Basic Study Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 21, 2015; 21(7): 2011-2029
Published online Feb 21, 2015. doi: 10.3748/wjg.v21.i7.2011
Gene expression profiling of MYC-driven tumor signatures in porcine liver stem cells by transcriptome sequencing
Rajagopal N Aravalli, Department of Radiology, University of Minnesota Medical School, Minneapolis, MN 55455, United States
Neil C Talbot, Beltsville Agricultural Research Center, US Department of Agriculture, Beltsville, MD 20705, United States
Clifford J Steer, Departments of Medicine, and Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 54455, United States
Author contributions: Aravalli RN designed the study, performed all the experiments, and wrote the manuscript; Talbot NC provided PICM-19 cells; Steer CJ provided vital resources to carry out the proposed studies; all authors reviewed and revised the manuscript.
Supported by Departmental funds to Dr. Aravalli RN
Ethics approval: Institutional review board approval is not needed for this study since it does not involve any human subjects. Therefore, we do not have any documents to enclose with this revision.
Institutional animal care and use committee: All procedures involving mice were reviewed and approved by the institutional animal care and use Committee of the University of Minnesota (IACUC protocol number: 1107A01962).
Conflict-of-interest: All authors have no conflict of interest. We have included the funding source in the title page of the revised manuscript.
Data sharing: Additional information of RNA-seq dataset is available from the corresponding author at arava001@umn.edu.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Rajagopal N Aravalli, PhD, Department of Radiology, University of Minnesota Medical School, MMC 292 Mayo, 420 Delaware Street SE, Minneapolis, MN 55455, United States. arava001@umn.edu
Telephone: +1-61-26265540 Fax: +1-61-26265580
Received: September 4, 2014
Peer-review started: September 4, 2014
First decision: October 29, 2014
Revised: November 6, 2014
Accepted: December 14, 2014
Article in press: December 16, 2014
Published online: February 21, 2015

Abstract

AIM: To identify the genes induced and regulated by the MYC protein in generating tumors from liver stem cells.

METHODS: In this study, we have used an immortal porcine liver stem cell line, PICM-19, to study the role of c-MYC in hepatocarcinogenesis. PICM-19 cells were converted into cancer cells (PICM-19-CSCs) by overexpressing human MYC. To identify MYC-driven differential gene expression, transcriptome sequencing was carried out by RNA sequencing, and genes identified by this method were validated using real-time PCR. In vivo tumorigenicity studies were then conducted by injecting PICM-19-CSCs into the flanks of immunodeficient mice.

RESULTS: Our results showed that MYC-overexpressing PICM-19 stem cells formed tumors in immunodeficient mice demonstrating that a single oncogene was sufficient to convert them into cancer cells (PICM-19-CSCs). By using comparative bioinformatics analyses, we have determined that > 1000 genes were differentially expressed between PICM-19 and PICM-19-CSCs. Gene ontology analysis further showed that the MYC-induced, altered gene expression was primarily associated with various cellular processes, such as metabolism, cell adhesion, growth and proliferation, cell cycle, inflammation and tumorigenesis. Interestingly, six genes expressed by PICM-19 cells (CDO1, C22orf39, DKK2, ENPEP, GPX6, SRPX2) were completely silenced after MYC-induction in PICM-19-CSCs, suggesting that the absence of these genes may be critical for inducing tumorigenesis.

CONCLUSION: MYC-driven genes may serve as promising candidates for the development of hepatocellular carcinoma therapeutics that would not have deleterious effects on other cell types in the liver.

Key Words: Hepatocellular carcinoma, MYC, Stem cells, Gene expression, RNA sequencing

Core tip: It is well-established that cancer stem cells not only drive tumor growth in the liver, in general, but also that the proto-oncogene c-MYC plays a critical role in that process. However, little is known about genes induced and regulated by MYC to generate tumors, and, in particular, those involved in liver stem cells. In this study, we examined the role of MYC protein in hepatocarcinogenesis using an immortal porcine liver stem cell line, PICM-19. Interestingly, MYC-overexpression silenced the expression of six genes in PICM-19 cells (CDO1, C22orf39, DKK2, ENPEP, GPX6, SRPX2), suggesting that they may be critical in inducing tumorigenesis.



INTRODUCTION

Hepatocellular carcinoma (HCC) is the most common form of liver cancer, and is the third leading cause of cancer-related deaths worldwide[1]. It occurs as a consequence of exposure to a variety of risk factors, and appears primarily in liver disease patients with underlying cirrhosis[2]. While surgical resection and whole liver transplantation are the most effective treatment options currently available for the treatment of HCC, recurrence is quite common in patients who have had a resection, and the 5-year survival rate post-surgery is < 50%[3].

Hepatocarcinogenesis is a multistep process involving a number of cellular and molecular factors. Among these, the proto-oncogene c-MYC (herein, referred to as MYC) has been shown to be a critical regulator of malignant transformation during the early stages of liver cancer[4]. Dysregulation of MYC expression correlates with poor prognosis in human cancers, including HCC[5]. Its overexpression, and subsequent induction of its target genes, causes the malignant conversion of preneoplastic liver lesions[4]. Conversely, silencing of MYC results in the inhibition of migration, invasion and proliferation of human liver cancer cells[6]. Therefore, the study of MYC-expressing liver stem cells could be highly relevant to our understanding of the pathogenesis in human HCC. Due to the involvement of the MYC oncoprotein in angiogenesis and various cellular processes such as cell cycle regulation, differentiation, growth, metabolism, adhesion and motility, MYC, and its downstream target genes have become attractive therapeutic aims of cancer research[7].

It is now apparent that the capacity to sustain tumor growth resides in a small population of stem cells [termed “cancer stem cells” (CSCs)] present in most cancers, including HCC[8-10]. CSCs possess greater colony-forming efficiency, higher proliferation potential, and the necessary phenotype of “stemness” to form tumors in animals[5]. To support this, CD133-expressing progenitor cells were shown to drive the recurrence of liver cancer following chemotherapy, suggesting that CSCs are resistant to treatment[11]. A possible mechanism for the enhanced survival of CSCs in tumors is the activation of the Akt (also known as protein kinase B) and B-cell lymphoma 2 (Bcl-2) signaling pathways[11]. Therefore, a critical step towards developing effective cancer treatments is to understand cellular, molecular and biochemical differences between normal stem cells and CSCs.

To decipher the role of MYC and MYC-targeted genes in HCC, we have developed a liver cell model of MYC oncogene transformation based on the overexpression of MYC in a porcine liver stem cell line, PICM-19[12]. The PICM-19 cell line originated from the spontaneous differentiation of cultured pig epiblast tissue and was, therefore, derived from pig embryonic stem cells[13]. The cell line is unique in its ability to differentiate into either of the two cell types that comprise the parenchyma of the developing liver, i.e., fetal hepatocytes or bile duct epithelium, and grows indefinitely without change in stem cell character. In this study, we have MYC-transformed the PICM-19 cell line into CSCs, and studied their transcriptome profiles by RNA sequencing (RNA-seq). We determined that MYC induced the expression of a large number of genes with diverse cellular processes in PICM-19 cells, most of which were not previously implicated in HCC. Our data identified novel therapeutic targets while underscoring the importance of MYC in tumor generation in primary liver stem cells.

MATERIALS AND METHODS
Cell culture

The PICM-19 porcine liver stem cell line was established from a pig embryo’s inner cell mass, i.e., embryonic stem cells, and was cultured as described previously[12,13]. Briefly, cells were grown in 10% DMEM/199 medium and plated as feeder-free cultures using Matrigel (BD Biosciences, San Jose, CA). This growth medium is a 50:50 mixture of DMEM low glucose (Invitrogen, Carlsbad, CA) and Medium 199 (Invitrogen) supplemented with 10% fetal bovine serum, 2-mercaptoethanol and nucleosides. In subsequent steps, cells were fed daily with conditioned medium obtained from γ-irradiated STO feeder cells, a mouse embryonic fibroblast cell line (Cat. #CRL-1503, ATCC, Rockville, MD). PICM-19-CSCs, generated by transfecting the PICM-19 cell line (see below), were also maintained under these growth conditions. All cells were maintained at 37 °C in a 5% CO2 humidified incubator.

Plasmids

The expression vector pUNO1-hMYC was generated by cloning the human c-MYC open reading frame (ORF) into the multiple cloning site of the plasmid pUNO1-mcs (InvivoGen, San Diego, CA) downstream of a strong elongation factor (EF)-1α/human T-lymphotropic virus (HTLV) hybrid promoter active in most cell types. pUNO1-mcs contains the blasticidin resistance gene driven by a CMV promoter and enhancer in tandem with the bacterial EM7 promoter. This allows the amplification of the plasmid, and, after transfection into mammalian cells, the blasticidin selection of stable transfectants. Another plasmid, pUNO1-MYC-IRES-Luc was also constructed by cloning the firefly luciferase ORF downstream of MYC ORF separated by an internal ribosome entry site (IRES) sequence to maintain expression of both MYC and luciferase (luc) genes under the control of the hybrid promoter in this plasmid. Large scale preparations of both vectors were prepared by transforming them into competent cells of Escherichia coli strain DH5α, and by extracting them using the Qiagen Plasmid Maxi kit (Qiagen, Valencia, CA).

Luciferase assay

PICM-19 cells were successfully transfected with the pUNO1-MYC-IRES-Luc plasmid using the mouse macrophage nucleofection kit (Amaxa Biosystems, Gaithersburg, MD) and the program A-13 on the nucleofector I device (Amaxa). Following nucleofection, cells were plated in 12-well plates and incubated overnight at 37 °C. Growth medium was then replaced with the fresh medium containing 5 μg/mL blasticidin (InvivoGen) to select for positive transfectants. Individual colonies that formed were further grown, and assessed for MYC expression using reverse transcription polymerase chain reaction (RT-PCR) (data not shown). The clone that showed the highest MYC expression was then used in further experiments mentioned below. Next, cells of this clone were plated in 6-well plates, and 24 h post-plating they were resuspended in fresh medium and were treated with the Bright-Glo luciferase assay substrate (Promega, Madison, WI) to measure luciferase activity using the IVIS® Imaging System (Xenogen Corporation, Alameda, CA).

Western blotting

Western blot analysis of cellular proteins extracted from PICM-19 and PICM-19-CSCs was performed using mouse anti-human c-MYC antibody (Cat. #sc-40, Santa Cruz Biotechnology, Inc., Santa Cruz, CA). Ten μg of total protein from these cells was loaded onto a 10% denaturing SDS-PAGE gel. Following separation, proteins were transferred to a nitrocellulose filter and treated with blocking solution containing 5% milk powder. Primary antibody at a concentration of 1:500 was then added to the filter and incubated for 2 h in a rotating chamber at 4 °C. After several washes in buffer, the secondary anti-mouse IgG antibody (Santa Cruz Biotechnology) was added at a concentration of 1:1000 and the filter incubated for 2 h at RT. The blot was washed and probed with ECL solution to visualize protein bands (Thermo Fischer Scientific, Rockford, IL).

Tumorigenicity assay

Five-week old NOD/SCID mice were purchased from Charles River Breeding Laboratories (Wilmington, MA). Freshly cultured PICM-19 and PICM-19-CSCs were treated with trypsin to detach and harvest them in PBS. After washing twice in PBS, 1 × 106 cells were resuspended in 100 μL PBS, and were injected into the flanks of the immunodeficient mice (n = 3). This study was approved by the University of Minnesota Institutional Animal Care and Use Committee. Animals were observed on a daily basis for signs of declining health and lethargy. Tumor formation was monitored twice weekly by measuring the width and length of tumors. Animals with tumors that grew to a diameter of 1.5 cm, as measured by calipers, were sacrificed. To alleviate pain and stress in animals, carbon dioxide was administered in the fume hood prior to euthanasia. Tumors were then harvested and immediately fixed in 10% formalin for 24 h. These tissue samples were cut in 5 μm-thick sections on a microtome. Sections were deparaffinized with xylene, rehydrated in three five-minute steps with 100% ethanol, 90% ethanol, and 70% ethanol, and stained with hematoxylin and eosin (HE) for 1 h. Slides were rinsed with RNase-free water and mounted under glass coverslips with Clarion mounting medium (Santa Cruz Biotechnology). They were observed using a Nikon DXM1200C microscope.

RNA isolation and library preparation

Total cellular RNAs were isolated from PICM-19 and PICM-19-CSCs using the RNeasy Mini kit (Qiagen). They were quantified using a fluorimetric RiboGreen assay. Total RNA integrity was assessed by capillary electrophoresis using BioAnalyzer 2100 (Agilent Technologies, Santa Clara, CA, United States). For samples to pass the initial quality control (QC) step, it was necessary to quantify > 1 μg and to have a RNA Integrity Number of ≥ 8. They were then converted to Illumina sequencing libraries using the Truseq RNA Sample Preparation Kit (Cat. # RS-122-2001, Illumina, San Diego, CA). For amplification and library preparation, 1 μg of total RNA was purified using oligo-dT coated magnetic beads, fragmented and reverse transcribed into cDNA. The cDNA was fragmented, blunt-ended, and ligated to indexed (barcoded) adaptors and amplified using 15 cycles of PCR. Final library size distribution was validated using capillary electrophoresis and quantified using fluorimetry (PicoGreen) and by Q-PCR. Indexed libraries were normalized, pooled and size selected to 320bp +/- 5% using Caliper’s XT instrument.

Cluster generation and sequencing

Truseq libraries were hybridized to a paired-end flow cell and individual fragments were clonally amplified by bridge amplification on the Illumina cBot. Once clustering was complete, the flow cell was loaded onto the Illumina HiSeq 2000 sequencer and sequenced using Illumina’s SBS chemistry. Upon completion of read 1, a 7 base pair index read was performed. Finally, the library fragments were resynthesized in the reverse direction and sequenced from the opposite end of the read 1 fragment thus producing the template for paired end read 2.

Primary analysis and de-multiplexing

Base call (.bcl) files for each cycle of sequencing were generated by Illumina Real Time Analysis software. The base call files and run folders were then exported to servers maintained at the Minnesota Supercomputing Institute, where primary analysis and de-multiplexing were performed using Illumina’s CASAVA software 1.8.2. De-multiplexed FASTQ files generated by the CASAVA workflow were analyzed by the mapping and aligning software as described below.

Read mapping and RNA sequencing analysis

Raw paired-end RNA-seq reads in FASTQ format were assessed for base call quality, cycle uniformity and contamination using FastQC (http://www.bioinformatics.bbsrc.ac.uk/projects/fastqc/). The QC-filtered paired-end reads were mapped to pig genome (UCSC susScr3 assembly) via Tophat 2[14] using the UCSC pig annotation in GTF format. Resulting Binary Alignment and Mapping (BAM) files were then visualized using the Integrative Genomics Viewer (IGV)[15] to ensure reasonable behavior of expected control invariant genes [e.g., glyceraldehyde 3-phosphate dehydrogenase (GAPDH)], and to confirm expected behavior of positive control genes (e.g., MYC). BAM files were subsequently analyzed via Cuffdiff (default parameters using the iGenomes GTF file as shown previously[16]) to quantify the expression level(s) of each known gene in units of FPKM (Fragment mapped per kilobase of genes per million mapped reads), and tested for differential expression. A matrix of FPKM transcript abundance levels for each gene was extracted and downloaded into CummeRbund[17] to assess sample clustering using Multi-Dimensional Scaling and Principle Component Analysis, which revealed sample outliers. Differential gene sets were filtered to remove those with fold changes < 1.5 (up- or downregulated) and with a BH FDR-corrected P-value > 0.05.

Real time PCR

The cDNA from PICM-19 and PICM-19-CSCs was synthesized with 1 μg of total RNA using SuperScript II reverse transcriptase (Invitrogen Life Technologies, Carlsbad, CA) and oligo dT6-12 primers (Sigma-Genosys, The Woodlands, TX). Quantitative real-time PCR was performed using the FullVelocity SYBR Green QPCR master mix (Stratagene, La Jolla, CA) following the manufacturer’s specifications. The 25 μL final reaction volume consisted of pre-made reaction mix (SYBR Green I dye, reaction buffer, Taq DNA polymerase, and dNTPs), 0.3 mmol/L of each primer, and 0.5 ng cDNA in water. Reaction conditions for PCR for the Mx3000P QPCR System (Stratagene) were as follows: polymerase activation at 95 °C for 5 min, 40 denaturation cycles of 95 °C for 10 s, annealing at 60 °C for 10 s and elongation at 72 °C for 10 s. A melting curve was performed to assess primer specificity and product quality using the same conditions. The relative product levels were quantified using the 2(-∆∆CT) method[18]. Data are presented as relative induction of each porcine gene, normalized to β-actin, and are representative of three independent experiments.

RESULTS
Expression of MYC protein in PICM-19 stem cells

We initially determined that PICM-19 cells can be transfected with an expression vector driven by the EF-1α/HTLV hybrid promoter. pUNO1-MYC-IRES-Luc was constructed by cloning the open reading frame (ORF) of firefly luciferase (luc) gene downstream of the MYC ORF separated by an IRES sequence. This vector also contains the blasticidin resistance gene as a selectable marker. The function of the genetic construct was tested by assaying the expression of luciferase in transfected cells. After transfection, feeder-independent PICM-19 cells were cultured (Figure 1A) and transfectants were selected using blasticidin. Individual clones were then screened for MYC expression, and the clone that had the highest MYC expression as detected by RT-PCR (data not shown) was assayed for luciferase activity in 24-well plates using D-luciferin as the substrate. Transfected PICM-19 cells expressed luciferase demonstrating that they took up and incorporated the plasmid DNA (Figure 1B). This finding also indicated that the stem cells were expressing the MYC ORF located upstream of luc ORF in the expression vector. This was confirmed by Western blotting assay using anti-human MYC antibody (Figure 1C).

Figure 1
Figure 1 Generation of PICM-19-CSCs. A: Feeder-independent PICM-19 cells were grown in 10% DMEM/Medium 199 on Matrigel in 24-well plates (see Materials and Methods). They were transfected with pUNO1-MYC-IRES-Luc plasmid and tested for luciferase expression using luciferin as substrate; B: Untransfected PICM-19 cells did not show bioluminescence (left panel), whereas transfected cells expressed luciferase (right panel); C: Detection of MYC protein in PICM-19-CSCs by Western blotting using the anti-human mouse c-MYC monoclonal antibody. As expected, PICM-19-CSCs, but not the untransfected PICM-19 stem cells (control) expressed MYC.
Conversion of PICM-19 liver stem cells into cancer cells by MYC overexpression

We subcutaneously injected 1 × 106 cells into the back flanks of NOD-SCID mice to establish that MYC-overexpressing PICM-19 cells (PICM-19-CSCs) can generate tumors in mice. PICM-19-CSCs, but not the control PICM-19 stem cells, generated tumors in vivo (Figure 2). When the tumors reached 1.5 cm in diameter, animals were sacrificed and the tumors were sectioned to be analyzed by HE staining. The tumors were composed of a uniform mass of cells that were actively undergoing cell division. This experiment demonstrated the proof-of-principle that PICM-19-CSCs are capable of producing tumors in vivo.

Figure 2
Figure 2 PICM-19-CSCs generate tumors after implantation in mice. A: PICM-19 cells were transfected with 3 μg of pUNO1-hMYC by nucleofection. 1 x 106 PICM-19 cells expressing MYC were implanted into the back flanks of NOD-SCID mice (n = 3) by subcutaneous injections. Tumors that grew to 1.5 cm in diameter were harvested; B and C: Tumors were sectioned and HE staining was performed, and cells were visualized by confocal microscopy (B: × 40; C: × 400). These images show that the PICM-19-CSCs induced tumors were uniform, and the cells were actively dividing in the PICM-19-CSC tumors, as seen by mitotic cell division [indicated by arrows in (C)].
Molecular characterization of PICM-19 and PICM-19-CSCs

To understand the differences between PICM-19 and PICM-19-CSCs at the molecular level, we examined the RNA-seq profiles of both these cell types, and found sets of genes that were expressed in each cell line (Tables 1 and 2). These genes were normalized to the expression of housekeeping gene GAPDH, and were analyzed. Only a two-fold change or greater was considered significant. In this process, we identified a set of six genes that were expressed only in PICM-19 stem cells (Table 1). The expression of these six genes (CDO1, C22orf39, DKK2, ENPEP, GPX6 and SRPX2) was undetectable in PICM-19-CSCs, as a result of MYC protein overexpression. Their absence in PICM-19-CSCs suggested their potential use as therapeutic targets for liver cancers associated with MYC overexpression. Among these, CDO1 is a tumor suppressor[19], while Dickkopf-related protein 2 (DKK2) is an inhibitor of the Wnt signaling pathway[20]. Both these genes are epigenetically silenced in human cancers. While the overexpression of DKK2 led to an inhibition in renal cancer progression through apoptotic and cell cycle pathways[20], forced expression of cysteine dioxygenase type 1 (CDO1) decreased tumor cell growth of different tumor cell lines and in mice[19]. Lack of DDK2 expression in PICM-19-CSCs suggested that a putative epigenetic control mechanism to silence DKK2 also exists in MYC-induced liver cancer.

Table 1 Genes expressed by PICM-19 liver stem cells.
Gene IDGene abbreviationFunctionFold Δ1
C22ORF39Human chromosome 22 orf39 homologUnknown11.33
CDO1Cysteine dioxygenase type 1Tumor suppressor6.28
DKK2Dickkopf Wnt signaling pathway inhibitor 2Cell signaling12.20
ENPEPGlutamyl aminopeptidaseProteolysis4.82
GPX6Glutathione peroxidase 6Detoxification2.18
SRPX2Sushi-repeat containing protein, X-linked 2Angiogenesis17.22
Table 2 Genes expressed in PICM-19-CSCs but not in PICM-19 cells.
Gene IDGene abbreviationFold Δ1
VEGFVascular endothelial growth factor41718.7
snoZ40Small nucleolar RNA Z4016658.2
ssc-mir-195a-1MicroRNA mir-195a-16514.83
ssc-mir-125b-1MicroRNA mir-125b-13627.42
ssc-mir-423MicroRNA mir-4233627.42
SNORD53Small nucleolar RNA, C/D box532811.10
ssc-mir-421MicroRNA mir-4212811.10
ssc-mir-133a-2MicroRNA mir-133a-22558.12
ssc-mir-365-1MicroRNA mir-365-12558.12
SNORD16Small nucleolar RNA, C/D box 161497.27
ssc-mir-31MicroRNA mir-311144.09
SERP2Stress-associated ER protein family member 21137.16
SNORD94Small nucleolar RNA, C/D box 941126.85
SNORD67Small nucleolar RNA, C/D box 67816.01
Y_RNANon-coding RNA727.41
GRIAFAD-dependent oxygenase370.13
CH242-74M17.6GAS locus protein, porcine-specific322.43
CH242-74M17.6GNAS complex locus, porcine-specific322.43
SNORA36Small nucleolar RNA, C/D box 36199.10
RBP1Retinol-binding protein 1182.72
TIMP-3TIMP metallopeptidase inhibitor 3173.55
RNF130Ring finger protein 130147.15
MT-ND6Mitochondrial NADH dehydrogenase 6141.82
PCDH7Protocadherin 790.60
ND4LNADH dehydrogenase subunit 4L89.11
U12Non-coding RNA85.94
MT1AMetallothionein 1A84.09
ZFXZinc finger protein X-linked79.94
CH242-17J7.1GNAS complex locus, porcine-specific65.88
RNPC1RNA binding motif protein 3865.10
GEM1Gremlin 154.41
CADM4Cell adhesion molecule 439.92
CYP1A1Cytochrome P450, family 1, subfamily A139.89
TFAP2ATranscription factor AP-2 alpha39.27
AFPAlpha-fetoprotein33.23
CFDComplement factor D26.62
GNGTwo-component system response regulator GnG/NtrC24.20
RNF128Ring finger protein 12823.79
MIFMacrophage migration inhibitory factor23.69
IFI27l2Interferon, alpha-inducible protein 27-like 222.72
PDE6HPhosphodiesterase 6H, cGMP-specific22.54
KLF5Krüppel-like factor 522.53
PRKD1Protein kinase D121.27
SPINT2Serpin peptidase inhibitor, Kunitz type, 218.54
MYCMyelocytomatosis oncogene17.64
CD44Cell surface glycoprotein 4417.14
GPC3Glypican 316.54
ECHS1Enoyl-coA hydratase, short-chain, 115.42
S100A14S100 calcium binding protein A1415.41
LONRF3LON peptidase N-terminal domain and ring finger 315.25
CU463271.1Protein encoded by CU463271.1 locus14.86
MMP16Matrix metalloproteinase 1614.28
S100A16S100 calcium binding protein A1614.14
SLC22A17Solute carrier family 22 member A1714.07
ID1Inhibitor of DNA binding 113.34
ATP5EATP synthase H+ transporting epsilon13.14
ISG12(a)ISG12 protein-like13.03
UQCRHUniquinol-cytochrome c reductase hinge protein12.44
DKK2Dickkopf homolog 212.20
IL17RAInterleukin 17 receptor A12.13
STXBP6Syntaxin binding protein 611.63
F2Coagulation factor II11.57
COX17Cytochrome c oxidase copper chaperone10.05
MGMTO-6-methylguanine-DNA methyltransferase9.96
SDF4Stromal cell derived factor 49.50
PDPNPodoplanin9.46
DLK2Delta-like 2 homolog9.24
WNT1Wingless-related MMTV integration site family member 19.03
CISD1CDGSH iron sulfur domain 19.00
MYD88Myeloid differentiation primary response 888.71
SDC4Syndecan 48.68
EPCAMEpithelial cell adhesion molecule8.59
RPS29Ribosomal protein S298.32
P14ARFp14 ARF protein (porcine-specific)8.29
ELAVL2ELAV-like neuron-specific RNA binding protein 28.23
FZD3Frizzled homolog 38.17
XBP1X-box binding protein 18.09
TRIAP1TP53 regulated inhibitor of apoptosis 18.08
ATP5OATP synthase H+ transporting O subunit8.02
VKORC1Vitamin K epoxide reductase complex, subunit 17.80
FAM84BFamily with sequence similarity 84, member B7.70
ECI2Enoyl-coA delta isomerase 27.69
ARHGDIBRho GDP dissociation inhibitor beta7.41
C20ORF27Human chromosome 22 orf39 homolog7.31
RAB6BRAS oncogene family member7.28
DDTD-dopachrome tautomerase7.14
AGMATAgmatine ureohydrolase (Agmatinase)7.12
CHCHD2Coiled-coil-helix-coil-helix domain 26.92
EPHA7Eph receptor A76.72
OSR2Odd-skipped related 26.71
CFIComplement factor I6.55
NDUFB11NADH dehydrogenase 1 alpha subcomplex 116.37
SLC35F1Solute carrier family 35 member F16.37
NUDT14Nucleoside diphosphate linked moiety X-type motif 146.35
SH3GL3SH3 domain GRB2-like 36.32
NDUFA12NADH dehydrogenase 1 alpha subcomplex 126.30
CDO1Cysteine dioxygenase type 16.28
DYNLT1Dynein, light-chain, Tctex-type6.27
HSBP1Heat shock factor binding protein 16.20
CCKChloecystokinin5.98
ANXA1Annexin A25.85
FCGRTFc fragment of IgG, receptor, transporter, alpha5.84
AK5Adenylate kinase 55.83
RDH11Retinol dehydrogenase 115.58
RPS18Ribosomal protein S185.56
TGOLN2Trans-golgi network protein 25.55
RNF150Ring finger protein 1505.54
HIFXHypoxia-inducible factor X5.47
CFHComplement factor H5.43
NPC2Niemann-Pick disease, type C25.34
DCTPP1dCTP pyrophosphatase 15.25
CLVS1Clavesin 15.24
HSF4Heat shock transcription factor 45.23
SERINC2Serine incorporator 25.08
MSMBMicroseminoprotein, beta5.03
CHID1Chitinase domain containing 14.98
LYSMD2LysM, putative peptidoglycan-binding, domain 24.95
SH3BGRL2SH3 domain binding glutamic acid-rich protein-like 24.86
POU3F1POU domain, class 3, transcription factor 14.77
TCN2Transcobalamin II4.76
SORT1Sortilin 14.75
PTGESProstaglandin E synthase4.65
CCND2Cyclin D24.63
GXYLT2Glucoside xylosyltransferase 24.50
CRTAC1Cartilage acidic protein 14.33
MFGE8Milk fat globule-EGF factor 84.29
TNCTenascin C4.28
PCBD1Pterin-4 alpha-carbinolamine4.27
PDE10APhosphodiesterase 10A4.22
RAB27BRAS oncogene family member4.18
INCENPInner centromere protein antigens 135/155kDa4.14
GJB1Gap junction protein, beta 14.09
CEBPGCCAAT/enhancer binding protein, gamma4.04
DUROC-SLA1Src-like adaptor 1 (pig-specific)4.04
ITIH4Inter-alpha-trypsin inhibitor heavy chain family, member 44.03
PEBP1Phosphatidylethanolamine binding protein 14.02
ABTB2Ankyrin repeat and BTB (POZ) domain containing 24.00
GJB5Gap junction protein, beta 53.95
CREB3cAMP responsive element binding protein 33.86
DPM2Dolichyl-phasphate mannosyltranferase polypeptide 23.85
GEMGTP binding protein overexpressed in skeletal muscle3.85
DOCK8Dedicator of cytokinesis 83.84
TMP-SLA-2Transmembrane protein, pig-specific3.83
CYB5BCytochrome b5 type B3.80
TSAN6Tsan6, Pig-specific protein3.75
RNASEH2CRibonuclease H2, subunit C3.71
NARSAsparaginyl-tRNA synthetase3.70
LSM6U6 snRNP-associated protein3.69
MTFP1Mitochondrial fission process 13.66
SLC16A1Solute carrier family 16 member A13.65
RARBRetinoic acid receptor, beta3.64
SDSLSerine dehydrarase-like3.64
MPC2Mitochondrial pyruvate carrier 23.63
PARVBParvin, beta3.60
RENBPRenin binding protein3.55
POC1APOC1 centrolar protein A3.54
GUSBGlucuronidase, beta3.53
CH242-9E10.1GNAS complex locus, porcine-specific3.52
REEP3Receptor accessory protein 33.51
CPT1ACarnitine palmitoyltransferase 1A3.40
BHLHB9Basis helix-loop-helix domain containing B93.37
FAAHFatty acid amide hydrolase3.37
MRPL20Mitochondrial ribosomal protein L203.37
HTR2A5-hydroxytryptamine (serotonin) receptor 2A, G protein-coupled3.36
TRERF1Transcriptional regulating factor 13.35
XYLBXylulokinase3.32
RBPMS2RNA binding protein with multiple splicing 23.28
FGFR3Fibroblast growth factor receptor 33.27
SLC22A23Solute carrier family 22 member A233.21
NDUFB1NADH dehydrogenase 1 alpha subcomplex 13.18
ZFAND2AZinc finger, AN1-type domain 2A3.16
EPB41Erythrocyte membrane protein band 4.13.11
IGSF23Immunoglobulin superfamily, member 233.11
FAM122CFamily with sequence similarity 122, member C3.10
AMIGO2Adhesion molecule with Ig-like domain3.03
SHROOM4Shroom family member 43.02
MAT1AMethionine adenosyltransferase 1A3.01
PARNPoly(A)-specific ribonuclease3.00
PFKFB16-phosphofructo-2-kinase/fructose-2,6-biphosphatase 13.00
TCF25Transcription factor 252.98
PRODH2Proline dehydrogenase (oxidase) 22.95
CHCHD10Coiled-coil-helix-coil-helix domain 102.91
CH242-217H9.3GNAS complex locus, porcine-specific2.85
CASC5Cancer susceptibility candidate 52.84
IGLON5IgLON family member 52.84
CDH17Cadherin 172.82
SLC44A4Solute carrier family 44 member A42.82
LRCH2Leucine-rich repeats and calponin homology domain 22.76
DUROC-NEU1Sialidase 1 (pig-specific)2.75
CDC45Cell division cycle 452.71
BCKDHBBranched chain keto acid dehydrogenase E12.69
HNF4AHepatocyte nuclear factor 4, alpha2.67
SCG5Secretogranin V2.66
BOLA1BolA family member 12.65
PYGLPhosphorylase, glycogen, liver-specific2.64
SCARB1Scavenger receptor class B, member 12.64
SERPIND1Serpin peptidase inhibitor, clade D, member 12.63
GAS2L3Growth arrest-specific 2 like 32.61
EFNA2Ephrin A22.60
PDZK1IP1PDZK1 interacting protein 12.59
VIMPVCP-interacting membrane protein2.59
PRR5LProline-rich 5-like2.57
ACVR2BActivin A receptor, type IIB2.56
ATP9AATPase, class II, type 9A2.55
PLAURPlasminogen activator, urokinase receptor2.53
LDLRLow density lipoprotein receptor2.51
FLRT3Fibronectin leucine-rich transmembrane protein 32.50
ADAMTSL3ADAMTS-like 32.49
CINPCyclin-dependent kinase 2 interacting protein2.48
WBSCR22Williams Beuren syndrome chromosome region 222.48
ALDH9A1Aldehyde dehydrogenase 9 family member 12.46
SSC.82438Sus scrofa Amphiregulin2.44
NME3NME/NME23 nucleoside diphosphate kinase 32.43
CCSCopper chaperone for superoxide dismutase2.40
PITX2Paired-like homeodomain transcription factor 22.38
MRPL3Mitochondrial ribosomal protein L32.33
BNC1Basonuclin 12.32
RPS16Ribosomal protein S162.32
TM7SF2Transmembrane 7 superfamily member 22.30
CRELD2Cystein-rich with EGF-like domains 22.22
HHEXHematopoietically expressed homeobox2.20
RPL31Ribosomal protein L312.20
TGFATransforming growth factor, alpha2.20
IDSIduronate 2-sulfatase2.18
NRG4Neuregulin 42.17
DGAT1Diacylglycerol O-acyltransferase 12.16
GNAZGuanine nucleotide binding protein2.14
ARL3ADP-ribosylation factor-like 32.12
FABP5Fatty acid binding protein 52.12
KIF12Kinesin family member 122.12
IKBKGInhibitor of kappa B2.09
PDZRN3PDZ domain containing ring finger 32.09
PHF19PHD finger protein 192.09
MRPL24Mitochondrial ribosomal protein L242.07
ARAP2ArfGAP with RhoGAP domain, ankyrin repeat and PH domain 22.04
ITGF3ItgF3-like protein2.02
POLA2Polymerase (DNA directed), alpha 22.02
ADAMTS7ADAM metallopeptidase with thrombo- Spondin type 1 motif 71.45

In contrast, ectopic expression of MYC protein upregulated the expression of 237 genes in PICM-19-CSCs not found in the PICM-19 stem cell profile (Table 3). New genes upregulated by MYC included those involved in cell growth and proliferation, adhesion, migration, protein transport, cell cycle, metabolism, inflammation, and tumorigenesis underscoring the broad reach of MYC protein in regulating diverse cellular processes. However, genes coding RNAs involved in transcriptional and post-transcriptional regulatory mechanisms dominated the top 25 MYC-upregulated genes (Table 2), consistent with the active cell growth and proliferative nature of PICM-19-CSCs.

Table 3 Genes upregulated by MYC in PICM-19-CSCs.
Gene IDGene abbreviationFunctionlog2 ratio
CH242-278B18.2GNAS Complex, porcine-specificUnknown10.25
NFKB1Nuclear factor κBInflammation8.94
SEMA3FSemaphorin 3FCell signaling8.27
NME1-NME2NME1-NME2 read-throughInflammation8.22
SERPINA1Serpin protease inhibitor, clade A1Inflammation8.17
VLDLRVery low density lipoprotein receptorMetabolism8.01
APOEApolipoprotein EMetabolism7.57
ITGA3Integrin, alpha 3Migration, Cell adhesion, Invasion7.41
IGF2BP2Insulin-like growth factor 2 mRNA binding protein 2Gene regulation7.33
MT-ATP6Mitochondrial ATP synthase 6Metabolism7.21
HIF1AHypoxia-inducible factor 1αAngiogenesis7.12
APOA1Apolipoprotein A1Metabolism7.06
ITGB7Integrin, beta 7Cell adhesion7.03
NKAIN1Na+/K+ transporting ATPaseIon transport7.00
FGGFibrinogen gamma chainCell adhesion6.79
CTSBCathepsin BCarcinogenesis6.74
NTN1Netrin 1Inflammation6.65
JUPJunction plakoglobinCell adhesion6.61
PLGPlasminogenCell proliferation6.60
MT-ND2Mitochondrial NADH dehydrogenase 2Metabolism6.37
SERPINA3-2Serpin protease inhibitor, clade A3-2Inflammation6.31
RBP4Retinol binding protein 4Metabolism6.18
FTLFerritin, light polypeptideIron metabolism6.08
DUSP4Dual specificity phosphatase 4Cell proliferation6.02
MT-CO1Mitochondrial cytochrome c oxidase IInflammation6.00

The farnesoid X receptor (FXR), a member of the nuclear receptor superfamily, plays an important role in regulating the progression of several cancers including HCC; and miR-421, which was highly upregulated in PICM-19-CSCs, suppresses the expression of FXR to induce tumorigenesis and migration in HCC cells[21]. MiR-421 also regulates deleted in pancreatic cancer 4 (DPC4; also known as Smad4), a critical tumor suppressor shown to be involved in the progression of pancreatic cancer[22].

In addition to these regulatory RNAs, protocadherin-7, tissue inhibitor of metalloproteinase 3 and vascular endothelial growth factor (VEGF) were highly expressed in PICM-19-CSCs compared to PICM-19 parental stem cells. The ratio between the matrix metalloproteinsases (MMPs) and their inhibitors (TIMPs) is an important factor in balancing tissue homeostasis. Recent studies have shown that the TIMP family of proteins not only inhibits several disintegrin-metalloproteinases (ADAMs and ADAMTSs) but also has contrasting functions including anti- and pro-apoptotic[23]. The sequestration of TIMP-3 by binding to the extracellular matrix (ECM) or through endocytosis may help to protect cells from apoptosis while still allowing it to control pericellular proteolysis via its potent inhibition of MMPs, ADAMs and ADAMTSs[23]. In PICM-19-CSCs, only TIMP-3 was upregulated (Table 2), and ADAM23 and ADAMTS2 were suppressed (Table 4).

Table 4 Genes downregulated by MYC in PICM-19-CSCs.
Gene IDGene abbreviationFunctionlog2 ratio
ASPNAsporinExtracellular matrix8.20
IGF1Insulin-like growth factor 1Cell growth7.44
OGNOsteoglycinMetabolism6.52
GPM6BGlycoprotein M6BMembrane trafficking, cell-to-cell communication6.23
ISLRImmunoglobulin superfamily containing leucine-rich repeatUnknown6.19
ANGPT1Angiopoietin 1Angiogenesis5.53
WDR66WD repeat domain 66EMT modulator5.49
RSPO2R-spondin 2 homologWnt modulator5.46
COL6A2Collagen, type VI, alpha 2Fibrosis5.14
TNRTenascin RExtracellular matrix5.03
FAM5CFamily with sequence similarity 5, member CUnknown4.97
FIGFc-Fos iduced growth factorAngiogenesis4.92
ADAM23ADAM metallopeptidase domain 23Metabolism4.84
MAB21L2Mab-21-like 2Cell survival4.83
PDE1APhosphodiesterase 1AMetabolism, Apoptosis4.83
CLMPCXADR-like membrane proteinCell adhesion4.74
COL28A1Collagen, type XXVIII, alpha 1Fibrosis4.74
GJA1Gap junction protein, alpha 1Oxidative stress4.67
SELENBP1Selenium binding protein 1Tumor suppressor4.58
CPED1Cadherin-like and PC-esterase domain containing 1Unknown4.52
COL5A3Collagen, type V, alpha 3Fibrosis4.51
ADAMTS2ADAM metallopeptidase with thrombospondin typ1 motif 2Metabolism4.50
PDE8BPhosphodiesterase 8BMetabolism4.47
SGCESarcoglycan, epsilonCytoskeleton4.42
GDPD2Glycerophosphodiester phosphodiesterase domain containing 2Cell growth4.00

It has been well established that VEGF is highly produced in HCC, and the inhibition of VEGF results in the suppression of angiogenesis[24]. Moreover, miR-26a targets VEGF and hepatocyte growth factor (HGF) to cause this effect via the HGF-cMet pathway[24] as well as the phosphatidylinositol 3-kinase (PIK3C2α)/Akt/hypoxia-inducible factor (HIF)-1α pathway in HCC cells[25]. In our study, downregulation of miR-26a and upregulation of HIF-1α in PICM-19-CSCs demonstrates that MYC is also a player in this tumor-inducing molecular mechanism of HCC.

Genes not regulated by the MYC protein in PICM-19-CSCs

Next, we studied the profiles of PICM-19 and PICM-19-CSCs to identify the genes whose expression was not altered in stem cells due to MYC overexpression. For this, we selected genes that showed more than a two-fold increase when normalized with the GAPDH. We found that the expression of 28 genes was not affected by the MYC protein (Table 5). These genes included a number of microRNAs (miRNAs) and small nucleolar RNAs (SnoRNAs) that regulate post-transcriptional gene expression, and are expressed at very high levels. As MYC has no influence on their expression, it appears that the genes of this 28-gene set may be primarily involved in maintaining the cellular integrity and stemness.

Table 5 Genes expressed by both PICM-19 and PICM-19-CSCs that were not affected by MYC overexpression.
Gene IDGene abbreviationFunctionFold Δ1
SSC-MIR-425MicroRNA mir-425Gene regulation7925.28
SNORD53Small nucleolar RNA, C/D box 53Gene regulation4969.50
SSC-MIR-378-1MicroRNA mir-378-1Gene regulation3962.64
SSC-MIR-99AMicroRNA mir-99aGene regulation3761.27
SSC-MIR-101-2MicroRNA mir-101-2Gene regulation3400.56
SSC-LET-7F-2MicroRNA let-7f-2Gene regulation2814.68
5s rRNA5S ribosomal RNAProtein synthesis2255.63
SSC-MIR-210MicroRNA mir-210Gene regulation1730.53
U6Small nucleolar RNA U6RNA interference1187.43
SSC-MIR-196A-2MicroRNA mir-196a-2Gene regulation1127.81
SCARNA15Small Cajal body-specific RNA 15Gene regulation287.24
SNORA72Small nucleolar RNA, H/ACA box 72Gene regulation255.25
SCARNA11Small Cajal body-specific RNA 11Gene regulation163.50
RAMP2Receptor (G protein-coupled) activity modifying protein 2Protein transport56.66
CCNI2Cyclin I family member 2Cell cycle10.67
P4HTMProlyl 4-hydroxylase, transmembraneHypoxia6.25
SPDYASpeedy/RINGO cell cycle regulator family member ACell cycle5.56
PARK2Parkin RBR E3 ubiquitin ligaseUnknown5.09
GRPRGastrin-releasing peptide receptorCell proliferation4.52
ATG10Autophagy related 10Autophagy3.76
PHYHIPPhytanoyl-CoA hydroxylase interacting proteinCell proliferation3.41
DPEP1Dipeptidase 1Metabolism2.72
DUROC-C2Nuclear receptor corepressor 2DNA binding2.64
HIST1H2BHHistone cluster 1, H2bhHistone modification2.58
PHKG1Phosphorylase kinase, gamma 1Glycogenolysis2.52
TMEM79Transmembrane protein 79Protein secretion2.04
APH1BAPH1B gamma secretase subunitProtein transport2.01
IRAK4Interleukin-1 receptor-associated kinase 4Cell signaling2.01
Differential expression of MYC-regulated genes in PICM-19-CSCs

In order to identify genes that are involved in MYC-induced oncogenesis, we studied global transciptome profiles of PICM-19-CSCs and compared them with those obtained with PICM-19 stem cells. While these genes are expressed by both PICM-19 and PICM-19-CSCs, a significant number of genes were differentially regulated by MYC. While MYC upregulated 4173 genes in PICM-19-CSCs, it downregulated 4248 genes. Statistical analysis of the data has revealed 616 upregulated and 391 downregulated genes that had a log2 ratio of at least 2. Analysis of gene expression of these two cell types showed that MYC overexpression resulted in changes in gene expression among genes involved in diverse cellular processes. This is not surprising as it is known that MYC regulates about 15% of the genes in mammalian genomes[26].

Among the genes upregulated by MYC are ribosomal genes demonstrating active protein synthesis in CSCs which is associated with cell growth, proliferation, and metabolism. While our data show only 8 of these ribosomal genes, others such as the initiation factors (elF-2a, eIF4A, eIF4E, eIF4G, eIF5A), MrDb, nucleolin, and ribosomal proteins (S11, S16) were also induced > 1.5 fold (data not shown). These genes were previously shown to be regulated by the MYC protein[27].

MYC is also considered an important regulator of the cell cycle. We found that the expression of several cell cycle genes (cdc45, CCND2, CIPN) was enhanced in PICM-19-CSCs. Adhesion molecule with Ig-like domain 2 (AMIGO2), a leucine-rich repeat family member implicated in decreased cell adhesion and migration, and an increase in chromosomal instability and tumorigenicity in human gastric adenocarcinoma[28], was also upregulated in PICM-19-CSCs.

The majority of the top 25 genes upregulated by MYC are those involved in metabolism, adhesion and inflammation (Table 3). Integrin genes ITGA3 and ITGB4, and plakoglobin JUP were upregulated in PICM-19-CSCs. These three genes were recently found to be responsible for metastasis seen in oral squamous cell carcinoma[29]. Their expression in CSCs is significant because integrin adhesion receptors link the ECM to actin cytoskeleton and transmit biochemical signals across the plasma membrane[30]. Binding of integrins to their cognitive receptors trigger signaling cascades to induce diverse cellular processes such as cell survival, apoptosis, proliferation, and motility[29]. JUP expression was also associated with colorectal tumor metastasis[31].

Interestingly, markers of inflammation in the liver, apolipoproteins (APOA1 and APOE) and the SERPINA1 gene encoding alpha1-antitypsin (AAT), and another member of the AAT family, SERPINA3-2 were also induced in PICM-19-CSCs. While APOE was shown to confer protection against liver damage caused by hepatitis C virus (HCV) infection[32], its expression level was found to be significantly higher in HBV associated liver cirrhosis[33]. In a recent HCC study on familial aggregation, APOA1 and APOE were identified by mass spectroscopy of the proteins as being among nine highly overexpressed in individuals with HCC[34]. Previous studies have shown that the protein level of APOE was increased in human HCC[35], and that human hepatoma-derived cell lines express high levels of APOE and very low density lipoprotein (VLDL). Consistent with this, PICM-19-CSCs displayed an upregulated expression of VLDLR (Table 3). AAT is the most abundant liver-derived glycoprotein in the plasma, and a diagnostic and prognostic marker of HCC[36,37]. It is associated with the progression from liver cirrhosis to cancer in HCV-infected individuals[38]. Collectively, upregulation of APOA1, APOE, VLDLR and AAT in PICM-19-CSCs suggested their involvement in MYC-induced liver cancer.

We found that MYC overexpression resulted in downregulation of a number of genes (Table 4) that were previously implicated in tumor reduction and/or inhibition of human cancers. These genes differ from those that were silenced; and the reduced expression could be a result of either direct or indirect MYC targeting (Table 1). Among these, c-Fos induced growth factor, FIGF (also known as vascular endothelial growth factor D), is active in angiogenesis and endothelial cell growth. Its putative role in human cancers is unclear. While it was shown to enhance lymphatic node metastasis in tissue sarcomas[39], its expression was significantly lower in the serum of metastatic patients with differentiated thyroid sarcoma suggesting that FIGF may prevent metastasis. Interestingly, MYC caused the downregulation of FIGF in PICM-19-CSCs (Table 4) indicating that, in liver stem cells, FIGF has an anti-tumor function. Osteoglycin, a protein that decreases gelatinase activity and suppresses lymphatic metastasis[40], was also downregulated in PICM-19-CSCs.

Collagens are structural components of the ECM and are primarily synthesized by hepatic stellate cells in the liver to induce fibrosis[41]. A recent proteomic study with platelet-derived growth factor C transgenic and phosphatase and tensin homolog null mouse models of liver fibrosis and its progression to HCC demonstrated that the protein levels of collagens COL6A2 and COL28A1 were downregulated in fibrosis, and COL5A3 was upregulated in fibrosis; whereas only COL6A2, but not COL5A3 and COL28A1, was increased in tumors generated in both mouse models[42]. We found that COL5A3, COL6A2, COL28A1 were significantly downregulated in PICM-19-CSCs. Thus, in liver cells, MYC may be involved in the regulation of COL5A3 and COL28A1.

Among regulatory RNAs upregulated in PICM-19-CSCs, miR-125b was recently shown as a negative regulator of p53[43]. While the knockdown of miR-125b elevated p53 levels and induced apoptosis in zebra fish and human cells[43], its overexpression promoted human glioma cell proliferation and inhibited apoptosis by targeting the pro-apoptotic Bcl-2 modifying factor[44].

MiR-31 is an oncomir that is upregulated in PICM-19-CSCs. In colorectal cancer cells, miR-31 activated the rat sarcoma (RAS) pathway and inhibited the function of the RAS p21 GTPase activating protein 1 (RASA1) in promoting tumorigenesis and cancer cell growth[45]. It was also shown to promote oncogenesis in the cholangiocarcinoma HCCC-9810 cell line by inhibiting RASA1, thereby downregulating the mitogen-activated protein kinase signaling pathway[46]. Based on these findings, it appears that miR-31 may also be inhibiting the RASA1 in liver cancers. Paradoxically, miR-195, which was shown to suppress tumorigenicity and cause cell cycle arrest by blocking the G1/S transition via the repression of retinoblastoma-E2 transcription factor signaling through cyclin D1, CDK6, and E2F3 in HCC cell lines[47], was upregulated in PICM-19-CSCs.

High levels of miR-365 were found in PICM-19-CSCs. The expression levels of miR-365 vary in different malignancies. It is was overexpressed in both cells and clinical specimens of cutaneous squamous cell carcinoma[48], where it was found to suppress to Mybl2 activity[49]. siRNA silencing of Mybl2 in proliferating Caco-2 cells increased the expression of the cell-cycle regulators cdk2, cyclin D2, and c-MYC; and decreased the expression of cdc25B and cyclin B2[50]. Thus, it is possible that MYC protein suppresses the expression of Mybl2 through the upregulation of miR-365, as seen in the PICM-19-CSCs.

Another key and common feature of CSCs was activation of NF-κB signaling, which is known to increase stress resistance and survival[10]. PICM-19-CSCs had a high level of NF-κB expression when compared to PICM-19 cells. Similarly, the expression of anti-apoptotic genes Bcl-xl and Mcl-1 were elevated in PICM-19-CSCs. Thus, the blocking of anti-apoptotic effects of these proteins may be a viable strategy to eliminate tumor cells. Sabutoclax, a pan-inhibitor of Bcl-2 family of protein that was shown recently to target Mcl-1[51], may be a suitable drug for this purpose.

Selenium-binding protein (SELENBP1) is a tumor suppressor and lower SELENBP1 levels are associated with poor prognosis of liver, lung, colorectal and stomach cancers[52,53]. Decreased SELENBP1 expression has been associated with increased cell motility, proliferation and inhibition of apoptosis, and vascular invasion in HCC[53]. Its expression was suppressed in PICM-19-CSCs suggesting that MYC regulates this gene in the liver to promote cell proliferation and invasion. In a recent proteomic study conducted with serum and tumor tissue specimens of c-MYC transgenic mice, it was shown that APOE, SELENBP1 and retinol binding protein 4 are differentially regulated[54]. Our data support these findings and confirms that MYC differentially regulates the expression of these proteins in liver cells.

Previous studies have also identified MYC as a downstream target of the Wnt signaling, a pathway that is highly active in HCC[55,56]. Dysregulated Wnt signaling occurs in 30% of HCCs. Also, deletion of adenomatous polyposis coli (APC), a negative regulator of Wnt, in the adult mouse liver, leads to an increase in the nuclear β-catenin and MYC[57]. Gene expression array analysis of colorectal cancer cells has shown that MYC is required for the majority of Wnt targeted gene activation after the APC loss[58]. In our study, we found a high level of Wnt expression (Table 2) demonstrating that the Wnt pathway was activated in PICM-19-CSCs.

Validation of MYC-regulated genes in PICM-19-CSCs

To further validate these RNA-seq data, four genes that were upregulated, four genes downregulated, and two genes that were silenced in PICM-19-CSCs, when compared with PICM-19 cells (Tables 1, 3-4) were selected for quantitative real-time PCR studies on the basis of their roles in diverse cellular processes. PCR was performed using porcine-specific primers for CTSB, HIF-1A α-subunit, SERPINA3-2, ITGA3, SELENBP1, RSPO2, FIGF, ANGPT1, CDO1 and DKK2 (Table 6). While two of them (HIF-1α and SELENBP1) were suggested as biomarkers for HCC, others were identified in studies with multiple human cancers. As shown in Figure 3, induced expression of MYC in PICM-19-CSCs resulted in the alteration of gene expression for all ten genes studies. These findings clearly illustrate differences in the expression of these genes between PICM-19 stem cells and PICM-19-CSCs.

Figure 3
Figure 3 RNA sequencing of PICM-19 liver stem cells and PICM-19-CSCs. A: Quality of the RNA extracted from PICM-19 and PICM-19-CSCs was assessed by electrophoresis (left panel) and by Nanodrop measurement to determine (RIN) for each cell type. RIN for PICM-19 and PICM-19-CSCs was 9.50 and 9.30, respectively. After the completion of sequencing, data was analyzed with TopHat and Cufflinks (see Materials and Methods), differential gene expression was calculated as fragments per kilobase of transcripts per million mapped reads (FPKM). Data thus obtained was plotted as a B: Density plot; C: Scatter Matrix plot; D: Box plot. Differentially expressed genes for each cell type were shown as log10 fold change. log10 fold change of 2 or greater was considered significant. In total, 25096 genes were differentially expressed in both cell types, whereas PICM-19 and PICM-19-CSCs displayed unique expression of 6 and 237 genes, respectively.
Table 6 Procine-specific primers used in real-time polymerase chain reaction.
GeneGene IDGenBank#Primer sequencesProduct size (bp)
CTSB100370961NM_001097458.1F: AGCCTGGAACTTCTGGACAA190
R: TTTGTAGGACGGGGTGTAGC
HIF-1A α-subunit396696NM_001123124.1F: GCCAGAACCTCCTGTAACCA204
R: CCTTTTCCTGCTCTGTTTGG
SERPINA3-2396686NM_213787.1F: TGAAAGTTTCCCAGGTGGTC200
R: CTAGGGCTTGGTGACTTTGC
ITGA3100517053XM_005668879.1F: CTTCAAACGGAACCAGAGGA203
R: AGAAGCTTTGTAGCCGGTGA
SELENBP1100152724XM_001929643.3F: GGTACCAGCCTCGACACAAT203
R: GTTGTGCAGGAATCGGATCT
RSPO2100154008NM_001293141.1F: GTCCAGATGGTTTTGCACCT202
R: CTCCTGGATTCAGCAATGGT
FIGF100155670XM_001928382.3F: TCCCCAACCCTGTCATTTTA202
R: TCCTTCTCCATTCCTTGGTG
ANGPT1397009NM_213959.1F: GGGGGAGGTTGGACTGTAAT203
R: TGTGAATAGGCTCGGTTTCC
CDO1100312964NM_001167643.1F: TCTGAAGATGCTGCAAGGAA208
R: CGTATCGAAGGGTGGACTGT
DKK2100519672XM_003129269.2F: CTGGTACCCGCTGCAATAAT198
R: GACAGGGATCTCCTTCATGC
β-actin45269029AY550069.1F: GGACCTGACCGACTACCTCA180
R: GGCAGCTCGTAGCTCTTCTC

Collectively, the vast repertoire of genes that are either directly or indirectly induced by MYC protein expression in PICM-19-CSCs underscores the importance of its involvement in the induction of diverse cellular pathways that ultimately may lead to liver cancer.

DISCUSSION

CSCs have been identified in a number of cancers, including HCC, as a unique population of cells that possess self-renewing and tumor-initiating properties[5,8]. Mounting evidence strongly suggests that CSCs are resistant to anti-cancer therapies[11,59]. Therefore, targeting CSCs may be an effective strategy to eliminate the drug resistant and metastatic tumor cells[60]. Here, we have developed CSCs that are capable of forming tumors in mice by overexpressing human MYC in the liver stem cell line, PICM-19. The involvement of MYC in murine and human HCC malignant transformation has been well documented[2,4,7]. Cellular targets of MYC include genes from almost every biochemical and regulatory pathway, including growth, metabolism, cell cycle progression, differentiation and apoptosis[26,61]. Our results provide a snapshot of differential gene expression induced by the MYC protein in an in vitro model of liver stem cells. PICM-19-CSCs were able to form tumors in immunodeficient mice within three weeks, demonstrating their elevated proliferation and tumor-producing potential in vivo. Analysis of gene expression profiles in PICM-19-CSCs has revealed novel “molecular signatures” that were either up- or downregulated as a consequence of MYC overexpression. A number of genes were both up- and downregulated by MYC overexpression in PICM-19 cells demonstrating their potential involvement in MYC-induced tumorigenesis. We also found that MYC expression silenced six genes completely (Table 1), suggesting that the inhibition of these genes is potentially crucial for tumor induction by MYC. To our knowledge, this is the first report to investigate transcriptome profiles driven by a single oncogene in liver stem cells, and cells of porcine origin, an important animal for modeling human physiology and disease.

The use of MYC to generate CSCs is highly relevant to study HCC because MYC is a key component of hepatocarcinogenesis. High-level expression of MYC was also shown to be essential for the proliferation and survival of other CSCs, such as in glioma[62]. In a recent report, chemoresistant CSCs were isolated from MYC-driven mouse liver tumors[63]. Side-population (SP) cells in these tumors expressed CD44, a marker of hepatic progenitor cells, and displayed an upregulation in the expression of multidrug resistance gene 1 (MDR1), which conferred resistance to doxorubicin, paclitaxel and SN38[63]. We found that PICM-19-CSCs had a significant upregulated expression of MDR1 when compared to PICM-19 cells, suggesting that MDR1 can be an effective therapeutic target for MYC-driven liver cancers. Interestingly, CD44 expression was found only in PICM-19-CSCs but not in PICM-19 cells, demonstrating that MYC regulates CD44.

We discovered several notable similarities in comparing our RNA-seq data with those recently published from CD90+ liver CSCs isolated from HCC patients[64]. Among these, upregulation of glypican 3 (GPC3), apolipoprotein E (APOE), Polo-like kinase 2 and peroxisome proliferator-activated receptor γ, and downregulation of interleukins 8 and 12, suppressor of cytokine signaling 3, and chemokine ligands CXCL12 and CCL2 also occurred in PICM-19-CSCs. Interestingly, neither PICM-19 nor PICM-19-CSCs expressed the progenitor cell markers CD90 and CD133. This observation suggests species-specific differences between cells of human and porcine origin.

MYC also induced the expression of hepatic stem cell marker EpCAM, a target of the Wnt/β-catenin signaling pathway, in PICM-19-CSCs. EpCAM-expressing cells in HCC tumor specimens were previously shown to possess stem-like features and were able to initiate tumors within the liver[65]. PICM-19 cells expressed MYC, Oct4, Sox2, Klf4, and Nanog, which are factors that are commonly used to generate induced pluripotent stem cells. When MYC protein was overexpressed in PICM-19 cells the expression of these proteins was elevated, suggesting that such reprogramming factors are critical for the proliferation of CSCs, and may play a role in tumor-initiation, as shown recently in Ewing sarcoma[66] and breast cancer[67].

By comparing global transcriptome sequencing data between PICM-19 and PICM-19-CSCs, we have confirmed the involvement of a number of MYC-regulated genes involved in development (Sox2, Klf4, Nanog, Oct4, Amd1, Med12), stemness and self-renewal (Wnt/β-catenin), cell cycle (Cyclin D1, GPC3), metabolism (APOA1, APOE), and markers of hepatic stem/progenitor cells (EpCAM, KRT19). In fact, some of these were found to be expressed by side-population (SP) cells isolated from liver cancer cell lines[68]. Their upregulation in CSCs makes them potential candidates as therapeutic targets in liver cancers where MYC is overexpressed. In addition to these genes, PICM-19-CSCs also displayed high levels of a novel protein Sal-like protein 2 (SALL2), a member of the zinc finger transcription factor family. Recently, a family member SALL4 was found to be expressed in liver cancers[69], where it regulates the stemness of EpCAM+ CSCs[70]. While we did not detect SALL4 in PICM-19-CSCs, the induction of SALL2 suggests a certain degree of redundancy in the function of these proteins.

Hypoxia plays an important role in the recurrence of HCC, and HIF-1α is critical part of this process. After insufficient radiofrequency ablation of HCC or sustained treatment with sorafenib, tumor recurrence occurs via signaling pathways involving HIF-1α[71,72], and the expression level of HIF-1α was recently shown to correlate with unfavorable outcomes in patients with HCC[73]. We found that the expression of HIF-1α was significantly upregulated in CSCs. Another protein overexpressed in CSCs is cathepsin B (CTSB). Even though the role of CTSB in HCC is unclear, it has been shown to interact directly with the hepatitis B spliced protein-encoded by the HBV in hepatoma cell lines promoting tumor cell migration and invasion[74]. The alpha-1-antichymotrypsin 2 (SERPINA3-2) is a porcine-specific gene that is also significantly upregulated in CSCs (Figure 4). It shares 71% identity with human SERPINA3, a serine protease inhibitor[75]. While its involvement in HCC is unknown, it has been reported recently that the expression of SEPRINA3 is elevated in cholangiocarcinoma[76].

Figure 4
Figure 4 Validation of RNA-seq data using real-time polymerase chain reaction. Expression of ten genes was tested by real-time polymerase chain reaction using total RNA isolated from PICM-19 and PICM-19-CSCs (see Materials and Methods). Data are presented as mean values from three independent experiments with a variance of < 10%.

Epithelial-mesenchymal transition is a crucial event in the tumor progression of HCC. It has been reported recently that the expression of integrin α3 (ITGA3) is a molecular marker of cells undergoing EMT and of cancer cells with aggressive phenotypes[77]. ITGA3 expression is upregulated in breast cancer[77], and colorectal cancer[78]; and ITGA3-knockout mice were found to be considerably less susceptible to skin carcinogenesis[79], suggesting that ITGA3 is a critical gene in multiple cancers. While its involvement has not been demonstrated in HCC, we observed that its expression is enhanced in CSCs. This result indicated that ITGA3 may play a role in MYC-induced liver cancer.

The R-spondin proteins are secreted agonists of the canonical Wnt/β-catenin signaling pathway. Although they are unable to initiate Wnt signaling, they can enhance responses to low-dose Wnt proteins[80]. One of these proteins, R-spondin 2 (RSPO2) possesses tumor-suppressive activity and was recently shown to inhibit tumor growth in colorectal cancer cells by inhibiting Wnt signaling[81]. Induction of Wnt signaling and downregulation of RSPO2 was also observed in human neurofibromas and malignant peripheral nerve sheath tumors[82]. We found that RSPO2 was downregulated in CSCs, suggesting that it might act as a tumor suppressor in liver cells as well.

The expression of SELENBP1 is markedly reduced in multiple tumor types. Downregulation of SELENBP1 correlated with increased DNA damage and oxidative stress in tissue samples of patients with HCC[83]. Recent studies have shown that the reduced expression of SELENBP1 correlated with poor survival in breast cancer patients[84], and with the degree of colorectal cancer differentiation[85], suggesting that its expression could serve as a potential biomarker for these cancers. We found down-regulation of SELENBP1 in CSCs, demonstrating that that MYC-overexpression can result in this effect.

Tumor metastasis is associated with accelerated cell proliferation, dedifferentiation and loss of cell-cell contacts. FIGF and ANGPT1, two genes involved in angiogenesis, were found to be down-regulated in canine mammary carcinoma[86], breast cancer[87], and prostate cancer[88]. In our studies, the expression of both these genes was significantly lowered in CSCs, suggesting that their reduced expression is a factor in MYC-induced tumorigenesis in liver cells.

Aberrant DNA methylation was shown to play a role in carcinogenesis and disease progression in many cancers[89], and DNA methylation analysis of cancer genes might reveal novel prognostic biomarkers[90]. Cysteine dioxygenase 1 (CDO1), a tumor suppressor, is one such gene that is silenced by promoter methylation in multiple human cancers[19]. For instance, in lymph node-positive breast cancer patients, promoter DNA methylation of CDO1 causes its downregulation, making it an ideal biomarker for disease outcome prediction[91]. A recent DNA methylation study on human lung squamous cell carcinoma has also shown that CDO1 is hypermethylated and downregulated in lung cancer[92]. Even though forced expression of CDO1 resulted in markedly decreased tumor cells growth in human cancer cell lines and in mice[19], little is known about its role in HCC. In our studies, we found that CDO1 is silenced in CSCs, indicating that CDO1 may play a role in MYC-induced carcinogenesis.

Among Wnt antagonists, the Dickkopf (DKK) family of proteins act as tumor suppressors and are frequently silenced in various cancers[93,94]. It has been previously shown that DKK2 is epigenetically silenced in renal cancer, where it inhibits tumor progression through apoptotic and cell cycle pathways involving Bcl2 and cyclin D1[20]. In our study, we found that the expression of DKK2 is silenced in CSCs after MYC-overexpression (Figure 4), suggesting that the inhibition of Wnt antagonist is a mechanism of carcinogenesis mediated by the MYC protein.

Approximately, 70000 cancer deaths in the United States each year are associated with changes in the MYC gene or its expression[61]. Despite enormous progress achieved during the past several decades, long-term patient survival remains very low. Advances in stem cell biology provide opportunities to develop novel approaches to reduce morbidity and mortality associated with liver cancer. Our study is an important contribution to understanding the potential of CSC-targeted therapy for HCC. Novel genes identified in our investigation may serve as promising candidates to develop effective therapeutic options for HCC.

ACKNOWLEDGMENTS

We thank Dr. Aaron Becker (BioMedical Genomics Center, University of Minnesota) for RNA library preparation and sequencing, and Drs. John Garbe and Ying Zhang (Minnesota Supercomputing Institute) for help with Galaxy Suite software analysis.

COMMENTS
Background

It is now well-established that cancer stem cells (CSCs) not only drive tumor growth in the liver, in general, but also that the proto-oncogene c-Myc plays a critical role in that process. However, little is known about the genes that are induced and regulated by the MYC protein to generate tumors, and, in particular, those involved in liver stem cells.

Research frontiers

Hepatocellular carcinoma is a multifactorial disease in which the MYC protein plays a central role. However, a number of different proteins and signaling pathways are activated in liver cancer. Cancer stem cells were isolated from hepatocellular carcinoma (HCC) specimens and by culturing human HCC cell lines. But it is unclear as to which cellular pathways are activated by the MYC protein.

Innovations and breakthroughs

This is the first study to demonstrate the regulation of global gene expression by a single oncoprotein (MYC) in porcine liver stem cells using a novel stem cell line, PICM-19, that is capable of propagating in cell culture for a prolonged time period without losing its phenotypic characteristics. By overexpressing the MYC protein, these stem cells were converted into cancer cells. By using a cutting-edge deep sequencing technique (RNA-seq), the authors have identified six genes (CDO1, C22orf39, DKK2, ENPEP, GPX6, SRPX2) that might be critical in carcinogenesis induced by the MYC protein in PICM-19 cells.

Applications

MYC-driven genes identified in this study may serve as promising candidates for the development of HCC therapeutics without the deleterious effects on non-cancerous stem cells and on other cells present in the liver.

Terminology

CSC - cancer stem cells; RNA-seq - RNA sequencing.

Peer-review

In this study, the authors identify the overall genes expression in a liver stem cell after induction by c-MYC plasmid transfection in generating hepatic tumors. The authors used an immortal porcine liver stem cell line, PICM-19, to identify MYC-driven differential gene expression. Transcriptome sequencing was carried out by RNA sequencing and genes identified by this method were validated using real-time PCR. MYC-driven genes identified in this study may serve as promising candidates for the development of HCC therapeutics that would not have deleterious effects on other cell types in the liver.

Footnotes

P- Reviewer: Cao GW, Leardkamolkarn V, Welling TH, Wu JL S- Editor: Ma YJ L- Editor: A E- Editor: Liu XM

References
1.  Nordenstedt H, White DL, El-Serag HB. The changing pattern of epidemiology in hepatocellular carcinoma. Dig Liver Dis. 2010;42 Suppl 3:S206-S214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 367]  [Cited by in F6Publishing: 394]  [Article Influence: 28.1]  [Reference Citation Analysis (0)]
2.  Aravalli RN, Cressman EN, Steer CJ. Cellular and molecular mechanisms of hepatocellular carcinoma: an update. Arch Toxicol. 2013;87:227-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 166]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
3.  Blum HE. Hepatocellular carcinoma: therapy and prevention. World J Gastroenterol. 2005;11:7391-7400.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Kaposi-Novak P, Libbrecht L, Woo HG, Lee YH, Sears NC, Coulouarn C, Conner EA, Factor VM, Roskams T, Thorgeirsson SS. Central role of c-Myc during malignant conversion in human hepatocarcinogenesis. Cancer Res. 2009;69:2775-2782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 143]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
5.  Ma S, Chan KW, Hu L, Lee TK, Wo JY, Ng IO, Zheng BJ, Guan XY. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology. 2007;132:2542-2556.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Zhao Y, Jian W, Gao W, Zheng YX, Wang YK, Zhou ZQ, Zhang H, Wang CJ. RNAi silencing of c-Myc inhibits cell migration, invasion, and proliferation in HepG2 human hepatocellular carcinoma cell line: c-Myc silencing in hepatocellular carcinoma cell. Cancer Cell Int. 2013;13:23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
7.  Lin CP, Liu CR, Lee CN, Chan TS, Liu HE. Targeting c-Myc as a novel approach for hepatocellular carcinoma. World J Hepatol. 2010;2:16-20.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105-111.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Wang B, Jacob ST. Role of cancer stem cells in hepatocarcinogenesis. Genome Med. 2011;3:11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
10.  Alison MR. Liver stem cells: implications for hepatocarcinogenesis. Stem Cell Rev. 2005;1:253-260.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Ma S, Lee TK, Zheng BJ, Chan KW, Guan XY. CD133+ HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway. Oncogene. 2008;27:1749-1758.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Talbot NC, Blomberg Le Ann, Garrett WM, Caperna TJ. Feeder-independent continuous culture of the PICM-19 pig liver stem cell line. In Vitro Cell Dev Biol Anim. 2010;46:746-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
13.  Talbot NC, Caperna TJ, Garrett WM. Growth and Development Symposium: Development, characterization, and use of a porcine epiblast-derived liver stem cell line: ARS-PICM-19. J Anim Sci. 2013;91:66-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
14.  Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25:1105-1111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9298]  [Cited by in F6Publishing: 9091]  [Article Influence: 606.1]  [Reference Citation Analysis (0)]
15.  Thorvaldsdóttir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14:178-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5308]  [Cited by in F6Publishing: 5529]  [Article Influence: 460.8]  [Reference Citation Analysis (0)]
16.  Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, Salzberg SL, Wold BJ, Pachter L. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28:511-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12168]  [Cited by in F6Publishing: 10845]  [Article Influence: 774.6]  [Reference Citation Analysis (0)]
17.  Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H, Salzberg SL, Rinn JL, Pachter L. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012;7:562-578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8810]  [Cited by in F6Publishing: 8790]  [Article Influence: 732.5]  [Reference Citation Analysis (0)]
18.  Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402-408.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Brait M, Ling S, Nagpal JK, Chang X, Park HL, Lee J, Okamura J, Yamashita K, Sidransky D, Kim MS. Cysteine dioxygenase 1 is a tumor suppressor gene silenced by promoter methylation in multiple human cancers. PLoS One. 2012;7:e44951.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 97]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
20.  Hirata H, Hinoda Y, Nakajima K, Kawamoto K, Kikuno N, Kawakami K, Yamamura S, Ueno K, Majid S, Saini S. Wnt antagonist gene DKK2 is epigenetically silenced and inhibits renal cancer progression through apoptotic and cell cycle pathways. Clin Cancer Res. 2009;15:5678-5687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 70]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
21.  Zhang Y, Gong W, Dai S, Huang G, Shen X, Gao M, Xu Z, Zeng Y, He F. Downregulation of human farnesoid X receptor by miR-421 promotes proliferation and migration of hepatocellular carcinoma cells. Mol Cancer Res. 2012;10:516-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
22.  Hao J, Zhang S, Zhou Y, Liu C, Hu X, Shao C. MicroRNA 421 suppresses DPC4/Smad4 in pancreatic cancer. Biochem Biophys Res Commun. 2011;406:552-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 90]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
23.  Brew K, Nagase H. The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and functional diversity. Biochim Biophys Acta. 2010;1803:55-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 799]  [Cited by in F6Publishing: 861]  [Article Influence: 61.5]  [Reference Citation Analysis (0)]
24.  Yang X, Zhang XF, Lu X, Jia HL, Liang L, Dong QZ, Ye QH, Qin LX. MicroRNA-26a suppresses angiogenesis in human hepatocellular carcinoma by targeting hepatocyte growth factor-cMet pathway. Hepatology. 2014;59:1874-1885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 161]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
25.  Chai ZT, Kong J, Zhu XD, Zhang YY, Lu L, Zhou JM, Wang LR, Zhang KZ, Zhang QB, Ao JY. MicroRNA-26a inhibits angiogenesis by down-regulating VEGFA through the PIK3C2α/Akt/HIF-1α pathway in hepatocellular carcinoma. PLoS One. 2013;8:e77957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 93]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
26.  Eilers M, Eisenman RN. Myc’s broad reach. Genes Dev. 2008;22:2755-2766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 681]  [Cited by in F6Publishing: 726]  [Article Influence: 45.4]  [Reference Citation Analysis (0)]
27.  Guo QM, Malek RL, Kim S, Chiao C, He M, Ruffy M, Sanka K, Lee NH, Dang CV, Liu ET. Identification of c-myc responsive genes using rat cDNA microarray. Cancer Res. 2000;60:5922-5928.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Rabenau KE, O’Toole JM, Bassi R, Kotanides H, Witte L, Ludwig DL, Pereira DS. DEGA/AMIGO-2, a leucine-rich repeat family member, differentially expressed in human gastric adenocarcinoma: effects on ploidy, chromosomal stability, cell adhesion/migration and tumorigenicity. Oncogene. 2004;23:5056-5067.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Nagata M, Noman AA, Suzuki K, Kurita H, Ohnishi M, Ohyama T, Kitamura N, Kobayashi T, Uematsu K, Takahashi K. ITGA3 and ITGB4 expression biomarkers estimate the risks of locoregional and hematogenous dissemination of oral squamous cell carcinoma. BMC Cancer. 2013;13:410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
30.  Calderwood DA, Shattil SJ, Ginsberg MH. Integrins and actin filaments: reciprocal regulation of cell adhesion and signaling. J Biol Chem. 2000;275:22607-22610.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Luque-García JL, Martínez-Torrecuadrada JL, Epifano C, Cañamero M, Babel I, Casal JI. Differential protein expression on the cell surface of colorectal cancer cells associated to tumor metastasis. Proteomics. 2010;10:940-952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 74]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
32.  Wozniak MA, Itzhaki RF, Faragher EB, James MW, Ryder SD, Irving WL. Apolipoprotein E-epsilon 4 protects against severe liver disease caused by hepatitis C virus. Hepatology. 2002;36:456-463.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Ahn SJ, Kim DK, Kim SS, Bae CB, Cho HJ, Kim HG, Kim YJ, Lee JH, Lee HJ, Lee MY. Association between apolipoprotein E genotype, chronic liver disease, and hepatitis B virus. Clin Mol Hepatol. 2012;18:295-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
34.  Zhong DN, Ning QY, Wu JZ, Zang N, Wu JL, Hu DF, Luo SY, Huang AC, Li LL, Li GJ. Comparative proteomic profiles indicating genetic factors may involve in hepatocellular carcinoma familial aggregation. Cancer Sci. 2012;103:1833-1838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
35.  Yokoyama Y, Kuramitsu Y, Takashima M, Iizuka N, Terai S, Oka M, Nakamura K, Okita K, Sakaida I. Protein level of apolipoprotein E increased in human hepatocellular carcinoma. Int J Oncol. 2006;28:625-631.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Topic A, Ljujic M, Radojkovic D. Alpha-1-antitrypsin in pathogenesis of hepatocellular carcinoma. Hepat Mon. 2012;12:e7042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
37.  Pirisi M, Fabris C, Soardo G, Toniutto P, Vitulli D, Bartoli E. Prognostic value of serum alpha-1-antitrypsin in hepatocellular carcinoma. Eur J Cancer. 1996;32A:221-225.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Comunale MA, Rodemich-Betesh L, Hafner J, Wang M, Norton P, Di Bisceglie AM, Block T, Mehta A. Linkage specific fucosylation of alpha-1-antitrypsin in liver cirrhosis and cancer patients: implications for a biomarker of hepatocellular carcinoma. PLoS One. 2010;5:e12419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 106]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
39.  Yanagawa T, Shinozaki T, Watanabe H, Saito K, Raz A, Takagishi K. Vascular endothelial growth factor-D is a key molecule that enhances lymphatic metastasis of soft tissue sarcomas. Exp Cell Res. 2012;318:800-808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
40.  Cui X, Song B, Hou L, Wei Z, Tang J. High expression of osteoglycin decreases the metastatic capability of mouse hepatocarcinoma Hca-F cells to lymph nodes. Acta Biochim Biophys Sin (Shanghai). 2008;40:349-355.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Kisseleva T, Brenner DA. Hepatic stellate cells and the reversal of fibrosis. J Gastroenterol Hepatol. 2006;21 Suppl 3:S84-S87.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Lai KK, Shang S, Lohia N, Booth GC, Masse DJ, Fausto N, Campbell JS, Beretta L. Extracellular matrix dynamics in hepatocarcinogenesis: a comparative proteomics study of PDGFC transgenic and Pten null mouse models. PLoS Genet. 2011;7:e1002147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 70]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
43.  Le MT, Teh C, Shyh-Chang N, Xie H, Zhou B, Korzh V, Lodish HF, Lim B. MicroRNA-125b is a novel negative regulator of p53. Genes Dev. 2009;23:862-876.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 490]  [Cited by in F6Publishing: 509]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
44.  Xia HF, He TZ, Liu CM, Cui Y, Song PP, Jin XH, Ma X. MiR-125b expression affects the proliferation and apoptosis of human glioma cells by targeting Bmf. Cell Physiol Biochem. 2009;23:347-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 130]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
45.  Sun D, Yu F, Ma Y, Zhao R, Chen X, Zhu J, Zhang CY, Chen J, Zhang J. MicroRNA-31 activates the RAS pathway and functions as an oncogenic MicroRNA in human colorectal cancer by repressing RAS p21 GTPase activating protein 1 (RASA1). J Biol Chem. 2013;288:9508-9518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 145]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
46.  Hu C, Huang F, Deng G, Nie W, Huang W, Zeng X. miR-31 promotes oncogenesis in intrahepatic cholangiocarcinoma cells via the direct suppression of RASA1. Exp Ther Med. 2013;6:1265-1270.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Xu T, Zhu Y, Xiong Y, Ge YY, Yun JP, Zhuang SM. MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology. 2009;50:113-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 310]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
48.  Zhou M, Liu W, Ma S, Cao H, Peng X, Guo L, Zhou X, Zheng L, Guo L, Wan M. A novel onco-miR-365 induces cutaneous squamous cell carcinoma. Carcinogenesis. 2013;34:1653-1659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
49.  Papetti M, Augenlicht LH. Mybl2, downregulated during colon epithelial cell maturation, is suppressed by miR-365. Am J Physiol Gastrointest Liver Physiol. 2011;301:G508-G518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
50.  Papetti M, Augenlicht LH. MYBL2, a link between proliferation and differentiation in maturing colon epithelial cells. J Cell Physiol. 2011;226:785-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
51.  Jackson RS, Placzek W, Fernandez A, Ziaee S, Chu CY, Wei J, Stebbins J, Kitada S, Fritz G, Reed JC. Sabutoclax, a Mcl-1 antagonist, inhibits tumorigenesis in transgenic mouse and human xenograft models of prostate cancer. Neoplasia. 2012;14:656-665.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Yang W, Diamond AM. Selenium-binding protein 1 as a tumor suppressor and a prognostic indicator of clinical outcome. Biomark Res. 2013;1:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
53.  Huang C, Ding G, Gu C, Zhou J, Kuang M, Ji Y, He Y, Kondo T, Fan J. Decreased selenium-binding protein 1 enhances glutathione peroxidase 1 activity and downregulates HIF-1α to promote hepatocellular carcinoma invasiveness. Clin Cancer Res. 2012;18:3042-3053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 71]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
54.  Ritorto MS, Borlak J. Combined serum and tissue proteomic study applied to a c-Myc transgenic mouse model of hepatocellular carcinoma identified novel disease regulated proteins suitable for diagnosis and therapeutic intervention strategies. J Proteome Res. 2011;10:3012-3030.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Merle P, Kim M, Herrmann M, Gupte A, Lefrançois L, Califano S, Trépo C, Tanaka S, Vitvitski L, de la Monte S. Oncogenic role of the frizzled-7/beta-catenin pathway in hepatocellular carcinoma. J Hepatol. 2005;43:854-862.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Quinn LM, Secombe J, Hime GR. Myc in stem cell behaviour: insights from Drosophila. Adv Exp Med Biol. 2013;786:269-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
57.  Reed KR, Athineos D, Meniel VS, Wilkins JA, Ridgway RA, Burke ZD, Muncan V, Clarke AR, Sansom OJ. B-catenin deficiency, but not Myc deletion, suppresses the immediate phenotypes of APC loss in the liver. Proc Natl Acad Sci USA. 2008;105:18919-18923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
58.  Sansom OJ, Meniel VS, Muncan V, Phesse TJ, Wilkins JA, Reed KR, Vass JK, Athineos D, Clevers H, Clarke AR. Myc deletion rescues Apc deficiency in the small intestine. Nature. 2007;446:676-679.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Perona R, López-Ayllón BD, de Castro Carpeño J, Belda-Iniesta C. A role for cancer stem cells in drug resistance and metastasis in non-small-cell lung cancer. Clin Transl Oncol. 2011;13:289-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
60.  Clevers H. The cancer stem cell: premises, promises and challenges. Nat Med. 2011;17:313-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1349]  [Cited by in F6Publishing: 1379]  [Article Influence: 106.1]  [Reference Citation Analysis (0)]
61.  Dang CV. c-Myc target genes involved in cell growth, apoptosis, and metabolism. Mol Cell Biol. 1999;19:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Wang J, Wang H, Li Z, Wu Q, Lathia JD, McLendon RE, Hjelmeland AB, Rich JN. c-Myc is required for maintenance of glioma cancer stem cells. PLoS One. 2008;3:e3769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 316]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
63.  Chow EK, Fan LL, Chen X, Bishop JM. Oncogene-specific formation of chemoresistant murine hepatic cancer stem cells. Hepatology. 2012;56:1331-1341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 77]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
64.  Ho DW, Yang ZF, Yi K, Lam CT, Ng MN, Yu WC, Lau J, Wan T, Wang X, Yan Z. Gene expression profiling of liver cancer stem cells by RNA-sequencing. PLoS One. 2012;7:e37159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
65.  Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, Jia H, Ye Q, Qin LX, Wauthier E. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 2009;136:1012-1024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 891]  [Cited by in F6Publishing: 905]  [Article Influence: 60.3]  [Reference Citation Analysis (0)]
66.  Riggi N, Suvà ML, De Vito C, Provero P, Stehle JC, Baumer K, Cironi L, Janiszewska M, Petricevic T, Suvà D. EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell reprogramming toward Ewing sarcoma cancer stem cells. Genes Dev. 2010;24:916-932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 213]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
67.  Leis O, Eguiara A, Lopez-Arribillaga E, Alberdi MJ, Hernandez-Garcia S, Elorriaga K, Pandiella A, Rezola R, Martin AG. Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene. 2012;31:1354-1365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 394]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
68.  Marquardt JU, Raggi C, Andersen JB, Seo D, Avital I, Geller D, Lee YH, Kitade M, Holczbauer A, Gillen MC. Human hepatic cancer stem cells are characterized by common stemness traits and diverse oncogenic pathways. Hepatology. 2011;54:1031-1042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 69]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
69.  Oikawa T, Kamiya A, Zeniya M, Chikada H, Hyuck AD, Yamazaki Y, Wauthier E, Tajiri H, Miller LD, Wang XW. Sal-like protein 4 (SALL4), a stem cell biomarker in liver cancers. Hepatology. 2013;57:1469-1483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 159]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
70.  Zeng SS, Yamashita T, Kondo M, Nio K, Hayashi T, Hara Y, Nomura Y, Yoshida M, Hayashi T, Oishi N. The transcription factor SALL4 regulates stemness of EpCAM-positive hepatocellular carcinoma. J Hepatol. 2014;60:127-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
71.  Xu M, Zheng YL, Xie XY, Liang JY, Pan FS, Zheng SG, Lü MD. Sorafenib blocks the HIF-1α/VEGFA pathway, inhibits tumor invasion, and induces apoptosis in hepatoma cells. DNA Cell Biol. 2014;33:275-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
72.  Liang Y, Zheng T, Song R, Wang J, Yin D, Wang L, Liu H, Tian L, Fang X, Meng X. Hypoxia-mediated sorafenib resistance can be overcome by EF24 through Von Hippel-Lindau tumor suppressor-dependent HIF-1α inhibition in hepatocellular carcinoma. Hepatology. 2013;57:1847-1857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 203]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
73.  Yang SL, Liu LP, Jiang JX, Xiong ZF, He QJ, Wu C. The correlation of expression levels of HIF-1α and HIF-2α in hepatocellular carcinoma with capsular invasion, portal vein tumor thrombi and patients’ clinical outcome. Jpn J Clin Oncol. 2014;44:159-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
74.  Chen WN, Chen JY, Jiao BY, Lin WS, Wu YL, Liu LL, Lin X. Interaction of the hepatitis B spliced protein with cathepsin B promotes hepatoma cell migration and invasion. J Virol. 2012;86:13533-13541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
75.  Stratil A, Peelman LJ, Mattheeuws M, Van Poucke M, Reiner G, Geldermann H. A novel porcine gene, alpha-1-antichymotrypsin 2 (SERPINA3-2): sequence, genomic organization, polymorphism and mapping. Gene. 2002;292:113-119.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Chapman MH, Tidswell R, Dooley JS, Sandanayake NS, Cerec V, Deheragoda M, Lee AJ, Swanton C, Andreola F, Pereira SP. Whole genome RNA expression profiling of endoscopic biliary brushings provides data suitable for biomarker discovery in cholangiocarcinoma. J Hepatol. 2012;56:877-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
77.  Shirakihara T, Kawasaki T, Fukagawa A, Semba K, Sakai R, Miyazono K, Miyazawa K, Saitoh M. Identification of integrin α3 as a molecular marker of cells undergoing epithelial-mesenchymal transition and of cancer cells with aggressive phenotypes. Cancer Sci. 2013;104:1189-1197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
78.  Denadai MV, Viana LS, Affonso RJ, Silva SR, Oliveira ID, Toledo SR, Matos D. Expression of integrin genes and proteins in progression and dissemination of colorectal adenocarcinoma. BMC Clin Pathol. 2013;13:16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
79.  Sachs N, Secades P, van Hulst L, Kreft M, Song JY, Sonnenberg A. Loss of integrin α3 prevents skin tumor formation by promoting epidermal turnover and depletion of slow-cycling cells. Proc Natl Acad Sci USA. 2012;109:21468-21473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
80.  de Lau WB, Snel B, Clevers HC. The R-spondin protein family. Genome Biol. 2012;13:242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 204]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
81.  Wu C, Qiu S, Lu L, Zou J, Li WF, Wang O, Zhao H, Wang H, Tang J, Chen L. RSPO2-LGR5 signaling has tumour-suppressive activity in colorectal cancer. Nat Commun. 2014;5:3149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 86]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
82.  Watson AL, Rahrmann EP, Moriarity BS, Choi K, Conboy CB, Greeley AD, Halfond AL, Anderson LK, Wahl BR, Keng VW. Canonical Wnt/β-catenin signaling drives human Schwann cell transformation, progression, and tumor maintenance. Cancer Discov. 2013;3:674-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
83.  DI Stasio M, Volpe MG, Colonna G, Nazzaro M, Polimeno M, Scala S, Castello G, Costantini S. A possible predictive marker of progression for hepatocellular carcinoma. Oncol Lett. 2011;2:1247-1251.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Zhang S, Li F, Younes M, Liu H, Chen C, Yao Q. Reduced selenium-binding protein 1 in breast cancer correlates with poor survival and resistance to the anti-proliferative effects of selenium. PLoS One. 2013;8:e63702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
85.  Wang N, Chen Y, Yang X, Jiang Y. Selenium-binding protein 1 is associated with the degree of colorectal cancer differentiation and is regulated by histone modification. Oncol Rep. 2014;31:2506-2514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
86.  Klopfleisch R, Lenze D, Hummel M, Gruber AD. The metastatic cascade is reflected in the transcriptome of metastatic canine mammary carcinomas. Vet J. 2011;190:236-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
87.  Roberti MP, Arriaga JM, Bianchini M, Quintá HR, Bravo AI, Levy EM, Mordoh J, Barrio MM. Protein expression changes during human triple negative breast cancer cell line progression to lymph node metastasis in a xenografted model in nude mice. Cancer Biol Ther. 2012;13:1123-1140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
88.  Wong L, Gipp J, Carr J, Loftus CJ, Benck M, Lee S, Mehta V, Vezina CM, Bushman W. Prostate angiogenesis in development and inflammation. Prostate. 2014;74:346-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
89.  Ting AH, McGarvey KM, Baylin SB. The cancer epigenome-components and functional correlates. Genes Dev. 2006;20:3215-3231.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Duffy MJ, Napieralski R, Martens JW, Span PN, Spyratos F, Sweep FC, Brunner N, Foekens JA, Schmitt M. Methylated genes as new cancer biomarkers. Eur J Cancer. 2009;45:335-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 76]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
91.  Dietrich D, Krispin M, Dietrich J, Fassbender A, Lewin J, Harbeck N, Schmitt M, Eppenberger-Castori S, Vuaroqueaux V, Spyratos F. CDO1 promoter methylation is a biomarker for outcome prediction of anthracycline treated, estrogen receptor-positive, lymph node-positive breast cancer patients. BMC Cancer. 2010;10:247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 36]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
92.  Kwon YJ, Lee SJ, Koh JS, Kim SH, Lee HW, Kang MC, Bae JB, Kim YJ, Park JH. Genome-wide analysis of DNA methylation and the gene expression change in lung cancer. J Thorac Oncol. 2012;7:20-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 71]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
93.  Maehata T, Taniguchi H, Yamamoto H, Nosho K, Adachi Y, Miyamoto N, Miyamoto C, Akutsu N, Yamaoka S, Itoh F. Transcriptional silencing of Dickkopf gene family by CpG island hypermethylation in human gastrointestinal cancer. World J Gastroenterol. 2008;14:2702-2714.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Sato H, Suzuki H, Toyota M, Nojima M, Maruyama R, Sasaki S, Takagi H, Sogabe Y, Sasaki Y, Idogawa M. Frequent epigenetic inactivation of DICKKOPF family genes in human gastrointestinal tumors. Carcinogenesis. 2007;28:2459-2466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 166]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]