zhao F, Ge C, et al. Disruption of xCTinhibits cell growth via the ROS/autophagy pathway in hepatocellularcarcinoma. Cancer Lett 2011;312:55–61.

45. Harris IS, Treloar AE, Inoue S, Sasaki M, Gorrini C, Lee KC, et al. Gluta-thione and thioredoxin antioxidant pathways synergize to drive cancerinitiation and progression. Cancer Cell 2015;27:211–22.

www.aacrjournals.org Clin Cancer Res; 22(12) June 15, 2016 3117

NDUFA4L2 in HCC Development

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987

Page 14: NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma · Biology of Human Tumors NDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular Carcinoma Robin Kit-Ho Lai1, Iris

2016;22:3105-3117. Published OnlineFirst January 27, 2016.Clin Cancer Res   Robin Kit-Ho Lai, Iris Ming-Jing Xu, David Kung-Chun Chiu, et al.   CarcinomaNDUFA4L2 Fine-tunes Oxidative Stress in Hepatocellular

  Updated version

  10.1158/1078-0432.CCR-15-1987doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2016/01/27/1078-0432.CCR-15-1987.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/22/12/3105.full#ref-list-1

This article cites 45 articles, 11 of which you can access for free at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/22/12/3105To request permission to re-use all or part of this article, use this link

Research. on September 27, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 27, 2016; DOI: 10.1158/1078-0432.CCR-15-1987