You are on page 1of 9

Computational Inferring of Risk

Subpathways Mediated by Dysfunctional 9


Non-coding RNAs

Yanjun Xu, Yunpeng Zhang, and Xia Li

Abstract Keywords
Non-coding RNAs mediated core elements of Non-coding RNA · Subpathway · Pathway
pathways contributes to the disorder of topology · Integration · Network
biological function in diseases. Identification
of non-coding RNAs mediated subpathways
not only can help for deciphering the patho- 9.1 Introduction
genic mechanism of complex diseases, but
also can gain insight into the functional roles Recently, non-coding RNAs have received
of non-coding RNAs in human diseases. Here, increased attention with the development of
we summarized the general steps for whole genome and transcriptome sequencing
identifying non-coding RNA mediated technologies [1, 2]. A large number of studies
subpathways and overviewed two of our pre- have demonstrated the important roles of
viously developed methods, Subpathway- non-coding RNAs in human diseases.
GMir and Subpathway-LNCE, which were MicroRNAs mediated the transcription processes
designed to identify miRNAs and lncRNAs and thus can impact the initiation, progression,
mediated risk subpathways respectively. We and prognosis of tumors [3–6]. LncRNAs also
identified the key subpathway regions by have be confirmed as key regulators of diverse
integrating non-coding RNA-target gene cellular processes [7–9], such as genomic
associations, interesting genes and imprinting [10], cell differentiation [11] and
non-coding RNAs and pathway topologies. immune responses [12]. Especially, lncRNAs
By applying methods to several disease can regulate the expression of mRNAs by com-
datasets, we confirmed that our methods is peting common miRNA binding sites with
effective in identifying risk subpathways and mRNAs [13, 14]. PTEN, a critical tumor suppres-
also can help uncover key non-coding RNAs sor gene, has been successfully validated that it
in diseases. Additionally, reproducibility and can act as ceRNA of PTENP1 which is a PTEN
robustness analysis demonstrated our methods pseudogene [15, 16]. H19 is an oncogene which
are reliable. has been shown as a ceRNA to adsorb miR-138
and miR-200a and then impact the core gene of
mesenchymal cells (e.g., ZEB1/ZEB2) in colo-
Y. Xu · Y. Zhang · X. Li (*)
rectal tumor [17]. As non-coding RNAs play
College of Bioinformatics Science and Technology, such critical roles in biological system, it is essen-
Harbin Medical University, Harbin, Heilongjiang, China tial to develop methods which is helpful for
e-mail: zhangyp@hrbmu.edu.cn; lixia@hrbmu.edu.cn

# Springer Nature Singapore Pte Ltd. 2018 87


X. Li et al. (eds.), Non-coding RNAs in Complex Diseases, Advances in Experimental Medicine and
Biology 1094, https://doi.org/10.1007/978-981-13-0719-5_9
88 Y. Xu et al.

revealing the action mechanism and functional identifying key dysfunctional sub-regions
roles of non-coding RNAs and enhance the mediated by non-coding RNAs in diseases.
understanding of etiology in human diseases. Here, we introduced two of our previously
Recently, a series of databases and methods developed methods including Subpathway-GMir
have been provided. TarBase [18], miR2Disease [26] and Subpathway-LNCE [27], both of which
[19], mirTarBase [20] and miRecords [21] are aim to identify dysregulated subpathways
databases that aim to collect experimentally mediated by non-coding RNAs. Specifically,
validated associations between miRNAs and tar- Subpathway-GMir was designed for identifying
get genes. Naamati et al. provided method to miRNA mediated risk subpathways by consider-
investigate the coordinated action of miRNAs to ing interesting genes, miRNAs and pathway
impact biological pathways [22]. Guttman et al. topologies; while Subpathway-LNCE aims to
inferred the putative functions for lincRNAs locate key subpathway regions that competitively
based on their co-expressed mRNAs regulated by lncRNAs in diseases. These two
[1]. LncRNA2Function web tool annotated the methods are not only helpful for deciphering the
function of lncRNAs based on the expression functional roles of lncRNAs, but also can gain
correlation of lncRNA and mRNA, which is insight into the underlying mechanism.
extracted from the RNA-Seq data for 19 normal
tissues [23]. These databases and methods are all
useful for investigating the function of
9.2 Methods
non-coding RNAs. However, our currently
knowledge relevant with the functional roles and
The general steps of non-coding RNA mediated
regulation mechanism of non-coding RNAs con-
subpathway identification processes is depicted in
tribute to disease pathologies is still limit.
Fig. 9.1. It contains four main components:
Identifying non-coding RNAs mediated
(I) Extraction of pathway topology;
biological pathways is an effective strategy to
(II) Reconstructing pathways that involved
address this challenge. Moreover, it have been
non-coding RNAs by integrating non-coding
demonstrated that abnormalities in “subpathway
RNA-gene association data and pathway
regions” may be more relevant with the disease
topologies; (III) Locating candidate key
etiology comparing with entire pathways
subpathway regions based on interesting genes,
[24, 25]. Thus, it is reasonable to focus on
non-coding RNAs and/or molecular profiles, etc.;

Fig. 9.1 The pipeline overview of identifying dysregulated subpathways mediated by non-coding RNAs
9 Identification of Risk Subpathways 89

(IV) Evaluating the significance of candidate nodes and non-coding RNA-gene interaction
subpathways. The details of the processes are edges. For example, in Subpathway-GMir
described below. method, the experimentally verified miRNA-
target interactions were used for reconstructing
KEGG pathway structures. These miRNA-target
9.2.1 Extraction of Pathway interactions were integrated from miRTarBase
Topology [20], mir2Disease [19], miRecords (V4.0) [21]
and TarBase (V6.0) [18] databases. In this
We converted pathway structures of KEGG into method, the reconstructed KEGG pathways con-
undirected graphs with nodes represent genes and tain miRNA nodes and miRNA-gene interaction
edges represent interactions between genes by the edges. In the Subpathway-LNCE method, the
use of “iSubpathwayMiner”, an R package devel- lncRNA-gene associations were constructed
oped by us previously [24, 25]. In details, the based on the competing endogenous RNA
corresponding XML file of pathways were hypothesis [28]. Firstly, the lncRNA-miRNA-
downloaded from the KEGG KGML (http:// mRNA competing relationships were
www.genome.jp/kegg/) which represent the path- constructed. We collected the miRNA and its
way information, firstly. Then, the pathway struc- target mRNA from four public databases
ture (gene-gene interaction information) were (TarBase, mirTarBase, mir2Disease, miRecords
extracted from these KGML files. For each meta- (V4.0)), and lncRNA-miRNA interactions were
bolic pathway, the relationship of enzymes were obtained from our previously work and StarBase
extracted from each XML file. Two enzymes are [29]. For each lncRNA, we recognize its compet-
connected by an edge if their corresponding itive regulated mRNA(s) based on their shared
reactions have a common compound. The meta- miRNAs, meeting the following two conditions.
bolic pathways were simplified to an undirected (i) hypergeometric test p-value of shared miRNAs
graph with enzymes (genes) as nodes. Two genes under a threshold 0.05 (ii) Jaccard Coefficient of
are connected by an edge if they share a common shared miRNAs rank at top 20%. Then, we
compound in the corresponding reactions. For mapped lncRNAs into pathway graphs as nodes
each of other pathways (e.g. signaling pathways), by linking to their regulated-mRNAs, whose
two genes are connected with each other if their Pearson Correlation Coefficient had reached a
annotated proteins were interacted in the original significant threshold (p < 0.05). Finally, we
KEGG pathway map. Finally, the gene-gene obtained condition-specific lncRNA competi-
interaction (pathway structure) were extracted tively regulated signal pathways (LRSP), which
for each pathway as an undirected graph. included lncRNA nodes and lncRNA-mRNA
competitively regulated edges.

9.2.2 Reconstruction of Pathway


Topology Embedded with Non- 9.2.3 Locating Dysfunctional
coding RNAs Subpathway Regions Mediated
by Non-coding RNAs
In this section, the non-coding RNAs were
embedded into each pathways based on the Then, the dysfunctional subpathway regions were
non-coding RNA-gene associations. Briefly, for located within these reconstructed pathways
each pathway, non-coding RNAs with at least one based on interesting nodes (genes and/or
associated target were embedded as non-coding non-coding RNAs) or molecule profiles. In
RNA node into the pathway by connecting it to its Subpathway-GMir [26] and SubpathwayLNCE
verified targets contained in the corresponding [27] method, we used “lenient distance” similar-
pathway. Finally, we obtained the reconstructed ity combined with network topology structure
KEGG pathways which contain non-coding RNA [25] to locate the dysfunctional subpathway
90 Y. Xu et al.

regions based on interesting nodes. Briefly, we In Subpathway-LNCE method [27], We used


first mapped the interesting nodes into the above the Wallenius approximation methods to estimate
reconstructed pathways which were embedded whether the candidate subpathway was signifi-
with non-coding RNAs. For each signature node cantly competing regulated by lncRNAs compar-
pairs, we extracted the shortest path between ing with random. The following parameters were
them. If the number of non-interesting molecules required: (i) the number of interesting genes (x);
in the shortest path of each signature pairs was no (ii) the number of background genes (n); (iii) the
longer than n, we merge the nodes which among number of background genes annotated in
the shortest path between any two signature nodes subpathway (m1); (iv) the number of interesting
into modules. While, the number of nodes in the genes involved in subpathway (m2); (v) the
molecule sets within pathway no less than s were weight of subpathway (w), which was computed
regarded as candidate subpahtways regions that as the following formula, suggested the intensity
mediated by non-coding RNAs. Here, the param- of lncRNAs regulated for the corresponding
eter n and s control the intensity of regulated subpathway. The parameter Gp and Gl represent
signal and the size of the candidate subpathways, the number of genes and genes that competitively
respectively. regulated by lncRNAs in subpathway, respec-
tively. In the Subpathway-LNCE method, we set
β ¼ 1. The Wallenius approximation method was
performed using R package BiasedUrn [30].
9.2.4 Calculating Significance
of Subpathway Regions
  
Gl
After located the subpathway regions within each W ¼ 1 þ β log2 ð9:2Þ
Gp
entire pathway, some strategies were used to eval-
uate the significance of each candidate
subpathways. Some mostly used statistical
methods including hypergeometric test, Fisher
9.3 Case study
Exact test, etc. Furthermore, the random pertur-
bation strategy was also one of the mostly used
9.3.1 Identifying Metabolic
method for evaluating the significance of candi-
Subpathways Mediated by
date subpathways. In the Subpathway-GMir
miRNAs in LIHC
method [26], the following formula based on
hypergeometric method was used to evaluate the
We used Subpathway-GMir method to identify
statistic significance of the difference between
liver hepatocellular carcinoma (LIHC) associated
two study conditions for located subpathways.
subpathways that mediated by miRNAs. First, we
identified risk genes and miRNAs of LIHC based
  
t g þt mir 1
r g þr mir mg þmmir r g r mir on the gene and miRNA expression profile from
X k ng þnmir k
P¼1   ð9:1Þ TCGA database (https://cancergenome.nih.gov/).
mgþmmir ng þnmir ngþnmir
k¼0 ngþnmir The miRNA expression profile contains
100 tumor and 50 normal samples; while
Where mg (mmir) is the number of genes 17 tumor and 9 normal samples were involved
(miRNAs) in entire genome (miRNAome), and in the gene expression profile. We obtained 3357
ng (nmir) is the number of all differentially differential genes and 394 differential miRNAs,
expressed genes (miRNAs). For single which regarded as risk genes/miRNAs, by com-
subpathway containing rg genes (rmir miRNAs), bining the fold change and edgeR methods. Then,
there are tg (tmir) genes (miRNAs) participated we applied Subpathway-GMir method to identify
in it. miRNA mediated risk subpathways for LIHC.
The results shown that there were 29 subpathways
9 Identification of Risk Subpathways 91

Fig. 9.2 MiRNA mediated risk subpathway region of miRNAs and metabolites, respectively. Red nodes repre-
arachidonic acid metabolism in LIHC. Rectangle, circle sent differential genes/miRNAs in the corresponding
and triangle nodes represent protein coding genes, LIHC dataset

that belonged to 29 distinct complete pathways at expression profiles for prostate cancer were
FDR <0.01, 18 of which have been reported to be extracted from SRP002628 which stored in SRA
related with cancers. For example, the database [36]. Most of these dysregulated
path:00590_1 (Fig. 9.2) shows the most signifi- subpathways were associated with the initiation
cance and takes an important part in arachidonic and development of tumor. Specifically, path:
acid metabolic (AAM) pathway which can sup- 04510_1 is an important sub-region within focal
press the development and apoptosis of adhesion pathway (Fig. 9.3). In this subpathway,
hepatomas in light of many studies [31, 32]. The extracellular matrix (ECM) was an upstream pro-
path:00590_1 contains ten genes and three tein, which has been reported to implicate with
miRNAs, including Cyclooxygenase-2 (COX-2), the metastasis of tumors [37, 38]. Interestingly,
Prostaglandin H2 (PGH2) and miR-101, etc. In this protein coding gene was coordinately
vitro and animal models experiments regulated by four lncRNAs (Fig. 9.3), in particu-
demonstrated that overexpression of COX-2 lar, MEG8 and AC078937.4.1 were differentially
which is a differential gene in this LIHC dataset expressed in the prostate cancer dataset. Another
can promote LIHC cell growth [33]. Moreover, dysregulated lncRNA, LINC00087, which com-
miR-101 inhibits COX-2 expression and its petitively regulated two critical cancer genes
downstream metabolite PGH2 to promote pro- including VEGF and CCND2. This indicates
gression of multiple cancers [34, 35]. These that LINC00087 may play an important role in
findings indicate that Subpathway-GMir can prostate cancer. Furthermore, DLEU2 and
effectively identify key sub-regions that are rele- HOTAIRM1 were also competitively regulated
vant with study condition. this subpathway, which had been reported play
important role in the leukemia cells
[39, 40]. DLEU2 was the host gene of miR-15a
9.3.2 Identifying Signal Subpathways and miR-16-1 which are important in
Competitively Regulated by tumorigenesis [41]. In summary, these results
LncRNAs for Prostate Cancer shown that Subpathway-LNCE method can iden-
tify lncRNAs competitively regulated
We used Subpathway-LNCE method success- subpathways and potential key lncRNA underly-
fully identified dysregulated subpathways that ing disease condition.
competitively regulated by lncRNAs in prostate
cancer. The matched lncRNA and mRNA
92 Y. Xu et al.

Fig. 9.3 LncRNA


competitively regulated
subpathway region of Focal
adhesion pathway in
prostate cancer. Triangle
and circle nodes represent
lncRNAs and protein
coding genes, respectively.
Red node label represent
differential genes/lncRNAs
in the corresponding
prostate cancer dataset

9.4 Evaluation of Method 9.4.2 Reproducibility and Robustness


Analysis
9.4.1 Comparison with Other
Methods 9.4.2.1 Reproducibility Analysis
As non-coding RNA/gene expression profiles that
from different experimental platforms exist tech-
After developed the method, an important step is
nical difference, a reliable method should be
to evaluate and demonstrate the superiority of
resistant for this. Reproducibility analysis aims
it. Comparison with previous methods is one of
to evaluate whether pathway identification
the mostly used strategies. For example, we com-
method could have a high concordance when
pared Subpathway-GMir method with the ORA
applied to datasets from different platforms but
[42] and the Kretschmann et al. method [43, 44]
which is underlying the same study condition.
based on the LIHC dataset. Specifically, the ORA
Both Subpathway-GMir and Subpathway-LNCE
method only focuses on genes while the
methods were all have a relative high degree of
Kretschmann et al. method only focuses on
reproducibility. In order to evaluate the reproduc-
miRNA. We found that pathways identified by
ibility of Subpathway-GMir method, we collected
these two previous methods were all included in
another LIHC related dataset which was
the pathway list that identified by Subpathway-
downloaded from GEO database [45] and
GMir method. And many pathways that closely
provided by a different research group. The
related with the initiation and progression of
dataset accession number for miRNA and gene
tumor were missed by these two methods
is GSE36915 and GSE46408, respectively. Then,
(Table 9.1). This indicates that Subpathway-
we applied Subpathway-GMir to this dataset. As
GMir is more effective to detect LIHC-relevant
a result, up to 83% pathways that identified based
pathways than other methods.
the second LIHC dataset were also identified in
the original LIHC dataset; while these two LIHC
9 Identification of Risk Subpathways 93

Table 9.1 Comparison of subpathway-GMir with other methods


Pathway name Subpathway-GMir ORA Kretschmann et al.
Glycolysis/Gluconeogenesis √ √
Purine metabolism √ √
Pyrimidine metabolism √ √
Inositol phosphate metabolism √ √
Glycerophospholipid metabolism √ √
Metabolism of xenobiotics by cytochrome P450 √ √
Lysine degradation √ √
Arachidonic acid metabolism √
Fatty acid metabolism √ √
Glycerolipid metabolism √
Retinol metabolism √
Fructose and mannose metabolism √
Drug metabolism – cytochrome P450 √
Sphingolipid metabolism √
Pentose phosphate pathway √
Steroid hormone biosynthesis √
Tryptophan metabolism √
Starch and sucrose metabolism √
Arginine and proline metabolism √ √
Ether lipid metabolism √
Alanine, aspartate and glutamate metabolism √
Pyruvate metabolism √
beta-Alanine metabolism √
Tyrosine metabolism √
Valine, leucine and isoleucine degradation √
Glutathione metabolism √
Glycine, serine and threonine metabolism √
Galactose metabolism √
Histidine metabolism √
Note: pathway name marked with bold represent it has been reported to associate with cancer. ‘√’ represents pathway
was identified by the corresponding method

related datasets only shared 30% of the differen- factors that critical for method is a useful strategy
tial genes and 5% of the differential miRNAs. to evaluate the stability of method. For example,
This indicates that the high reproducibility of the noise from expression profiles and false posi-
Subpathway-GMir was not due to common dif- tive interactions within pathway topology may
ferential genes and miRNAs. Similarly, we also impact the performance of both Subpathway-
evaluate the reproducibility of Subpathway- GMir and Subpathway-LNCE methods. Thus, in
LNCE method based on three independent pros- the evaluation process of these two methods, we
tate cancer datasets which were obtained from randomly deleted different percentage (5%, 10%,
SRA,TCGA and GEO databases, respectively. . . ., 30%) of genes/non-coding RNAs from
The result demonstrated that Subpathway-LNCE expression profiles and different percentage
also has a high reproducibility. (5%, 10%, . . ., 30%) of edges within each path-
way, respectively. The results shown that our
9.4.2.2 Robustness Analysis method have a relative strong robustness in dis-
Robustness analysis is to evaluate whether turbance of molecular profiles and pathway
method is stable for the disturbance from topology.
biological data that used in method. Disturb
94 Y. Xu et al.

9.5 Concluding Remarks 9. Ponting CP, Oliver PL, Reik W (2009) Evolution and
functions of long noncoding RNAs. Cell 136:629–641
10. Koerner MV, Pauler FM, Huang R et al (2009) The
We summarized two our previously developed function of non-coding RNAs in genomic imprinting.
methods (Subpathway-GMir and Subpathway- Development 136:1771–1783
LNCE) that devoted to identify non-coding 11. Chen ZH, Wang WT, Huang W et al (2017) The
lncRNA HOTAIRM1 regulates the degradation of
RNA mediated dysregulated subpathways for PML-RARA oncoprotein and myeloid cell differentia-
complex diseases. In our methods, we focused tion by enhancing the autophagy pathway. Cell Death
on identifying the key sub-regions within entire Differ 24:212–224
pathway, which is consistent with the concept that 12. Mercer TR, Dinger ME, Mattick JS (2009) Long
non-coding RNAs: insights into functions. Nat Rev
key local sub-regions, but not the entire Genet 10:155–159
pathways, were dysregulated in diseases. More- 13. Ebert MS, Sharp PA (2010) Emerging roles for natural
over, we incorporated pathway topologies and microRNA sponges. Curr Biol 20:R858–R861
non-coding RNA-target gene interaction data 14. Tay Y, Rinn J, Pandolfi PP (2014) The multilayered
complexity of ceRNA crosstalk and competition.
and thus these two methods could provide the Nature 505:344–352
detail interaction information between genes and 15. Karreth FA, Pandolfi PP (2013) ceRNA cross-talk in
non-coding RNAs that associated with the disor- cancer: when ce-bling rivalries go awry. Cancer
dered subpathway. In summary, our methods will Discov 3:1113–1121
16. Poliseno L, Salmena L, Zhang J et al (2010) A coding-
not only be helpful for understanding the mecha- independent function of gene and pseudogene mRNAs
nism of complex diseases, but also will greatly regulates tumour biology. Nature 465:1033–1038
facilitate the exploring of non-coding RNA func- 17. Liang WC, Fu WM, Wong CW et al (2015) The
tion and their roles in human diseases. Especially, lncRNA H19 promotes epithelial to mesenchymal
transition by functioning as miRNA sponges in colo-
it is more precision to characterize the functional rectal cancer. Oncotarget 6:22513–22525
roles of non-coding RNAs at subpathway level. 18. Vergoulis T, Vlachos IS, Alexiou P et al (2012)
TarBase 6.0: capturing the exponential growth of
miRNA targets with experimental support. Nucleic
Acids Res 40:D222–D229
References 19. Jiang Q, Wang Y, Hao Y et al (2009) miR2Disease: a
manually curated database for microRNA deregulation
1. Guttman M, Amit I, Garber M et al (2009) Chromatin in human disease. Nucleic Acids Res 37:D98–D104
signature reveals over a thousand highly conserved 20. Hsu SD, Lin FM, Wu WY et al (2011) miRTarBase: a
large non-coding RNAs in mammals. Nature database curates experimentally validated microRNA-
458:223–227 target interactions. Nucleic Acids Res 39:D163–D169
2. Iyer MK, Niknafs YS, Malik R et al (2015) The land- 21. Xiao F, Zuo Z, Cai G et al (2009) miRecords: an
scape of long noncoding RNAs in the human integrated resource for microRNA-target interactions.
transcriptome. Nat Genet 47:199–208 Nucleic Acids Res 37:D105–D110
3. Rottiers V, Naar AM (2012) MicroRNAs in metabo- 22. Naamati G, Friedman Y, Balaga O et al (2012) Sus-
lism and metabolic disorders. Nat Rev Mol Cell Biol ceptibility of the human pathways graphs to fragmen-
13:239–250 tation by small sets of microRNAs. Bioinformatics
4. Chen B, Li H, Zeng X et al (2012) Roles of microRNA 28:983–990
on cancer cell metabolism. J Transl Med 10:228 23. Jiang Q, Ma R, Wang J et al (2015)
5. Tomasetti M, Santarelli L, Neuzil J et al (2014) LncRNA2Function: a comprehensive resource for
MicroRNA regulation of cancer metabolism: role in functional investigation of human lncRNAs based on
tumour suppression. Mitochondrion 19 Pt A:29–38 RNA-seq data. BMC Genomics 16(Suppl 3):S2
6. Zhang C, Li C, Li J et al (2014) Identification of 24. Li C, Li X, Miao Y et al (2009) SubpathwayMiner: a
miRNA-mediated core gene module for glioma patient software package for flexible identification of
prediction by integrating high-throughput miRNA, pathways. Nucleic Acids Res 37:e131
mRNA expression and pathway structure. PLoS One 25. Li C, Han J, Yao Q et al (2013) Subpathway-GM:
9:e96908 identification of metabolic subpathways via joint
7. Wang KC, Chang HY (2011) Molecular mechanisms power of interesting genes and metabolites and their
of long noncoding RNAs. Mol Cell 43:904–914 topologies within pathways. Nucleic Acids Res 41:
8. Wilusz JE, Sunwoo H, Spector DL (2009) Long non- e101
coding RNAs: functional surprises from the RNA 26. Feng L, Xu Y, Zhang Y et al (2015) Subpathway-
world. Genes Dev 23:1494–1504 GMir: identifying miRNA-mediated metabolic
9 Identification of Risk Subpathways 95

subpathways by integrating condition-specific genes, 36. Kannan K, Wang L, Wang J et al (2011) Recurrent
microRNAs, and pathway topologies. Oncotarget chimeric RNAs enriched in human prostate cancer
6:39151–39164 identified by deep sequencing. Proc Natl Acad Sci U
27. Shi X, Xu Y, Zhang C et al (2016) Subpathway- S A 108:9172–9177
LNCE: Identify dysfunctional subpathways competi- 37. Insua-Rodriguez J, Oskarsson T (2016) The extracel-
tively regulated by lncRNAs through integrating lular matrix in breast cancer. Adv Drug Deliv Rev
lncRNA-mRNA expression profile and pathway 97:41–55
topologies. Oncotarget 7:69857–69870 38. Alfano M, Canducci F, Nebuloni M et al (2016) The
28. Liu K, Yan Z, Li Y et al (2013) Linc2GO: a human interplay of extracellular matrix and microbiome in
LincRNA function annotation resource based on urothelial bladder cancer. Nat Rev Urol 13:77–90
ceRNA hypothesis. Bioinformatics 29:2221–2222 39. Zhang X, Weissman SM, Newburger PE (2014) Long
29. Li JH, Liu S, Zhou H et al (2014) starBase v2.0: intergenic non-coding RNA HOTAIRM1 regulates
decoding miRNA-ceRNA, miRNA-ncRNA and cell cycle progression during myeloid maturation in
protein-RNA interaction networks from large-scale NB4 human promyelocytic leukemia cells. RNA Biol
CLIP-Seq data. Nucleic Acids Res 42:D92–D97 11:777–787
30. Epstein MP, Duncan R, Jiang Y et al (2012) A permu- 40. Kasar S, Underbayev C, Yuan Y et al (2014) Thera-
tation procedure to correct for confounders in case- peutic implications of activation of the host gene
control studies, including tests of rare variation. Am J (Dleu2) promoter for miR-15a/16-1 in chronic lym-
Hum Genet 91:215–223 phocytic leukemia. Oncogene 33:3307–3315
31. Xu Z, Zhang M, Lv X et al (2010) The inhibitory effect 41. Lerner M, Harada M, Loven J et al (2009) DLEU2,
of celecoxib on mouse hepatoma H22 cell line on the frequently deleted in malignancy, functions as a criti-
arachidonic acid metabolic pathway. Biochem Cell cal host gene of the cell cycle inhibitory microRNAs
Biol 88:603–609 miR-15a and miR-16-1. Exp Cell Res 315:2941–2952
32. Dymkowska D, Wojtczak L (2009) Arachidonic acid- 42. Breitling R, Amtmann A, Herzyk P (2004) Iterative
induced apoptosis in rat hepatoma AS-30D cells is Group Analysis (iGA): a simple tool to enhance sensi-
mediated by reactive oxygen species. Acta Biochim tivity and facilitate interpretation of microarray
Pol 56:711–715 experiments. BMC Bioinformatics 5:34
33. Leng J, Han C, Demetris AJ et al (2003) 43. Godard P, van Eyll J (2015) Pathway analysis from
Cyclooxygenase-2 promotes hepatocellular carcinoma lists of microRNAs: common pitfalls and alternative
cell growth through Akt activation: evidence for Akt strategy. Nucleic Acids Res 43:3490–3497
inhibition in celecoxib-induced apoptosis. Hepatology 44. Kretschmann A, Danis B, Andonovic L et al (2015)
38:756–768 Different microRNA profiles in chronic epilepsy ver-
34. Chakrabarty A, Tranguch S, Daikoku T et al (2007) sus acute seizure mouse models. J Mol Neurosci
MicroRNA regulation of cyclooxygenase-2 during 55:466–479
embryo implantation. Proc Natl Acad Sci U S A 45. Barrett T, Troup DB, Wilhite SE et al (2009) NCBI
104:15144–15149 GEO: archive for high-throughput functional genomic
35. Lewis BP, Shih IH, Jones-Rhoades MW et al (2003) data. Nucleic Acids Res 37:D885–D890
Prediction of mammalian microRNA targets. Cell
115:787–798

You might also like